A signal-transducing adaptor protein that associates with TNF RECEPTOR complexes. It contains a death effector domain that can interact with death effector domains found on INITIATOR CASPASES such as CASPASE 8 and CASPASE 10. Activation of CASPASES via interaction with this protein plays a role in the signaling cascade that leads to APOPTOSIS.
A 34 kDa signal transducing adaptor protein that associates with TUMOR NECROSIS FACTOR RECEPTOR TYPE 1. It facilitates the recruitment of signaling proteins such as TNF RECEPTOR-ASSOCIATED FACTOR 2 and FAS ASSOCIATED DEATH DOMAIN PROTEIN to the receptor complex.
Intracellular signaling adaptor proteins that bind to the cytoplasmic death domain region found on DEATH DOMAIN RECEPTORS. Many of the proteins in this class take part in intracellular signaling from TUMOR NECROSIS FACTOR RECEPTORS.
Cell surface receptors that bind TUMOR NECROSIS FACTORS and trigger changes which influence the behavior of cells.
A tumor necrosis factor receptor subtype that has specificity for TUMOR NECROSIS FACTOR ALPHA and LYMPHOTOXIN ALPHA. It is constitutively expressed in most tissues and is a key mediator of tumor necrosis factor signaling in the vast majority of cells. The activated receptor signals via a conserved death domain that associates with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
A signal transducing tumor necrosis factor receptor associated factor that is involved in TNF RECEPTOR feedback regulation. It is similar in structure and appears to work in conjunction with TNF RECEPTOR-ASSOCIATED FACTOR 2 to inhibit APOPTOSIS.
A long pro-domain caspase that contains a death effector domain in its pro-domain region. Caspase 8 plays a role in APOPTOSIS by cleaving and activating EFFECTOR CASPASES. Activation of this enzyme can occur via the interaction of its N-terminal death effector domain with DEATH DOMAIN RECEPTOR SIGNALING ADAPTOR PROTEINS.
A tumor necrosis factor receptor subtype found in a variety of tissues and on activated LYMPHOCYTES. It has specificity for FAS LIGAND and plays a role in regulation of peripheral immune responses and APOPTOSIS. Multiple isoforms of the protein exist due to multiple ALTERNATIVE SPLICING. The activated receptor signals via a conserved death domain that associates with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
A broad category of carrier proteins that play a role in SIGNAL TRANSDUCTION. They generally contain several modular domains, each of which having its own binding activity, and act by forming complexes with other intracellular-signaling molecules. Signal-transducing adaptor proteins lack enzyme activity, however their activity can be modulated by other signal-transducing enzymes
A family of serine-threonine kinases that plays a role in intracellular signal transduction by interacting with a variety of signaling adaptor proteins such as CRADD SIGNALING ADAPTOR PROTEIN; TNF RECEPTOR-ASSOCIATED FACTOR 2; and TNF RECEPTOR-ASSOCIATED DEATH DOMAIN PROTEIN. Although they were initially described as death domain-binding adaptor proteins, members of this family may contain other protein-binding domains such as those involving caspase activation and recruitment.
One of the mechanisms by which CELL DEATH occurs (compare with NECROSIS and AUTOPHAGOCYTOSIS). Apoptosis is the mechanism responsible for the physiological deletion of cells and appears to be intrinsically programmed. It is characterized by distinctive morphologic changes in the nucleus and cytoplasm, chromatin cleavage at regularly spaced sites, and the endonucleolytic cleavage of genomic DNA; (DNA FRAGMENTATION); at internucleosomal sites. This mode of cell death serves as a balance to mitosis in regulating the size of animal tissues and in mediating pathologic processes associated with tumor growth.
A long pro-domain caspase that contains a caspase recruitment domain in its pro-domain region. Caspase 9 is activated during cell stress by mitochondria-derived proapoptotic factors and by CARD SIGNALING ADAPTOR PROTEINS such as APOPTOTIC PROTEASE-ACTIVATING FACTOR 1. It activates APOPTOSIS by cleaving and activating EFFECTOR CASPASES.
A family of intracellular CYSTEINE ENDOPEPTIDASES that play a role in regulating INFLAMMATION and APOPTOSIS. They specifically cleave peptides at a CYSTEINE amino acid that follows an ASPARTIC ACID residue. Caspases are activated by proteolytic cleavage of a precursor form to yield large and small subunits that form the enzyme. Since the cleavage site within precursors matches the specificity of caspases, sequential activation of precursors by activated caspases can occur.
Serum glycoprotein produced by activated MACROPHAGES and other mammalian MONONUCLEAR LEUKOCYTES. It has necrotizing activity against tumor cell lines and increases ability to reject tumor transplants. Also known as TNF-alpha, it is only 30% homologous to TNF-beta (LYMPHOTOXIN), but they share TNF RECEPTORS.
A tumor necrosis factor receptor subtype that is expressed primarily in IMMUNE SYSTEM cells. It has specificity for membrane-bound form of TUMOR NECROSIS FACTORS and mediates intracellular-signaling through TNF RECEPTOR ASSOCIATED FACTORS.
Transport proteins that carry specific substances in the blood or across cell membranes.
A tumor necrosis factor receptor-associated factor that acts as a specific signaling adaptor protein for the EDAR RECEPTOR and plays an important role in ectodermal development. It binds to edar receptor via its C-terminal death domain region and to other specific TNF receptor-associated factors via its N-terminal domain. Loss of function of edar-associated death domain protein is associated with AUTOSOMAL RECESSIVE ANHIDROTIC ECTODERMAL DYSPLASIA.
The intracellular transfer of information (biological activation/inhibition) through a signal pathway. In each signal transduction system, an activation/inhibition signal from a biologically active molecule (hormone, neurotransmitter) is mediated via the coupling of a receptor/enzyme to a second messenger system or to an ion channel. Signal transduction plays an important role in activating cellular functions, cell differentiation, and cell proliferation. Examples of signal transduction systems are the GAMMA-AMINOBUTYRIC ACID-postsynaptic receptor-calcium ion channel system, the receptor-mediated T-cell activation pathway, and the receptor-mediated activation of phospholipases. Those coupled to membrane depolarization or intracellular release of calcium include the receptor-mediated activation of cytotoxic functions in granulocytes and the synaptic potentiation of protein kinase activation. Some signal transduction pathways may be part of larger signal transduction pathways; for example, protein kinase activation is part of the platelet activation signal pathway.
A signal transducing tumor necrosis factor receptor associated factor that is involved in TNF RECEPTOR feedback regulation. It is similar in structure and appears to work in conjunction with TNF RECEPTOR-ASSOCIATED FACTOR 1 to inhibit APOPTOSIS.
Intracellular signaling peptides and proteins that bind directly or indirectly to the cytoplasmic portion of TUMOR NECROSIS FACTOR RECEPTORS.
The termination of the cell's ability to carry out vital functions such as metabolism, growth, reproduction, responsiveness, and adaptability.
Ubiquitous, inducible, nuclear transcriptional activator that binds to enhancer elements in many different cell types and is activated by pathogenic stimuli. The NF-kappa B complex is a heterodimer composed of two DNA-binding subunits: NF-kappa B1 and relA.
An APOPTOSIS-regulating protein that is structurally related to CASPASE 8 and competes with CASPASE 8 for binding to FAS ASSOCIATED DEATH DOMAIN PROTEIN. Two forms of CASP8 and FADD-like apoptosis regulating protein exist, a long form containing a caspase-like enzymatically inactive domain and a short form which lacks the caspase-like domain.
Inflammation of the tonsils, especially the PALATINE TONSILS but the ADENOIDS (pharyngeal tonsils) and lingual tonsils may also be involved. Tonsillitis usually is caused by bacterial infection. Tonsillitis may be acute, chronic, or recurrent.
Inflammation of the lymph nodes.
A long pro-domain caspase that contains a death effector domain in its pro-domain region. Activation of this enzyme can occur via the interaction of its N-terminal death effector domain with DEATH DOMAIN RECEPTOR SIGNALING ADAPTOR PROTEINS. Caspase 10 plays a role in APOPTOSIS by cleaving and activating EFFECTOR CASPASES. Several isoforms of this protein exist due to multiple alternative splicing of its MESSENGER RNA.
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
A transmembrane protein belonging to the tumor necrosis factor superfamily that was originally discovered on cells of the lymphoid-myeloid lineage, including activated T-LYMPHOCYTES and NATURAL KILLER CELLS. It plays an important role in immune homeostasis and cell-mediated toxicity by binding to the FAS RECEPTOR and triggering APOPTOSIS.
A CELL LINE derived from human T-CELL LEUKEMIA and used to determine the mechanism of differential susceptibility to anti-cancer drugs and radiation.
Established cell cultures that have the potential to propagate indefinitely.
Linear POLYPEPTIDES that are synthesized on RIBOSOMES and may be further modified, crosslinked, cleaved, or assembled into complex proteins with several subunits. The specific sequence of AMINO ACIDS determines the shape the polypeptide will take, during PROTEIN FOLDING, and the function of the protein.
A large group of proteins that control APOPTOSIS. This family of proteins includes many ONCOGENE PROTEINS as well as a wide variety of classes of INTRACELLULAR SIGNALING PEPTIDES AND PROTEINS such as CASPASES.
Tumor necrosis factor receptor family members that are widely expressed and play a role in regulation of peripheral immune responses and APOPTOSIS. The receptors are specific for TNF-RELATED APOPTOSIS-INDUCING LIGAND and signal via conserved death domains that associate with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
A transmembrane-protein belonging to the TNF family of intercellular signaling proteins. It is a widely expressed ligand that activates APOPTOSIS by binding to TNF-RELATED APOPTOSIS-INDUCING LIGAND RECEPTORS. The membrane-bound form of the protein can be cleaved by specific CYSTEINE ENDOPEPTIDASES to form a soluble ligand form.
Factors which produce cessation of all vital bodily functions. They can be analyzed from an epidemiologic viewpoint.
Proteins and peptides that are involved in SIGNAL TRANSDUCTION within the cell. Included here are peptides and proteins that regulate the activity of TRANSCRIPTION FACTORS and cellular processes in response to signals from CELL SURFACE RECEPTORS. Intracellular signaling peptide and proteins may be part of an enzymatic signaling cascade or act through binding to and modifying the action of other signaling factors.
The level of protein structure in which combinations of secondary protein structures (alpha helices, beta sheets, loop regions, and motifs) pack together to form folded shapes called domains. Disulfide bridges between cysteines in two different parts of the polypeptide chain along with other interactions between the chains play a role in the formation and stabilization of tertiary structure. Small proteins usually consist of only one domain but larger proteins may contain a number of domains connected by segments of polypeptide chain which lack regular secondary structure.
Conversion of an inactive form of an enzyme to one possessing metabolic activity. It includes 1, activation by ions (activators); 2, activation by cofactors (coenzymes); and 3, conversion of an enzyme precursor (proenzyme or zymogen) to an active enzyme.
A family of cell surface receptors that signal via a conserved domain that extends into the cell CYTOPLASM. The conserved domain is referred to as a death domain due to the fact that many of these receptors are involved in signaling APOPTOSIS. Several DEATH DOMAIN RECEPTOR SIGNALING ADAPTOR PROTEINS can bind to the death domains of the activated receptors and through a complex series of interactions activate apoptotic mediators such as CASPASES.
Glycoproteins found on the membrane or surface of cells.
The degree of similarity between sequences of amino acids. This information is useful for the analyzing genetic relatedness of proteins and species.
A short pro-domain caspase that plays an effector role in APOPTOSIS. It is activated by INITIATOR CASPASES such as CASPASE 9. Isoforms of this protein exist due to multiple alternative splicing of its MESSENGER RNA.
The process in which substances, either endogenous or exogenous, bind to proteins, peptides, enzymes, protein precursors, or allied compounds. Specific protein-binding measures are often used as assays in diagnostic assessments.
Endogenous and exogenous compounds and that either inhibit CASPASES or prevent their activation.
The uptake of naked or purified DNA by CELLS, usually meaning the process as it occurs in eukaryotic cells. It is analogous to bacterial transformation (TRANSFORMATION, BACTERIAL) and both are routinely employed in GENE TRANSFER TECHNIQUES.
A large class of structurally-related proteins that contain one or more LIM zinc finger domains. Many of the proteins in this class are involved in intracellular signaling processes and mediate their effects via LIM domain protein-protein interactions. The name LIM is derived from the first three proteins in which the motif was found: LIN-11, Isl1 and Mec-3.
Cells propagated in vitro in special media conducive to their growth. Cultured cells are used to study developmental, morphologic, metabolic, physiologic, and genetic processes, among others.
Recombinant proteins produced by the GENETIC TRANSLATION of fused genes formed by the combination of NUCLEIC ACID REGULATORY SEQUENCES of one or more genes with the protein coding sequences of one or more genes.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
A family of enzymes that catalyze the stereoselective, regioselective, or chemoselective syn-dehydrogenation reactions. They function by a mechanism that is linked directly to reduction of molecular OXYGEN.
Differentiation antigens residing on mammalian leukocytes. CD stands for cluster of differentiation, which refers to groups of monoclonal antibodies that show similar reactivity with certain subpopulations of antigens of a particular lineage or differentiation stage. The subpopulations of antigens are also known by the same CD designation.
A tumor necrosis factor receptor subtype with specificity for TUMOR NECROSIS FACTOR LIGAND SUPERFAMILY MEMBER 15. It is found in tissues containing LYMPHOCYTES and may play a role in regulating lymphocyte homeostasis and APOPTOSIS. The activated receptor signals via a conserved death domain that associates with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
A death domain receptor signaling adaptor protein that plays a role in signaling the activation of INITIATOR CASPASES such as CASPASE 2. It contains a death domain that is specific for RIP SERINE-THEONINE KINASES and a caspase-binding domain that binds to and activates CASPASES such as CASPASE 2.
Proteins prepared by recombinant DNA technology.
A subgroup of mitogen-activated protein kinases that activate TRANSCRIPTION FACTOR AP-1 via the phosphorylation of C-JUN PROTEINS. They are components of intracellular signaling pathways that regulate CELL PROLIFERATION; APOPTOSIS; and CELL DIFFERENTIATION.
The introduction of a phosphoryl group into a compound through the formation of an ester bond between the compound and a phosphorus moiety.
The parts of a macromolecule that directly participate in its specific combination with another molecule.
A superfamily of PROTEIN-SERINE-THREONINE KINASES that are activated by diverse stimuli via protein kinase cascades. They are the final components of the cascades, activated by phosphorylation by MITOGEN-ACTIVATED PROTEIN KINASE KINASES, which in turn are activated by mitogen-activated protein kinase kinase kinases (MAP KINASE KINASE KINASES).
The arrangement of two or more amino acid or base sequences from an organism or organisms in such a way as to align areas of the sequences sharing common properties. The degree of relatedness or homology between the sequences is predicted computationally or statistically based on weights assigned to the elements aligned between the sequences. This in turn can serve as a potential indicator of the genetic relatedness between the organisms.
Within a eukaryotic cell, a membrane-limited body which contains chromosomes and one or more nucleoli (CELL NUCLEOLUS). The nuclear membrane consists of a double unit-type membrane which is perforated by a number of pores; the outermost membrane is continuous with the ENDOPLASMIC RETICULUM. A cell may contain more than one nucleus. (From Singleton & Sainsbury, Dictionary of Microbiology and Molecular Biology, 2d ed)
Protein modules with conserved ligand-binding surfaces which mediate specific interaction functions in SIGNAL TRANSDUCTION PATHWAYS and the specific BINDING SITES of their cognate protein LIGANDS.
Proteins which are found in membranes including cellular and intracellular membranes. They consist of two types, peripheral and integral proteins. They include most membrane-associated enzymes, antigenic proteins, transport proteins, and drug, hormone, and lectin receptors.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
A subclass of tumor necrosis family receptors that lack cell signaling domains. They bind to specific TNF RECEPTOR LIGANDS and are believed to play a modulating role in the TNF signaling pathway. Some of the decoy receptors are products of distinct genes, while others are products of ALTERNATIVE SPLICING of the MRNA for the active receptor.
The insertion of recombinant DNA molecules from prokaryotic and/or eukaryotic sources into a replicating vehicle, such as a plasmid or virus vector, and the introduction of the resultant hybrid molecules into recipient cells without altering the viability of those cells.
Proteins which bind to DNA. The family includes proteins which bind to both double- and single-stranded DNA and also includes specific DNA binding proteins in serum which can be used as markers for malignant diseases.
The first continuously cultured human malignant CELL LINE, derived from the cervical carcinoma of Henrietta Lacks. These cells are used for VIRUS CULTIVATION and antitumor drug screening assays.
Irreversible cessation of all bodily functions, manifested by absence of spontaneous breathing and total loss of cardiovascular and cerebral functions.
Proteins associated with the inner surface of the lipid bilayer of the viral envelope. These proteins have been implicated in control of viral transcription and may possibly serve as the "glue" that binds the nucleocapsid to the appropriate membrane site during viral budding from the host cell.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
A superfamily of proteins that share a highly conserved MADS domain sequence motif. The term MADS refers to the first four members which were MCM1 PROTEIN; AGAMOUS 1 PROTEIN; DEFICIENS PROTEIN; and SERUM RESPONSE FACTOR. Many MADS domain proteins have been found in species from all eukaryotic kingdoms. They play an important role in development, especially in plants where they have an important role in flower development.
Endogenous substances, usually proteins, which are effective in the initiation, stimulation, or termination of the genetic transcription process.
Screening techniques first developed in yeast to identify genes encoding interacting proteins. Variations are used to evaluate interplay between proteins and other molecules. Two-hybrid techniques refer to analysis for protein-protein interactions, one-hybrid for DNA-protein interactions, three-hybrid interactions for RNA-protein interactions or ligand-based interactions. Reverse n-hybrid techniques refer to analysis for mutations or other small molecules that dissociate known interactions.
Regions of AMINO ACID SEQUENCE similarity in the SRC-FAMILY TYROSINE KINASES that fold into specific functional tertiary structures. The SH1 domain is a CATALYTIC DOMAIN. SH2 and SH3 domains are protein interaction domains. SH2 usually binds PHOSPHOTYROSINE-containing proteins and SH3 interacts with CYTOSKELETAL PROTEINS.
Strains of mice in which certain GENES of their GENOMES have been disrupted, or "knocked-out". To produce knockouts, using RECOMBINANT DNA technology, the normal DNA sequence of the gene being studied is altered to prevent synthesis of a normal gene product. Cloned cells in which this DNA alteration is successful are then injected into mouse EMBRYOS to produce chimeric mice. The chimeric mice are then bred to yield a strain in which all the cells of the mouse contain the disrupted gene. Knockout mice are used as EXPERIMENTAL ANIMAL MODELS for diseases (DISEASE MODELS, ANIMAL) and to clarify the functions of the genes.
Inbred C57BL mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings, resulting in a high degree of genetic uniformity and homozygosity, making them widely used for biomedical research, including studies on genetics, immunology, cancer, and neuroscience.
A family of proteins that were originally identified by their ability to cause NECROSIS of NEOPLASMS. Their necrotic effect on cells is mediated through TUMOR NECROSIS FACTOR RECEPTORS which induce APOPTOSIS.
Models used experimentally or theoretically to study molecular shape, electronic properties, or interactions; includes analogous molecules, computer-generated graphics, and mechanical structures.
The characteristic 3-dimensional shape of a protein, including the secondary, supersecondary (motifs), tertiary (domains) and quaternary structure of the peptide chain. PROTEIN STRUCTURE, QUATERNARY describes the conformation assumed by multimeric proteins (aggregates of more than one polypeptide chain).
Genetically engineered MUTAGENESIS at a specific site in the DNA molecule that introduces a base substitution, or an insertion or deletion.
Commonly observed structural components of proteins formed by simple combinations of adjacent secondary structures. A commonly observed structure may be composed of a CONSERVED SEQUENCE which can be represented by a CONSENSUS SEQUENCE.
RNA sequences that serve as templates for protein synthesis. Bacterial mRNAs are generally primary transcripts in that they do not require post-transcriptional processing. Eukaryotic mRNA is synthesized in the nucleus and must be exported to the cytoplasm for translation. Most eukaryotic mRNAs have a sequence of polyadenylic acid at the 3' end, referred to as the poly(A) tail. The function of this tail is not known for certain, but it may play a role in the export of mature mRNA from the nucleus as well as in helping stabilize some mRNA molecules by retarding their degradation in the cytoplasm.
The level of protein structure in which regular hydrogen-bond interactions within contiguous stretches of polypeptide chain give rise to alpha helices, beta strands (which align to form beta sheets) or other types of coils. This is the first folding level of protein conformation.
Proteins found in the nucleus of a cell. Do not confuse with NUCLEOPROTEINS which are proteins conjugated with nucleic acids, that are not necessarily present in the nucleus.
Protein interaction domains of about 70-90 amino acid residues, named after a common structure found in PSD-95, Discs Large, and Zona Occludens 1 proteins. PDZ domains are involved in the recruitment and interaction of proteins, and aid the formation of protein scaffolds and signaling networks. This is achieved by sequence-specific binding between a PDZ domain in one protein and a PDZ motif in another protein.
A member of the TNF receptor family that was initially identified as a DEXAMETHASONE-induced protein from a T-CELL line. It may play a role in regulating APOPTOSIS and modulating immune response by T-lymphocytes. Signaling by the activated receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS.
Cell surface receptors that bind NERVE GROWTH FACTOR; (NGF) and a NGF-related family of neurotrophic factors that includes neurotrophins, BRAIN-DERIVED NEUROTROPHIC FACTOR and CILIARY NEUROTROPHIC FACTOR.
Single-stranded complementary DNA synthesized from an RNA template by the action of RNA-dependent DNA polymerase. cDNA (i.e., complementary DNA, not circular DNA, not C-DNA) is used in a variety of molecular cloning experiments as well as serving as a specific hybridization probe.
A tumor necrosis factor family member that is released by activated LYMPHOCYTES. Soluble lymphotoxin is specific for TUMOR NECROSIS FACTOR RECEPTOR TYPE I; TUMOR NECROSIS FACTOR RECEPTOR TYPE II; and TUMOR NECROSIS FACTOR RECEPTOR SUPERFAMILY, MEMBER 14. Lymphotoxin-alpha can form a membrane-bound heterodimer with LYMPHOTOXIN-BETA that has specificity for the LYMPHOTOXIN BETA RECEPTOR.
The process by which two molecules of the same chemical composition form a condensation product or polymer.
Identification of proteins or peptides that have been electrophoretically separated by blot transferring from the electrophoresis gel to strips of nitrocellulose paper, followed by labeling with antibody probes.
The phenotypic manifestation of a gene or genes by the processes of GENETIC TRANSCRIPTION and GENETIC TRANSLATION.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control (induction or repression) of gene action at the level of transcription or translation.
The lipid- and protein-containing, selectively permeable membrane that surrounds the cytoplasm in prokaryotic and eukaryotic cells.
The span of viability of a cell characterized by the capacity to perform certain functions such as metabolism, growth, reproduction, some form of responsiveness, and adaptability.
Proteins that originate from insect species belonging to the genus DROSOPHILA. The proteins from the most intensely studied species of Drosophila, DROSOPHILA MELANOGASTER, are the subject of much interest in the area of MORPHOGENESIS and development.
Theoretical representations that simulate the behavior or activity of biological processes or diseases. For disease models in living animals, DISEASE MODELS, ANIMAL is available. Biological models include the use of mathematical equations, computers, and other electronic equipment.
The study of crystal structure using X-RAY DIFFRACTION techniques. (McGraw-Hill Dictionary of Scientific and Technical Terms, 4th ed)
A molecule that binds to another molecule, used especially to refer to a small molecule that binds specifically to a larger molecule, e.g., an antigen binding to an antibody, a hormone or neurotransmitter binding to a receptor, or a substrate or allosteric effector binding to an enzyme. Ligands are also molecules that donate or accept a pair of electrons to form a coordinate covalent bond with the central metal atom of a coordination complex. (From Dorland, 27th ed)
The part of a cell that contains the CYTOSOL and small structures excluding the CELL NUCLEUS; MITOCHONDRIA; and large VACUOLES. (Glick, Glossary of Biochemistry and Molecular Biology, 1990)
A low affinity receptor that binds NERVE GROWTH FACTOR; BRAIN-DERIVED NEUROTROPHIC FACTOR; NEUROTROPHIN 3; and neurotrophin 4.
A cell line derived from cultured tumor cells.
Deletion of sequences of nucleic acids from the genetic material of an individual.
Short sequences (generally about 10 base pairs) of DNA that are complementary to sequences of messenger RNA and allow reverse transcriptases to start copying the adjacent sequences of mRNA. Primers are used extensively in genetic and molecular biology techniques.
The process of moving proteins from one cellular compartment (including extracellular) to another by various sorting and transport mechanisms such as gated transport, protein translocation, and vesicular transport.
Elements of limited time intervals, contributing to particular results or situations.
The rate dynamics in chemical or physical systems.
A sequence of amino acids in a polypeptide or of nucleotides in DNA or RNA that is similar across multiple species. A known set of conserved sequences is represented by a CONSENSUS SEQUENCE. AMINO ACID MOTIFS are often composed of conserved sequences.
The relationship between the chemical structure of a compound and its biological or pharmacological activity. Compounds are often classed together because they have structural characteristics in common including shape, size, stereochemical arrangement, and distribution of functional groups.
Partial proteins formed by partial hydrolysis of complete proteins or generated through PROTEIN ENGINEERING techniques.
The biosynthesis of RNA carried out on a template of DNA. The biosynthesis of DNA from an RNA template is called REVERSE TRANSCRIPTION.
Proteins that originate from plants species belonging to the genus ARABIDOPSIS. The most intensely studied species of Arabidopsis, Arabidopsis thaliana, is commonly used in laboratory experiments.
The aggregation of soluble ANTIGENS with ANTIBODIES, alone or with antibody binding factors such as ANTI-ANTIBODIES or STAPHYLOCOCCAL PROTEIN A, into complexes large enough to fall out of solution.
Serologic tests in which a positive reaction manifested by visible CHEMICAL PRECIPITATION occurs when a soluble ANTIGEN reacts with its precipitins, i.e., ANTIBODIES that can form a precipitate.
A variation of the PCR technique in which cDNA is made from RNA via reverse transcription. The resultant cDNA is then amplified using standard PCR protocols.
A negative regulatory effect on physiological processes at the molecular, cellular, or systemic level. At the molecular level, the major regulatory sites include membrane receptors, genes (GENE EXPRESSION REGULATION), mRNAs (RNA, MESSENGER), and proteins.
Membrane proteins encoded by the BCL-2 GENES and serving as potent inhibitors of cell death by APOPTOSIS. The proteins are found on mitochondrial, microsomal, and NUCLEAR MEMBRANE sites within many cell types. Overexpression of bcl-2 proteins, due to a translocation of the gene, is associated with follicular lymphoma.
A family of calcium/calmodulin-dependent PROETIN-SERINE-THREONINE KINASES. They are ubiquitously expressed in adult and embryonic mammalian tissues, and their functions are tightly related to the early stages of eukaryotic programmed cell death.
A long pro-domain caspase that contains a caspase recruitment domain in its pro-domain region. Activation of this enzyme can occur via the interaction of its caspase recruitment domain with CARD SIGNALING ADAPTOR PROTEINS. Caspase 2 plays a role in APOPTOSIS by cleaving and activating effector pro-caspases. Several isoforms of this protein exist due to multiple alternative splicing of its MESSENGER RNA.
A positive regulatory effect on physiological processes at the molecular, cellular, or systemic level. At the molecular level, the major regulatory sites include membrane receptors, genes (GENE EXPRESSION REGULATION), mRNAs (RNA, MESSENGER), and proteins.
Cell surface proteins that bind signalling molecules external to the cell with high affinity and convert this extracellular event into one or more intracellular signals that alter the behavior of the target cell (From Alberts, Molecular Biology of the Cell, 2nd ed, pp693-5). Cell surface receptors, unlike enzymes, do not chemically alter their ligands.
Antibodies produced by a single clone of cells.
A protein serine-threonine kinase that catalyzes the PHOSPHORYLATION of I KAPPA B PROTEINS. This enzyme also activates the transcription factor NF-KAPPA B and is composed of alpha and beta catalytic subunits, which are protein kinases and gamma, a regulatory subunit.
A human cell line established from a diffuse histiocytic lymphoma (HISTIOCYTIC LYMPHOMA, DIFFUSE) and displaying many monocytic characteristics. It serves as an in vitro model for MONOCYTE and MACROPHAGE differentiation.
Unexpected rapid natural death due to cardiovascular collapse within one hour of initial symptoms. It is usually caused by the worsening of existing heart diseases. The sudden onset of symptoms, such as CHEST PAIN and CARDIAC ARRHYTHMIAS, particularly VENTRICULAR TACHYCARDIA, can lead to the loss of consciousness and cardiac arrest followed by biological death. (from Braunwald's Heart Disease: A Textbook of Cardiovascular Medicine, 7th ed., 2005)
A group of enzymes that catalyzes the phosphorylation of serine or threonine residues in proteins, with ATP or other nucleotides as phosphate donors.
Extrachromosomal, usually CIRCULAR DNA molecules that are self-replicating and transferable from one organism to another. They are found in a variety of bacterial, archaeal, fungal, algal, and plant species. They are used in GENETIC ENGINEERING as CLONING VECTORS.
A genetic rearrangement through loss of segments of DNA or RNA, bringing sequences which are normally separated into close proximity. This deletion may be detected using cytogenetic techniques and can also be inferred from the phenotype, indicating a deletion at one specific locus.
Lymphocytes responsible for cell-mediated immunity. Two types have been identified - cytotoxic (T-LYMPHOCYTES, CYTOTOXIC) and helper T-lymphocytes (T-LYMPHOCYTES, HELPER-INDUCER). They are formed when lymphocytes circulate through the THYMUS GLAND and differentiate to thymocytes. When exposed to an antigen, they divide rapidly and produce large numbers of new T cells sensitized to that antigen.
Technique using an instrument system for making, processing, and displaying one or more measurements on individual cells obtained from a cell suspension. Cells are usually stained with one or more fluorescent dyes specific to cell components of interest, e.g., DNA, and fluorescence of each cell is measured as it rapidly transverses the excitation beam (laser or mercury arc lamp). Fluorescence provides a quantitative measure of various biochemical and biophysical properties of the cell, as well as a basis for cell sorting. Other measurable optical parameters include light absorption and light scattering, the latter being applicable to the measurement of cell size, shape, density, granularity, and stain uptake.
A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
The pathological process occurring in cells that are dying from irreparable injuries. It is caused by the progressive, uncontrolled action of degradative ENZYMES, leading to MITOCHONDRIAL SWELLING, nuclear flocculation, and cell lysis. It is distinct it from APOPTOSIS, which is a normal, regulated cellular process.
A species of the genus SACCHAROMYCES, family Saccharomycetaceae, order Saccharomycetales, known as "baker's" or "brewer's" yeast. The dried form is used as a dietary supplement.
Immunologic method used for detecting or quantifying immunoreactive substances. The substance is identified by first immobilizing it by blotting onto a membrane and then tagging it with labeled antibodies.
Small double-stranded, non-protein coding RNAs (21-31 nucleotides) involved in GENE SILENCING functions, especially RNA INTERFERENCE (RNAi). Endogenously, siRNAs are generated from dsRNAs (RNA, DOUBLE-STRANDED) by the same ribonuclease, Dicer, that generates miRNAs (MICRORNAS). The perfect match of the siRNAs' antisense strand to their target RNAs mediates RNAi by siRNA-guided RNA cleavage. siRNAs fall into different classes including trans-acting siRNA (tasiRNA), repeat-associated RNA (rasiRNA), small-scan RNA (scnRNA), and Piwi protein-interacting RNA (piRNA) and have different specific gene silencing functions.
Proteins which maintain the transcriptional quiescence of specific GENES or OPERONS. Classical repressor proteins are DNA-binding proteins that are normally bound to the OPERATOR REGION of an operon, or the ENHANCER SEQUENCES of a gene until a signal occurs that causes their release.
A genus of small, two-winged flies containing approximately 900 described species. These organisms are the most extensively studied of all genera from the standpoint of genetics and cytology.
Different forms of a protein that may be produced from different GENES, or from the same gene by ALTERNATIVE SPLICING.
A cell line generated from human embryonic kidney cells that were transformed with human adenovirus type 5.
Genes whose expression is easily detectable and therefore used to study promoter activity at many positions in a target genome. In recombinant DNA technology, these genes may be attached to a promoter region of interest.
Microscopy of specimens stained with fluorescent dye (usually fluorescein isothiocyanate) or of naturally fluorescent materials, which emit light when exposed to ultraviolet or blue light. Immunofluorescence microscopy utilizes antibodies that are labeled with fluorescent dye.
The relationship between the dose of an administered drug and the response of the organism to the drug.
The outward appearance of the individual. It is the product of interactions between genes, and between the GENOTYPE and the environment.
Products of proto-oncogenes. Normally they do not have oncogenic or transforming properties, but are involved in the regulation or differentiation of cell growth. They often have protein kinase activity.
Laboratory mice that have been produced from a genetically manipulated EGG or EMBRYO, MAMMALIAN.
A member of the Bcl-2 protein family that reversibly binds MEMBRANES. It is a pro-apoptotic protein that is activated by caspase cleavage.
Proteins found in any species of bacterium.
A soluble factor produced by MONOCYTES; MACROPHAGES, and other cells which activates T-lymphocytes and potentiates their response to mitogens or antigens. Interleukin-1 is a general term refers to either of the two distinct proteins, INTERLEUKIN-1ALPHA and INTERLEUKIN-1BETA. The biological effects of IL-1 include the ability to replace macrophage requirements for T-cell activation.
DNA sequences which are recognized (directly or indirectly) and bound by a DNA-dependent RNA polymerase during the initiation of transcription. Highly conserved sequences within the promoter include the Pribnow box in bacteria and the TATA BOX in eukaryotes.
Process of generating a genetic MUTATION. It may occur spontaneously or be induced by MUTAGENS.
'Nerve tissue proteins' are specialized proteins found within the nervous system's biological tissue, including neurofilaments, neuronal cytoskeletal proteins, and neural cell adhesion molecules, which facilitate structural support, intracellular communication, and synaptic connectivity essential for proper neurological function.
The naturally occurring or experimentally induced replacement of one or more AMINO ACIDS in a protein with another. If a functionally equivalent amino acid is substituted, the protein may retain wild-type activity. Substitution may also diminish, enhance, or eliminate protein function. Experimentally induced substitution is often used to study enzyme activities and binding site properties.
Chemically stimulated aggregation of cell surface receptors, which potentiates the action of the effector cell.
A mutation caused by the substitution of one nucleotide for another. This results in the DNA molecule having a change in a single base pair.
Non-antibody proteins secreted by inflammatory leukocytes and some non-leukocytic cells, that act as intercellular mediators. They differ from classical hormones in that they are produced by a number of tissue or cell types rather than by specialized glands. They generally act locally in a paracrine or autocrine rather than endocrine manner.
Phosphoproteins are proteins that have been post-translationally modified with the addition of a phosphate group, usually on serine, threonine or tyrosine residues, which can play a role in their regulation, function, interaction with other molecules, and localization within the cell.
A state of prolonged irreversible cessation of all brain activity, including lower brain stem function with the complete absence of voluntary movements, responses to stimuli, brain stem reflexes, and spontaneous respirations. Reversible conditions which mimic this clinical state (e.g., sedative overdose, hypothermia, etc.) are excluded prior to making the determination of brain death. (From Adams et al., Principles of Neurology, 6th ed, pp348-9)
A CALMODULIN-dependent enzyme that catalyzes the phosphorylation of proteins. This enzyme is also sometimes dependent on CALCIUM. A wide range of proteins can act as acceptor, including VIMENTIN; SYNAPSINS; GLYCOGEN SYNTHASE; MYOSIN LIGHT CHAINS; and the MICROTUBULE-ASSOCIATED PROTEINS. (From Enzyme Nomenclature, 1992, p277)
The characteristic 3-dimensional shape and arrangement of multimeric proteins (aggregates of more than one polypeptide chain).
Cell lines whose original growing procedure consisted being transferred (T) every 3 days and plated at 300,000 cells per plate (J Cell Biol 17:299-313, 1963). Lines have been developed using several different strains of mice. Tissues are usually fibroblasts derived from mouse embryos but other types and sources have been developed as well. The 3T3 lines are valuable in vitro host systems for oncogenic virus transformation studies, since 3T3 cells possess a high sensitivity to CONTACT INHIBITION.
A species of gram-negative, facultatively anaerobic, rod-shaped bacteria (GRAM-NEGATIVE FACULTATIVELY ANAEROBIC RODS) commonly found in the lower part of the intestine of warm-blooded animals. It is usually nonpathogenic, but some strains are known to produce DIARRHEA and pyogenic infections. Pathogenic strains (virotypes) are classified by their specific pathogenic mechanisms such as toxins (ENTEROTOXIGENIC ESCHERICHIA COLI), etc.
Protein analogs and derivatives of the Aequorea victoria green fluorescent protein that emit light (FLUORESCENCE) when excited with ULTRAVIOLET RAYS. They are used in REPORTER GENES in doing GENETIC TECHNIQUES. Numerous mutants have been made to emit other colors or be sensitive to pH.
A family of transcription factors characterized by the presence of a bipartite DNA-binding domain known as the POU domain. The POU domain contains two subdomains, a POU-specific domain and a POU-homeodomain. The POU domain was originally identified as a region of approximately 150 amino acids shared between the Pit-1, Oct-1, Oct-2, and Unc-86 transcription factors.
Members of the class of compounds composed of AMINO ACIDS joined together by peptide bonds between adjacent amino acids into linear, branched or cyclical structures. OLIGOPEPTIDES are composed of approximately 2-12 amino acids. Polypeptides are composed of approximately 13 or more amino acids. PROTEINS are linear polypeptides that are normally synthesized on RIBOSOMES.
Processes that stimulate the GENETIC TRANSCRIPTION of a gene or set of genes.
Semiautonomous, self-reproducing organelles that occur in the cytoplasm of all cells of most, but not all, eukaryotes. Each mitochondrion is surrounded by a double limiting membrane. The inner membrane is highly invaginated, and its projections are called cristae. Mitochondria are the sites of the reactions of oxidative phosphorylation, which result in the formation of ATP. They contain distinctive RIBOSOMES, transfer RNAs (RNA, TRANSFER); AMINO ACYL T RNA SYNTHETASES; and elongation and termination factors. Mitochondria depend upon genes within the nucleus of the cells in which they reside for many essential messenger RNAs (RNA, MESSENGER). Mitochondria are believed to have arisen from aerobic bacteria that established a symbiotic relationship with primitive protoeukaryotes. (King & Stansfield, A Dictionary of Genetics, 4th ed)
Inhibitors of SERINE ENDOPEPTIDASES and sulfhydryl group-containing enzymes. They act as alkylating agents and are known to interfere in the translation process.
A signal transducing tumor necrosis factor receptor associated factor that is involved in regulation of NF-KAPPA B signalling and activation of JNK MITOGEN-ACTIVATED PROTEIN KINASES.
A process whereby multiple RNA transcripts are generated from a single gene. Alternative splicing involves the splicing together of other possible sets of EXONS during the processing of some, but not all, transcripts of the gene. Thus a particular exon may be connected to any one of several alternative exons to form a mature RNA. The alternative forms of mature MESSENGER RNA produce PROTEIN ISOFORMS in which one part of the isoforms is common while the other parts are different.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control of gene action during the developmental stages of an organism.
All of the processes involved in increasing CELL NUMBER including CELL DIVISION.
Lipid-containing polysaccharides which are endotoxins and important group-specific antigens. They are often derived from the cell wall of gram-negative bacteria and induce immunoglobulin secretion. The lipopolysaccharide molecule consists of three parts: LIPID A, core polysaccharide, and O-specific chains (O ANTIGENS). When derived from Escherichia coli, lipopolysaccharides serve as polyclonal B-cell mitogens commonly used in laboratory immunology. (From Dorland, 28th ed)
A plant genus of the family BRASSICACEAE that contains ARABIDOPSIS PROTEINS and MADS DOMAIN PROTEINS. The species A. thaliana is used for experiments in classical plant genetics as well as molecular genetic studies in plant physiology, biochemistry, and development.
Eukaryotic cell line obtained in a quiescent or stationary phase which undergoes conversion to a state of unregulated growth in culture, resembling an in vitro tumor. It occurs spontaneously or through interaction with viruses, oncogenes, radiation, or drugs/chemicals.
Splitting the DNA into shorter pieces by endonucleolytic DNA CLEAVAGE at multiple sites. It includes the internucleosomal DNA fragmentation, which along with chromatin condensation, are considered to be the hallmarks of APOPTOSIS.
Diffusible gene products that act on homologous or heterologous molecules of viral or cellular DNA to regulate the expression of proteins.
Processes involved in the formation of TERTIARY PROTEIN STRUCTURE.
The relationships of groups of organisms as reflected by their genetic makeup.
Proteins that bind to RNA molecules. Included here are RIBONUCLEOPROTEINS and other proteins whose function is to bind specifically to RNA.
Connective tissue cells which secrete an extracellular matrix rich in collagen and other macromolecules.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Proteins from the nematode species CAENORHABDITIS ELEGANS. The proteins from this species are the subject of scientific interest in the area of multicellular organism MORPHOGENESIS.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
Motifs in DNA- and RNA-binding proteins whose amino acids are folded into a single structural unit around a zinc atom. In the classic zinc finger, one zinc atom is bound to two cysteines and two histidines. In between the cysteines and histidines are 12 residues which form a DNA binding fingertip. By variations in the composition of the sequences in the fingertip and the number and spacing of tandem repeats of the motif, zinc fingers can form a large number of different sequence specific binding sites.
Progressive restriction of the developmental potential and increasing specialization of function that leads to the formation of specialized cells, tissues, and organs.
A family of intracellular signaling kinases that were identified by their ability to signal from the activated INTERLEUKIN-1 RECEPTORS. Signaling from these kinases involves their interaction with SIGNAL TRANSDUCING ADAPTOR PROTEINS such as MYELOID DIFFERENTIATION FACTOR 88 and TNF RECEPTOR-ASSOCIATED FACTOR 6.
Proteins found in any species of fungus.
Proteins found in plants (flowers, herbs, shrubs, trees, etc.). The concept does not include proteins found in vegetables for which VEGETABLE PROTEINS is available.
A cytokine that stimulates the growth and differentiation of B-LYMPHOCYTES and is also a growth factor for HYBRIDOMAS and plasmacytomas. It is produced by many different cells including T-LYMPHOCYTES; MONOCYTES; and FIBROBLASTS.
A species of nematode that is widely used in biological, biochemical, and genetic studies.
Compounds or agents that combine with an enzyme in such a manner as to prevent the normal substrate-enzyme combination and the catalytic reaction.
A member of the tumor necrosis factor receptor superfamily. It has specificity for LYMPHOTOXIN ALPHA1, BETA2 HETEROTRIMER and TUMOR NECROSIS FACTOR LIGAND SUPERFAMILY MEMBER 14. The receptor plays a role in regulating lymphoid ORGANOGENESIS and the differentiation of certain subsets of NATURAL KILLER T-CELLS. Signaling of the receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS.
The basic cellular units of nervous tissue. Each neuron consists of a body, an axon, and dendrites. Their purpose is to receive, conduct, and transmit impulses in the NERVOUS SYSTEM.
An in situ method for detecting areas of DNA which are nicked during APOPTOSIS. Terminal deoxynucleotidyl transferase is used to add labeled dUTP, in a template-independent manner, to the 3 prime OH ends of either single- or double-stranded DNA. The terminal deoxynucleotidyl transferase nick end labeling, or TUNEL, assay labels apoptosis on a single-cell level, making it more sensitive than agarose gel electrophoresis for analysis of DNA FRAGMENTATION.
Dioxygenase enzymes that specifically hydroxylate a PROLINE residue on the HYPOXIA-INDUCIBLE FACTOR 1, ALPHA SUBUNIT. They are OXYGEN-dependent enzymes that play an important role in mediating cellular adaptive responses to HYPOXIA.
ENDOPEPTIDASES which have a cysteine involved in the catalytic process. This group of enzymes is inactivated by CYSTEINE PROTEINASE INHIBITORS such as CYSTATINS and SULFHYDRYL REAGENTS.
The assembly of the QUATERNARY PROTEIN STRUCTURE of multimeric proteins (MULTIPROTEIN COMPLEXES) from their composite PROTEIN SUBUNITS.
Death of the developing young in utero. BIRTH of a dead FETUS is STILLBIRTH.
In vitro method for producing large amounts of specific DNA or RNA fragments of defined length and sequence from small amounts of short oligonucleotide flanking sequences (primers). The essential steps include thermal denaturation of the double-stranded target molecules, annealing of the primers to their complementary sequences, and extension of the annealed primers by enzymatic synthesis with DNA polymerase. The reaction is efficient, specific, and extremely sensitive. Uses for the reaction include disease diagnosis, detection of difficult-to-isolate pathogens, mutation analysis, genetic testing, DNA sequencing, and analyzing evolutionary relationships.
Macromolecular complexes formed from the association of defined protein subunits.
An enzyme that catalyzes the methylation of the epsilon-amino group of lysine residues in proteins to yield epsilon mono-, di-, and trimethyllysine. EC 2.1.1.43.

The zinc finger protein A20 inhibits TNF-induced NF-kappaB-dependent gene expression by interfering with an RIP- or TRAF2-mediated transactivation signal and directly binds to a novel NF-kappaB-inhibiting protein ABIN. (1/76)

The zinc finger protein A20 is a tumor necrosis factor (TNF)- and interleukin 1 (IL-1)-inducible protein that negatively regulates nuclear factor-kappa B (NF-kappaB)-dependent gene expression. However, the molecular mechanism by which A20 exerts this effect is still unclear. We show that A20 does not inhibit TNF- induced nuclear translocation and DNA binding of NF-kappaB, although it completely prevents the TNF- induced activation of an NF-kappaB-dependent reporter gene, as well as TNF-induced IL-6 and granulocyte macrophage-colony stimulating factor gene expression. Moreover, NF-kappaB activation induced by overexpression of the TNF receptor-associated proteins TNF receptor-associated death domain protein (TRADD), receptor interacting protein (RIP), and TNF recep- tor-associated factor 2 (TRAF2) was also inhibited by expression of A20, whereas NF-kappaB activation induced by overexpression of NF-kappaB-inducing kinase (NIK) or the human T cell leukemia virus type 1 (HTLV-1) Tax was unaffected. These results demonstrate that A20 inhibits NF-kappaB-dependent gene expression by interfering with a novel TNF-induced and RIP- or TRAF2-mediated pathway that is different from the NIK-IkappaB kinase pathway and that is specifically involved in the transactivation of NF-kappaB. Via yeast two-hybrid screening, we found that A20 binds to a novel protein, ABIN, which mimics the NF-kappaB inhibiting effects of A20 upon overexpression, suggesting that the effect of A20 is mediated by its interaction with this NF-kappaB inhibiting protein, ABIN.  (+info)

Activation of the small GTPase Cdc42 by the inflammatory cytokines TNF(alpha) and IL-1, and by the Epstein-Barr virus transforming protein LMP1. (2/76)

Cdc42, a Rho-family GTPase, has been implicated in several signal transduction pathways, including organization of the actin cytoskeleton, activation of the c-Jun N-terminal MAP kinase (JNK) and stimulation of the nuclear transcription factor kappa B (NF(kappa)B). We report here that exposure of fibroblasts to the inflammatory cytokines tumor necrosis factor (alpha) (TNF(alpha)) and interleukin-1 (IL-1) triggers the activation of Cdc42 leading first to filopodia formation and subsequently to Rac and Rho activation. Inhibition of Cdc42 completely suppresses cytokine-induced actin polymerization, but not activation of JNK or NF(kappa)B. The latent membrane protein 1 of Epstein-Barr virus, LMP1, is thought to mimic constitutively activated TNF family receptors. When expressed in fibroblasts, LMP1 stimulates Cdc42-dependent filopodia formation as well as JNK and NF(kappa)B activation. Using LMP1 mutants, we show that activation of Cdc42 and JNK/NF(kappa)B occur through distinct pathways and that Cdc42 activation is independent of LMP1's interaction with TRADD and TRAF proteins.  (+info)

The A20 protein interacts with the Epstein-Barr virus latent membrane protein 1 (LMP1) and alters the LMP1/TRAF1/TRADD complex. (3/76)

The Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) interacts with the tumor necrosis factor receptor (TNFR)-associated factor (TRAF) molecules, which are important for LMP1-mediated signaling. Two domains of LMP1 can independently activate NF-kB, carboxyl-terminal activating region 1 (CTAR1) and CTAR2. The activation of NF-kB by CTAR1 occurs through direct interaction of LMP1 with the TRAF molecules, whereas CTAR2 interacts with the TNFR-associated death domain protein (TRADD) to activate NF-kB and the c-Jun N-terminal kinase (JNK). A20, which is induced by LMP1 through NF-kB, can block NF-kB activation from both domains of LMP1 and inhibit JNK activation from CTAR2. A20 also has been shown to associate with TRAF1 and TRAF2. In this study, an interaction between LMP1 and A20 was detected that was increased by TRAF2 overexpression. A20 did not affect the association of TRAF1 with TRAF2 but did displace TRAF1 from the LMP1 complex. The interaction of LMP1 and TRADD was decreased in the presence of A20, and the LMP1-A20 association was decreased by TRADD, suggesting that A20 and TRADD both interact with LMP1 and may compete for binding. These data indicate that A20 alters the interactions between LMP1 and the TRAF molecules and TRADD, affecting the activation of NF-kB, JNK, and perhaps other TRAF-mediated signaling events.  (+info)

The residues between the two transformation effector sites of Epstein-Barr virus latent membrane protein 1 are not critical for B-lymphocyte growth transformation. (4/76)

Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is essential for EBV-mediated transformation of primary B lymphocytes. LMP1 spontaneously aggregates in the plasma membrane and enables two transformation effector sites (TES1 and TES2) within the 200-amino-acid cytoplasmic carboxyl terminus to constitutively engage the tumor necrosis factor receptor (TNFR)-associated factors TRAF1, TRAF2, TRAF3, and TRAF5 and the TNFR-associated death domain proteins TRADD and RIP, thereby activating NF-kappaB and c-Jun N-terminal kinase (JNK). To investigate the importance of the 60% of the LMP1 carboxyl terminus that lies between the TES1-TRAF and TES2-TRADD and -RIP binding sites, an EBV recombinant was made that contains a specific deletion of LMP1 codons 232 to 351. Surprisingly, the deletion mutant was similar to wild-type (wt) LMP1 EBV recombinants in its efficiency in transforming primary B lymphocytes into lymphoblastoid cell lines (LCLs). Mutant and wt EBV-transformed LCLs were similarly efficient in long-term outgrowth and in regrowth after endpoint dilution. Mutant and wt LMP1 proteins were also similar in their constitutive association with TRAF1, TRAF2, TRAF3, TRADD, and RIP. Mutant and wt EBV-transformed LCLs were similar in steady-state levels of Bcl2, JNK, and activated JNK proteins. The wt phenotype of recombinants with LMP1 codons 232 to 351 deleted further demarcates TES1 and TES2, underscores their central importance in B-lymphocyte growth transformation, and provides a new perspective on LMP1 sequence variation between TES1 and TES2.  (+info)

A novel mechanism of TRAF signaling revealed by structural and functional analyses of the TRADD-TRAF2 interaction. (5/76)

TRAF proteins are major mediators for the cell activation, cell survival, and antiapoptotic functions of the TNF receptor superfamily. They can be recruited to activated TNF receptors either by direct interactions with the receptors or indirectly via the adaptor protein TRADD. We now report the structure of the TRADD-TRAF2 complex, which is highly distinct from receptor-TRAF2 interactions. This interaction is significantly stronger and we show by an in vivo signaling assay that TRAF2 signaling is more readily initiated by TRADD than by direct receptor-TRAF2 interactions. TRADD is specific for TRAF1 and TRAF2, which ensures the recruitment of clAPs for the direct inhibition of caspase activation in the signaling complex. The stronger affinity and unique specificity of the TRADD-TRAF2 interaction are crucial for the suppression of apoptosis and provide a mechanistic basis for the perturbation of TRAF recruitment in sensitizing cell death induction.  (+info)

Mechanism of chronic obstructive uropathy: increased expression of apoptosis-promoting molecules. (6/76)

BACKGROUND: We have demonstrated that renal tubular and interstitial cells undergo pronounced apoptosis during the course of chronic obstructive uropathy (COU). Apoptosis is a complex cellular process consisting of multiple steps, each of which is mediated by families of related molecules. These families may include receptor/ligand molecules such as Fas, Fas ligand, tumor necrosis factor receptor-1 (TNFR-1), and TNF-related apoptosis inducing ligand (TRAIL); signal transduction adapter molecules such as Fas-associated death domain (FADD), TNFR-1 associated death domain (TRADD), receptor-interacting protein (RIP), Fas-associated factor (FAF), and Fas-associated phosphatase (FAP); or effector molecules such as caspases. However, the mechanism of tubular cell apoptosis, as well as the pathogenetic relevance of these apoptosis-related molecules in COU, remains poorly understood. METHODS: Kidneys were harvested from sham-operated control mice and mice with COU created by left ureter ligation sacrificed in groups of three at days 4, 15, 30, and 45. To detect apoptotic tubular and interstitial cells, in situ end labeling of fragmented DNA was performed. To detect the expression of apoptosis-related molecules, ribonuclease protection assay was used with specific antisense RNA probes for Fas, Fas ligand, TNFR-1, TRAIL, FADD, TRADD, RIP, FAF, FAP, and caspase-8. Immunostaining for Fas, Fas ligand, TRAIL, TRADD, RIP, and caspase-8 was also performed. To assess the role of these molecules in COU-associated renal cell apoptosis, the frequencies of apoptotic tubular and interstitial cells were separately quantitated for each experimental time point, and their patterns of variation were correlated with those of apoptosis-related molecules. RESULTS: The obstructed kidneys displayed increased apoptosis of both tubular and interstitial cells. Tubular cell apoptosis appeared at day 4 after ureter ligation, peaked (fivefold of control) at day 15, and decreased gradually until the end of the experiment. In contrast, interstitial cell apoptosis sustained a progressive increase throughout the experiment. Apoptosis was minimal at all experimental time points for control and contralateral kidneys. Compared with control and contralateral kidneys, the ligated kidneys displayed a dynamic expression of mRNAs for many apoptosis-related molecules, which included an up to threefold increase for Fas, Fas ligand, TNF-R1, TRAIL, TRADD, RIP, and caspase-8, and an up to twofold increase for FADD and FAP, but there was little change for FAF. These mRNAs increased between days 4 and 15, decreased until day 30, but then increased again until day 45. The rise and fall of mRNAs between days 4 and 30 paralleled a similar fluctuation in tubular cell apoptosis in that period. The subsequent increase of mRNAs was correlated with a continuous rise of interstitial cell apoptosis. We demonstrated a positive immunostaining for Fas and Fas ligand in the tubular cells at early time points as well as in interstitial inflammatory cells at later time points. Although increased expression of TRAIL, TRADD, RIP, and caspase-8 was noted in tubular cells, there was no staining for these molecules in interstitial cells. CONCLUSION: The current study documents a dynamic expression of several molecules that are known to mediate the most crucial steps of apoptosis. It implicates these molecules in COU-associated renal cell apoptosis and in the pathogenesis of this condition. It also lays the foundation for interventional studies, including genetic engineering, to evaluate the molecular control of apoptosis associated with COU.  (+info)

TRADD domain of Epstein-Barr virus transforming protein LMP1 is essential for inducing immortalization and suppressing senescence of primary rodent fibroblasts. (7/76)

Mutation analysis of latent membrane protein 1 (LMP1) in Epstein-Barr virus (EBV)-induced B-cell immortalization revealed two transformation effector sites, TES1 and TES2. TES2 mediates the interaction with tumor necrosis factor receptor-associated death domain protein (TRADD) and plays a key role in transactivating NF-kappa B and AP-1. Recombinant EBV containing LMP1 with TES2 deleted induces a limited proliferation of B cells. The present study shows that a mutant with an LMP1 site-specific mutation at TES2, LMP1(TRADD), initially stimulates cell growth and significantly extends the life span of MEF. However, it is not sufficient for the immortalization of MEF, and MEF-LMP1(TRADD) cells eventually enter growth arrest. Further analysis reveals that although LMP1(TRADD) promotes cell growth, it does not prevent the eventual onset of senescence and the expression of tumor suppressor p16(Ink4a).  (+info)

Hepatitis C virus core protein enhances FADD-mediated apoptosis and suppresses TRADD signaling of tumor necrosis factor receptor. (8/76)

Hepatitis C virus (HCV) core protein has been shown to interact with the death domain (DD) of tumor necrosis factor receptor-1 (TNFR1). In this study, we further examined the interaction of the core protein with the signaling molecules of TNFR1, including FADD, TRADD, and TRAF2, in a human embryonic kidney cell line, HEK-293, that overexpresses the HCV core protein. This core protein-expressing cell line exhibited enhanced sensitivity to TNF-induced apoptosis. By in vitro binding and in vivo coimmunoprecipitation assays, we showed that the HCV core protein interacted with the DD of FADD and enhanced apoptosis induced by FADD overexpression. This enhancement could be blocked by a dominant-negative mutant of FADD. In contrast, the core protein did not directly interact with the DD of TRADD, but could disrupt the binding of TRADD to TNFR1. TRAF2 recruitment to the TNFR1 signaling complex was also disrupted by the core protein. Correspondingly, TRAF2-dependent activation of the protein kinase JNK was suppressed in the core protein-expressing cells. However, NF kappa B activation by TNF was not significantly altered by the HCV core protein, suggesting the existence of TRAF2-independent pathways for NF kappa B activation. These results combined indicate that the HCV core protein sensitizes cells to TNF-induced apoptosis primarily by facilitating FADD recruitment to TNFR1. The inhibition of JNK activation by the HCV core protein may also contribute to the increased propensity of cells for apoptosis. These results, in comparison with other published studies, suggest that the effects of the HCV core protein and their underlying mechanisms vary significantly among cells of different origins.  (+info)

The Fas-Associated Death Domain Protein (FADD), also known as Mort1 or MORT1, is a protein that plays a crucial role in the programmed cell death pathway, also known as apoptosis. It is composed of an N-terminal death effector domain (DED), a middle domain, and a C-terminal death domain (DD).

FADD functions as an adaptor protein that links the Fas receptor to downstream signaling molecules in the extrinsic pathway of apoptosis. When the Fas receptor is activated by its ligand (FasL), it recruits FADD through homotypic interactions between their DED domains. This recruitment leads to the formation of the death-inducing signaling complex (DISC) and the activation of caspase-8, which subsequently activates downstream effector caspases that ultimately lead to cell death.

FADD is essential for maintaining tissue homeostasis by eliminating damaged or potentially harmful cells, and its dysregulation has been implicated in various pathological conditions, including cancer, neurodegenerative diseases, and autoimmune disorders.

TNF Receptor-Associated Death Domain Protein (TRADD) is a type of adaptor protein that plays a crucial role in the intracellular signaling pathways associated with the tumor necrosis factor (TNF) receptor superfamily. TRADD is composed of several functional domains, including a death domain (DD), a really interesting new gene (RING) finger domain, and multiple protein-protein interaction motifs.

When TNF ligands bind to their respective receptors, they induce the formation of a signaling complex, which includes TRADD. The DD of TRADD interacts with the DD of the TNFR1, leading to the recruitment of other signaling proteins such as TNF receptor-associated factor 2 (TRAF2), Fas-associated death domain protein (FADD), and receptor-interacting serine/threonine-protein kinase 1 (RIPK1).

The assembly of this complex triggers two major signaling cascades: the pro-survival NF-κB pathway and the pro-apoptotic caspase activation pathway. TRADD is a key player in both these pathways, acting as a scaffold to facilitate protein-protein interactions and downstream signal transduction events.

In the NF-κB pathway, TRADD recruits TRAF2, which subsequently activates the IKK complex, leading to the nuclear translocation of NF-κB and the induction of target genes involved in cell survival, proliferation, and inflammation. In the caspase activation pathway, TRADD interacts with FADD, forming a death-inducing signaling complex (DISC) that activates caspases 8 and 10, ultimately leading to apoptosis or programmed cell death.

Dysregulation of TRADD-mediated signaling has been implicated in various pathological conditions, including cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, understanding the molecular mechanisms underlying TRADD function is essential for developing novel therapeutic strategies to target these diseases.

Death domain receptor signaling adaptor proteins are a group of intracellular signaling molecules that play a crucial role in the transduction of signals from death receptors, which are a type of cell surface receptor involved in programmed cell death or apoptosis. These adaptor proteins contain a protein-protein interaction module called the death domain (DD), which allows them to interact with other DD-containing proteins and initiate downstream signaling pathways leading to apoptosis.

Some of the key death domain receptor signaling adaptor proteins include Fas-associated death domain protein (FADD), receptor-interacting protein (RIP) kinases, and TNF receptor-associated death domain protein (TRADD). These proteins help to recruit and activate various downstream effectors, such as caspases, which are a family of cysteine proteases that play an essential role in the execution of apoptosis.

Abnormalities in death domain receptor signaling adaptor protein function have been implicated in a variety of diseases, including cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, understanding the mechanisms underlying their regulation and activity is an important area of research with potential therapeutic implications.

Tumor Necrosis Factor (TNF) Receptors are cell surface receptors that bind to tumor necrosis factor cytokines. They play crucial roles in the regulation of a variety of immune cell functions, including inflammation, immunity, and cell survival or death (apoptosis).

There are two major types of TNF receptors: TNFR1 (also known as p55 or CD120a) and TNFR2 (also known as p75 or CD120b). TNFR1 is widely expressed in most tissues, while TNFR2 has a more restricted expression pattern and is mainly found on immune cells.

TNF receptors have an intracellular domain called the death domain, which can trigger signaling pathways leading to apoptosis when activated by TNF ligands. However, they can also activate other signaling pathways that promote cell survival, differentiation, and inflammation. Dysregulation of TNF receptor signaling has been implicated in various diseases, including cancer, autoimmune disorders, and neurodegenerative conditions.

Tumor Necrosis Factor Receptor 1 (TNFR1), also known as p55 or CD120a, is a type I transmembrane protein that belongs to the tumor necrosis factor receptor superfamily. It is widely expressed in various tissues and cells, including immune cells, endothelial cells, and fibroblasts. TNFR1 plays a crucial role in regulating inflammation, immunity, cell survival, differentiation, and apoptosis (programmed cell death).

TNFR1 is activated by its ligand, Tumor Necrosis Factor-alpha (TNF-α), which is a potent proinflammatory cytokine produced mainly by activated macrophages and monocytes. Upon binding of TNF-α to TNFR1, a series of intracellular signaling events are initiated through the recruitment of adaptor proteins, such as TNF receptor-associated death domain (TRADD), receptor-interacting protein kinase 1 (RIPK1), and TNF receptor-associated factor 2 (TRAF2). These interactions lead to the activation of several downstream signaling pathways, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs), which ultimately regulate gene expression and cellular responses.

TNFR1 has been implicated in various physiological and pathological processes, such as inflammation, infection, autoimmunity, cancer, and neurodegenerative disorders. Dysregulation of TNFR1 signaling can contribute to the development and progression of several diseases, making it an attractive target for therapeutic interventions.

TNF Receptor-Associated Factor 1 (TRAF1) is a protein in humans that plays a crucial role in the signaling pathways of tumor necrosis factor (TNF) receptors. TRAF1 is a member of the TRAF family, which includes TRAF1-6. These proteins function as adaptors to mediate signal transduction from the cell surface to the nucleus, ultimately leading to the activation of various transcription factors and the regulation of gene expression.

TRAF1 is primarily associated with the TNFR2 receptor and contributes to the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. These pathways are essential for immune cell activation, differentiation, and survival, as well as inflammatory responses. Dysregulation of TRAF1 function has been implicated in several diseases, including autoimmune disorders and cancer.

In summary, TNF Receptor-Associated Factor 1 (TRAF1) is a protein involved in the signaling pathways of tumor necrosis factor (TNF) receptors, primarily associated with TNFR2, contributing to immune cell activation, differentiation, and survival, as well as inflammatory responses.

Caspase 8 is a type of protease enzyme that plays a crucial role in programmed cell death, also known as apoptosis. It is a key component of the extrinsic pathway of apoptosis, which can be initiated by the binding of death ligands to their respective death receptors on the cell surface.

Once activated, Caspase 8 cleaves and activates other downstream effector caspases, which then go on to degrade various cellular proteins, leading to the characteristic morphological changes associated with apoptosis, such as cell shrinkage, membrane blebbing, and DNA fragmentation.

In addition to its role in apoptosis, Caspase 8 has also been implicated in other cellular processes, including inflammation, differentiation, and proliferation. Dysregulation of Caspase 8 activity has been linked to various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases.

CD95 (also known as Fas or APO-1) is a type of cell surface receptor that can bind to specific proteins and trigger programmed cell death, also known as apoptosis. It is an important regulator of the immune system and helps to control the activation and deletion of immune cells. CD95 ligand (CD95L), the protein that binds to CD95, is expressed on activated T-cells and can induce apoptosis in other cells that express CD95, including other T-cells and tumor cells.

An antigen is any substance that can stimulate an immune response, leading to the production of antibodies or activation of immune cells. In the context of CD95, antigens may refer to substances that can induce the expression of CD95 on the surface of cells, making them susceptible to CD95L-mediated apoptosis. These antigens could include viral proteins, tumor antigens, or other substances that trigger an immune response.

Therefore, the medical definition of 'antigens, CD95' may refer to substances that can induce the expression of CD95 on the surface of cells and make them targets for CD95L-mediated apoptosis.

Adaptor proteins are a type of protein that play a crucial role in intracellular signaling pathways by serving as a link between different components of the signaling complex. Specifically, "signal transducing adaptor proteins" refer to those adaptor proteins that are involved in signal transduction processes, where they help to transmit signals from the cell surface receptors to various intracellular effectors. These proteins typically contain modular domains that allow them to interact with multiple partners, thereby facilitating the formation of large signaling complexes and enabling the integration of signals from different pathways.

Signal transducing adaptor proteins can be classified into several families based on their structural features, including the Src homology 2 (SH2) domain, the Src homology 3 (SH3) domain, and the phosphotyrosine-binding (PTB) domain. These domains enable the adaptor proteins to recognize and bind to specific motifs on other signaling molecules, such as receptor tyrosine kinases, G protein-coupled receptors, and cytokine receptors.

One well-known example of a signal transducing adaptor protein is the growth factor receptor-bound protein 2 (Grb2), which contains an SH2 domain that binds to phosphotyrosine residues on activated receptor tyrosine kinases. Grb2 also contains an SH3 domain that interacts with proline-rich motifs on other signaling proteins, such as the guanine nucleotide exchange factor SOS. This interaction facilitates the activation of the Ras small GTPase and downstream signaling pathways involved in cell growth, differentiation, and survival.

Overall, signal transducing adaptor proteins play a critical role in regulating various cellular processes by modulating intracellular signaling pathways in response to extracellular stimuli. Dysregulation of these proteins has been implicated in various diseases, including cancer and inflammatory disorders.

Receptor-Interacting Protein Serine-Threonine Kinases (RIPKs) are a family of serine-threonine kinases that play crucial roles in the regulation of cell death, inflammation, and immune response. In humans, there are seven known members of this family, RIPK1 to RIPK7, which share a conserved N-terminal kinase domain and C-terminal domains involved in protein-protein interactions.

RIPKs can be activated by various stimuli, including cytokines, pathogens, and stress signals, leading to the phosphorylation of downstream substrates that modulate cellular processes such as apoptosis (programmed cell death), necroptosis (a programmed form of necrosis), and inflammation.

RIPK1 is one of the most well-studied members, acting as a key regulator of both cell survival and death pathways. In response to tumor necrosis factor (TNF) receptor engagement, RIPK1 can form complexes with other proteins that either promote cell survival through the activation of nuclear factor kappa B (NF-κB) or induce cell death via apoptosis or necroptosis.

Dysregulation of RIPKs has been implicated in several pathological conditions, including neurodegenerative diseases, inflammatory disorders, and cancer. Therefore, targeting RIPKs with small molecule inhibitors is an area of active research for the development of novel therapeutic strategies to treat these diseases.

Apoptosis is a programmed and controlled cell death process that occurs in multicellular organisms. It is a natural process that helps maintain tissue homeostasis by eliminating damaged, infected, or unwanted cells. During apoptosis, the cell undergoes a series of morphological changes, including cell shrinkage, chromatin condensation, and fragmentation into membrane-bound vesicles called apoptotic bodies. These bodies are then recognized and engulfed by neighboring cells or phagocytic cells, preventing an inflammatory response. Apoptosis is regulated by a complex network of intracellular signaling pathways that involve proteins such as caspases, Bcl-2 family members, and inhibitors of apoptosis (IAPs).

Caspase-9 is a type of protease enzyme that plays a crucial role in the execution phase of programmed cell death, also known as apoptosis. It is a member of the cysteine-aspartic acid protease (caspase) family, which are characterized by their ability to cleave proteins after an aspartic acid residue. Caspase-9 is activated through a process called cytochrome c-mediated caspase activation, which occurs in the mitochondria during apoptosis. Once activated, caspase-9 cleaves and activates other downstream effector caspases, such as caspase-3 and caspase-7, leading to the proteolytic degradation of cellular structures and ultimately resulting in cell death. Dysregulation of caspase-9 activity has been implicated in various diseases, including neurodegenerative disorders and cancer.

Caspases are a family of protease enzymes that play essential roles in programmed cell death, also known as apoptosis. These enzymes are produced as inactive precursors and are activated when cells receive signals to undergo apoptosis. Once activated, caspases cleave specific protein substrates, leading to the characteristic morphological changes and DNA fragmentation associated with apoptotic cell death. Caspases also play roles in other cellular processes, including inflammation and differentiation. There are two types of caspases: initiator caspases (caspase-2, -8, -9, and -10) and effector caspases (caspase-3, -6, and -7). Initiator caspases are activated in response to various apoptotic signals and then activate the effector caspases, which carry out the proteolytic cleavage of cellular proteins. Dysregulation of caspase activity has been implicated in a variety of diseases, including neurodegenerative disorders, ischemic injury, and cancer.

Tumor Necrosis Factor-alpha (TNF-α) is a cytokine, a type of small signaling protein involved in immune response and inflammation. It is primarily produced by activated macrophages, although other cell types such as T-cells, natural killer cells, and mast cells can also produce it.

TNF-α plays a crucial role in the body's defense against infection and tissue injury by mediating inflammatory responses, activating immune cells, and inducing apoptosis (programmed cell death) in certain types of cells. It does this by binding to its receptors, TNFR1 and TNFR2, which are found on the surface of many cell types.

In addition to its role in the immune response, TNF-α has been implicated in the pathogenesis of several diseases, including autoimmune disorders such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis, as well as cancer, where it can promote tumor growth and metastasis.

Therapeutic agents that target TNF-α, such as infliximab, adalimumab, and etanercept, have been developed to treat these conditions. However, these drugs can also increase the risk of infections and other side effects, so their use must be carefully monitored.

Tumor Necrosis Factor (TNF) Receptor II, also known as TNFRSF1B or CD120b, is a type of receptor that binds to the TNF-alpha cytokine and plays a crucial role in the immune system. It is a transmembrane protein mainly expressed on the surface of various cells including immune cells, fibroblasts, and endothelial cells.

The activation of TNFRII by TNF-alpha leads to the initiation of intracellular signaling pathways that regulate inflammatory responses, cell survival, differentiation, and apoptosis (programmed cell death). Dysregulation of this receptor's function has been implicated in several pathological conditions such as autoimmune diseases, cancer, and neurodegenerative disorders.

TNFRII is a member of the TNF receptor superfamily (TNFRSF) and consists of an extracellular domain containing multiple cysteine-rich motifs that facilitate ligand binding, a transmembrane domain, and an intracellular domain responsible for signal transduction. Upon ligand binding, TNFRII forms complexes with various adaptor proteins to activate downstream signaling cascades, ultimately leading to the activation of nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPKs), and other signaling molecules.

In summary, Tumor Necrosis Factor Receptor II is a key regulator of immune responses and cell fate decisions, with its dysregulation contributing to various pathological conditions.

Carrier proteins, also known as transport proteins, are a type of protein that facilitates the movement of molecules across cell membranes. They are responsible for the selective and active transport of ions, sugars, amino acids, and other molecules from one side of the membrane to the other, against their concentration gradient. This process requires energy, usually in the form of ATP (adenosine triphosphate).

Carrier proteins have a specific binding site for the molecule they transport, and undergo conformational changes upon binding, which allows them to move the molecule across the membrane. Once the molecule has been transported, the carrier protein returns to its original conformation, ready to bind and transport another molecule.

Carrier proteins play a crucial role in maintaining the balance of ions and other molecules inside and outside of cells, and are essential for many physiological processes, including nerve impulse transmission, muscle contraction, and nutrient uptake.

Edar-associated death domain protein (EDARADD) is a gene that encodes for a protein involved in the signaling pathway of the ectodysplasin A receptor (EDAR). The EDAR signaling pathway plays crucial roles in the development of various organs, including skin, hair, teeth, and sweat glands.

The EDARADD protein contains a death domain that interacts with the death domain of EDAR upon activation by ectodysplasin A (EDA). This interaction leads to the recruitment of additional signaling proteins and ultimately activates downstream targets, which regulate cellular processes such as proliferation, differentiation, and apoptosis.

Mutations in the EDARADD gene have been associated with several human genetic disorders, including ectodermal dysplasias, hypohidrotic ectodermal dysplasia (HED), and an autosomal recessive form of cleft lip/palate. These conditions are characterized by abnormalities in the development of structures derived from the ectoderm, such as skin, hair, teeth, nails, and sweat glands.

Signal transduction is the process by which a cell converts an extracellular signal, such as a hormone or neurotransmitter, into an intracellular response. This involves a series of molecular events that transmit the signal from the cell surface to the interior of the cell, ultimately resulting in changes in gene expression, protein activity, or metabolism.

The process typically begins with the binding of the extracellular signal to a receptor located on the cell membrane. This binding event activates the receptor, which then triggers a cascade of intracellular signaling molecules, such as second messengers, protein kinases, and ion channels. These molecules amplify and propagate the signal, ultimately leading to the activation or inhibition of specific cellular responses.

Signal transduction pathways are highly regulated and can be modulated by various factors, including other signaling molecules, post-translational modifications, and feedback mechanisms. Dysregulation of these pathways has been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

TNF Receptor-Associated Factor 2 (TRAF2) is a protein that plays a crucial role in the signaling pathways of tumor necrosis factor (TNF) receptors. TRAF2 is a member of the TRAF family, which includes TRAF1, TRAF2-6, and CD40TRAF. These proteins function as adaptors that mediate signal transduction from the cell surface to the nucleus by interacting with various signaling molecules.

TRAF2 is primarily associated with the TNFR1 receptor, where it binds to the intracellular death domain of the receptor upon TNF-α binding. The formation of this complex leads to the activation of several downstream signaling pathways, including the NF-κB and MAPK pathways, which regulate various cellular processes such as inflammation, immune response, differentiation, and apoptosis.

TRAF2 also plays a role in the regulation of cell death and survival by modulating the activity of caspases, which are protease enzymes that play a central role in programmed cell death or apoptosis. TRAF2 can inhibit caspase activation and promote cell survival by interacting with other proteins such as cIAP1 and cIAP2, which are E3 ubiquitin ligases that target caspases for degradation.

Mutations in the TRAF2 gene have been associated with various diseases, including immunodeficiency, autoimmunity, and cancer. Dysregulation of TRAF2 signaling has been implicated in the pathogenesis of several inflammatory and degenerative disorders, making it a potential therapeutic target for the development of novel drugs to treat these conditions.

Tumor Necrosis Factor Receptor-Associated Proteins (TRAPs) and Peptides are a group of proteins and peptides that interact with the tumor necrosis factor (TNF) receptors. TNF is a cytokine involved in inflammation, immune response, and cell death. TRAPs modulate the signals generated by TNF receptors, thereby regulating various cellular responses such as proliferation, differentiation, survival, and apoptosis (programmed cell death).

TRAPs include adaptor proteins, regulatory proteins, and signaling molecules that are recruited to the TNF receptor complex upon TNF ligand binding. They can have both positive and negative effects on TNF-induced signaling pathways, depending on the specific TRAP involved and the cellular context.

Examples of TRAPs include TNF receptor-associated death domain (TRADD), Fas-associated death domain protein (FADD), receptor-interacting protein (RIP), TNF receptor-associated factor (TRAF) proteins, and cellular inhibitor of apoptosis proteins (cIAPs).

Abnormal regulation of TRAPs has been implicated in various pathological conditions, including cancer, autoimmune diseases, and neurodegenerative disorders. Therefore, understanding the function and regulation of TRAPs is crucial for developing novel therapeutic strategies to target these diseases.

Cell death is the process by which cells cease to function and eventually die. There are several ways that cells can die, but the two most well-known and well-studied forms of cell death are apoptosis and necrosis.

Apoptosis is a programmed form of cell death that occurs as a normal and necessary process in the development and maintenance of healthy tissues. During apoptosis, the cell's DNA is broken down into small fragments, the cell shrinks, and the membrane around the cell becomes fragmented, allowing the cell to be easily removed by phagocytic cells without causing an inflammatory response.

Necrosis, on the other hand, is a form of cell death that occurs as a result of acute tissue injury or overwhelming stress. During necrosis, the cell's membrane becomes damaged and the contents of the cell are released into the surrounding tissue, causing an inflammatory response.

There are also other forms of cell death, such as autophagy, which is a process by which cells break down their own organelles and proteins to recycle nutrients and maintain energy homeostasis, and pyroptosis, which is a form of programmed cell death that occurs in response to infection and involves the activation of inflammatory caspases.

Cell death is an important process in many physiological and pathological processes, including development, tissue homeostasis, and disease. Dysregulation of cell death can contribute to the development of various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases.

NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells) is a protein complex that plays a crucial role in regulating the immune response to infection and inflammation, as well as in cell survival, differentiation, and proliferation. It is composed of several subunits, including p50, p52, p65 (RelA), c-Rel, and RelB, which can form homodimers or heterodimers that bind to specific DNA sequences called κB sites in the promoter regions of target genes.

Under normal conditions, NF-κB is sequestered in the cytoplasm by inhibitory proteins known as IκBs (inhibitors of κB). However, upon stimulation by various signals such as cytokines, bacterial or viral products, and stress, IκBs are phosphorylated, ubiquitinated, and degraded, leading to the release and activation of NF-κB. Activated NF-κB then translocates to the nucleus, where it binds to κB sites and regulates the expression of target genes involved in inflammation, immunity, cell survival, and proliferation.

Dysregulation of NF-κB signaling has been implicated in various pathological conditions such as cancer, chronic inflammation, autoimmune diseases, and neurodegenerative disorders. Therefore, targeting NF-κB signaling has emerged as a potential therapeutic strategy for the treatment of these diseases.

CASP8 and FADD-Like Apoptosis Regulating Protein, also known as CFLAR or FLIP, is a protein that plays a role in regulating cell death (apoptosis). It is a member of the inhibitor of apoptosis protein (IAP) family. The protein contains a death effector domain (DED), which allows it to interact with other proteins that contain DEDs, such as FADD and caspase-8.

CFLAR can function as an inhibitor or a promoter of apoptosis, depending on the context. When CFLAR is present in high levels, it can bind to and inhibit the activity of caspase-8, preventing the initiation of the apoptotic signaling pathway. However, when CFLAR is present in low levels or is cleaved by proteases, it can promote apoptosis by facilitating the activation of caspase-8.

Mutations in the gene that encodes CFLAR have been associated with an increased risk of developing certain types of cancer, such as Hodgkin lymphoma and diffuse large B-cell lymphoma.

Tonsillitis is a medical condition characterized by inflammation and infection of the tonsils, which are two masses of lymphoid tissue located on either side of the back of the throat. The tonsils serve as a defense mechanism against inhaled or ingested pathogens; however, they can become infected themselves, leading to tonsillitis.

The inflammation of the tonsils is often accompanied by symptoms such as sore throat, difficulty swallowing, fever, swollen and tender lymph nodes in the neck, cough, headache, and fatigue. In severe or recurrent cases, a tonsillectomy (surgical removal of the tonsils) may be recommended to alleviate symptoms and prevent complications.

Tonsillitis can be caused by both viral and bacterial infections, with group A streptococcus being one of the most common bacterial causes. It is typically diagnosed based on a physical examination and medical history, and sometimes further confirmed through laboratory tests such as a throat swab or rapid strep test. Treatment may include antibiotics for bacterial tonsillitis, pain relievers, and rest to aid in recovery.

Lymphadenitis is a medical term that refers to the inflammation of one or more lymph nodes, which are small, bean-shaped glands that are part of the body's immune system. Lymph nodes contain white blood cells called lymphocytes, which help fight infection and disease.

Lymphadenitis can occur as a result of an infection in the area near the affected lymph node or as a result of a systemic infection that has spread through the bloodstream. The inflammation causes the lymph node to become swollen, tender, and sometimes painful to the touch.

The symptoms of lymphadenitis may include fever, fatigue, and redness or warmth in the area around the affected lymph node. In some cases, the overlying skin may also appear red and inflamed. Lymphadenitis can occur in any part of the body where there are lymph nodes, including the neck, armpits, groin, and abdomen.

The underlying cause of lymphadenitis must be diagnosed and treated promptly to prevent complications such as the spread of infection or the formation of an abscess. Treatment may include antibiotics, pain relievers, and warm compresses to help reduce swelling and discomfort.

Caspase-10 is a type of protease enzyme that plays a crucial role in programmed cell death, also known as apoptosis. It is a member of the cysteine-aspartic acid protease (caspase) family, which are proteases that specifically cleave their substrates after an aspartic acid residue. Caspase-10 is activated in response to various cellular signals, such as those triggered by immune responses or DNA damage, and it contributes to the execution of apoptosis by cleaving and activating other downstream effector caspases. Additionally, caspase-10 has been implicated in the regulation of inflammatory responses.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Fas Ligand Protein (FasL or CD95L) is a type II transmembrane protein belonging to the tumor necrosis factor (TNF) superfamily. It plays a crucial role in programmed cell death, also known as apoptosis. The FasL protein binds to its receptor, Fas (CD95 or APO-1), which is found on the surface of various cells including immune cells. This binding triggers a signaling cascade that leads to apoptosis, helping to regulate the immune response and maintain homeostasis in tissues.

FasL can also be produced as a soluble protein (sFasL) through alternative splicing or proteolytic cleavage of the membrane-bound form. Soluble FasL may have different functions compared to its membrane-bound counterpart, and its role in physiology and disease is still under investigation.

Dysregulation of the Fas/FasL system has been implicated in various pathological conditions, including autoimmune diseases, neurodegenerative disorders, and cancer.

Jurkat cells are a type of human immortalized T lymphocyte (a type of white blood cell) cell line that is commonly used in scientific research. They were originally isolated from the peripheral blood of a patient with acute T-cell leukemia. Jurkat cells are widely used as a model system to study T-cell activation, signal transduction, and apoptosis (programmed cell death). They are also used in the study of HIV infection and replication, as they can be infected with the virus and used to investigate viral replication and host cell responses.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

Proteins are complex, large molecules that play critical roles in the body's functions. They are made up of amino acids, which are organic compounds that are the building blocks of proteins. Proteins are required for the structure, function, and regulation of the body's tissues and organs. They are essential for the growth, repair, and maintenance of body tissues, and they play a crucial role in many biological processes, including metabolism, immune response, and cellular signaling. Proteins can be classified into different types based on their structure and function, such as enzymes, hormones, antibodies, and structural proteins. They are found in various foods, especially animal-derived products like meat, dairy, and eggs, as well as plant-based sources like beans, nuts, and grains.

Apoptosis regulatory proteins are a group of proteins that play an essential role in the regulation and execution of apoptosis, also known as programmed cell death. This process is a normal part of development and tissue homeostasis, allowing for the elimination of damaged or unnecessary cells. The balance between pro-apoptotic and anti-apoptotic proteins determines whether a cell will undergo apoptosis.

Pro-apoptotic proteins, such as BAX, BID, and PUMA, promote apoptosis by neutralizing or counteracting the effects of anti-apoptotic proteins or by directly activating the apoptotic pathway. These proteins can be activated in response to various stimuli, including DNA damage, oxidative stress, and activation of the death receptor pathway.

Anti-apoptotic proteins, such as BCL-2, BCL-XL, and MCL-1, inhibit apoptosis by binding and neutralizing pro-apoptotic proteins or by preventing the release of cytochrome c from the mitochondria, which is a key step in the intrinsic apoptotic pathway.

Dysregulation of apoptosis regulatory proteins has been implicated in various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, understanding the role of these proteins in apoptosis regulation is crucial for developing new therapeutic strategies to treat these conditions.

TNF-related apoptosis-inducing ligand (TRAIL) receptors are a group of cell surface proteins that belong to the tumor necrosis factor (TNF) receptor superfamily. There are four known TRAIL receptors, referred to as TRAIL-R1, TRAIL-R2, TRAIL-R3, and TRAIL-R4.

TRAIL receptors play a crucial role in the regulation of programmed cell death, also known as apoptosis. TRAIL binding to its receptors TRAIL-R1 and TRAIL-R2 can trigger the activation of intracellular signaling pathways that lead to apoptotic cell death. This is an important mechanism for eliminating damaged or abnormal cells, including cancer cells.

On the other hand, TRAIL receptors TRAIL-R3 and TRAIL-R4 do not transmit apoptotic signals because they lack functional death domains. Instead, they act as decoy receptors that can bind to TRAIL and prevent it from interacting with TRAIL-R1 and TRAIL-R2, thereby inhibiting TRAIL-induced apoptosis.

Abnormalities in the regulation of TRAIL receptor signaling have been implicated in various pathological conditions, including cancer, autoimmune diseases, and neurodegenerative disorders. Therefore, targeting TRAIL receptors has emerged as a promising therapeutic strategy for the treatment of these diseases.

TNF-Related Apoptosis-Inducing Ligand (TRAIL) is a type II transmembrane protein and a member of the tumor necrosis factor (TNF) ligand family. It binds to death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5), leading to the activation of extrinsic apoptosis pathway in sensitive cells. This protein is involved in immune surveillance against tumor cells, as it can selectively induce apoptosis in malignant or virus-infected cells while sparing normal cells. TRAIL also plays a role in inflammation and innate immunity.

The "cause of death" is a medical determination of the disease, injury, or event that directly results in a person's death. This information is typically documented on a death certificate and may be used for public health surveillance, research, and legal purposes. The cause of death is usually determined by a physician based on their clinical judgment and any available medical evidence, such as laboratory test results, autopsy findings, or eyewitness accounts. In some cases, the cause of death may be uncertain or unknown, and the death may be classified as "natural," "accidental," "homicide," or "suicide" based on the available information.

Intracellular signaling peptides and proteins are molecules that play a crucial role in transmitting signals within cells, which ultimately lead to changes in cell behavior or function. These signals can originate from outside the cell (extracellular) or within the cell itself. Intracellular signaling molecules include various types of peptides and proteins, such as:

1. G-protein coupled receptors (GPCRs): These are seven-transmembrane domain receptors that bind to extracellular signaling molecules like hormones, neurotransmitters, or chemokines. Upon activation, they initiate a cascade of intracellular signals through G proteins and secondary messengers.
2. Receptor tyrosine kinases (RTKs): These are transmembrane receptors that bind to growth factors, cytokines, or hormones. Activation of RTKs leads to autophosphorylation of specific tyrosine residues, creating binding sites for intracellular signaling proteins such as adapter proteins, phosphatases, and enzymes like Ras, PI3K, and Src family kinases.
3. Second messenger systems: Intracellular second messengers are small molecules that amplify and propagate signals within the cell. Examples include cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), diacylglycerol (DAG), inositol triphosphate (IP3), calcium ions (Ca2+), and nitric oxide (NO). These second messengers activate or inhibit various downstream effectors, leading to changes in cellular responses.
4. Signal transduction cascades: Intracellular signaling proteins often form complex networks of interacting molecules that relay signals from the plasma membrane to the nucleus. These cascades involve kinases (protein kinases A, B, C, etc.), phosphatases, and adapter proteins, which ultimately regulate gene expression, cell cycle progression, metabolism, and other cellular processes.
5. Ubiquitination and proteasome degradation: Intracellular signaling pathways can also control protein stability by modulating ubiquitin-proteasome degradation. E3 ubiquitin ligases recognize specific substrates and conjugate them with ubiquitin molecules, targeting them for proteasomal degradation. This process regulates the abundance of key signaling proteins and contributes to signal termination or amplification.

In summary, intracellular signaling pathways involve a complex network of interacting proteins that relay signals from the plasma membrane to various cellular compartments, ultimately regulating gene expression, metabolism, and other cellular processes. Dysregulation of these pathways can contribute to disease development and progression, making them attractive targets for therapeutic intervention.

Tertiary protein structure refers to the three-dimensional arrangement of all the elements (polypeptide chains) of a single protein molecule. It is the highest level of structural organization and results from interactions between various side chains (R groups) of the amino acids that make up the protein. These interactions, which include hydrogen bonds, ionic bonds, van der Waals forces, and disulfide bridges, give the protein its unique shape and stability, which in turn determines its function. The tertiary structure of a protein can be stabilized by various factors such as temperature, pH, and the presence of certain ions. Any changes in these factors can lead to denaturation, where the protein loses its tertiary structure and thus its function.

Enzyme activation refers to the process by which an enzyme becomes biologically active and capable of carrying out its specific chemical or biological reaction. This is often achieved through various post-translational modifications, such as proteolytic cleavage, phosphorylation, or addition of cofactors or prosthetic groups to the enzyme molecule. These modifications can change the conformation or structure of the enzyme, exposing or creating a binding site for the substrate and allowing the enzymatic reaction to occur.

For example, in the case of proteolytic cleavage, an inactive precursor enzyme, known as a zymogen, is cleaved into its active form by a specific protease. This is seen in enzymes such as trypsin and chymotrypsin, which are initially produced in the pancreas as inactive precursors called trypsinogen and chymotrypsinogen, respectively. Once they reach the small intestine, they are activated by enteropeptidase, a protease that cleaves a specific peptide bond, releasing the active enzyme.

Phosphorylation is another common mechanism of enzyme activation, where a phosphate group is added to a specific serine, threonine, or tyrosine residue on the enzyme by a protein kinase. This modification can alter the conformation of the enzyme and create a binding site for the substrate, allowing the enzymatic reaction to occur.

Enzyme activation is a crucial process in many biological pathways, as it allows for precise control over when and where specific reactions take place. It also provides a mechanism for regulating enzyme activity in response to various signals and stimuli, such as hormones, neurotransmitters, or changes in the intracellular environment.

'Death domain receptors' (also known as 'death receptors') are a type of transmembrane receptor proteins that play a crucial role in activating programmed cell death, or apoptosis, in response to specific signals. These receptors have an intracellular domain called the 'death domain,' which can interact with other proteins to initiate the signaling cascade leading to cell death. This process is essential for maintaining tissue homeostasis and eliminating damaged, infected, or potentially cancerous cells. Examples of death domain receptors include Fas (CD95), TNFR1 (Tumor Necrosis Factor Receptor 1), and DR3 (Death Receptor 3).

Membrane glycoproteins are proteins that contain oligosaccharide chains (glycans) covalently attached to their polypeptide backbone. They are integral components of biological membranes, spanning the lipid bilayer and playing crucial roles in various cellular processes.

The glycosylation of these proteins occurs in the endoplasmic reticulum (ER) and Golgi apparatus during protein folding and trafficking. The attached glycans can vary in structure, length, and composition, which contributes to the diversity of membrane glycoproteins.

Membrane glycoproteins can be classified into two main types based on their orientation within the lipid bilayer:

1. Type I (N-linked): These glycoproteins have a single transmembrane domain and an extracellular N-terminus, where the oligosaccharides are predominantly attached via asparagine residues (Asn-X-Ser/Thr sequon).
2. Type II (C-linked): These glycoproteins possess two transmembrane domains and an intracellular C-terminus, with the oligosaccharides linked to tryptophan residues via a mannose moiety.

Membrane glycoproteins are involved in various cellular functions, such as:

* Cell adhesion and recognition
* Receptor-mediated signal transduction
* Enzymatic catalysis
* Transport of molecules across membranes
* Cell-cell communication
* Immunological responses

Some examples of membrane glycoproteins include cell surface receptors (e.g., growth factor receptors, cytokine receptors), adhesion molecules (e.g., integrins, cadherins), and transporters (e.g., ion channels, ABC transporters).

Sequence homology, amino acid, refers to the similarity in the order of amino acids in a protein or a portion of a protein between two or more species. This similarity can be used to infer evolutionary relationships and functional similarities between proteins. The higher the degree of sequence homology, the more likely it is that the proteins are related and have similar functions. Sequence homology can be determined through various methods such as pairwise alignment or multiple sequence alignment, which compare the sequences and calculate a score based on the number and type of matching amino acids.

Caspase-3 is a type of protease enzyme that plays a central role in the execution-phase of cell apoptosis, or programmed cell death. It's also known as CPP32 (CPP for ced-3 protease precursor) or apopain. Caspase-3 is produced as an inactive protein that is activated when cleaved by other caspases during the early stages of apoptosis. Once activated, it cleaves a variety of cellular proteins, including structural proteins, enzymes, and signal transduction proteins, leading to the characteristic morphological and biochemical changes associated with apoptotic cell death. Caspase-3 is often referred to as the "death protease" because of its crucial role in executing the cell death program.

Protein binding, in the context of medical and biological sciences, refers to the interaction between a protein and another molecule (known as the ligand) that results in a stable complex. This process is often reversible and can be influenced by various factors such as pH, temperature, and concentration of the involved molecules.

In clinical chemistry, protein binding is particularly important when it comes to drugs, as many of them bind to proteins (especially albumin) in the bloodstream. The degree of protein binding can affect a drug's distribution, metabolism, and excretion, which in turn influence its therapeutic effectiveness and potential side effects.

Protein-bound drugs may be less available for interaction with their target tissues, as only the unbound or "free" fraction of the drug is active. Therefore, understanding protein binding can help optimize dosing regimens and minimize adverse reactions.

Caspase inhibitors are substances or molecules that block the activity of caspases, which are a family of enzymes involved in programmed cell death, also known as apoptosis. Caspases play a crucial role in the execution phase of apoptosis by cleaving various proteins and thereby bringing about characteristic changes in the cell, such as cell shrinkage, membrane blebbing, and DNA fragmentation.

Caspase inhibitors can be synthetic or natural compounds that bind to caspases and prevent them from carrying out their function. These inhibitors have been used in research to study the role of caspases in various biological processes and have also been explored as potential therapeutic agents for conditions associated with excessive apoptosis, such as neurodegenerative diseases and ischemia-reperfusion injury.

It's important to note that while caspase inhibitors can prevent apoptotic cell death, they may also have unintended consequences, such as promoting the survival of damaged or cancerous cells. Therefore, their use as therapeutic agents must be carefully evaluated and balanced against potential risks.

Transfection is a term used in molecular biology that refers to the process of deliberately introducing foreign genetic material (DNA, RNA or artificial gene constructs) into cells. This is typically done using chemical or physical methods, such as lipofection or electroporation. Transfection is widely used in research and medical settings for various purposes, including studying gene function, producing proteins, developing gene therapies, and creating genetically modified organisms. It's important to note that transfection is different from transduction, which is the process of introducing genetic material into cells using viruses as vectors.

LIM domain proteins are a group of transcription factors that contain LIM domains, which are cysteine-rich zinc-binding motifs. These proteins play crucial roles in various cellular processes such as gene regulation, cell proliferation, differentiation, and migration. They are involved in the development and functioning of several organ systems including the nervous system, cardiovascular system, and musculoskeletal system. LIM domain proteins can interact with other proteins and DNA to regulate gene expression and have been implicated in various diseases such as cancer and neurological disorders.

"Cells, cultured" is a medical term that refers to cells that have been removed from an organism and grown in controlled laboratory conditions outside of the body. This process is called cell culture and it allows scientists to study cells in a more controlled and accessible environment than they would have inside the body. Cultured cells can be derived from a variety of sources, including tissues, organs, or fluids from humans, animals, or cell lines that have been previously established in the laboratory.

Cell culture involves several steps, including isolation of the cells from the tissue, purification and characterization of the cells, and maintenance of the cells in appropriate growth conditions. The cells are typically grown in specialized media that contain nutrients, growth factors, and other components necessary for their survival and proliferation. Cultured cells can be used for a variety of purposes, including basic research, drug development and testing, and production of biological products such as vaccines and gene therapies.

It is important to note that cultured cells may behave differently than they do in the body, and results obtained from cell culture studies may not always translate directly to human physiology or disease. Therefore, it is essential to validate findings from cell culture experiments using additional models and ultimately in clinical trials involving human subjects.

Recombinant fusion proteins are artificially created biomolecules that combine the functional domains or properties of two or more different proteins into a single protein entity. They are generated through recombinant DNA technology, where the genes encoding the desired protein domains are linked together and expressed as a single, chimeric gene in a host organism, such as bacteria, yeast, or mammalian cells.

The resulting fusion protein retains the functional properties of its individual constituent proteins, allowing for novel applications in research, diagnostics, and therapeutics. For instance, recombinant fusion proteins can be designed to enhance protein stability, solubility, or immunogenicity, making them valuable tools for studying protein-protein interactions, developing targeted therapies, or generating vaccines against infectious diseases or cancer.

Examples of recombinant fusion proteins include:

1. Etaglunatide (ABT-523): A soluble Fc fusion protein that combines the heavy chain fragment crystallizable region (Fc) of an immunoglobulin with the extracellular domain of the human interleukin-6 receptor (IL-6R). This fusion protein functions as a decoy receptor, neutralizing IL-6 and its downstream signaling pathways in rheumatoid arthritis.
2. Etanercept (Enbrel): A soluble TNF receptor p75 Fc fusion protein that binds to tumor necrosis factor-alpha (TNF-α) and inhibits its proinflammatory activity, making it a valuable therapeutic option for treating autoimmune diseases like rheumatoid arthritis, ankylosing spondylitis, and psoriasis.
3. Abatacept (Orencia): A fusion protein consisting of the extracellular domain of cytotoxic T-lymphocyte antigen 4 (CTLA-4) linked to the Fc region of an immunoglobulin, which downregulates T-cell activation and proliferation in autoimmune diseases like rheumatoid arthritis.
4. Belimumab (Benlysta): A monoclonal antibody that targets B-lymphocyte stimulator (BLyS) protein, preventing its interaction with the B-cell surface receptor and inhibiting B-cell activation in systemic lupus erythematosus (SLE).
5. Romiplostim (Nplate): A fusion protein consisting of a thrombopoietin receptor agonist peptide linked to an immunoglobulin Fc region, which stimulates platelet production in patients with chronic immune thrombocytopenia (ITP).
6. Darbepoetin alfa (Aranesp): A hyperglycosylated erythropoiesis-stimulating protein that functions as a longer-acting form of recombinant human erythropoietin, used to treat anemia in patients with chronic kidney disease or cancer.
7. Palivizumab (Synagis): A monoclonal antibody directed against the F protein of respiratory syncytial virus (RSV), which prevents RSV infection and is administered prophylactically to high-risk infants during the RSV season.
8. Ranibizumab (Lucentis): A recombinant humanized monoclonal antibody fragment that binds and inhibits vascular endothelial growth factor A (VEGF-A), used in the treatment of age-related macular degeneration, diabetic retinopathy, and other ocular disorders.
9. Cetuximab (Erbitux): A chimeric monoclonal antibody that binds to epidermal growth factor receptor (EGFR), used in the treatment of colorectal cancer and head and neck squamous cell carcinoma.
10. Adalimumab (Humira): A fully humanized monoclonal antibody that targets tumor necrosis factor-alpha (TNF-α), used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn's disease.
11. Bevacizumab (Avastin): A recombinant humanized monoclonal antibody that binds to VEGF-A, used in the treatment of various cancers, including colorectal, lung, breast, and kidney cancer.
12. Trastuzumab (Herceptin): A humanized monoclonal antibody that targets HER2/neu receptor, used in the treatment of breast cancer.
13. Rituximab (Rituxan): A chimeric monoclonal antibody that binds to CD20 antigen on B cells, used in the treatment of non-Hodgkin's lymphoma and rheumatoid arthritis.
14. Palivizumab (Synagis): A humanized monoclonal antibody that binds to the F protein of respiratory syncytial virus, used in the prevention of respiratory syncytial virus infection in high-risk infants.
15. Infliximab (Remicade): A chimeric monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including Crohn's disease, ulcerative colitis, rheumatoid arthritis, and ankylosing spondylitis.
16. Natalizumab (Tysabri): A humanized monoclonal antibody that binds to α4β1 integrin, used in the treatment of multiple sclerosis and Crohn's disease.
17. Adalimumab (Humira): A fully human monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
18. Golimumab (Simponi): A fully human monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and ulcerative colitis.
19. Certolizumab pegol (Cimzia): A PEGylated Fab' fragment of a humanized monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and Crohn's disease.
20. Ustekinumab (Stelara): A fully human monoclonal antibody that targets IL-12 and IL-23, used in the treatment of psoriasis, psoriatic arthritis, and Crohn's disease.
21. Secukinumab (Cosentyx): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis.
22. Ixekizumab (Taltz): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis and psoriatic arthritis.
23. Brodalumab (Siliq): A fully human monoclonal antibody that targets IL-17 receptor A, used in the treatment of psoriasis.
24. Sarilumab (Kevzara): A fully human monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis.
25. Tocilizumab (Actemra): A humanized monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis, systemic juvenile idiopathic arthritis, polyarticular juvenile idiopathic arthritis, giant cell arteritis, and chimeric antigen receptor T-cell-induced cytokine release syndrome.
26. Siltuximab (Sylvant): A chimeric monoclonal antibody that targets IL-6, used in the treatment of multicentric Castleman disease.
27. Satralizumab (Enspryng): A humanized monoclonal antibody that targets IL-6 receptor alpha, used in the treatment of neuromyelitis optica spectrum disorder.
28. Sirukumab (Plivensia): A human monoclonal antibody that targets IL-6, used in the treatment

An amino acid sequence is the specific order of amino acids in a protein or peptide molecule, formed by the linking of the amino group (-NH2) of one amino acid to the carboxyl group (-COOH) of another amino acid through a peptide bond. The sequence is determined by the genetic code and is unique to each type of protein or peptide. It plays a crucial role in determining the three-dimensional structure and function of proteins.

Fatty acid desaturases are enzymes that introduce double bonds into fatty acid molecules, thereby reducing their saturation level. These enzymes play a crucial role in the synthesis of unsaturated fatty acids, which are essential components of cell membranes and precursors for various signaling molecules.

The position of the introduced double bond is specified by the type of desaturase enzyme. For example, Δ-9 desaturases introduce a double bond at the ninth carbon atom from the methyl end of the fatty acid chain. This enzyme is responsible for converting saturated fatty acids like stearic acid (18:0) to monounsaturated fatty acids like oleic acid (18:1n-9).

In humans, there are several fatty acid desaturases, including Δ-5 and Δ-6 desaturases, which introduce double bonds at the fifth and sixth carbon atoms from the methyl end, respectively. These enzymes are essential for the synthesis of long-chain polyunsaturated fatty acids (LC-PUFAs) such as arachidonic acid (20:4n-6), eicosapentaenoic acid (EPA, 20:5n-3), and docosahexaenoic acid (DHA, 22:6n-3).

Disorders in fatty acid desaturase activity or expression have been linked to various diseases, including cardiovascular disease, cancer, and metabolic disorders. Therefore, understanding the regulation and function of these enzymes is crucial for developing strategies to modulate fatty acid composition in cells and tissues, which may have therapeutic potential.

CD (cluster of differentiation) antigens are cell-surface proteins that are expressed on leukocytes (white blood cells) and can be used to identify and distinguish different subsets of these cells. They are important markers in the field of immunology and hematology, and are commonly used to diagnose and monitor various diseases, including cancer, autoimmune disorders, and infectious diseases.

CD antigens are designated by numbers, such as CD4, CD8, CD19, etc., which refer to specific proteins found on the surface of different types of leukocytes. For example, CD4 is a protein found on the surface of helper T cells, while CD8 is found on cytotoxic T cells.

CD antigens can be used as targets for immunotherapy, such as monoclonal antibody therapy, in which antibodies are designed to bind to specific CD antigens and trigger an immune response against cancer cells or infected cells. They can also be used as markers to monitor the effectiveness of treatments and to detect minimal residual disease (MRD) after treatment.

It's important to note that not all CD antigens are exclusive to leukocytes, some can be found on other cell types as well, and their expression can vary depending on the activation state or differentiation stage of the cells.

Tumor Necrosis Factor Receptor Superfamily Member 25 (TNFRSF25), also known as Death Receptor 3 (DR3) or APO-3, is a type of cell surface receptor that belongs to the Tumor Necrosis Factor Receptor Superfamily (TNFRSF). These receptors are involved in various biological processes such as immune regulation, inflammation, and apoptosis (programmed cell death).

TNFRSF25 is composed of an extracellular domain that binds to its ligand, Tumor Necrosis Factor-like protein 1A (TL1A), and an intracellular domain that mediates signal transduction. The binding of TL1A to TNFRSF25 can activate several signaling pathways, including the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, which regulate cell survival, proliferation, differentiation, and apoptosis.

In the context of tumors, TNFRSF25 has been found to be expressed in various types of cancer cells, including colorectal, gastric, and breast cancer. The activation of TNFRSF25 by TL1A can induce apoptosis in some cancer cells, suggesting that it may have potential as a therapeutic target for cancer treatment. However, the role of TNFRSF25 in tumor development and progression is complex and context-dependent, and further research is needed to fully understand its functions and clinical relevance.

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

CRADD, or Cav-1 related death domain protein, is a signaling adaptor protein that plays a role in regulating cell death and survival pathways. It contains a death domain that allows it to interact with other proteins involved in these pathways, including the tumor suppressor protein p53 and the death receptor Fas. CRADD has been implicated in a number of cellular processes, including apoptosis (programmed cell death), autophagy, and inflammation. Mutations in the CRADD gene have been associated with various diseases, including neurodevelopmental disorders and cancer.

Recombinant proteins are artificially created proteins produced through the use of recombinant DNA technology. This process involves combining DNA molecules from different sources to create a new set of genes that encode for a specific protein. The resulting recombinant protein can then be expressed, purified, and used for various applications in research, medicine, and industry.

Recombinant proteins are widely used in biomedical research to study protein function, structure, and interactions. They are also used in the development of diagnostic tests, vaccines, and therapeutic drugs. For example, recombinant insulin is a common treatment for diabetes, while recombinant human growth hormone is used to treat growth disorders.

The production of recombinant proteins typically involves the use of host cells, such as bacteria, yeast, or mammalian cells, which are engineered to express the desired protein. The host cells are transformed with a plasmid vector containing the gene of interest, along with regulatory elements that control its expression. Once the host cells are cultured and the protein is expressed, it can be purified using various chromatography techniques.

Overall, recombinant proteins have revolutionized many areas of biology and medicine, enabling researchers to study and manipulate proteins in ways that were previously impossible.

JNK (c-Jun N-terminal kinase) Mitogen-Activated Protein Kinases are a subgroup of the Ser/Thr protein kinases that are activated by stress stimuli and play important roles in various cellular processes, including inflammation, apoptosis, and differentiation. They are involved in the regulation of gene expression through phosphorylation of transcription factors such as c-Jun. JNKs are activated by a variety of upstream kinases, including MAP2Ks (MKK4/SEK1 and MKK7), which are in turn activated by MAP3Ks (such as ASK1, MEKK1, MLKs, and TAK1). JNK signaling pathways have been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory diseases.

Phosphorylation is the process of adding a phosphate group (a molecule consisting of one phosphorus atom and four oxygen atoms) to a protein or other organic molecule, which is usually done by enzymes called kinases. This post-translational modification can change the function, localization, or activity of the target molecule, playing a crucial role in various cellular processes such as signal transduction, metabolism, and regulation of gene expression. Phosphorylation is reversible, and the removal of the phosphate group is facilitated by enzymes called phosphatases.

In the context of medical and biological sciences, a "binding site" refers to a specific location on a protein, molecule, or cell where another molecule can attach or bind. This binding interaction can lead to various functional changes in the original protein or molecule. The other molecule that binds to the binding site is often referred to as a ligand, which can be a small molecule, ion, or even another protein.

The binding between a ligand and its target binding site can be specific and selective, meaning that only certain ligands can bind to particular binding sites with high affinity. This specificity plays a crucial role in various biological processes, such as signal transduction, enzyme catalysis, or drug action.

In the case of drug development, understanding the location and properties of binding sites on target proteins is essential for designing drugs that can selectively bind to these sites and modulate protein function. This knowledge can help create more effective and safer therapeutic options for various diseases.

Mitogen-Activated Protein Kinases (MAPKs) are a family of serine/threonine protein kinases that play crucial roles in various cellular processes, including proliferation, differentiation, transformation, and apoptosis, in response to diverse stimuli such as mitogens, growth factors, hormones, cytokines, and environmental stresses. They are highly conserved across eukaryotes and consist of a three-tiered kinase module composed of MAPK kinase kinases (MAP3Ks), MAPK kinases (MKKs or MAP2Ks), and MAPKs.

Activation of MAPKs occurs through a sequential phosphorylation and activation cascade, where MAP3Ks phosphorylate and activate MKKs, which in turn phosphorylate and activate MAPKs at specific residues (Thr-X-Tyr or Ser-Pro motifs). Once activated, MAPKs can further phosphorylate and regulate various downstream targets, including transcription factors and other protein kinases.

There are four major groups of MAPKs in mammals: extracellular signal-regulated kinases (ERK1/2), c-Jun N-terminal kinases (JNK1/2/3), p38 MAPKs (p38α/β/γ/δ), and ERK5/BMK1. Each group of MAPKs has distinct upstream activators, downstream targets, and cellular functions, allowing for a high degree of specificity in signal transduction and cellular responses. Dysregulation of MAPK signaling pathways has been implicated in various human diseases, including cancer, diabetes, neurodegenerative disorders, and inflammatory diseases.

In genetics, sequence alignment is the process of arranging two or more DNA, RNA, or protein sequences to identify regions of similarity or homology between them. This is often done using computational methods to compare the nucleotide or amino acid sequences and identify matching patterns, which can provide insight into evolutionary relationships, functional domains, or potential genetic disorders. The alignment process typically involves adjusting gaps and mismatches in the sequences to maximize the similarity between them, resulting in an aligned sequence that can be visually represented and analyzed.

The cell nucleus is a membrane-bound organelle found in the eukaryotic cells (cells with a true nucleus). It contains most of the cell's genetic material, organized as DNA molecules in complex with proteins, RNA molecules, and histones to form chromosomes.

The primary function of the cell nucleus is to regulate and control the activities of the cell, including growth, metabolism, protein synthesis, and reproduction. It also plays a crucial role in the process of mitosis (cell division) by separating and protecting the genetic material during this process. The nuclear membrane, or nuclear envelope, surrounding the nucleus is composed of two lipid bilayers with numerous pores that allow for the selective transport of molecules between the nucleoplasm (nucleus interior) and the cytoplasm (cell exterior).

The cell nucleus is a vital structure in eukaryotic cells, and its dysfunction can lead to various diseases, including cancer and genetic disorders.

Protein interaction domains and motifs refer to specific regions or sequences within proteins that are involved in mediating interactions between two or more proteins. These elements can be classified into two main categories: domains and motifs.

Domains are structurally conserved regions of a protein that can fold independently and perform specific functions, such as binding to other molecules like DNA, RNA, or other proteins. They typically range from 25 to 500 amino acids in length and can be found in multiple copies within a single protein or shared among different proteins.

Motifs, on the other hand, are shorter sequences of 3-10 amino acids that mediate more localized interactions with other molecules. Unlike domains, motifs may not have well-defined structures and can be found in various contexts within a protein.

Together, these protein interaction domains and motifs play crucial roles in many biological processes, including signal transduction, gene regulation, enzyme function, and protein complex formation. Understanding the specificity and dynamics of these interactions is essential for elucidating cellular functions and developing therapeutic strategies.

Membrane proteins are a type of protein that are embedded in the lipid bilayer of biological membranes, such as the plasma membrane of cells or the inner membrane of mitochondria. These proteins play crucial roles in various cellular processes, including:

1. Cell-cell recognition and signaling
2. Transport of molecules across the membrane (selective permeability)
3. Enzymatic reactions at the membrane surface
4. Energy transduction and conversion
5. Mechanosensation and signal transduction

Membrane proteins can be classified into two main categories: integral membrane proteins, which are permanently associated with the lipid bilayer, and peripheral membrane proteins, which are temporarily or loosely attached to the membrane surface. Integral membrane proteins can further be divided into three subcategories based on their topology:

1. Transmembrane proteins, which span the entire width of the lipid bilayer with one or more alpha-helices or beta-barrels.
2. Lipid-anchored proteins, which are covalently attached to lipids in the membrane via a glycosylphosphatidylinositol (GPI) anchor or other lipid modifications.
3. Monotopic proteins, which are partially embedded in the membrane and have one or more domains exposed to either side of the bilayer.

Membrane proteins are essential for maintaining cellular homeostasis and are targets for various therapeutic interventions, including drug development and gene therapy. However, their structural complexity and hydrophobicity make them challenging to study using traditional biochemical methods, requiring specialized techniques such as X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and single-particle cryo-electron microscopy (cryo-EM).

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

Tumor Necrosis Factor (TNF) Decoy Receptors are soluble forms of TNF receptors that act as decoy molecules to neutralize the activity of TNF-α, a pro-inflammatory cytokine. They function by binding to TNF-α and preventing it from interacting with its cell surface receptors (TNFR1 and TNFR2), thereby inhibiting the downstream signaling cascades that lead to inflammation and tissue damage.

There are two main types of TNF decoy receptors:

1. TNF Receptor 1 (TNFR1, also known as p55 or p60) - This type of decoy receptor is produced by alternative splicing of the TNFR1 gene and can be found in both membrane-bound and soluble forms. The soluble form of TNFR1 acts as a decoy receptor for TNF-α, preventing it from binding to its cell surface receptors.
2. TNF Receptor 2 (TNFR2, also known as p75 or p80) - This type of decoy receptor is primarily found in the soluble form and is produced by proteolytic cleavage of the membrane-bound TNFR2. Soluble TNFR2 can bind to TNF-α with higher affinity than TNFR1, making it a more effective decoy receptor.

TNF decoy receptors have been implicated in various physiological and pathological processes, including inflammation, immune regulation, and cancer. They are being investigated as potential therapeutic targets for the treatment of various inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis.

Molecular cloning is a laboratory technique used to create multiple copies of a specific DNA sequence. This process involves several steps:

1. Isolation: The first step in molecular cloning is to isolate the DNA sequence of interest from the rest of the genomic DNA. This can be done using various methods such as PCR (polymerase chain reaction), restriction enzymes, or hybridization.
2. Vector construction: Once the DNA sequence of interest has been isolated, it must be inserted into a vector, which is a small circular DNA molecule that can replicate independently in a host cell. Common vectors used in molecular cloning include plasmids and phages.
3. Transformation: The constructed vector is then introduced into a host cell, usually a bacterial or yeast cell, through a process called transformation. This can be done using various methods such as electroporation or chemical transformation.
4. Selection: After transformation, the host cells are grown in selective media that allow only those cells containing the vector to grow. This ensures that the DNA sequence of interest has been successfully cloned into the vector.
5. Amplification: Once the host cells have been selected, they can be grown in large quantities to amplify the number of copies of the cloned DNA sequence.

Molecular cloning is a powerful tool in molecular biology and has numerous applications, including the production of recombinant proteins, gene therapy, functional analysis of genes, and genetic engineering.

DNA-binding proteins are a type of protein that have the ability to bind to DNA (deoxyribonucleic acid), the genetic material of organisms. These proteins play crucial roles in various biological processes, such as regulation of gene expression, DNA replication, repair and recombination.

The binding of DNA-binding proteins to specific DNA sequences is mediated by non-covalent interactions, including electrostatic, hydrogen bonding, and van der Waals forces. The specificity of binding is determined by the recognition of particular nucleotide sequences or structural features of the DNA molecule.

DNA-binding proteins can be classified into several categories based on their structure and function, such as transcription factors, histones, and restriction enzymes. Transcription factors are a major class of DNA-binding proteins that regulate gene expression by binding to specific DNA sequences in the promoter region of genes and recruiting other proteins to modulate transcription. Histones are DNA-binding proteins that package DNA into nucleosomes, the basic unit of chromatin structure. Restriction enzymes are DNA-binding proteins that recognize and cleave specific DNA sequences, and are widely used in molecular biology research and biotechnology applications.

HeLa cells are a type of immortalized cell line used in scientific research. They are derived from a cancer that developed in the cervical tissue of Henrietta Lacks, an African-American woman, in 1951. After her death, cells taken from her tumor were found to be capable of continuous division and growth in a laboratory setting, making them an invaluable resource for medical research.

HeLa cells have been used in a wide range of scientific studies, including research on cancer, viruses, genetics, and drug development. They were the first human cell line to be successfully cloned and are able to grow rapidly in culture, doubling their population every 20-24 hours. This has made them an essential tool for many areas of biomedical research.

It is important to note that while HeLa cells have been instrumental in numerous scientific breakthroughs, the story of their origin raises ethical questions about informed consent and the use of human tissue in research.

Death is the cessation of all biological functions that sustain a living organism. It is characterized by the loss of brainstem reflexes, unresponsiveness, and apnea (no breathing). In medical terms, death can be defined as:

1. Cardiopulmonary Death: The irreversible cessation of circulatory and respiratory functions.
2. Brain Death: The irreversible loss of all brain function, including the brainstem. This is often used as a definition of death when performing organ donation.

It's important to note that the exact definition of death can vary somewhat based on cultural, religious, and legal perspectives.

Viral matrix proteins are structural proteins that play a crucial role in the morphogenesis and life cycle of many viruses. They are often located between the viral envelope and the viral genome, serving as a scaffold for virus assembly and budding. These proteins also interact with other viral components, such as the viral genome, capsid proteins, and envelope proteins, to form an infectious virion. Additionally, matrix proteins can have regulatory functions, influencing viral transcription, replication, and host cell responses. The specific functions of viral matrix proteins vary among different virus families.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

MADS domain proteins are a family of transcription factors that play crucial roles in various developmental processes in plants, including flower development and organ formation. The name "MADS" is an acronym derived from the initial letters of four founding members: MCM1 from Saccharomyces cerevisiae, AGAMOUS from Arabidopsis thaliana, DEFICIENS from Antirrhinum majus, and SRF from Homo sapiens.

These proteins share a highly conserved DNA-binding domain called the MADS-box, which binds to specific sequences in the promoter regions of their target genes. The MADS domain proteins often form higher-order complexes through protein-protein interactions, leading to the regulation of gene expression involved in developmental transitions and cell fate determination. In plants, MADS domain proteins have been implicated in various aspects of reproductive development, such as floral meristem identity, floral organ specification, and ovule development.

Transcription factors are proteins that play a crucial role in regulating gene expression by controlling the transcription of DNA to messenger RNA (mRNA). They function by binding to specific DNA sequences, known as response elements, located in the promoter region or enhancer regions of target genes. This binding can either activate or repress the initiation of transcription, depending on the properties and interactions of the particular transcription factor. Transcription factors often act as part of a complex network of regulatory proteins that determine the precise spatiotemporal patterns of gene expression during development, differentiation, and homeostasis in an organism.

A two-hybrid system technique is a type of genetic screening method used in molecular biology to identify protein-protein interactions within an organism, most commonly baker's yeast (Saccharomyces cerevisiae) or Escherichia coli. The name "two-hybrid" refers to the fact that two separate proteins are being examined for their ability to interact with each other.

The technique is based on the modular nature of transcription factors, which typically consist of two distinct domains: a DNA-binding domain (DBD) and an activation domain (AD). In a two-hybrid system, one protein of interest is fused to the DBD, while the second protein of interest is fused to the AD. If the two proteins interact, the DBD and AD are brought in close proximity, allowing for transcriptional activation of a reporter gene that is linked to a specific promoter sequence recognized by the DBD.

The main components of a two-hybrid system include:

1. Bait protein (fused to the DNA-binding domain)
2. Prey protein (fused to the activation domain)
3. Reporter gene (transcribed upon interaction between bait and prey proteins)
4. Promoter sequence (recognized by the DBD when brought in proximity due to interaction)

The two-hybrid system technique has several advantages, including:

1. Ability to screen large libraries of potential interacting partners
2. High sensitivity for detecting weak or transient interactions
3. Applicability to various organisms and protein types
4. Potential for high-throughput analysis

However, there are also limitations to the technique, such as false positives (interactions that do not occur in vivo) and false negatives (lack of detection of true interactions). Additionally, the fusion proteins may not always fold or localize correctly, leading to potential artifacts. Despite these limitations, two-hybrid system techniques remain a valuable tool for studying protein-protein interactions and have contributed significantly to our understanding of various cellular processes.

SRC homology domains, often abbreviated as SH domains, are conserved protein modules that were first identified in the SRC family of non-receptor tyrosine kinases. These domains are involved in various intracellular signaling processes and mediate protein-protein interactions. There are several types of SH domains, including:

1. SH2 domain: This domain is approximately 100 amino acids long and binds to specific phosphotyrosine-containing motifs in other proteins, thereby mediating signal transduction.
2. SH3 domain: This domain is about 60 amino acids long and recognizes proline-rich sequences in target proteins, playing a role in protein-protein interactions and intracellular signaling.
3. SH1 domain: Also known as the tyrosine kinase catalytic domain, this region contains the active site responsible for transferring a phosphate group from ATP to specific tyrosine residues on target proteins.
4. SH4 domain: This domain is present in some SRC family members and serves as a membrane-targeting module by interacting with lipids or transmembrane proteins.

These SH domains allow SRC kinases and other proteins containing them to participate in complex signaling networks that regulate various cellular processes, such as proliferation, differentiation, survival, and migration.

A "knockout" mouse is a genetically engineered mouse in which one or more genes have been deleted or "knocked out" using molecular biology techniques. This allows researchers to study the function of specific genes and their role in various biological processes, as well as potential associations with human diseases. The mice are generated by introducing targeted DNA modifications into embryonic stem cells, which are then used to create a live animal. Knockout mice have been widely used in biomedical research to investigate gene function, disease mechanisms, and potential therapeutic targets.

C57BL/6 (C57 Black 6) is an inbred strain of laboratory mouse that is widely used in biomedical research. The term "inbred" refers to a strain of animals where matings have been carried out between siblings or other closely related individuals for many generations, resulting in a population that is highly homozygous at most genetic loci.

The C57BL/6 strain was established in 1920 by crossing a female mouse from the dilute brown (DBA) strain with a male mouse from the black strain. The resulting offspring were then interbred for many generations to create the inbred C57BL/6 strain.

C57BL/6 mice are known for their robust health, longevity, and ease of handling, making them a popular choice for researchers. They have been used in a wide range of biomedical research areas, including studies of cancer, immunology, neuroscience, cardiovascular disease, and metabolism.

One of the most notable features of the C57BL/6 strain is its sensitivity to certain genetic modifications, such as the introduction of mutations that lead to obesity or impaired glucose tolerance. This has made it a valuable tool for studying the genetic basis of complex diseases and traits.

Overall, the C57BL/6 inbred mouse strain is an important model organism in biomedical research, providing a valuable resource for understanding the genetic and molecular mechanisms underlying human health and disease.

Tumor Necrosis Factor (TNF) is a type of cytokine, which is a category of proteins that are crucial to cell signaling. TNF plays a significant role in the body's immune response and inflammation process. Specifically, it's primarily produced by activated macrophages as a defensive response against infection, but it can also be produced by other cells such as T-cells and NK cells.

TNF has two types of receptors, TNFR1 and TNFR2, through which it exerts its biological effects. These effects include:

1. Activation of immune cells: TNF helps in the activation of other inflammatory cells like more macrophages and stimulates the release of other cytokines.
2. Cell survival or death: Depending on the context, TNF can promote cell survival or induce programmed cell death (apoptosis), particularly in cancer cells.
3. Fever and acute phase response: TNF is one of the mediators that cause fever and the acute phase reaction during an infection.

The term 'Tumor Necrosis Factor' comes from its historical discovery where it was noted to cause necrosis (death) of tumor cells in certain conditions, although this is not its primary function in the body. Overproduction or dysregulation of TNF has been implicated in several diseases such as rheumatoid arthritis, inflammatory bowel disease, and some types of cancer.

Molecular models are three-dimensional representations of molecular structures that are used in the field of molecular biology and chemistry to visualize and understand the spatial arrangement of atoms and bonds within a molecule. These models can be physical or computer-generated and allow researchers to study the shape, size, and behavior of molecules, which is crucial for understanding their function and interactions with other molecules.

Physical molecular models are often made up of balls (representing atoms) connected by rods or sticks (representing bonds). These models can be constructed manually using materials such as plastic or wooden balls and rods, or they can be created using 3D printing technology.

Computer-generated molecular models, on the other hand, are created using specialized software that allows researchers to visualize and manipulate molecular structures in three dimensions. These models can be used to simulate molecular interactions, predict molecular behavior, and design new drugs or chemicals with specific properties. Overall, molecular models play a critical role in advancing our understanding of molecular structures and their functions.

Protein conformation refers to the specific three-dimensional shape that a protein molecule assumes due to the spatial arrangement of its constituent amino acid residues and their associated chemical groups. This complex structure is determined by several factors, including covalent bonds (disulfide bridges), hydrogen bonds, van der Waals forces, and ionic bonds, which help stabilize the protein's unique conformation.

Protein conformations can be broadly classified into two categories: primary, secondary, tertiary, and quaternary structures. The primary structure represents the linear sequence of amino acids in a polypeptide chain. The secondary structure arises from local interactions between adjacent amino acid residues, leading to the formation of recurring motifs such as α-helices and β-sheets. Tertiary structure refers to the overall three-dimensional folding pattern of a single polypeptide chain, while quaternary structure describes the spatial arrangement of multiple folded polypeptide chains (subunits) that interact to form a functional protein complex.

Understanding protein conformation is crucial for elucidating protein function, as the specific three-dimensional shape of a protein directly influences its ability to interact with other molecules, such as ligands, nucleic acids, or other proteins. Any alterations in protein conformation due to genetic mutations, environmental factors, or chemical modifications can lead to loss of function, misfolding, aggregation, and disease states like neurodegenerative disorders and cancer.

Site-directed mutagenesis is a molecular biology technique used to introduce specific and targeted changes to a specific DNA sequence. This process involves creating a new variant of a gene or a specific region of interest within a DNA molecule by introducing a planned, deliberate change, or mutation, at a predetermined site within the DNA sequence.

The methodology typically involves the use of molecular tools such as PCR (polymerase chain reaction), restriction enzymes, and/or ligases to introduce the desired mutation(s) into a plasmid or other vector containing the target DNA sequence. The resulting modified DNA molecule can then be used to transform host cells, allowing for the production of large quantities of the mutated gene or protein for further study.

Site-directed mutagenesis is a valuable tool in basic research, drug discovery, and biotechnology applications where specific changes to a DNA sequence are required to understand gene function, investigate protein structure/function relationships, or engineer novel biological properties into existing genes or proteins.

Amino acid motifs are recurring patterns or sequences of amino acids in a protein molecule. These motifs can be identified through various sequence analysis techniques and often have functional or structural significance. They can be as short as two amino acids in length, but typically contain at least three to five residues.

Some common examples of amino acid motifs include:

1. Active site motifs: These are specific sequences of amino acids that form the active site of an enzyme and participate in catalyzing chemical reactions. For example, the catalytic triad in serine proteases consists of three residues (serine, histidine, and aspartate) that work together to hydrolyze peptide bonds.
2. Signal peptide motifs: These are sequences of amino acids that target proteins for secretion or localization to specific organelles within the cell. For example, a typical signal peptide consists of a positively charged n-region, a hydrophobic h-region, and a polar c-region that directs the protein to the endoplasmic reticulum membrane for translocation.
3. Zinc finger motifs: These are structural domains that contain conserved sequences of amino acids that bind zinc ions and play important roles in DNA recognition and regulation of gene expression.
4. Transmembrane motifs: These are sequences of hydrophobic amino acids that span the lipid bilayer of cell membranes and anchor transmembrane proteins in place.
5. Phosphorylation sites: These are specific serine, threonine, or tyrosine residues that can be phosphorylated by protein kinases to regulate protein function.

Understanding amino acid motifs is important for predicting protein structure and function, as well as for identifying potential drug targets in disease-associated proteins.

Messenger RNA (mRNA) is a type of RNA (ribonucleic acid) that carries genetic information copied from DNA in the form of a series of three-base code "words," each of which specifies a particular amino acid. This information is used by the cell's machinery to construct proteins, a process known as translation. After being transcribed from DNA, mRNA travels out of the nucleus to the ribosomes in the cytoplasm where protein synthesis occurs. Once the protein has been synthesized, the mRNA may be degraded and recycled. Post-transcriptional modifications can also occur to mRNA, such as alternative splicing and addition of a 5' cap and a poly(A) tail, which can affect its stability, localization, and translation efficiency.

Secondary protein structure refers to the local spatial arrangement of amino acid chains in a protein, typically described as regular repeating patterns held together by hydrogen bonds. The two most common types of secondary structures are the alpha-helix (α-helix) and the beta-pleated sheet (β-sheet). In an α-helix, the polypeptide chain twists around itself in a helical shape, with each backbone atom forming a hydrogen bond with the fourth amino acid residue along the chain. This forms a rigid rod-like structure that is resistant to bending or twisting forces. In β-sheets, adjacent segments of the polypeptide chain run parallel or antiparallel to each other and are connected by hydrogen bonds, forming a pleated sheet-like arrangement. These secondary structures provide the foundation for the formation of tertiary and quaternary protein structures, which determine the overall three-dimensional shape and function of the protein.

Nuclear proteins are a category of proteins that are primarily found in the nucleus of a eukaryotic cell. They play crucial roles in various nuclear functions, such as DNA replication, transcription, repair, and RNA processing. This group includes structural proteins like lamins, which form the nuclear lamina, and regulatory proteins, such as histones and transcription factors, that are involved in gene expression. Nuclear localization signals (NLS) often help target these proteins to the nucleus by interacting with importin proteins during active transport across the nuclear membrane.

PDZ domains are protein interaction modules, which are named after the first letters of three proteins in which they were originally discovered: PSD-95, DLG, and ZO-1. These domains are typically located at the C-terminal region of a protein and have a length of approximately 80-90 amino acids. They play a crucial role in organizing and assembling signaling complexes by binding to specific motifs found on other proteins, such as C-terminal PDZ-binding motifs or internal PDZ-binding sites. This ability to interact with multiple partners enables PDZ domains to function as molecular scaffolds that help regulate various cellular processes, including signal transduction, cell adhesion, and trafficking of proteins to specific subcellular locations.

Glucocorticoid-induced TNFR-related protein (GITRP) is not a widely recognized or established medical term in the field of glucocorticoids, tumor necrosis factor receptors (TNFRs), or related proteins. It's possible that there is some confusion with the term, and it might be referring to TNF-related apoptosis-inducing ligand receptor (TRAIL-R) or a specific isoform of this receptor, such as TRAIL-R2/DR5, which can be upregulated by glucocorticoids.

To provide some context, TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs) are a group of death receptors that play a role in the regulation of cell survival and apoptosis (programmed cell death). Glucocorticoids, which are frequently used anti-inflammatory and immunosuppressive agents, have been shown to modulate the expression of TRAIL-Rs on the cell surface. This modulation can potentially influence the sensitivity of cells to TRAIL-induced apoptosis, although the exact mechanisms and clinical relevance are still a subject of ongoing research.

If you require more specific information about 'Glucocorticoid-induced TNFR-related protein' or need clarification on the topic, please provide additional context or details to help better understand the question.

Nerve Growth Factor (NGF) receptors are a type of protein molecule found on the surface of certain cells, specifically those associated with the nervous system. They play a crucial role in the development, maintenance, and survival of neurons (nerve cells). There are two main types of NGF receptors:

1. Tyrosine Kinase Receptor A (TrkA): This is a high-affinity receptor for NGF and is primarily found on sensory neurons and sympathetic neurons. TrkA activation by NGF leads to the initiation of various intracellular signaling pathways that promote neuronal survival, differentiation, and growth.
2. P75 Neurotrophin Receptor (p75NTR): This is a low-affinity receptor for NGF and other neurotrophins. It can function as a coreceptor with Trk receptors to modulate their signals or act independently to mediate cell death under certain conditions.

Together, these two types of NGF receptors help regulate the complex interactions between neurons and their targets during development and throughout adult life.

Complementary DNA (cDNA) is a type of DNA that is synthesized from a single-stranded RNA molecule through the process of reverse transcription. In this process, the enzyme reverse transcriptase uses an RNA molecule as a template to synthesize a complementary DNA strand. The resulting cDNA is therefore complementary to the original RNA molecule and is a copy of its coding sequence, but it does not contain non-coding regions such as introns that are present in genomic DNA.

Complementary DNA is often used in molecular biology research to study gene expression, protein function, and other genetic phenomena. For example, cDNA can be used to create cDNA libraries, which are collections of cloned cDNA fragments that represent the expressed genes in a particular cell type or tissue. These libraries can then be screened for specific genes or gene products of interest. Additionally, cDNA can be used to produce recombinant proteins in heterologous expression systems, allowing researchers to study the structure and function of proteins that may be difficult to express or purify from their native sources.

Lymphotoxin-alpha (LT-alpha), also known as Tumor Necrosis Factor-beta (TNF-beta), is a cytokine that belongs to the TNF superfamily. It is primarily produced by activated CD4+ and CD8+ T cells, and to some extent by B cells, natural killer (NK) cells, and neutrophils. LT-alpha can form homotrimers or heterotrimers with Lymphotoxin-beta (LT-beta), which bind to the LT-beta receptor (LTβR) and herceptin-resistant tumor cells (HRT) on the surface of various cell types, including immune cells, fibroblasts, and endothelial cells.

The activation of the LTβR signaling pathway plays a crucial role in the development and organization of secondary lymphoid organs, such as lymph nodes, Peyer's patches, and spleen. Additionally, LT-alpha has proinflammatory effects, inducing apoptosis in susceptible cells, activating immune cells, and contributing to the pathogenesis of several inflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn's disease.

Dimerization is a process in which two molecules, usually proteins or similar structures, bind together to form a larger complex. This can occur through various mechanisms, such as the formation of disulfide bonds, hydrogen bonding, or other non-covalent interactions. Dimerization can play important roles in cell signaling, enzyme function, and the regulation of gene expression.

In the context of medical research and therapy, dimerization is often studied in relation to specific proteins that are involved in diseases such as cancer. For example, some drugs have been developed to target and inhibit the dimerization of certain proteins, with the goal of disrupting their function and slowing or stopping the progression of the disease.

Western blotting is a laboratory technique used in molecular biology to detect and quantify specific proteins in a mixture of many different proteins. This technique is commonly used to confirm the expression of a protein of interest, determine its size, and investigate its post-translational modifications. The name "Western" blotting distinguishes this technique from Southern blotting (for DNA) and Northern blotting (for RNA).

The Western blotting procedure involves several steps:

1. Protein extraction: The sample containing the proteins of interest is first extracted, often by breaking open cells or tissues and using a buffer to extract the proteins.
2. Separation of proteins by electrophoresis: The extracted proteins are then separated based on their size by loading them onto a polyacrylamide gel and running an electric current through the gel (a process called sodium dodecyl sulfate-polyacrylamide gel electrophoresis or SDS-PAGE). This separates the proteins according to their molecular weight, with smaller proteins migrating faster than larger ones.
3. Transfer of proteins to a membrane: After separation, the proteins are transferred from the gel onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric current in a process called blotting. This creates a replica of the protein pattern on the gel but now immobilized on the membrane for further analysis.
4. Blocking: The membrane is then blocked with a blocking agent, such as non-fat dry milk or bovine serum albumin (BSA), to prevent non-specific binding of antibodies in subsequent steps.
5. Primary antibody incubation: A primary antibody that specifically recognizes the protein of interest is added and allowed to bind to its target protein on the membrane. This step may be performed at room temperature or 4°C overnight, depending on the antibody's properties.
6. Washing: The membrane is washed with a buffer to remove unbound primary antibodies.
7. Secondary antibody incubation: A secondary antibody that recognizes the primary antibody (often coupled to an enzyme or fluorophore) is added and allowed to bind to the primary antibody. This step may involve using a horseradish peroxidase (HRP)-conjugated or alkaline phosphatase (AP)-conjugated secondary antibody, depending on the detection method used later.
8. Washing: The membrane is washed again to remove unbound secondary antibodies.
9. Detection: A detection reagent is added to visualize the protein of interest by detecting the signal generated from the enzyme-conjugated or fluorophore-conjugated secondary antibody. This can be done using chemiluminescent, colorimetric, or fluorescent methods.
10. Analysis: The resulting image is analyzed to determine the presence and quantity of the protein of interest in the sample.

Western blotting is a powerful technique for identifying and quantifying specific proteins within complex mixtures. It can be used to study protein expression, post-translational modifications, protein-protein interactions, and more. However, it requires careful optimization and validation to ensure accurate and reproducible results.

Gene expression is the process by which the information encoded in a gene is used to synthesize a functional gene product, such as a protein or RNA molecule. This process involves several steps: transcription, RNA processing, and translation. During transcription, the genetic information in DNA is copied into a complementary RNA molecule, known as messenger RNA (mRNA). The mRNA then undergoes RNA processing, which includes adding a cap and tail to the mRNA and splicing out non-coding regions called introns. The resulting mature mRNA is then translated into a protein on ribosomes in the cytoplasm through the process of translation.

The regulation of gene expression is a complex and highly controlled process that allows cells to respond to changes in their environment, such as growth factors, hormones, and stress signals. This regulation can occur at various stages of gene expression, including transcriptional activation or repression, RNA processing, mRNA stability, and translation. Dysregulation of gene expression has been implicated in many diseases, including cancer, genetic disorders, and neurological conditions.

'Gene expression regulation' refers to the processes that control whether, when, and where a particular gene is expressed, meaning the production of a specific protein or functional RNA encoded by that gene. This complex mechanism can be influenced by various factors such as transcription factors, chromatin remodeling, DNA methylation, non-coding RNAs, and post-transcriptional modifications, among others. Proper regulation of gene expression is crucial for normal cellular function, development, and maintaining homeostasis in living organisms. Dysregulation of gene expression can lead to various diseases, including cancer and genetic disorders.

A cell membrane, also known as the plasma membrane, is a thin semi-permeable phospholipid bilayer that surrounds all cells in animals, plants, and microorganisms. It functions as a barrier to control the movement of substances in and out of the cell, allowing necessary molecules such as nutrients, oxygen, and signaling molecules to enter while keeping out harmful substances and waste products. The cell membrane is composed mainly of phospholipids, which have hydrophilic (water-loving) heads and hydrophobic (water-fearing) tails. This unique structure allows the membrane to be flexible and fluid, yet selectively permeable. Additionally, various proteins are embedded in the membrane that serve as channels, pumps, receptors, and enzymes, contributing to the cell's overall functionality and communication with its environment.

Cell survival refers to the ability of a cell to continue living and functioning normally, despite being exposed to potentially harmful conditions or treatments. This can include exposure to toxins, radiation, chemotherapeutic drugs, or other stressors that can damage cells or interfere with their normal processes.

In scientific research, measures of cell survival are often used to evaluate the effectiveness of various therapies or treatments. For example, researchers may expose cells to a particular drug or treatment and then measure the percentage of cells that survive to assess its potential therapeutic value. Similarly, in toxicology studies, measures of cell survival can help to determine the safety of various chemicals or substances.

It's important to note that cell survival is not the same as cell proliferation, which refers to the ability of cells to divide and multiply. While some treatments may promote cell survival, they may also inhibit cell proliferation, making them useful for treating diseases such as cancer. Conversely, other treatments may be designed to specifically target and kill cancer cells, even if it means sacrificing some healthy cells in the process.

'Drosophila proteins' refer to the proteins that are expressed in the fruit fly, Drosophila melanogaster. This organism is a widely used model system in genetics, developmental biology, and molecular biology research. The study of Drosophila proteins has contributed significantly to our understanding of various biological processes, including gene regulation, cell signaling, development, and aging.

Some examples of well-studied Drosophila proteins include:

1. HSP70 (Heat Shock Protein 70): A chaperone protein involved in protein folding and protection from stress conditions.
2. TUBULIN: A structural protein that forms microtubules, important for cell division and intracellular transport.
3. ACTIN: A cytoskeletal protein involved in muscle contraction, cell motility, and maintenance of cell shape.
4. BETA-GALACTOSIDASE (LACZ): A reporter protein often used to monitor gene expression patterns in transgenic flies.
5. ENDOGLIN: A protein involved in the development of blood vessels during embryogenesis.
6. P53: A tumor suppressor protein that plays a crucial role in preventing cancer by regulating cell growth and division.
7. JUN-KINASE (JNK): A signaling protein involved in stress response, apoptosis, and developmental processes.
8. DECAPENTAPLEGIC (DPP): A member of the TGF-β (Transforming Growth Factor Beta) superfamily, playing essential roles in embryonic development and tissue homeostasis.

These proteins are often studied using various techniques such as biochemistry, genetics, molecular biology, and structural biology to understand their functions, interactions, and regulation within the cell.

Biological models, also known as physiological models or organismal models, are simplified representations of biological systems, processes, or mechanisms that are used to understand and explain the underlying principles and relationships. These models can be theoretical (conceptual or mathematical) or physical (such as anatomical models, cell cultures, or animal models). They are widely used in biomedical research to study various phenomena, including disease pathophysiology, drug action, and therapeutic interventions.

Examples of biological models include:

1. Mathematical models: These use mathematical equations and formulas to describe complex biological systems or processes, such as population dynamics, metabolic pathways, or gene regulation networks. They can help predict the behavior of these systems under different conditions and test hypotheses about their underlying mechanisms.
2. Cell cultures: These are collections of cells grown in a controlled environment, typically in a laboratory dish or flask. They can be used to study cellular processes, such as signal transduction, gene expression, or metabolism, and to test the effects of drugs or other treatments on these processes.
3. Animal models: These are living organisms, usually vertebrates like mice, rats, or non-human primates, that are used to study various aspects of human biology and disease. They can provide valuable insights into the pathophysiology of diseases, the mechanisms of drug action, and the safety and efficacy of new therapies.
4. Anatomical models: These are physical representations of biological structures or systems, such as plastic models of organs or tissues, that can be used for educational purposes or to plan surgical procedures. They can also serve as a basis for developing more sophisticated models, such as computer simulations or 3D-printed replicas.

Overall, biological models play a crucial role in advancing our understanding of biology and medicine, helping to identify new targets for therapeutic intervention, develop novel drugs and treatments, and improve human health.

X-ray crystallography is a technique used in structural biology to determine the three-dimensional arrangement of atoms in a crystal lattice. In this method, a beam of X-rays is directed at a crystal and diffracts, or spreads out, into a pattern of spots called reflections. The intensity and angle of each reflection are measured and used to create an electron density map, which reveals the position and type of atoms in the crystal. This information can be used to determine the molecular structure of a compound, including its shape, size, and chemical bonds. X-ray crystallography is a powerful tool for understanding the structure and function of biological macromolecules such as proteins and nucleic acids.

A ligand, in the context of biochemistry and medicine, is a molecule that binds to a specific site on a protein or a larger biomolecule, such as an enzyme or a receptor. This binding interaction can modify the function or activity of the target protein, either activating it or inhibiting it. Ligands can be small molecules, like hormones or neurotransmitters, or larger structures, like antibodies. The study of ligand-protein interactions is crucial for understanding cellular processes and developing drugs, as many therapeutic compounds function by binding to specific targets within the body.

Cytoplasm is the material within a eukaryotic cell (a cell with a true nucleus) that lies between the nuclear membrane and the cell membrane. It is composed of an aqueous solution called cytosol, in which various organelles such as mitochondria, ribosomes, endoplasmic reticulum, Golgi apparatus, lysosomes, and vacuoles are suspended. Cytoplasm also contains a variety of dissolved nutrients, metabolites, ions, and enzymes that are involved in various cellular processes such as metabolism, signaling, and transport. It is where most of the cell's metabolic activities take place, and it plays a crucial role in maintaining the structure and function of the cell.

A nerve growth factor (NGF) receptor is a type of protein found on the surface of certain cells that selectively binds to NGF, a neurotrophin or a small signaling protein that promotes the growth and survival of nerve cells. There are two main types of NGF receptors: tyrosine kinase receptor A (TrkA) and p75 neurotrophin receptor (p75NTR). TrkA is a high-affinity receptor that activates various signaling pathways leading to the survival, differentiation, and growth of nerve cells. In contrast, p75NTR has lower affinity for NGF and can either promote or inhibit NGF signaling depending on its interactions with other proteins. Together, these two types of receptors help regulate the development, maintenance, and function of the nervous system.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

A sequence deletion in a genetic context refers to the removal or absence of one or more nucleotides (the building blocks of DNA or RNA) from a specific region in a DNA or RNA molecule. This type of mutation can lead to the loss of genetic information, potentially resulting in changes in the function or expression of a gene. If the deletion involves a critical portion of the gene, it can cause diseases, depending on the role of that gene in the body. The size of the deleted sequence can vary, ranging from a single nucleotide to a large segment of DNA.

DNA primers are short single-stranded DNA molecules that serve as a starting point for DNA synthesis. They are typically used in laboratory techniques such as the polymerase chain reaction (PCR) and DNA sequencing. The primer binds to a complementary sequence on the DNA template through base pairing, providing a free 3'-hydroxyl group for the DNA polymerase enzyme to add nucleotides and synthesize a new strand of DNA. This allows for specific and targeted amplification or analysis of a particular region of interest within a larger DNA molecule.

Protein transport, in the context of cellular biology, refers to the process by which proteins are actively moved from one location to another within or between cells. This is a crucial mechanism for maintaining proper cell function and regulation.

Intracellular protein transport involves the movement of proteins within a single cell. Proteins can be transported across membranes (such as the nuclear envelope, endoplasmic reticulum, Golgi apparatus, or plasma membrane) via specialized transport systems like vesicles and transport channels.

Intercellular protein transport refers to the movement of proteins from one cell to another, often facilitated by exocytosis (release of proteins in vesicles) and endocytosis (uptake of extracellular substances via membrane-bound vesicles). This is essential for communication between cells, immune response, and other physiological processes.

It's important to note that any disruption in protein transport can lead to various diseases, including neurological disorders, cancer, and metabolic conditions.

In the field of medicine, "time factors" refer to the duration of symptoms or time elapsed since the onset of a medical condition, which can have significant implications for diagnosis and treatment. Understanding time factors is crucial in determining the progression of a disease, evaluating the effectiveness of treatments, and making critical decisions regarding patient care.

For example, in stroke management, "time is brain," meaning that rapid intervention within a specific time frame (usually within 4.5 hours) is essential to administering tissue plasminogen activator (tPA), a clot-busting drug that can minimize brain damage and improve patient outcomes. Similarly, in trauma care, the "golden hour" concept emphasizes the importance of providing definitive care within the first 60 minutes after injury to increase survival rates and reduce morbidity.

Time factors also play a role in monitoring the progression of chronic conditions like diabetes or heart disease, where regular follow-ups and assessments help determine appropriate treatment adjustments and prevent complications. In infectious diseases, time factors are crucial for initiating antibiotic therapy and identifying potential outbreaks to control their spread.

Overall, "time factors" encompass the significance of recognizing and acting promptly in various medical scenarios to optimize patient outcomes and provide effective care.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

A conserved sequence in the context of molecular biology refers to a pattern of nucleotides (in DNA or RNA) or amino acids (in proteins) that has remained relatively unchanged over evolutionary time. These sequences are often functionally important and are highly conserved across different species, indicating strong selection pressure against changes in these regions.

In the case of protein-coding genes, the corresponding amino acid sequence is deduced from the DNA sequence through the genetic code. Conserved sequences in proteins may indicate structurally or functionally important regions, such as active sites or binding sites, that are critical for the protein's activity. Similarly, conserved non-coding sequences in DNA may represent regulatory elements that control gene expression.

Identifying conserved sequences can be useful for inferring evolutionary relationships between species and for predicting the function of unknown genes or proteins.

A Structure-Activity Relationship (SAR) in the context of medicinal chemistry and pharmacology refers to the relationship between the chemical structure of a drug or molecule and its biological activity or effect on a target protein, cell, or organism. SAR studies aim to identify patterns and correlations between structural features of a compound and its ability to interact with a specific biological target, leading to a desired therapeutic response or undesired side effects.

By analyzing the SAR, researchers can optimize the chemical structure of lead compounds to enhance their potency, selectivity, safety, and pharmacokinetic properties, ultimately guiding the design and development of novel drugs with improved efficacy and reduced toxicity.

A peptide fragment is a short chain of amino acids that is derived from a larger peptide or protein through various biological or chemical processes. These fragments can result from the natural breakdown of proteins in the body during regular physiological processes, such as digestion, or they can be produced experimentally in a laboratory setting for research or therapeutic purposes.

Peptide fragments are often used in research to map the structure and function of larger peptides and proteins, as well as to study their interactions with other molecules. In some cases, peptide fragments may also have biological activity of their own and can be developed into drugs or diagnostic tools. For example, certain peptide fragments derived from hormones or neurotransmitters may bind to receptors in the body and mimic or block the effects of the full-length molecule.

Genetic transcription is the process by which the information in a strand of DNA is used to create a complementary RNA molecule. This process is the first step in gene expression, where the genetic code in DNA is converted into a form that can be used to produce proteins or functional RNAs.

During transcription, an enzyme called RNA polymerase binds to the DNA template strand and reads the sequence of nucleotide bases. As it moves along the template, it adds complementary RNA nucleotides to the growing RNA chain, creating a single-stranded RNA molecule that is complementary to the DNA template strand. Once transcription is complete, the RNA molecule may undergo further processing before it can be translated into protein or perform its functional role in the cell.

Transcription can be either "constitutive" or "regulated." Constitutive transcription occurs at a relatively constant rate and produces essential proteins that are required for basic cellular functions. Regulated transcription, on the other hand, is subject to control by various intracellular and extracellular signals, allowing cells to respond to changing environmental conditions or developmental cues.

Arabidopsis proteins refer to the proteins that are encoded by the genes in the Arabidopsis thaliana plant, which is a model organism commonly used in plant biology research. This small flowering plant has a compact genome and a short life cycle, making it an ideal subject for studying various biological processes in plants.

Arabidopsis proteins play crucial roles in many cellular functions, such as metabolism, signaling, regulation of gene expression, response to environmental stresses, and developmental processes. Research on Arabidopsis proteins has contributed significantly to our understanding of plant biology and has provided valuable insights into the molecular mechanisms underlying various agronomic traits.

Some examples of Arabidopsis proteins include transcription factors, kinases, phosphatases, receptors, enzymes, and structural proteins. These proteins can be studied using a variety of techniques, such as biochemical assays, protein-protein interaction studies, and genetic approaches, to understand their functions and regulatory mechanisms in plants.

Immunoprecipitation (IP) is a research technique used in molecular biology and immunology to isolate specific antigens or antibodies from a mixture. It involves the use of an antibody that recognizes and binds to a specific antigen, which is then precipitated out of solution using various methods, such as centrifugation or chemical cross-linking.

In this technique, an antibody is first incubated with a sample containing the antigen of interest. The antibody specifically binds to the antigen, forming an immune complex. This complex can then be captured by adding protein A or G agarose beads, which bind to the constant region of the antibody. The beads are then washed to remove any unbound proteins, leaving behind the precipitated antigen-antibody complex.

Immunoprecipitation is a powerful tool for studying protein-protein interactions, post-translational modifications, and signal transduction pathways. It can also be used to detect and quantify specific proteins in biological samples, such as cells or tissues, and to identify potential biomarkers of disease.

A precipitin test is a type of immunodiagnostic test used to detect and measure the presence of specific antibodies or antigens in a patient's serum. The test is based on the principle of antigen-antibody interaction, where the addition of an antigen to a solution containing its corresponding antibody results in the formation of an insoluble immune complex known as a precipitin.

In this test, a small amount of the patient's serum is added to a solution containing a known antigen or antibody. If the patient has antibodies or antigens that correspond to the added reagent, they will bind and form a visible precipitate. The size and density of the precipitate can be used to quantify the amount of antibody or antigen present in the sample.

Precipitin tests are commonly used in the diagnosis of various infectious diseases, autoimmune disorders, and allergies. They can also be used in forensic science to identify biological samples. However, they have largely been replaced by more modern immunological techniques such as enzyme-linked immunosorbent assays (ELISAs) and radioimmunoassays (RIAs).

Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) is a laboratory technique used in molecular biology to amplify and detect specific DNA sequences. This technique is particularly useful for the detection and quantification of RNA viruses, as well as for the analysis of gene expression.

The process involves two main steps: reverse transcription and polymerase chain reaction (PCR). In the first step, reverse transcriptase enzyme is used to convert RNA into complementary DNA (cDNA) by reading the template provided by the RNA molecule. This cDNA then serves as a template for the PCR amplification step.

In the second step, the PCR reaction uses two primers that flank the target DNA sequence and a thermostable polymerase enzyme to repeatedly copy the targeted cDNA sequence. The reaction mixture is heated and cooled in cycles, allowing the primers to anneal to the template, and the polymerase to extend the new strand. This results in exponential amplification of the target DNA sequence, making it possible to detect even small amounts of RNA or cDNA.

RT-PCR is a sensitive and specific technique that has many applications in medical research and diagnostics, including the detection of viruses such as HIV, hepatitis C virus, and SARS-CoV-2 (the virus that causes COVID-19). It can also be used to study gene expression, identify genetic mutations, and diagnose genetic disorders.

Down-regulation is a process that occurs in response to various stimuli, where the number or sensitivity of cell surface receptors or the expression of specific genes is decreased. This process helps maintain homeostasis within cells and tissues by reducing the ability of cells to respond to certain signals or molecules.

In the context of cell surface receptors, down-regulation can occur through several mechanisms:

1. Receptor internalization: After binding to their ligands, receptors can be internalized into the cell through endocytosis. Once inside the cell, these receptors may be degraded or recycled back to the cell surface in smaller numbers.
2. Reduced receptor synthesis: Down-regulation can also occur at the transcriptional level, where the expression of genes encoding for specific receptors is decreased, leading to fewer receptors being produced.
3. Receptor desensitization: Prolonged exposure to a ligand can lead to a decrease in receptor sensitivity or affinity, making it more difficult for the cell to respond to the signal.

In the context of gene expression, down-regulation refers to the decreased transcription and/or stability of specific mRNAs, leading to reduced protein levels. This process can be induced by various factors, including microRNA (miRNA)-mediated regulation, histone modification, or DNA methylation.

Down-regulation is an essential mechanism in many physiological processes and can also contribute to the development of several diseases, such as cancer and neurodegenerative disorders.

Proto-oncogene proteins c-bcl-2 are a group of proteins that play a role in regulating cell death (apoptosis). The c-bcl-2 gene produces one of these proteins, which helps to prevent cells from undergoing apoptosis. This protein is located on the membrane of mitochondria and endoplasmic reticulum and it can inhibit the release of cytochrome c, a key player in the activation of caspases, which are enzymes that trigger apoptosis.

In normal cells, the regulation of c-bcl-2 protein helps to maintain a balance between cell proliferation and cell death, ensuring proper tissue homeostasis. However, when the c-bcl-2 gene is mutated or its expression is dysregulated, it can contribute to cancer development by allowing cancer cells to survive and proliferate. High levels of c-bcl-2 protein have been found in many types of cancer, including leukemia, lymphoma, and carcinomas, and are often associated with a poor prognosis.

Death-associated protein kinases (DAPKs) are a group of serine/threonine protein kinases that have been implicated in the regulation of programmed cell death, also known as apoptosis. There are several isoforms of DAPKs, including DAPK1, DAPK2, and DAPK3, each with distinct functions and regulatory mechanisms.

DAPK1 was the first to be identified and is perhaps the best studied. It plays a critical role in various forms of programmed cell death, including apoptosis, autophagy, and necroptosis. DAPK1 can be activated by various stimuli, such as calcium influx, oxidative stress, and DNA damage, and its activation leads to the phosphorylation of several downstream targets that contribute to the execution of cell death.

DAPK2 and DAPK3 have also been shown to regulate programmed cell death, although their functions are less well understood than those of DAPK1. DAPK2 has been implicated in the regulation of autophagy, while DAPK3 has been suggested to play a role in the regulation of both apoptosis and necroptosis.

Overall, DAPKs are important regulators of programmed cell death and have been implicated in various physiological and pathological processes, including development, neurodegeneration, ischemia-reperfusion injury, and cancer.

Caspase-2 is a type of protease enzyme that plays a role in programmed cell death, also known as apoptosis. It is a member of the cysteine-aspartic acid protease (caspase) family, which are characterized by their ability to cleave proteins at specific aspartate residues. Caspase-2 is activated in response to various signals that trigger apoptosis and helps to carry out the ordered dismantling of the cell. It also has roles in other cellular processes such as cell cycle regulation, DNA repair, and inflammation.

Up-regulation is a term used in molecular biology and medicine to describe an increase in the expression or activity of a gene, protein, or receptor in response to a stimulus. This can occur through various mechanisms such as increased transcription, translation, or reduced degradation of the molecule. Up-regulation can have important functional consequences, for example, enhancing the sensitivity or response of a cell to a hormone, neurotransmitter, or drug. It is a normal physiological process that can also be induced by disease or pharmacological interventions.

Cell surface receptors, also known as membrane receptors, are proteins located on the cell membrane that bind to specific molecules outside the cell, known as ligands. These receptors play a crucial role in signal transduction, which is the process of converting an extracellular signal into an intracellular response.

Cell surface receptors can be classified into several categories based on their structure and mechanism of action, including:

1. Ion channel receptors: These receptors contain a pore that opens to allow ions to flow across the cell membrane when they bind to their ligands. This ion flux can directly activate or inhibit various cellular processes.
2. G protein-coupled receptors (GPCRs): These receptors consist of seven transmembrane domains and are associated with heterotrimeric G proteins that modulate intracellular signaling pathways upon ligand binding.
3. Enzyme-linked receptors: These receptors possess an intrinsic enzymatic activity or are linked to an enzyme, which becomes activated when the receptor binds to its ligand. This activation can lead to the initiation of various signaling cascades within the cell.
4. Receptor tyrosine kinases (RTKs): These receptors contain intracellular tyrosine kinase domains that become activated upon ligand binding, leading to the phosphorylation and activation of downstream signaling molecules.
5. Integrins: These receptors are transmembrane proteins that mediate cell-cell or cell-matrix interactions by binding to extracellular matrix proteins or counter-receptors on adjacent cells. They play essential roles in cell adhesion, migration, and survival.

Cell surface receptors are involved in various physiological processes, including neurotransmission, hormone signaling, immune response, and cell growth and differentiation. Dysregulation of these receptors can contribute to the development of numerous diseases, such as cancer, diabetes, and neurological disorders.

Monoclonal antibodies are a type of antibody that are identical because they are produced by a single clone of cells. They are laboratory-produced molecules that act like human antibodies in the immune system. They can be designed to attach to specific proteins found on the surface of cancer cells, making them useful for targeting and treating cancer. Monoclonal antibodies can also be used as a therapy for other diseases, such as autoimmune disorders and inflammatory conditions.

Monoclonal antibodies are produced by fusing a single type of immune cell, called a B cell, with a tumor cell to create a hybrid cell, or hybridoma. This hybrid cell is then able to replicate indefinitely, producing a large number of identical copies of the original antibody. These antibodies can be further modified and engineered to enhance their ability to bind to specific targets, increase their stability, and improve their effectiveness as therapeutic agents.

Monoclonal antibodies have several mechanisms of action in cancer therapy. They can directly kill cancer cells by binding to them and triggering an immune response. They can also block the signals that promote cancer growth and survival. Additionally, monoclonal antibodies can be used to deliver drugs or radiation directly to cancer cells, increasing the effectiveness of these treatments while minimizing their side effects on healthy tissues.

Monoclonal antibodies have become an important tool in modern medicine, with several approved for use in cancer therapy and other diseases. They are continuing to be studied and developed as a promising approach to treating a wide range of medical conditions.

I-kappa B kinase (IKK) is a protein complex that plays a crucial role in the activation of NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells), a transcription factor involved in the regulation of immune response, inflammation, cell survival, and proliferation.

The IKK complex is composed of two catalytic subunits, IKKα and IKKβ, and a regulatory subunit, IKKγ (also known as NEMO). Upon stimulation by various signals such as cytokines, pathogens, or stress, the IKK complex becomes activated and phosphorylates I-kappa B (IkB), an inhibitor protein that keeps NF-kB in an inactive state in the cytoplasm.

Once IkB is phosphorylated by the IKK complex, it undergoes ubiquitination and degradation, leading to the release and nuclear translocation of NF-kB, where it can bind to specific DNA sequences and regulate gene expression. Dysregulation of IKK activity has been implicated in various pathological conditions, including chronic inflammation, autoimmune diseases, and cancer.

U937 cells are a type of human histiocytic lymphoma cell line that is commonly used in scientific research and studies. They are derived from the peripheral blood of a patient with histiocytic lymphoma, which is a rare type of cancer that affects the immune system's cells called histiocytes.

U937 cells have a variety of uses in research, including studying the mechanisms of cancer cell growth and proliferation, testing the effects of various drugs and treatments on cancer cells, and investigating the role of different genes and proteins in cancer development and progression. These cells are easy to culture and maintain in the laboratory, making them a popular choice for researchers in many fields.

It is important to note that while U937 cells can provide valuable insights into the behavior of cancer cells, they do not necessarily reflect the complexity and diversity of human cancers. Therefore, findings from studies using these cells should be validated in more complex models or clinical trials before being applied to patient care.

Sudden cardiac death (SCD) is a sudden, unexpected natural death caused by the cessation of cardiac activity. It is often caused by cardiac arrhythmias, particularly ventricular fibrillation, and is often associated with underlying heart disease, although it can occur in people with no known heart condition. SCD is typically defined as a natural death due to cardiac causes that occurs within one hour of the onset of symptoms, or if the individual was last seen alive in a normal state of health, it can be defined as occurring within 24 hours.

It's important to note that sudden cardiac arrest (SCA) is different from SCD, although they are related. SCA refers to the sudden cessation of cardiac activity, which if not treated immediately can lead to SCD.

Protein-Serine-Threonine Kinases (PSTKs) are a type of protein kinase that catalyzes the transfer of a phosphate group from ATP to the hydroxyl side chains of serine or threonine residues on target proteins. This phosphorylation process plays a crucial role in various cellular signaling pathways, including regulation of metabolism, gene expression, cell cycle progression, and apoptosis. PSTKs are involved in many physiological and pathological processes, and their dysregulation has been implicated in several diseases, such as cancer, diabetes, and neurodegenerative disorders.

A plasmid is a small, circular, double-stranded DNA molecule that is separate from the chromosomal DNA of a bacterium or other organism. Plasmids are typically not essential for the survival of the organism, but they can confer beneficial traits such as antibiotic resistance or the ability to degrade certain types of pollutants.

Plasmids are capable of replicating independently of the chromosomal DNA and can be transferred between bacteria through a process called conjugation. They often contain genes that provide resistance to antibiotics, heavy metals, and other environmental stressors. Plasmids have also been engineered for use in molecular biology as cloning vectors, allowing scientists to replicate and manipulate specific DNA sequences.

Plasmids are important tools in genetic engineering and biotechnology because they can be easily manipulated and transferred between organisms. They have been used to produce vaccines, diagnostic tests, and genetically modified organisms (GMOs) for various applications, including agriculture, medicine, and industry.

Gene deletion is a type of mutation where a segment of DNA, containing one or more genes, is permanently lost or removed from a chromosome. This can occur due to various genetic mechanisms such as homologous recombination, non-homologous end joining, or other types of genomic rearrangements.

The deletion of a gene can have varying effects on the organism, depending on the function of the deleted gene and its importance for normal physiological processes. If the deleted gene is essential for survival, the deletion may result in embryonic lethality or developmental abnormalities. However, if the gene is non-essential or has redundant functions, the deletion may not have any noticeable effects on the organism's phenotype.

Gene deletions can also be used as a tool in genetic research to study the function of specific genes and their role in various biological processes. For example, researchers may use gene deletion techniques to create genetically modified animal models to investigate the impact of gene deletion on disease progression or development.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a key role in the adaptive immune system's response to infection. They are produced in the bone marrow and mature in the thymus gland. There are several different types of T-cells, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, and regulatory T-cells (Tregs).

CD4+ helper T-cells assist in activating other immune cells, such as B-lymphocytes and macrophages. They also produce cytokines, which are signaling molecules that help coordinate the immune response. CD8+ cytotoxic T-cells directly kill infected cells by releasing toxic substances. Regulatory T-cells help maintain immune tolerance and prevent autoimmune diseases by suppressing the activity of other immune cells.

T-lymphocytes are important in the immune response to viral infections, cancer, and other diseases. Dysfunction or depletion of T-cells can lead to immunodeficiency and increased susceptibility to infections. On the other hand, an overactive T-cell response can contribute to autoimmune diseases and chronic inflammation.

Flow cytometry is a medical and research technique used to measure physical and chemical characteristics of cells or particles, one cell at a time, as they flow in a fluid stream through a beam of light. The properties measured include:

* Cell size (light scatter)
* Cell internal complexity (granularity, also light scatter)
* Presence or absence of specific proteins or other molecules on the cell surface or inside the cell (using fluorescent antibodies or other fluorescent probes)

The technique is widely used in cell counting, cell sorting, protein engineering, biomarker discovery and monitoring disease progression, particularly in hematology, immunology, and cancer research.

Deoxyribonucleic acid (DNA) is the genetic material present in the cells of organisms where it is responsible for the storage and transmission of hereditary information. DNA is a long molecule that consists of two strands coiled together to form a double helix. Each strand is made up of a series of four nucleotide bases - adenine (A), guanine (G), cytosine (C), and thymine (T) - that are linked together by phosphate and sugar groups. The sequence of these bases along the length of the molecule encodes genetic information, with A always pairing with T and C always pairing with G. This base-pairing allows for the replication and transcription of DNA, which are essential processes in the functioning and reproduction of all living organisms.

Necrosis is the premature death of cells or tissues due to damage or injury, such as from infection, trauma, infarction (lack of blood supply), or toxic substances. It's a pathological process that results in the uncontrolled and passive degradation of cellular components, ultimately leading to the release of intracellular contents into the extracellular space. This can cause local inflammation and may lead to further tissue damage if not treated promptly.

There are different types of necrosis, including coagulative, liquefactive, caseous, fat, fibrinoid, and gangrenous necrosis, each with distinct histological features depending on the underlying cause and the affected tissues or organs.

"Saccharomyces cerevisiae" is not typically considered a medical term, but it is a scientific name used in the field of microbiology. It refers to a species of yeast that is commonly used in various industrial processes, such as baking and brewing. It's also widely used in scientific research due to its genetic tractability and eukaryotic cellular organization.

However, it does have some relevance to medical fields like medicine and nutrition. For example, certain strains of S. cerevisiae are used as probiotics, which can provide health benefits when consumed. They may help support gut health, enhance the immune system, and even assist in the digestion of certain nutrients.

In summary, "Saccharomyces cerevisiae" is a species of yeast with various industrial and potential medical applications.

Immunoblotting, also known as western blotting, is a laboratory technique used in molecular biology and immunogenetics to detect and quantify specific proteins in a complex mixture. This technique combines the electrophoretic separation of proteins by gel electrophoresis with their detection using antibodies that recognize specific epitopes (protein fragments) on the target protein.

The process involves several steps: first, the protein sample is separated based on size through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Next, the separated proteins are transferred onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric field. The membrane is then blocked with a blocking agent to prevent non-specific binding of antibodies.

After blocking, the membrane is incubated with a primary antibody that specifically recognizes the target protein. Following this, the membrane is washed to remove unbound primary antibodies and then incubated with a secondary antibody conjugated to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). The enzyme catalyzes a colorimetric or chemiluminescent reaction that allows for the detection of the target protein.

Immunoblotting is widely used in research and clinical settings to study protein expression, post-translational modifications, protein-protein interactions, and disease biomarkers. It provides high specificity and sensitivity, making it a valuable tool for identifying and quantifying proteins in various biological samples.

Small interfering RNA (siRNA) is a type of short, double-stranded RNA molecule that plays a role in the RNA interference (RNAi) pathway. The RNAi pathway is a natural cellular process that regulates gene expression by targeting and destroying specific messenger RNA (mRNA) molecules, thereby preventing the translation of those mRNAs into proteins.

SiRNAs are typically 20-25 base pairs in length and are generated from longer double-stranded RNA precursors called hairpin RNAs or dsRNAs by an enzyme called Dicer. Once generated, siRNAs associate with a protein complex called the RNA-induced silencing complex (RISC), which uses one strand of the siRNA (the guide strand) to recognize and bind to complementary sequences in the target mRNA. The RISC then cleaves the target mRNA, leading to its degradation and the inhibition of protein synthesis.

SiRNAs have emerged as a powerful tool for studying gene function and have shown promise as therapeutic agents for a variety of diseases, including viral infections, cancer, and genetic disorders. However, their use as therapeutics is still in the early stages of development, and there are challenges associated with delivering siRNAs to specific cells and tissues in the body.

Repressor proteins are a type of regulatory protein in molecular biology that suppress the transcription of specific genes into messenger RNA (mRNA) by binding to DNA. They function as part of gene regulation processes, often working in conjunction with an operator region and a promoter region within the DNA molecule. Repressor proteins can be activated or deactivated by various signals, allowing for precise control over gene expression in response to changing cellular conditions.

There are two main types of repressor proteins:

1. DNA-binding repressors: These directly bind to specific DNA sequences (operator regions) near the target gene and prevent RNA polymerase from transcribing the gene into mRNA.
2. Allosteric repressors: These bind to effector molecules, which then cause a conformational change in the repressor protein, enabling it to bind to DNA and inhibit transcription.

Repressor proteins play crucial roles in various biological processes, such as development, metabolism, and stress response, by controlling gene expression patterns in cells.

"Drosophila" is a genus of small flies, also known as fruit flies. The most common species used in scientific research is "Drosophila melanogaster," which has been a valuable model organism for many areas of biological and medical research, including genetics, developmental biology, neurobiology, and aging.

The use of Drosophila as a model organism has led to numerous important discoveries in genetics and molecular biology, such as the identification of genes that are associated with human diseases like cancer, Parkinson's disease, and obesity. The short reproductive cycle, large number of offspring, and ease of genetic manipulation make Drosophila a powerful tool for studying complex biological processes.

Protein isoforms are different forms or variants of a protein that are produced from a single gene through the process of alternative splicing, where different exons (or parts of exons) are included in the mature mRNA molecule. This results in the production of multiple, slightly different proteins that share a common core structure but have distinct sequences and functions. Protein isoforms can also arise from genetic variations such as single nucleotide polymorphisms or mutations that alter the protein-coding sequence of a gene. These differences in protein sequence can affect the stability, localization, activity, or interaction partners of the protein isoform, leading to functional diversity and specialization within cells and organisms.

HEK293 cells, also known as human embryonic kidney 293 cells, are a line of cells used in scientific research. They were originally derived from human embryonic kidney cells and have been adapted to grow in a lab setting. HEK293 cells are widely used in molecular biology and biochemistry because they can be easily transfected (a process by which DNA is introduced into cells) and highly express foreign genes. As a result, they are often used to produce proteins for structural and functional studies. It's important to note that while HEK293 cells are derived from human tissue, they have been grown in the lab for many generations and do not retain the characteristics of the original embryonic kidney cells.

A "reporter gene" is a type of gene that is linked to a gene of interest in order to make the expression or activity of that gene detectable. The reporter gene encodes for a protein that can be easily measured and serves as an indicator of the presence and activity of the gene of interest. Commonly used reporter genes include those that encode for fluorescent proteins, enzymes that catalyze colorimetric reactions, or proteins that bind to specific molecules.

In the context of genetics and genomics research, a reporter gene is often used in studies involving gene expression, regulation, and function. By introducing the reporter gene into an organism or cell, researchers can monitor the activity of the gene of interest in real-time or after various experimental treatments. The information obtained from these studies can help elucidate the role of specific genes in biological processes and diseases, providing valuable insights for basic research and therapeutic development.

Fluorescence microscopy is a type of microscopy that uses fluorescent dyes or proteins to highlight and visualize specific components within a sample. In this technique, the sample is illuminated with high-energy light, typically ultraviolet (UV) or blue light, which excites the fluorescent molecules causing them to emit lower-energy, longer-wavelength light, usually visible light in the form of various colors. This emitted light is then collected by the microscope and detected to produce an image.

Fluorescence microscopy has several advantages over traditional brightfield microscopy, including the ability to visualize specific structures or molecules within a complex sample, increased sensitivity, and the potential for quantitative analysis. It is widely used in various fields of biology and medicine, such as cell biology, neuroscience, and pathology, to study the structure, function, and interactions of cells and proteins.

There are several types of fluorescence microscopy techniques, including widefield fluorescence microscopy, confocal microscopy, two-photon microscopy, and total internal reflection fluorescence (TIRF) microscopy, each with its own strengths and limitations. These techniques can provide valuable insights into the behavior of cells and proteins in health and disease.

A dose-response relationship in the context of drugs refers to the changes in the effects or symptoms that occur as the dose of a drug is increased or decreased. Generally, as the dose of a drug is increased, the severity or intensity of its effects also increases. Conversely, as the dose is decreased, the effects of the drug become less severe or may disappear altogether.

The dose-response relationship is an important concept in pharmacology and toxicology because it helps to establish the safe and effective dosage range for a drug. By understanding how changes in the dose of a drug affect its therapeutic and adverse effects, healthcare providers can optimize treatment plans for their patients while minimizing the risk of harm.

The dose-response relationship is typically depicted as a curve that shows the relationship between the dose of a drug and its effect. The shape of the curve may vary depending on the drug and the specific effect being measured. Some drugs may have a steep dose-response curve, meaning that small changes in the dose can result in large differences in the effect. Other drugs may have a more gradual dose-response curve, where larger changes in the dose are needed to produce significant effects.

In addition to helping establish safe and effective dosages, the dose-response relationship is also used to evaluate the potential therapeutic benefits and risks of new drugs during clinical trials. By systematically testing different doses of a drug in controlled studies, researchers can identify the optimal dosage range for the drug and assess its safety and efficacy.

A phenotype is the physical or biochemical expression of an organism's genes, or the observable traits and characteristics resulting from the interaction of its genetic constitution (genotype) with environmental factors. These characteristics can include appearance, development, behavior, and resistance to disease, among others. Phenotypes can vary widely, even among individuals with identical genotypes, due to differences in environmental influences, gene expression, and genetic interactions.

Proto-oncogene proteins are normal cellular proteins that play crucial roles in various cellular processes, such as signal transduction, cell cycle regulation, and apoptosis (programmed cell death). They are involved in the regulation of cell growth, differentiation, and survival under physiological conditions.

When proto-oncogene proteins undergo mutations or aberrations in their expression levels, they can transform into oncogenic forms, leading to uncontrolled cell growth and division. These altered proteins are then referred to as oncogene products or oncoproteins. Oncogenic mutations can occur due to various factors, including genetic predisposition, environmental exposures, and aging.

Examples of proto-oncogene proteins include:

1. Ras proteins: Involved in signal transduction pathways that regulate cell growth and differentiation. Activating mutations in Ras genes are found in various human cancers.
2. Myc proteins: Regulate gene expression related to cell cycle progression, apoptosis, and metabolism. Overexpression of Myc proteins is associated with several types of cancer.
3. EGFR (Epidermal Growth Factor Receptor): A transmembrane receptor tyrosine kinase that regulates cell proliferation, survival, and differentiation. Mutations or overexpression of EGFR are linked to various malignancies, such as lung cancer and glioblastoma.
4. Src family kinases: Intracellular tyrosine kinases that regulate signal transduction pathways involved in cell proliferation, survival, and migration. Dysregulation of Src family kinases is implicated in several types of cancer.
5. Abl kinases: Cytoplasmic tyrosine kinases that regulate various cellular processes, including cell growth, differentiation, and stress responses. Aberrant activation of Abl kinases, as seen in chronic myelogenous leukemia (CML), leads to uncontrolled cell proliferation.

Understanding the roles of proto-oncogene proteins and their dysregulation in cancer development is essential for developing targeted cancer therapies that aim to inhibit or modulate these aberrant signaling pathways.

Transgenic mice are genetically modified rodents that have incorporated foreign DNA (exogenous DNA) into their own genome. This is typically done through the use of recombinant DNA technology, where a specific gene or genetic sequence of interest is isolated and then introduced into the mouse embryo. The resulting transgenic mice can then express the protein encoded by the foreign gene, allowing researchers to study its function in a living organism.

The process of creating transgenic mice usually involves microinjecting the exogenous DNA into the pronucleus of a fertilized egg, which is then implanted into a surrogate mother. The offspring that result from this procedure are screened for the presence of the foreign DNA, and those that carry the desired genetic modification are used to establish a transgenic mouse line.

Transgenic mice have been widely used in biomedical research to model human diseases, study gene function, and test new therapies. They provide a valuable tool for understanding complex biological processes and developing new treatments for a variety of medical conditions.

BH3 Interacting Domain Death Agonist Protein, also known as BAD protein, is a member of the Bcl-2 family of proteins. This protein is involved in the regulation of programmed cell death, or apoptosis. The BH3 domain of BAD protein allows it to interact with other members of the Bcl-2 family and modulate their function. When activated, BAD protein can promote cell death by binding to and inhibiting anti-apoptotic proteins such as Bcl-2 and Bcl-xL. This helps to release pro-apoptotic proteins such as Bax and Bak, which can then trigger the intrinsic pathway of apoptosis. The activation of BAD protein is tightly regulated by post-translational modifications, including phosphorylation and dephosphorylation, which can be influenced by various signals within the cell.

Bacterial proteins are a type of protein that are produced by bacteria as part of their structural or functional components. These proteins can be involved in various cellular processes, such as metabolism, DNA replication, transcription, and translation. They can also play a role in bacterial pathogenesis, helping the bacteria to evade the host's immune system, acquire nutrients, and multiply within the host.

Bacterial proteins can be classified into different categories based on their function, such as:

1. Enzymes: Proteins that catalyze chemical reactions in the bacterial cell.
2. Structural proteins: Proteins that provide structural support and maintain the shape of the bacterial cell.
3. Signaling proteins: Proteins that help bacteria to communicate with each other and coordinate their behavior.
4. Transport proteins: Proteins that facilitate the movement of molecules across the bacterial cell membrane.
5. Toxins: Proteins that are produced by pathogenic bacteria to damage host cells and promote infection.
6. Surface proteins: Proteins that are located on the surface of the bacterial cell and interact with the environment or host cells.

Understanding the structure and function of bacterial proteins is important for developing new antibiotics, vaccines, and other therapeutic strategies to combat bacterial infections.

Interleukin-1 (IL-1) is a type of cytokine, which are proteins that play a crucial role in cell signaling. Specifically, IL-1 is a pro-inflammatory cytokine that is involved in the regulation of immune and inflammatory responses in the body. It is produced by various cells, including monocytes, macrophages, and dendritic cells, in response to infection or injury.

IL-1 exists in two forms, IL-1α and IL-1β, which have similar biological activities but are encoded by different genes. Both forms of IL-1 bind to the same receptor, IL-1R, and activate intracellular signaling pathways that lead to the production of other cytokines, chemokines, and inflammatory mediators.

IL-1 has a wide range of biological effects, including fever induction, activation of immune cells, regulation of hematopoiesis (the formation of blood cells), and modulation of bone metabolism. Dysregulation of IL-1 production or activity has been implicated in various inflammatory diseases, such as rheumatoid arthritis, gout, and inflammatory bowel disease. Therefore, IL-1 is an important target for the development of therapies aimed at modulating the immune response and reducing inflammation.

Promoter regions in genetics refer to specific DNA sequences located near the transcription start site of a gene. They serve as binding sites for RNA polymerase and various transcription factors that regulate the initiation of gene transcription. These regulatory elements help control the rate of transcription and, therefore, the level of gene expression. Promoter regions can be composed of different types of sequences, such as the TATA box and CAAT box, and their organization and composition can vary between different genes and species.

Mutagenesis is the process by which the genetic material (DNA or RNA) of an organism is changed in a way that can alter its phenotype, or observable traits. These changes, known as mutations, can be caused by various factors such as chemicals, radiation, or viruses. Some mutations may have no effect on the organism, while others can cause harm, including diseases and cancer. Mutagenesis is a crucial area of study in genetics and molecular biology, with implications for understanding evolution, genetic disorders, and the development of new medical treatments.

Nerve tissue proteins are specialized proteins found in the nervous system that provide structural and functional support to nerve cells, also known as neurons. These proteins include:

1. Neurofilaments: These are type IV intermediate filaments that provide structural support to neurons and help maintain their shape and size. They are composed of three subunits - NFL (light), NFM (medium), and NFH (heavy).

2. Neuronal Cytoskeletal Proteins: These include tubulins, actins, and spectrins that provide structural support to the neuronal cytoskeleton and help maintain its integrity.

3. Neurotransmitter Receptors: These are specialized proteins located on the postsynaptic membrane of neurons that bind neurotransmitters released by presynaptic neurons, triggering a response in the target cell.

4. Ion Channels: These are transmembrane proteins that regulate the flow of ions across the neuronal membrane and play a crucial role in generating and transmitting electrical signals in neurons.

5. Signaling Proteins: These include enzymes, receptors, and adaptor proteins that mediate intracellular signaling pathways involved in neuronal development, differentiation, survival, and death.

6. Adhesion Proteins: These are cell surface proteins that mediate cell-cell and cell-matrix interactions, playing a crucial role in the formation and maintenance of neural circuits.

7. Extracellular Matrix Proteins: These include proteoglycans, laminins, and collagens that provide structural support to nerve tissue and regulate neuronal migration, differentiation, and survival.

An amino acid substitution is a type of mutation in which one amino acid in a protein is replaced by another. This occurs when there is a change in the DNA sequence that codes for a particular amino acid in a protein. The genetic code is redundant, meaning that most amino acids are encoded by more than one codon (a sequence of three nucleotides). As a result, a single base pair change in the DNA sequence may not necessarily lead to an amino acid substitution. However, if a change does occur, it can have a variety of effects on the protein's structure and function, depending on the nature of the substituted amino acids. Some substitutions may be harmless, while others may alter the protein's activity or stability, leading to disease.

Receptor aggregation, also known as receptor clustering or patching, is a process that occurs when multiple receptor proteins, which are typically found dispersed on the cell membrane, come together and form a cluster or aggregate in response to a stimulus. This can occur through various mechanisms such as ligand-induced dimerization, conformational changes, or interactions with intracellular signaling molecules.

Receptor aggregation can lead to changes in receptor function, including increased sensitivity, altered signaling properties, and internalization of the receptors. This process plays an important role in various physiological processes such as cell signaling, immune response, and neuronal communication. However, abnormal receptor aggregation has also been implicated in several diseases, including cancer and neurological disorders.

A point mutation is a type of genetic mutation where a single nucleotide base (A, T, C, or G) in DNA is altered, deleted, or substituted with another nucleotide. Point mutations can have various effects on the organism, depending on the location of the mutation and whether it affects the function of any genes. Some point mutations may not have any noticeable effect, while others might lead to changes in the amino acids that make up proteins, potentially causing diseases or altering traits. Point mutations can occur spontaneously due to errors during DNA replication or be inherited from parents.

Cytokines are a broad and diverse category of small signaling proteins that are secreted by various cells, including immune cells, in response to different stimuli. They play crucial roles in regulating the immune response, inflammation, hematopoiesis, and cellular communication.

Cytokines mediate their effects by binding to specific receptors on the surface of target cells, which triggers intracellular signaling pathways that ultimately result in changes in gene expression, cell behavior, and function. Some key functions of cytokines include:

1. Regulating the activation, differentiation, and proliferation of immune cells such as T cells, B cells, natural killer (NK) cells, and macrophages.
2. Coordinating the inflammatory response by recruiting immune cells to sites of infection or tissue damage and modulating their effector functions.
3. Regulating hematopoiesis, the process of blood cell formation in the bone marrow, by controlling the proliferation, differentiation, and survival of hematopoietic stem and progenitor cells.
4. Modulating the development and function of the nervous system, including neuroinflammation, neuroprotection, and neuroregeneration.

Cytokines can be classified into several categories based on their structure, function, or cellular origin. Some common types of cytokines include interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, colony-stimulating factors (CSFs), and transforming growth factors (TGFs). Dysregulation of cytokine production and signaling has been implicated in various pathological conditions, such as autoimmune diseases, chronic inflammation, cancer, and neurodegenerative disorders.

Phosphoproteins are proteins that have been post-translationally modified by the addition of a phosphate group (-PO3H2) onto specific amino acid residues, most commonly serine, threonine, or tyrosine. This process is known as phosphorylation and is mediated by enzymes called kinases. Phosphoproteins play crucial roles in various cellular processes such as signal transduction, cell cycle regulation, metabolism, and gene expression. The addition or removal of a phosphate group can activate or inhibit the function of a protein, thereby serving as a switch to control its activity. Phosphoproteins can be detected and quantified using techniques such as Western blotting, mass spectrometry, and immunofluorescence.

Brain death is a legal and medical determination that an individual has died because their brain has irreversibly lost all functions necessary for life. It is characterized by the absence of brainstem reflexes, unresponsiveness to stimuli, and the inability to breathe without mechanical support. Brain death is different from a vegetative state or coma, where there may still be some brain activity.

The determination of brain death involves a series of tests and examinations to confirm the absence of brain function. These tests are typically performed by trained medical professionals and may include clinical assessments, imaging studies, and electroencephalograms (EEGs) to confirm the absence of electrical activity in the brain.

Brain death is an important concept in medicine because it allows for the organ donation process to proceed, potentially saving the lives of others. In many jurisdictions, brain death is legally equivalent to cardiopulmonary death, which means that once a person has been declared brain dead, they are considered deceased and their organs can be removed for transplantation.

Calcium-calmodulin-dependent protein kinases (CAMKs) are a family of enzymes that play a crucial role in intracellular signaling pathways. They are activated by the binding of calcium ions and calmodulin, a ubiquitous calcium-binding protein, to their regulatory domain.

Once activated, CAMKs phosphorylate specific serine or threonine residues on target proteins, thereby modulating their activity, localization, or stability. This post-translational modification is essential for various cellular processes, including synaptic plasticity, gene expression, metabolism, and cell cycle regulation.

There are several subfamilies of CAMKs, including CaMKI, CaMKII, CaMKIII (also known as CaMKIV), and CaMK kinase (CaMKK). Each subfamily has distinct structural features, substrate specificity, and regulatory mechanisms. Dysregulation of CAMK signaling has been implicated in various pathological conditions, such as neurodegenerative diseases, cancer, and cardiovascular disorders.

Quaternary protein structure refers to the arrangement and interaction of multiple folded protein molecules in a multi-subunit complex. These subunits can be identical or different forms of the same protein or distinctly different proteins that associate to form a functional complex. The quaternary structure is held together by non-covalent interactions, such as hydrogen bonds, ionic bonds, and van der Waals forces. Understanding quaternary structure is crucial for comprehending the function, regulation, and assembly of many protein complexes involved in various cellular processes.

3T3 cells are a type of cell line that is commonly used in scientific research. The name "3T3" is derived from the fact that these cells were developed by treating mouse embryo cells with a chemical called trypsin and then culturing them in a flask at a temperature of 37 degrees Celsius.

Specifically, 3T3 cells are a type of fibroblast, which is a type of cell that is responsible for producing connective tissue in the body. They are often used in studies involving cell growth and proliferation, as well as in toxicity tests and drug screening assays.

One particularly well-known use of 3T3 cells is in the 3T3-L1 cell line, which is a subtype of 3T3 cells that can be differentiated into adipocytes (fat cells) under certain conditions. These cells are often used in studies of adipose tissue biology and obesity.

It's important to note that because 3T3 cells are a type of immortalized cell line, they do not always behave exactly the same way as primary cells (cells that are taken directly from a living organism). As such, researchers must be careful when interpreting results obtained using 3T3 cells and consider any potential limitations or artifacts that may arise due to their use.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

Green Fluorescent Protein (GFP) is not a medical term per se, but a scientific term used in the field of molecular biology. GFP is a protein that exhibits bright green fluorescence when exposed to light, particularly blue or ultraviolet light. It was originally discovered in the jellyfish Aequorea victoria.

In medical and biological research, scientists often use recombinant DNA technology to introduce the gene for GFP into other organisms, including bacteria, plants, and animals, including humans. This allows them to track the expression and localization of specific genes or proteins of interest in living cells, tissues, or even whole organisms.

The ability to visualize specific cellular structures or processes in real-time has proven invaluable for a wide range of research areas, from studying the development and function of organs and organ systems to understanding the mechanisms of diseases and the effects of therapeutic interventions.

POU domain factors are a family of transcription factors that play crucial roles in the development and function of various organisms, including humans. The name "POU" is an acronym derived from the names of three genes in which these domains were first identified: Pit-1, Oct-1, and Unc-86.

The POU domain is a conserved DNA-binding motif that consists of two subdomains: a POU-specific domain (POUs) and a POU homeodomain (POUh). The POUs domain recognizes and binds to specific DNA sequences, while the POUh domain enhances the binding affinity and specificity.

POU domain factors regulate gene expression by binding to regulatory elements in the promoter or enhancer regions of their target genes. They are involved in various biological processes, such as cell fate determination, development, differentiation, and metabolism. Some examples of POU domain factors include Oct-1, Oct-2, Oct-3/4, Sox2, and Brn-2.

Mutations or dysregulation of POU domain factors have been implicated in several human diseases, such as cancer, diabetes, and neurological disorders. Therefore, understanding the function and regulation of these transcription factors is essential for developing new therapeutic strategies to treat these conditions.

Peptides are short chains of amino acid residues linked by covalent bonds, known as peptide bonds. They are formed when two or more amino acids are joined together through a condensation reaction, which results in the elimination of a water molecule and the formation of an amide bond between the carboxyl group of one amino acid and the amino group of another.

Peptides can vary in length from two to about fifty amino acids, and they are often classified based on their size. For example, dipeptides contain two amino acids, tripeptides contain three, and so on. Oligopeptides typically contain up to ten amino acids, while polypeptides can contain dozens or even hundreds of amino acids.

Peptides play many important roles in the body, including serving as hormones, neurotransmitters, enzymes, and antibiotics. They are also used in medical research and therapeutic applications, such as drug delivery and tissue engineering.

Transcriptional activation is the process by which a cell increases the rate of transcription of specific genes from DNA to RNA. This process is tightly regulated and plays a crucial role in various biological processes, including development, differentiation, and response to environmental stimuli.

Transcriptional activation occurs when transcription factors (proteins that bind to specific DNA sequences) interact with the promoter region of a gene and recruit co-activator proteins. These co-activators help to remodel the chromatin structure around the gene, making it more accessible for the transcription machinery to bind and initiate transcription.

Transcriptional activation can be regulated at multiple levels, including the availability and activity of transcription factors, the modification of histone proteins, and the recruitment of co-activators or co-repressors. Dysregulation of transcriptional activation has been implicated in various diseases, including cancer and genetic disorders.

Mitochondria are specialized structures located inside cells that convert the energy from food into ATP (adenosine triphosphate), which is the primary form of energy used by cells. They are often referred to as the "powerhouses" of the cell because they generate most of the cell's supply of chemical energy. Mitochondria are also involved in various other cellular processes, such as signaling, differentiation, and apoptosis (programmed cell death).

Mitochondria have their own DNA, known as mitochondrial DNA (mtDNA), which is inherited maternally. This means that mtDNA is passed down from the mother to her offspring through the egg cells. Mitochondrial dysfunction has been linked to a variety of diseases and conditions, including neurodegenerative disorders, diabetes, and aging.

Amino acid chloromethyl ketones (AACMKs) are a class of chemical compounds that are widely used in research and industry. They are derivatives of amino acids, which are the building blocks of proteins, with a chloromethyl ketone group (-CO-CH2Cl) attached to the side chain of the amino acid.

In the context of medical research, AACMKs are often used as irreversible inhibitors of enzymes, particularly those that contain active site serine or cysteine residues. The chloromethyl ketone group reacts with these residues to form a covalent bond, which permanently inactivates the enzyme. This makes AACMKs useful tools for studying the mechanisms of enzymes and for developing drugs that target specific enzymes.

However, it is important to note that AACMKs can also be highly reactive and toxic, and they must be handled with care in the laboratory. They have been shown to inhibit a wide range of enzymes, including some that are essential for normal cellular function, and prolonged exposure can lead to cell damage or death. Therefore, their use is typically restricted to controlled experimental settings.

TNF Receptor-Associated Factor 6 (TRAF6) is a protein that plays a crucial role in the signaling pathways of various cytokine receptors and pattern recognition receptors, including TNF receptors, IL-1 receptors, and TLRs. It functions as an E3 ubiquitin ligase, which adds ubiquitin molecules to other proteins, thereby modulating their activity, stability, or localization.

TRAF6 is involved in the activation of several downstream signaling pathways, such as NF-κB and MAPK pathways, leading to the induction of immune responses, inflammation, cell survival, differentiation, and proliferation. Mutations or dysregulation of TRAF6 have been implicated in various diseases, including immunodeficiencies, autoimmune disorders, and cancers.

Alternative splicing is a process in molecular biology that occurs during the post-transcriptional modification of pre-messenger RNA (pre-mRNA) molecules. It involves the removal of non-coding sequences, known as introns, and the joining together of coding sequences, or exons, to form a mature messenger RNA (mRNA) molecule that can be translated into a protein.

In alternative splicing, different combinations of exons are selected and joined together to create multiple distinct mRNA transcripts from a single pre-mRNA template. This process increases the diversity of proteins that can be produced from a limited number of genes, allowing for greater functional complexity in organisms.

Alternative splicing is regulated by various cis-acting elements and trans-acting factors that bind to specific sequences in the pre-mRNA molecule and influence which exons are included or excluded during splicing. Abnormal alternative splicing has been implicated in several human diseases, including cancer, neurological disorders, and cardiovascular disease.

Developmental gene expression regulation refers to the processes that control the activation or repression of specific genes during embryonic and fetal development. These regulatory mechanisms ensure that genes are expressed at the right time, in the right cells, and at appropriate levels to guide proper growth, differentiation, and morphogenesis of an organism.

Developmental gene expression regulation is a complex and dynamic process involving various molecular players, such as transcription factors, chromatin modifiers, non-coding RNAs, and signaling molecules. These regulators can interact with cis-regulatory elements, like enhancers and promoters, to fine-tune the spatiotemporal patterns of gene expression during development.

Dysregulation of developmental gene expression can lead to various congenital disorders and developmental abnormalities. Therefore, understanding the principles and mechanisms governing developmental gene expression regulation is crucial for uncovering the etiology of developmental diseases and devising potential therapeutic strategies.

Cell proliferation is the process by which cells increase in number, typically through the process of cell division. In the context of biology and medicine, it refers to the reproduction of cells that makes up living tissue, allowing growth, maintenance, and repair. It involves several stages including the transition from a phase of quiescence (G0 phase) to an active phase (G1 phase), DNA replication in the S phase, and mitosis or M phase, where the cell divides into two daughter cells.

Abnormal or uncontrolled cell proliferation is a characteristic feature of many diseases, including cancer, where deregulated cell cycle control leads to excessive and unregulated growth of cells, forming tumors that can invade surrounding tissues and metastasize to distant sites in the body.

Lipopolysaccharides (LPS) are large molecules found in the outer membrane of Gram-negative bacteria. They consist of a hydrophilic polysaccharide called the O-antigen, a core oligosaccharide, and a lipid portion known as Lipid A. The Lipid A component is responsible for the endotoxic activity of LPS, which can trigger a powerful immune response in animals, including humans. This response can lead to symptoms such as fever, inflammation, and septic shock, especially when large amounts of LPS are introduced into the bloodstream.

'Arabidopsis' is a genus of small flowering plants that are part of the mustard family (Brassicaceae). The most commonly studied species within this genus is 'Arabidopsis thaliana', which is often used as a model organism in plant biology and genetics research. This plant is native to Eurasia and Africa, and it has a small genome that has been fully sequenced. It is known for its short life cycle, self-fertilization, and ease of growth, making it an ideal subject for studying various aspects of plant biology, including development, metabolism, and response to environmental stresses.

A "cell line, transformed" is a type of cell culture that has undergone a stable genetic alteration, which confers the ability to grow indefinitely in vitro, outside of the organism from which it was derived. These cells have typically been immortalized through exposure to chemical or viral carcinogens, or by introducing specific oncogenes that disrupt normal cell growth regulation pathways.

Transformed cell lines are widely used in scientific research because they offer a consistent and renewable source of biological material for experimentation. They can be used to study various aspects of cell biology, including signal transduction, gene expression, drug discovery, and toxicity testing. However, it is important to note that transformed cells may not always behave identically to their normal counterparts, and results obtained using these cells should be validated in more physiologically relevant systems when possible.

DNA fragmentation is the breaking of DNA strands into smaller pieces. This process can occur naturally during apoptosis, or programmed cell death, where the DNA is broken down and packaged into apoptotic bodies to be safely eliminated from the body. However, excessive or abnormal DNA fragmentation can also occur due to various factors such as oxidative stress, exposure to genotoxic agents, or certain medical conditions. This can lead to genetic instability, cellular dysfunction, and increased risk of diseases such as cancer. In the context of reproductive medicine, high levels of DNA fragmentation in sperm cells have been linked to male infertility and poor assisted reproductive technology outcomes.

Trans-activators are proteins that increase the transcriptional activity of a gene or a set of genes. They do this by binding to specific DNA sequences and interacting with the transcription machinery, thereby enhancing the recruitment and assembly of the complexes needed for transcription. In some cases, trans-activators can also modulate the chromatin structure to make the template more accessible to the transcription machinery.

In the context of HIV (Human Immunodeficiency Virus) infection, the term "trans-activator" is often used specifically to refer to the Tat protein. The Tat protein is a viral regulatory protein that plays a critical role in the replication of HIV by activating the transcription of the viral genome. It does this by binding to a specific RNA structure called the Trans-Activation Response Element (TAR) located at the 5' end of all nascent HIV transcripts, and recruiting cellular cofactors that enhance the processivity and efficiency of RNA polymerase II, leading to increased viral gene expression.

Protein folding is the process by which a protein molecule naturally folds into its three-dimensional structure, following the synthesis of its amino acid chain. This complex process is determined by the sequence and properties of the amino acids, as well as various environmental factors such as temperature, pH, and the presence of molecular chaperones. The final folded conformation of a protein is crucial for its proper function, as it enables the formation of specific interactions between different parts of the molecule, which in turn define its biological activity. Protein misfolding can lead to various diseases, including neurodegenerative disorders such as Alzheimer's and Parkinson's disease.

Phylogeny is the evolutionary history and relationship among biological entities, such as species or genes, based on their shared characteristics. In other words, it refers to the branching pattern of evolution that shows how various organisms have descended from a common ancestor over time. Phylogenetic analysis involves constructing a tree-like diagram called a phylogenetic tree, which depicts the inferred evolutionary relationships among organisms or genes based on molecular sequence data or other types of characters. This information is crucial for understanding the diversity and distribution of life on Earth, as well as for studying the emergence and spread of diseases.

RNA-binding proteins (RBPs) are a class of proteins that selectively interact with RNA molecules to form ribonucleoprotein complexes. These proteins play crucial roles in the post-transcriptional regulation of gene expression, including pre-mRNA processing, mRNA stability, transport, localization, and translation. RBPs recognize specific RNA sequences or structures through their modular RNA-binding domains, which can be highly degenerate and allow for the recognition of a wide range of RNA targets. The interaction between RBPs and RNA is often dynamic and can be regulated by various post-translational modifications of the proteins or by environmental stimuli, allowing for fine-tuning of gene expression in response to changing cellular needs. Dysregulation of RBP function has been implicated in various human diseases, including neurological disorders and cancer.

Fibroblasts are specialized cells that play a critical role in the body's immune response and wound healing process. They are responsible for producing and maintaining the extracellular matrix (ECM), which is the non-cellular component present within all tissues and organs, providing structural support and biochemical signals for surrounding cells.

Fibroblasts produce various ECM proteins such as collagens, elastin, fibronectin, and laminins, forming a complex network of fibers that give tissues their strength and flexibility. They also help in the regulation of tissue homeostasis by controlling the turnover of ECM components through the process of remodeling.

In response to injury or infection, fibroblasts become activated and start to proliferate rapidly, migrating towards the site of damage. Here, they participate in the inflammatory response, releasing cytokines and chemokines that attract immune cells to the area. Additionally, they deposit new ECM components to help repair the damaged tissue and restore its functionality.

Dysregulation of fibroblast activity has been implicated in several pathological conditions, including fibrosis (excessive scarring), cancer (where they can contribute to tumor growth and progression), and autoimmune diseases (such as rheumatoid arthritis).

Cell division is the process by which a single eukaryotic cell (a cell with a true nucleus) divides into two identical daughter cells. This complex process involves several stages, including replication of DNA, separation of chromosomes, and division of the cytoplasm. There are two main types of cell division: mitosis and meiosis.

Mitosis is the type of cell division that results in two genetically identical daughter cells. It is a fundamental process for growth, development, and tissue repair in multicellular organisms. The stages of mitosis include prophase, prometaphase, metaphase, anaphase, and telophase, followed by cytokinesis, which divides the cytoplasm.

Meiosis, on the other hand, is a type of cell division that occurs in the gonads (ovaries and testes) during the production of gametes (sex cells). Meiosis results in four genetically unique daughter cells, each with half the number of chromosomes as the parent cell. This process is essential for sexual reproduction and genetic diversity. The stages of meiosis include meiosis I and meiosis II, which are further divided into prophase, prometaphase, metaphase, anaphase, and telophase.

In summary, cell division is the process by which a single cell divides into two daughter cells, either through mitosis or meiosis. This process is critical for growth, development, tissue repair, and sexual reproduction in multicellular organisms.

'Caenorhabditis elegans' (C. elegans) is a type of free-living, transparent nematode (roundworm) that is often used as a model organism in scientific research. C. elegans proteins refer to the various types of protein molecules that are produced by the organism's genes and play crucial roles in maintaining its biological functions.

Proteins are complex molecules made up of long chains of amino acids, and they are involved in virtually every cellular process, including metabolism, DNA replication, signal transduction, and transportation of molecules within the cell. In C. elegans, proteins are encoded by genes, which are transcribed into messenger RNA (mRNA) molecules that are then translated into protein sequences by ribosomes.

Studying C. elegans proteins is important for understanding the basic biology of this organism and can provide insights into more complex biological systems, including humans. Because C. elegans has a relatively simple nervous system and a short lifespan, it is often used to study neurobiology, aging, and development. Additionally, because many of the genes and proteins in C. elegans have counterparts in other organisms, including humans, studying them can provide insights into human disease processes and potential therapeutic targets.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Zinc fingers are a type of protein structural motif involved in specific DNA binding and, by extension, in the regulation of gene expression. They are so named because of their characteristic "finger-like" shape that is formed when a zinc ion binds to the amino acids within the protein. This structure allows the protein to interact with and recognize specific DNA sequences, thereby playing a crucial role in various biological processes such as transcription, repair, and recombination of genetic material.

Cell differentiation is the process by which a less specialized cell, or stem cell, becomes a more specialized cell type with specific functions and structures. This process involves changes in gene expression, which are regulated by various intracellular signaling pathways and transcription factors. Differentiation results in the development of distinct cell types that make up tissues and organs in multicellular organisms. It is a crucial aspect of embryonic development, tissue repair, and maintenance of homeostasis in the body.

Interleukin-1 Receptor-Associated Kinases (IRAKs) are a group of serine/threonine protein kinases that play a crucial role in the signaling pathways of Toll-like receptors (TLRs) and Interleukin-1 receptors (IL-1Rs). These receptors are involved in the recognition and response to various pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which are essential for the activation of innate immune responses.

There are four known members of the IRAK family, namely IRAK1, IRAK2, IRAK3 (also known as IRAK-M), and IRAK4. Among these, IRAK4 is an upstream kinase that gets recruited to the receptor complex upon IL-1R or TLR activation. Once recruited, IRAK4 phosphorylates and activates IRAK1 and IRAK2, which in turn recruit additional signaling proteins leading to the activation of various transcription factors such as NF-κB and AP-1. These transcription factors regulate the expression of genes involved in inflammation, immune response, and cell survival.

IRAK3, on the other hand, is a negative regulator of TLR and IL-1R signaling. It lacks kinase activity and inhibits IRAK1 and IRAK4 activation, thereby dampening the immune response and preventing excessive inflammation. Dysregulation of IRAKs has been implicated in various inflammatory diseases, making them attractive targets for drug development.

Fungal proteins are a type of protein that is specifically produced and present in fungi, which are a group of eukaryotic organisms that include microorganisms such as yeasts and molds. These proteins play various roles in the growth, development, and survival of fungi. They can be involved in the structure and function of fungal cells, metabolism, pathogenesis, and other cellular processes. Some fungal proteins can also have important implications for human health, both in terms of their potential use as therapeutic targets and as allergens or toxins that can cause disease.

Fungal proteins can be classified into different categories based on their functions, such as enzymes, structural proteins, signaling proteins, and toxins. Enzymes are proteins that catalyze chemical reactions in fungal cells, while structural proteins provide support and protection for the cell. Signaling proteins are involved in communication between cells and regulation of various cellular processes, and toxins are proteins that can cause harm to other organisms, including humans.

Understanding the structure and function of fungal proteins is important for developing new treatments for fungal infections, as well as for understanding the basic biology of fungi. Research on fungal proteins has led to the development of several antifungal drugs that target specific fungal enzymes or other proteins, providing effective treatment options for a range of fungal diseases. Additionally, further study of fungal proteins may reveal new targets for drug development and help improve our ability to diagnose and treat fungal infections.

"Plant proteins" refer to the proteins that are derived from plant sources. These can include proteins from legumes such as beans, lentils, and peas, as well as proteins from grains like wheat, rice, and corn. Other sources of plant proteins include nuts, seeds, and vegetables.

Plant proteins are made up of individual amino acids, which are the building blocks of protein. While animal-based proteins typically contain all of the essential amino acids that the body needs to function properly, many plant-based proteins may be lacking in one or more of these essential amino acids. However, by consuming a variety of plant-based foods throughout the day, it is possible to get all of the essential amino acids that the body needs from plant sources alone.

Plant proteins are often lower in calories and saturated fat than animal proteins, making them a popular choice for those following a vegetarian or vegan diet, as well as those looking to maintain a healthy weight or reduce their risk of chronic diseases such as heart disease and cancer. Additionally, plant proteins have been shown to have a number of health benefits, including improving gut health, reducing inflammation, and supporting muscle growth and repair.

Interleukin-6 (IL-6) is a cytokine, a type of protein that plays a crucial role in communication between cells, especially in the immune system. It is produced by various cells including T-cells, B-cells, fibroblasts, and endothelial cells in response to infection, injury, or inflammation.

IL-6 has diverse effects on different cell types. In the immune system, it stimulates the growth and differentiation of B-cells into plasma cells that produce antibodies. It also promotes the activation and survival of T-cells. Moreover, IL-6 plays a role in fever induction by acting on the hypothalamus to raise body temperature during an immune response.

In addition to its functions in the immune system, IL-6 has been implicated in various physiological processes such as hematopoiesis (the formation of blood cells), bone metabolism, and neural development. However, abnormal levels of IL-6 have also been associated with several diseases, including autoimmune disorders, chronic inflammation, and cancer.

'Caenorhabditis elegans' is a species of free-living, transparent nematode (roundworm) that is widely used as a model organism in scientific research, particularly in the fields of biology and genetics. It has a simple anatomy, short lifespan, and fully sequenced genome, making it an ideal subject for studying various biological processes and diseases.

Some notable features of C. elegans include:

* Small size: Adult hermaphrodites are about 1 mm in length.
* Short lifespan: The average lifespan of C. elegans is around 2-3 weeks, although some strains can live up to 4 weeks under laboratory conditions.
* Development: C. elegans has a well-characterized developmental process, with adults developing from eggs in just 3 days at 20°C.
* Transparency: The transparent body of C. elegans allows researchers to observe its internal structures and processes easily.
* Genetics: C. elegans has a fully sequenced genome, which contains approximately 20,000 genes. Many of these genes have human homologs, making it an excellent model for studying human diseases.
* Neurobiology: C. elegans has a simple nervous system, with only 302 neurons in the hermaphrodite and 383 in the male. This simplicity makes it an ideal organism for studying neural development, function, and behavior.

Research using C. elegans has contributed significantly to our understanding of various biological processes, including cell division, apoptosis, aging, learning, and memory. Additionally, studies on C. elegans have led to the discovery of many genes associated with human diseases such as cancer, neurodegenerative disorders, and metabolic conditions.

Enzyme inhibitors are substances that bind to an enzyme and decrease its activity, preventing it from catalyzing a chemical reaction in the body. They can work by several mechanisms, including blocking the active site where the substrate binds, or binding to another site on the enzyme to change its shape and prevent substrate binding. Enzyme inhibitors are often used as drugs to treat various medical conditions, such as high blood pressure, abnormal heart rhythms, and bacterial infections. They can also be found naturally in some foods and plants, and can be used in research to understand enzyme function and regulation.

The Lymphotoxin-beta receptor (LTβR) is a type III transmembrane protein and a member of the tumor necrosis factor receptor superfamily (TNFRSF). It is primarily expressed on the surface of various cell types, including immune cells such as lymphocytes, dendritic cells, and stromal cells in lymphoid organs.

LTβR binds to its ligands, Lymphotoxin-alpha (LTα) and Lymphotoxin-beta (LTβ), which are primarily produced by activated T-cells and B-cells. The binding of LTα/LTβ to LTβR triggers a signaling cascade that leads to the activation of various downstream signaling pathways, including NF-κB and MAPK pathways.

The activation of LTβR plays critical roles in the development and organization of lymphoid tissues, immune responses, and inflammation. Dysregulation of LTβR signaling has been implicated in various autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis.

Neurons, also known as nerve cells or neurocytes, are specialized cells that constitute the basic unit of the nervous system. They are responsible for receiving, processing, and transmitting information and signals within the body. Neurons have three main parts: the dendrites, the cell body (soma), and the axon. The dendrites receive signals from other neurons or sensory receptors, while the axon transmits these signals to other neurons, muscles, or glands. The junction between two neurons is called a synapse, where neurotransmitters are released to transmit the signal across the gap (synaptic cleft) to the next neuron. Neurons vary in size, shape, and structure depending on their function and location within the nervous system.

In situ nick-end labeling (ISEL, also known as TUNEL) is a technique used in pathology and molecular biology to detect DNA fragmentation, which is a characteristic of apoptotic cells (cells undergoing programmed cell death). The method involves labeling the 3'-hydroxyl termini of double or single stranded DNA breaks in situ (within tissue sections or individual cells) using modified nucleotides that are coupled to a detectable marker, such as a fluorophore or an enzyme. This technique allows for the direct visualization and quantification of apoptotic cells within complex tissues or cell populations.

Hypoxia-Inducible Factor (HIF) is a transcription factor that plays a crucial role in the body's response to low oxygen levels (hypoxia). HIF is composed of two subunits: an alpha subunit and a beta subunit. Under normal oxygen conditions, the alpha subunit is constantly being broken down by prolyl hydroxylase domain-containing proteins, which are a type of enzyme known as HIF-Proline Dioxygenases (HIF-PDOs).

HIF-PDOs post-translationally modify the HIF alpha subunit by adding a hydroxyl group to specific proline residues. This modification marks the HIF alpha subunit for degradation by the proteasome, a complex that breaks down unneeded or damaged proteins in the cell. However, under hypoxic conditions, the activity of HIF-PDOs is inhibited, leading to the stabilization and accumulation of HIF alpha subunits.

Once stabilized, HIF alpha subunits dimerize with HIF beta subunits and translocate to the nucleus where they bind to hypoxia response elements (HREs) in the DNA. This binding induces the expression of genes involved in various cellular responses to hypoxia, such as angiogenesis, metabolic reprogramming, and erythropoiesis. Therefore, HIF-PDOs play a critical role in regulating the body's response to low oxygen levels by controlling the stability and activity of HIF.

Cysteine endopeptidases are a type of enzymes that cleave peptide bonds within proteins. They are also known as cysteine proteases or cysteine proteinases. These enzymes contain a catalytic triad consisting of three amino acids: cysteine, histidine, and aspartate. The thiol group (-SH) of the cysteine residue acts as a nucleophile and attacks the carbonyl carbon of the peptide bond, leading to its cleavage.

Cysteine endopeptidases play important roles in various biological processes, including protein degradation, cell signaling, and inflammation. They are involved in many physiological and pathological conditions, such as apoptosis, immune response, and cancer. Some examples of cysteine endopeptidases include cathepsins, caspases, and calpains.

It is important to note that these enzymes require a reducing environment to maintain the reduced state of their active site cysteine residue. Therefore, they are sensitive to oxidizing agents and inhibitors that target the thiol group. Understanding the structure and function of cysteine endopeptidases is crucial for developing therapeutic strategies that target these enzymes in various diseases.

Protein multimerization refers to the process where multiple protein subunits assemble together to form a complex, repetitive structure called a multimer or oligomer. This can involve the association of identical or similar protein subunits through non-covalent interactions such as hydrogen bonding, ionic bonding, and van der Waals forces. The resulting multimeric structures can have various shapes, sizes, and functions, including enzymatic activity, transport, or structural support. Protein multimerization plays a crucial role in many biological processes and is often necessary for the proper functioning of proteins within cells.

Fetal death, also known as stillbirth or intrauterine fetal demise, is defined as the death of a fetus at 20 weeks of gestation or later. The criteria for defining fetal death may vary slightly by country and jurisdiction, but in general, it refers to the loss of a pregnancy after the point at which the fetus is considered viable outside the womb.

Fetal death can occur for a variety of reasons, including chromosomal abnormalities, placental problems, maternal health conditions, infections, and umbilical cord accidents. In some cases, the cause of fetal death may remain unknown.

The diagnosis of fetal death is typically made through ultrasound or other imaging tests, which can confirm the absence of a heartbeat or movement in the fetus. Once fetal death has been diagnosed, medical professionals will work with the parents to determine the best course of action for managing the pregnancy and delivering the fetus. This may involve waiting for labor to begin naturally, inducing labor, or performing a cesarean delivery.

Experiencing a fetal death can be a very difficult and emotional experience for parents, and it is important for them to receive supportive care from their healthcare providers, family members, and friends. Grief counseling and support groups may also be helpful in coping with the loss.

Polymerase Chain Reaction (PCR) is a laboratory technique used to amplify specific regions of DNA. It enables the production of thousands to millions of copies of a particular DNA sequence in a rapid and efficient manner, making it an essential tool in various fields such as molecular biology, medical diagnostics, forensic science, and research.

The PCR process involves repeated cycles of heating and cooling to separate the DNA strands, allow primers (short sequences of single-stranded DNA) to attach to the target regions, and extend these primers using an enzyme called Taq polymerase, resulting in the exponential amplification of the desired DNA segment.

In a medical context, PCR is often used for detecting and quantifying specific pathogens (viruses, bacteria, fungi, or parasites) in clinical samples, identifying genetic mutations or polymorphisms associated with diseases, monitoring disease progression, and evaluating treatment effectiveness.

Medical Definition of "Multiprotein Complexes" :

Multiprotein complexes are large molecular assemblies composed of two or more proteins that interact with each other to carry out specific cellular functions. These complexes can range from relatively simple dimers or trimers to massive structures containing hundreds of individual protein subunits. They are formed through a process known as protein-protein interaction, which is mediated by specialized regions on the protein surface called domains or motifs.

Multiprotein complexes play critical roles in many cellular processes, including signal transduction, gene regulation, DNA replication and repair, protein folding and degradation, and intracellular transport. The formation of these complexes is often dynamic and regulated in response to various stimuli, allowing for precise control of their function.

Disruption of multiprotein complexes can lead to a variety of diseases, including cancer, neurodegenerative disorders, and infectious diseases. Therefore, understanding the structure, composition, and regulation of these complexes is an important area of research in molecular biology and medicine.

Histone-Lysine N-Methyltransferase is a type of enzyme that transfers methyl groups to specific lysine residues on histone proteins. These histone proteins are the main protein components of chromatin, which is the complex of DNA and proteins that make up chromosomes.

Histone-Lysine N-Methyltransferases play a crucial role in the regulation of gene expression by modifying the structure of chromatin. The addition of methyl groups to histones can result in either the activation or repression of gene transcription, depending on the specific location and number of methyl groups added.

These enzymes are important targets for drug development, as their dysregulation has been implicated in various diseases, including cancer. Inhibitors of Histone-Lysine N-Methyltransferases have shown promise in preclinical studies for the treatment of certain types of cancer.

COS cells are a type of cell line that are commonly used in molecular biology and genetic research. The name "COS" is an acronym for "CV-1 in Origin," as these cells were originally derived from the African green monkey kidney cell line CV-1. COS cells have been modified through genetic engineering to express high levels of a protein called SV40 large T antigen, which allows them to efficiently take up and replicate exogenous DNA.

There are several different types of COS cells that are commonly used in research, including COS-1, COS-3, and COS-7 cells. These cells are widely used for the production of recombinant proteins, as well as for studies of gene expression, protein localization, and signal transduction.

It is important to note that while COS cells have been a valuable tool in scientific research, they are not without their limitations. For example, because they are derived from monkey kidney cells, there may be differences in the way that human genes are expressed or regulated in these cells compared to human cells. Additionally, because COS cells express SV40 large T antigen, they may have altered cell cycle regulation and other phenotypic changes that could affect experimental results. Therefore, it is important to carefully consider the choice of cell line when designing experiments and interpreting results.

Cricetinae is a subfamily of rodents that includes hamsters, gerbils, and relatives. These small mammals are characterized by having short limbs, compact bodies, and cheek pouches for storing food. They are native to various parts of the world, particularly in Europe, Asia, and Africa. Some species are popular pets due to their small size, easy care, and friendly nature. In a medical context, understanding the biology and behavior of Cricetinae species can be important for individuals who keep them as pets or for researchers studying their physiology.

Cysteine proteinase inhibitors are a type of molecule that bind to and inhibit the activity of cysteine proteases, which are enzymes that cleave proteins at specific sites containing the amino acid cysteine. These inhibitors play important roles in regulating various biological processes, including inflammation, immune response, and programmed cell death (apoptosis). They can also have potential therapeutic applications in diseases where excessive protease activity contributes to pathology, such as cancer, arthritis, and neurodegenerative disorders. Examples of cysteine proteinase inhibitors include cystatins, kininogens, and serpins.

Inhibitor of Apoptosis Proteins (IAPs) are a family of proteins that play a crucial role in regulating programmed cell death, also known as apoptosis. These proteins function by binding to and inhibiting the activity of caspases, which are enzymes that drive the execution phase of apoptosis.

There are eight known human IAPs, including X-linked IAP (XIAP), cellular IAP1 (cIAP1), cIAP2, survivin, melanoma IAP (ML-IAP), ILP-2, NAIP, and Bruce. Each IAP contains at least one baculoviral IAP repeat (BIR) domain, which is responsible for binding to caspases and other regulatory proteins.

In addition to inhibiting caspases, some IAPs have been shown to regulate other cellular processes, such as inflammation, innate immunity, and cell cycle progression. Dysregulation of IAP function has been implicated in various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, IAPs are considered important targets for the development of new therapeutic strategies aimed at modulating apoptosis and other cellular processes.

Octamer Transcription Factor-6 (OTF-6) is not a commonly used or widely accepted medical term. However, in the field of molecular biology, an octamer transcription factor refers to a protein that binds to specific octamer motifs in DNA and regulates gene transcription. The "6" likely refers to the specific isoform or variant of this transcription factor.

More specifically, OTF-6 may refer to the protein product of the SOX6 gene, which encodes a member of the SOX (SRY-related HMG box) family of transcription factors. These proteins contain a high mobility group (HMG) box DNA-binding domain and play critical roles in various developmental processes, including cell fate specification, organogenesis, and tumorigenesis.

The SOX6 protein can form homodimers or heterodimers with other SOX family members to bind to specific octamer motifs (consensus sequence: AACAAAG) in the regulatory regions of target genes. By modulating the expression of these target genes, OTF-6/SOX6 helps regulate various cellular processes, such as neurogenesis, chondrogenesis, and myogenesis.

It is essential to note that the term "Octamer Transcription Factor-6" may not be universally recognized or consistently used in scientific literature, so it is always best to refer to primary sources for precise definitions and contexts.

Cycloheximide is an antibiotic that is primarily used in laboratory settings to inhibit protein synthesis in eukaryotic cells. It is derived from the actinobacteria species Streptomyces griseus. In medical terms, it is not used as a therapeutic drug in humans due to its significant side effects, including liver toxicity and potential neurotoxicity. However, it remains a valuable tool in research for studying protein function and cellular processes.

The antibiotic works by binding to the 60S subunit of the ribosome, thereby preventing the transfer RNA (tRNA) from delivering amino acids to the growing polypeptide chain during translation. This inhibition of protein synthesis can be lethal to cells, making cycloheximide a useful tool in studying cellular responses to protein depletion or misregulation.

In summary, while cycloheximide has significant research applications due to its ability to inhibit protein synthesis in eukaryotic cells, it is not used as a therapeutic drug in humans because of its toxic side effects.

Homeodomain proteins are a group of transcription factors that play crucial roles in the development and differentiation of cells in animals and plants. They are characterized by the presence of a highly conserved DNA-binding domain called the homeodomain, which is typically about 60 amino acids long. The homeodomain consists of three helices, with the third helix responsible for recognizing and binding to specific DNA sequences.

Homeodomain proteins are involved in regulating gene expression during embryonic development, tissue maintenance, and organismal growth. They can act as activators or repressors of transcription, depending on the context and the presence of cofactors. Mutations in homeodomain proteins have been associated with various human diseases, including cancer, congenital abnormalities, and neurological disorders.

Some examples of homeodomain proteins include PAX6, which is essential for eye development, HOX genes, which are involved in body patterning, and NANOG, which plays a role in maintaining pluripotency in stem cells.

Cytoskeletal proteins are a type of structural proteins that form the cytoskeleton, which is the internal framework of cells. The cytoskeleton provides shape, support, and structure to the cell, and plays important roles in cell division, intracellular transport, and maintenance of cell shape and integrity.

There are three main types of cytoskeletal proteins: actin filaments, intermediate filaments, and microtubules. Actin filaments are thin, rod-like structures that are involved in muscle contraction, cell motility, and cell division. Intermediate filaments are thicker than actin filaments and provide structural support to the cell. Microtubules are hollow tubes that are involved in intracellular transport, cell division, and maintenance of cell shape.

Cytoskeletal proteins are composed of different subunits that polymerize to form filamentous structures. These proteins can be dynamically assembled and disassembled, allowing cells to change their shape and move. Mutations in cytoskeletal proteins have been linked to various human diseases, including cancer, neurological disorders, and muscular dystrophies.

Gene expression regulation in plants refers to the processes that control the production of proteins and RNA from the genes present in the plant's DNA. This regulation is crucial for normal growth, development, and response to environmental stimuli in plants. It can occur at various levels, including transcription (the first step in gene expression, where the DNA sequence is copied into RNA), RNA processing (such as alternative splicing, which generates different mRNA molecules from a single gene), translation (where the information in the mRNA is used to produce a protein), and post-translational modification (where proteins are chemically modified after they have been synthesized).

In plants, gene expression regulation can be influenced by various factors such as hormones, light, temperature, and stress. Plants use complex networks of transcription factors, chromatin remodeling complexes, and small RNAs to regulate gene expression in response to these signals. Understanding the mechanisms of gene expression regulation in plants is important for basic research, as well as for developing crops with improved traits such as increased yield, stress tolerance, and disease resistance.

'Cercopithecus aethiops' is the scientific name for the monkey species more commonly known as the green monkey. It belongs to the family Cercopithecidae and is native to western Africa. The green monkey is omnivorous, with a diet that includes fruits, nuts, seeds, insects, and small vertebrates. They are known for their distinctive greenish-brown fur and long tail. Green monkeys are also important animal models in biomedical research due to their susceptibility to certain diseases, such as SIV (simian immunodeficiency virus), which is closely related to HIV.

Guanine Nucleotide Exchange Factors (GEFs) are a group of regulatory proteins that play a crucial role in the activation of GTPases, which are enzymes that regulate various cellular processes such as signal transduction, cytoskeleton reorganization, and vesicle trafficking.

GEFs function by promoting the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on GTPases. GTP is the active form of the GTPase, and its binding to the GTPase leads to a conformational change that activates the enzyme's function.

In the absence of GEFs, GTPases remain in their inactive GDP-bound state, and cellular signaling pathways are not activated. Therefore, GEFs play a critical role in regulating the activity of GTPases and ensuring proper signal transduction in cells.

There are many different GEFs that are specific to various GTPase families, including Ras, Rho, and Arf families. Dysregulation of GEFs has been implicated in various diseases, including cancer and neurological disorders.

Bcl-x is a protein that belongs to the Bcl-2 family, which regulates programmed cell death (apoptosis). Specifically, Bcl-x has both pro-survival and pro-apoptotic functions, depending on its splice variants. The long form of Bcl-x (Bcl-xL) is a potent inhibitor of apoptosis, while the short form (Bcl-xS) promotes cell death. Bcl-x plays critical roles in various cellular processes, including development, homeostasis, and stress responses, by controlling the mitochondrial outer membrane permeabilization and the release of cytochrome c, which eventually leads to caspase activation and apoptosis. Dysregulation of Bcl-x has been implicated in several diseases, such as cancer and neurodegenerative disorders.

Glutathione transferases (GSTs) are a group of enzymes involved in the detoxification of xenobiotics and endogenous compounds. They facilitate the conjugation of these compounds with glutathione, a tripeptide consisting of cysteine, glutamic acid, and glycine, which results in more water-soluble products that can be easily excreted from the body.

GSTs play a crucial role in protecting cells against oxidative stress and chemical injury by neutralizing reactive electrophilic species and peroxides. They are found in various tissues, including the liver, kidneys, lungs, and intestines, and are classified into several families based on their structure and function.

Abnormalities in GST activity have been associated with increased susceptibility to certain diseases, such as cancer, neurological disorders, and respiratory diseases. Therefore, GSTs have become a subject of interest in toxicology, pharmacology, and clinical research.

A genetic vector is a vehicle, often a plasmid or a virus, that is used to introduce foreign DNA into a host cell as part of genetic engineering or gene therapy techniques. The vector contains the desired gene or genes, along with regulatory elements such as promoters and enhancers, which are needed for the expression of the gene in the target cells.

The choice of vector depends on several factors, including the size of the DNA to be inserted, the type of cell to be targeted, and the efficiency of uptake and expression required. Commonly used vectors include plasmids, adenoviruses, retroviruses, and lentiviruses.

Plasmids are small circular DNA molecules that can replicate independently in bacteria. They are often used as cloning vectors to amplify and manipulate DNA fragments. Adenoviruses are double-stranded DNA viruses that infect a wide range of host cells, including human cells. They are commonly used as gene therapy vectors because they can efficiently transfer genes into both dividing and non-dividing cells.

Retroviruses and lentiviruses are RNA viruses that integrate their genetic material into the host cell's genome. This allows for stable expression of the transgene over time. Lentiviruses, a subclass of retroviruses, have the advantage of being able to infect non-dividing cells, making them useful for gene therapy applications in post-mitotic tissues such as neurons and muscle cells.

Overall, genetic vectors play a crucial role in modern molecular biology and medicine, enabling researchers to study gene function, develop new therapies, and modify organisms for various purposes.

CD40 is a type of protein known as a tumor necrosis factor receptor that is found on the surface of various cells in the body, including B cells, dendritic cells, and activated T cells. It plays an important role in the immune system by interacting with another protein called CD154 (also known as CD40 ligand) to activate immune responses.

CD40 antigens are molecules that can stimulate an immune response when introduced into the body because they are recognized as foreign substances by the immune system. They may be used in vaccines or other immunotherapies to induce an immune response against specific targets, such as cancer cells or infectious agents.

CD40 antigens can also be found on some types of tumor cells, and activating CD40 with CD154 has been shown to enhance the anti-tumor immune response in preclinical models. Therefore, CD40 agonists are being investigated as potential cancer therapies.

In summary, CD40 antigens are proteins that can stimulate an immune response and are involved in activating immune cells. They have potential applications in vaccines, immunotherapies, and cancer treatments.

'Drosophila melanogaster' is the scientific name for a species of fruit fly that is commonly used as a model organism in various fields of biological research, including genetics, developmental biology, and evolutionary biology. Its small size, short generation time, large number of offspring, and ease of cultivation make it an ideal subject for laboratory studies. The fruit fly's genome has been fully sequenced, and many of its genes have counterparts in the human genome, which facilitates the understanding of genetic mechanisms and their role in human health and disease.

Here is a brief medical definition:

Drosophila melanogaster (droh-suh-fih-luh meh-lon-guh-ster): A species of fruit fly used extensively as a model organism in genetic, developmental, and evolutionary research. Its genome has been sequenced, revealing many genes with human counterparts, making it valuable for understanding genetic mechanisms and their role in human health and disease.

BALB/c is an inbred strain of laboratory mouse that is widely used in biomedical research. The strain was developed at the Institute of Cancer Research in London by Henry Baldwin and his colleagues in the 1920s, and it has since become one of the most commonly used inbred strains in the world.

BALB/c mice are characterized by their black coat color, which is determined by a recessive allele at the tyrosinase locus. They are also known for their docile and friendly temperament, making them easy to handle and work with in the laboratory.

One of the key features of BALB/c mice that makes them useful for research is their susceptibility to certain types of tumors and immune responses. For example, they are highly susceptible to developing mammary tumors, which can be induced by chemical carcinogens or viral infection. They also have a strong Th2-biased immune response, which makes them useful models for studying allergic diseases and asthma.

BALB/c mice are also commonly used in studies of genetics, neuroscience, behavior, and infectious diseases. Because they are an inbred strain, they have a uniform genetic background, which makes it easier to control for genetic factors in experiments. Additionally, because they have been bred in the laboratory for many generations, they are highly standardized and reproducible, making them ideal subjects for scientific research.

Macrophages are a type of white blood cell that are an essential part of the immune system. They are large, specialized cells that engulf and destroy foreign substances, such as bacteria, viruses, parasites, and fungi, as well as damaged or dead cells. Macrophages are found throughout the body, including in the bloodstream, lymph nodes, spleen, liver, lungs, and connective tissues. They play a critical role in inflammation, immune response, and tissue repair and remodeling.

Macrophages originate from monocytes, which are a type of white blood cell produced in the bone marrow. When monocytes enter the tissues, they differentiate into macrophages, which have a larger size and more specialized functions than monocytes. Macrophages can change their shape and move through tissues to reach sites of infection or injury. They also produce cytokines, chemokines, and other signaling molecules that help coordinate the immune response and recruit other immune cells to the site of infection or injury.

Macrophages have a variety of surface receptors that allow them to recognize and respond to different types of foreign substances and signals from other cells. They can engulf and digest foreign particles, bacteria, and viruses through a process called phagocytosis. Macrophages also play a role in presenting antigens to T cells, which are another type of immune cell that helps coordinate the immune response.

Overall, macrophages are crucial for maintaining tissue homeostasis, defending against infection, and promoting wound healing and tissue repair. Dysregulation of macrophage function has been implicated in a variety of diseases, including cancer, autoimmune disorders, and chronic inflammatory conditions.

Ubiquitin-protein ligases, also known as E3 ubiquitin ligases, are a group of enzymes that play a crucial role in the ubiquitination process. Ubiquitination is a post-translational modification where ubiquitin molecules are attached to specific target proteins, marking them for degradation by the proteasome or for other regulatory functions.

Ubiquitin-protein ligases catalyze the final step in this process by binding to both the ubiquitin protein and the target protein, facilitating the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to the target protein. There are several different types of ubiquitin-protein ligases, each with their own specificity for particular target proteins and regulatory functions.

Ubiquitin-protein ligases have been implicated in various cellular processes such as protein degradation, DNA repair, signal transduction, and regulation of the cell cycle. Dysregulation of ubiquitination has been associated with several diseases, including cancer, neurodegenerative disorders, and inflammatory responses. Therefore, understanding the function and regulation of ubiquitin-protein ligases is an important area of research in biology and medicine.

Magnetic Resonance Spectroscopy (MRS) is a non-invasive diagnostic technique that provides information about the biochemical composition of tissues, including their metabolic state. It is often used in conjunction with Magnetic Resonance Imaging (MRI) to analyze various metabolites within body tissues, such as the brain, heart, liver, and muscles.

During MRS, a strong magnetic field, radio waves, and a computer are used to produce detailed images and data about the concentration of specific metabolites in the targeted tissue or organ. This technique can help detect abnormalities related to energy metabolism, neurotransmitter levels, pH balance, and other biochemical processes, which can be useful for diagnosing and monitoring various medical conditions, including cancer, neurological disorders, and metabolic diseases.

There are different types of MRS, such as Proton (^1^H) MRS, Phosphorus-31 (^31^P) MRS, and Carbon-13 (^13^C) MRS, each focusing on specific elements or metabolites within the body. The choice of MRS technique depends on the clinical question being addressed and the type of information needed for diagnosis or monitoring purposes.

DNA Mutational Analysis is a laboratory test used to identify genetic variations or changes (mutations) in the DNA sequence of a gene. This type of analysis can be used to diagnose genetic disorders, predict the risk of developing certain diseases, determine the most effective treatment for cancer, or assess the likelihood of passing on an inherited condition to offspring.

The test involves extracting DNA from a patient's sample (such as blood, saliva, or tissue), amplifying specific regions of interest using polymerase chain reaction (PCR), and then sequencing those regions to determine the precise order of nucleotide bases in the DNA molecule. The resulting sequence is then compared to reference sequences to identify any variations or mutations that may be present.

DNA Mutational Analysis can detect a wide range of genetic changes, including single-nucleotide polymorphisms (SNPs), insertions, deletions, duplications, and rearrangements. The test is often used in conjunction with other diagnostic tests and clinical evaluations to provide a comprehensive assessment of a patient's genetic profile.

It is important to note that not all mutations are pathogenic or associated with disease, and the interpretation of DNA Mutational Analysis results requires careful consideration of the patient's medical history, family history, and other relevant factors.

Lymphocyte activation is the process by which B-cells and T-cells (types of lymphocytes) become activated to perform effector functions in an immune response. This process involves the recognition of specific antigens presented on the surface of antigen-presenting cells, such as dendritic cells or macrophages.

The activation of B-cells leads to their differentiation into plasma cells that produce antibodies, while the activation of T-cells results in the production of cytotoxic T-cells (CD8+ T-cells) that can directly kill infected cells or helper T-cells (CD4+ T-cells) that assist other immune cells.

Lymphocyte activation involves a series of intracellular signaling events, including the binding of co-stimulatory molecules and the release of cytokines, which ultimately result in the expression of genes involved in cell proliferation, differentiation, and effector functions. The activation process is tightly regulated to prevent excessive or inappropriate immune responses that can lead to autoimmunity or chronic inflammation.

"Attitude to Death" is not a medical term per se, but it does refer to an individual's perspective, feelings, and beliefs about death and dying. It can encompass various aspects such as fear, acceptance, curiosity, denial, or preparation. While not a medical definition, understanding a person's attitude to death can be relevant in healthcare settings, particularly in palliative and end-of-life care, as it can influence their decisions and experiences around their own mortality.

The EDA receptor (Ectodysplasin A receptor) is a gene that encodes a transmembrane protein involved in the development and maintenance of various tissues, including the skin and hair follicles. The Edar receptor plays a crucial role in the signaling pathway that regulates the formation and patterning of these structures during embryonic development. Mutations in this gene have been associated with several human genetic disorders, such as ectodermal dysplasia, which is characterized by abnormalities in the hair, teeth, nails, and sweat glands.

Ubiquitin is a small protein that is present in all eukaryotic cells and plays a crucial role in the regulation of various cellular processes, such as protein degradation, DNA repair, and stress response. It is involved in marking proteins for destruction by attaching to them, a process known as ubiquitination. This modification can target proteins for degradation by the proteasome, a large protein complex that breaks down unneeded or damaged proteins in the cell. Ubiquitin also has other functions, such as regulating the localization and activity of certain proteins. The ability of ubiquitin to modify many different proteins and play a role in multiple cellular processes makes it an essential player in maintaining cellular homeostasis.

I-kappa B (IκB) proteins are a family of inhibitory proteins that play a crucial role in regulating the activity of nuclear factor kappa B (NF-κB), a key transcription factor involved in inflammation, immune response, and cell survival. In resting cells, NF-κB is sequestered in the cytoplasm by binding to IκB proteins, which prevents NF-κB from translocating into the nucleus and activating its target genes.

Upon stimulation of various signaling pathways, such as those triggered by proinflammatory cytokines, bacterial or viral components, and stress signals, IκB proteins become phosphorylated, ubiquitinated, and subsequently degraded by the 26S proteasome. This process allows NF-κB to dissociate from IκB, translocate into the nucleus, and bind to specific DNA sequences, leading to the expression of various genes involved in immune response, inflammation, cell growth, differentiation, and survival.

There are several members of the IκB protein family, including IκBα, IκBβ, IκBε, IκBγ, and Bcl-3. Each member has distinct functions and regulatory mechanisms in controlling NF-κB activity. Dysregulation of IκB proteins and NF-κB signaling has been implicated in various pathological conditions, such as chronic inflammation, autoimmune diseases, and cancer.

Viral proteins are the proteins that are encoded by the viral genome and are essential for the viral life cycle. These proteins can be structural or non-structural and play various roles in the virus's replication, infection, and assembly process. Structural proteins make up the physical structure of the virus, including the capsid (the protein shell that surrounds the viral genome) and any envelope proteins (that may be present on enveloped viruses). Non-structural proteins are involved in the replication of the viral genome and modulation of the host cell environment to favor viral replication. Overall, a thorough understanding of viral proteins is crucial for developing antiviral therapies and vaccines.

Inflammation is a complex biological response of tissues to harmful stimuli, such as pathogens, damaged cells, or irritants. It is characterized by the following signs: rubor (redness), tumor (swelling), calor (heat), dolor (pain), and functio laesa (loss of function). The process involves the activation of the immune system, recruitment of white blood cells, and release of inflammatory mediators, which contribute to the elimination of the injurious stimuli and initiation of the healing process. However, uncontrolled or chronic inflammation can also lead to tissue damage and diseases.

Membrane microdomains, also known as lipid rafts, are specialized microenvironments within the cell membrane. They are characterized by the presence of sphingolipids, cholesterol, and specific proteins that cluster together, forming dynamic, heterogeneous, and highly organized domains. These microdomains are involved in various cellular processes such as signal transduction, membrane trafficking, and pathogen entry. However, it's important to note that the existence and function of membrane microdomains are still subjects of ongoing research and debate within the scientific community.

Transcription Factor RelA, also known as NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells) p65, is a protein complex that plays a crucial role in regulating the immune response to infection and inflammation, as well as cell survival, differentiation, and proliferation.

RelA is one of the five subunits that make up the NF-kB protein complex, and it is responsible for the transcriptional activation of target genes. In response to various stimuli such as cytokines, bacterial or viral antigens, and stress signals, RelA can be activated by phosphorylation and then translocate into the nucleus where it binds to specific DNA sequences called kB sites in the promoter regions of target genes. This binding leads to the recruitment of coactivators and the initiation of transcription.

RelA has been implicated in a wide range of biological processes, including inflammation, immunity, cell growth, and apoptosis. Dysregulation of NF-kB signaling and RelA activity has been associated with various diseases, such as cancer, autoimmune disorders, and neurodegenerative diseases.

Protein kinases are a group of enzymes that play a crucial role in many cellular processes by adding phosphate groups to other proteins, a process known as phosphorylation. This modification can activate or deactivate the target protein's function, thereby regulating various signaling pathways within the cell. Protein kinases are essential for numerous biological functions, including metabolism, signal transduction, cell cycle progression, and apoptosis (programmed cell death). Abnormal regulation of protein kinases has been implicated in several diseases, such as cancer, diabetes, and neurological disorders.

Immunoglobulin G (IgG) is a type of antibody, which is a protective protein produced by the immune system in response to foreign substances like bacteria or viruses. IgG is the most abundant type of antibody in human blood, making up about 75-80% of all antibodies. It is found in all body fluids and plays a crucial role in fighting infections caused by bacteria, viruses, and toxins.

IgG has several important functions:

1. Neutralization: IgG can bind to the surface of bacteria or viruses, preventing them from attaching to and infecting human cells.
2. Opsonization: IgG coats the surface of pathogens, making them more recognizable and easier for immune cells like neutrophils and macrophages to phagocytose (engulf and destroy) them.
3. Complement activation: IgG can activate the complement system, a group of proteins that work together to help eliminate pathogens from the body. Activation of the complement system leads to the formation of the membrane attack complex, which creates holes in the cell membranes of bacteria, leading to their lysis (destruction).
4. Antibody-dependent cellular cytotoxicity (ADCC): IgG can bind to immune cells like natural killer (NK) cells and trigger them to release substances that cause target cells (such as virus-infected or cancerous cells) to undergo apoptosis (programmed cell death).
5. Immune complex formation: IgG can form immune complexes with antigens, which can then be removed from the body through various mechanisms, such as phagocytosis by immune cells or excretion in urine.

IgG is a critical component of adaptive immunity and provides long-lasting protection against reinfection with many pathogens. It has four subclasses (IgG1, IgG2, IgG3, and IgG4) that differ in their structure, function, and distribution in the body.

The cell cycle is a series of events that take place in a cell leading to its division and duplication. It consists of four main phases: G1 phase, S phase, G2 phase, and M phase.

During the G1 phase, the cell grows in size and synthesizes mRNA and proteins in preparation for DNA replication. In the S phase, the cell's DNA is copied, resulting in two complete sets of chromosomes. During the G2 phase, the cell continues to grow and produces more proteins and organelles necessary for cell division.

The M phase is the final stage of the cell cycle and consists of mitosis (nuclear division) and cytokinesis (cytoplasmic division). Mitosis results in two genetically identical daughter nuclei, while cytokinesis divides the cytoplasm and creates two separate daughter cells.

The cell cycle is regulated by various checkpoints that ensure the proper completion of each phase before progressing to the next. These checkpoints help prevent errors in DNA replication and division, which can lead to mutations and cancer.

RNA interference (RNAi) is a biological process in which RNA molecules inhibit the expression of specific genes. This process is mediated by small RNA molecules, including microRNAs (miRNAs) and small interfering RNAs (siRNAs), that bind to complementary sequences on messenger RNA (mRNA) molecules, leading to their degradation or translation inhibition.

RNAi plays a crucial role in regulating gene expression and defending against foreign genetic elements, such as viruses and transposons. It has also emerged as an important tool for studying gene function and developing therapeutic strategies for various diseases, including cancer and viral infections.

A consensus sequence in genetics refers to the most common nucleotide (DNA or RNA) or amino acid at each position in a multiple sequence alignment. It is derived by comparing and analyzing several sequences of the same gene or protein from different individuals or organisms. The consensus sequence provides a general pattern or motif that is shared among these sequences and can be useful in identifying functional regions, conserved domains, or evolutionary relationships. However, it's important to note that not every sequence will exactly match the consensus sequence, as variations can occur naturally due to mutations or genetic differences among individuals.

Adenoviridae is a family of viruses that includes many species that can cause various types of illnesses in humans and animals. These viruses are non-enveloped, meaning they do not have a lipid membrane, and have an icosahedral symmetry with a diameter of approximately 70-90 nanometers.

The genome of Adenoviridae is composed of double-stranded DNA, which contains linear chromosomes ranging from 26 to 45 kilobases in length. The family is divided into five genera: Mastadenovirus, Aviadenovirus, Atadenovirus, Siadenovirus, and Ichtadenovirus.

Human adenoviruses are classified under the genus Mastadenovirus and can cause a wide range of illnesses, including respiratory infections, conjunctivitis, gastroenteritis, and upper respiratory tract infections. Some serotypes have also been associated with more severe diseases such as hemorrhagic cystitis, hepatitis, and meningoencephalitis.

Adenoviruses are highly contagious and can be transmitted through respiratory droplets, fecal-oral route, or by contact with contaminated surfaces. They can also be spread through contaminated water sources. Infections caused by adenoviruses are usually self-limiting, but severe cases may require hospitalization and supportive care.

A missense mutation is a type of point mutation in which a single nucleotide change results in the substitution of a different amino acid in the protein that is encoded by the affected gene. This occurs when the altered codon (a sequence of three nucleotides that corresponds to a specific amino acid) specifies a different amino acid than the original one. The function and/or stability of the resulting protein may be affected, depending on the type and location of the missense mutation. Missense mutations can have various effects, ranging from benign to severe, depending on the importance of the changed amino acid for the protein's structure or function.

Antibodies are proteins produced by the immune system in response to the presence of a foreign substance, such as a bacterium or virus. They are capable of identifying and binding to specific antigens (foreign substances) on the surface of these invaders, marking them for destruction by other immune cells. Antibodies are also known as immunoglobulins and come in several different types, including IgA, IgD, IgE, IgG, and IgM, each with a unique function in the immune response. They are composed of four polypeptide chains, two heavy chains and two light chains, that are held together by disulfide bonds. The variable regions of the heavy and light chains form the antigen-binding site, which is specific to a particular antigen.

Ectodysplasin receptors are a group of proteins that belong to the tumor necrosis factor (TNF) receptor superfamily. They play crucial roles in the development and function of ectodermal tissues, which include the skin, hair, nails, teeth, and sweat glands.

There are two main types of Ectodysplasin receptors: EDAR (Ectodysplasin A Receptor) and XEDAR (X-linked Ectodysplasin A Receptor). These receptors bind to their respective ligands, Ectodysplasin A (EDA) and Ectodysplasin A2 (EDA2), which are also members of the TNF family.

When EDA or EDA2 binds to EDAR or XEDAR, it activates a signaling pathway that involves several downstream molecules, including TRAF6 (TNF Receptor-Associated Factor 6) and NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells). This signaling cascade ultimately leads to the regulation of gene expression and cellular responses that are essential for ectodermal development.

Mutations in the genes encoding EDA, EDAR, or XEDAR have been associated with various genetic disorders, such as ectodermal dysplasias, which are characterized by abnormalities in the development of ectodermal tissues.

Caspases are a family of protease enzymes that play essential roles in programmed cell death, also known as apoptosis. There are two types of caspases: initiator caspases and effector (or executioner) caspases.

Effector caspases, also known as caspases-3, -6, and -7, are responsible for carrying out the proteolytic cleavage of various cellular substrates during apoptosis. Once activated by initiator caspases, effector caspases cleave key structural and regulatory proteins, leading to the characteristic morphological and biochemical changes associated with apoptotic cell death, such as chromatin condensation, DNA fragmentation, and membrane blebbing.

In summary, effector caspases are crucial components of the apoptotic machinery that mediate the execution phase of programmed cell death.

Receptor-Interacting Protein Serine-Threonine Kinase 2 (RIPK2) is a protein that plays a crucial role in the regulation of inflammatory and cell death pathways. It is a serine-threonine kinase that interacts with receptors involved in innate immune signaling, such as TNFR1 and TLRs. RIPK2 is essential for the activation of NF-κB, a transcription factor that regulates the expression of genes involved in inflammation, immunity, and cell survival. Additionally, RIPK2 has been implicated in the regulation of programmed cell death pathways such as necroptosis. Mutations in RIPK2 have been associated with various immune-related disorders, including inflammatory bowel disease and Blau syndrome.

Macromolecular substances, also known as macromolecules, are large, complex molecules made up of repeating subunits called monomers. These substances are formed through polymerization, a process in which many small molecules combine to form a larger one. Macromolecular substances can be naturally occurring, such as proteins, DNA, and carbohydrates, or synthetic, such as plastics and synthetic fibers.

In the context of medicine, macromolecular substances are often used in the development of drugs and medical devices. For example, some drugs are designed to bind to specific macromolecules in the body, such as proteins or DNA, in order to alter their function and produce a therapeutic effect. Additionally, macromolecular substances may be used in the creation of medical implants, such as artificial joints and heart valves, due to their strength and durability.

It is important for healthcare professionals to have an understanding of macromolecular substances and how they function in the body, as this knowledge can inform the development and use of medical treatments.

Interleukin-1 (IL-1) receptors are a type of cell surface receptor that bind to and mediate the effects of interleukin-1 cytokines, which are involved in the regulation of inflammatory and immune responses. There are two main types of IL-1 receptors:

1. Type I IL-1 receptor (IL-1R1): This is a transmembrane protein that consists of three domains - an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain contains the binding site for IL-1 cytokines, while the intracellular domain is involved in signal transduction and activation of downstream signaling pathways.
2. Type II IL-1 receptor (IL-1R2): This is a decoy receptor that lacks an intracellular signaling domain and functions to regulate IL-1 activity by preventing its interaction with IL-1R1.

IL-1 receptors are widely expressed in various tissues and cell types, including immune cells, endothelial cells, and nervous system cells. Activation of IL-1 receptors leads to the induction of a variety of biological responses, such as fever, production of acute phase proteins, activation of immune cells, and modulation of pain sensitivity. Dysregulation of IL-1 signaling has been implicated in various pathological conditions, including autoimmune diseases, chronic inflammation, and neurodegenerative disorders.

Nuclear Magnetic Resonance (NMR) Biomolecular is a research technique that uses magnetic fields and radio waves to study the structure and dynamics of biological molecules, such as proteins and nucleic acids. This technique measures the magnetic properties of atomic nuclei within these molecules, specifically their spin, which can be influenced by the application of an external magnetic field.

When a sample is placed in a strong magnetic field, the nuclei absorb and emit electromagnetic radiation at specific frequencies, known as resonance frequencies, which are determined by the molecular structure and environment of the nuclei. By analyzing these resonance frequencies and their interactions, researchers can obtain detailed information about the three-dimensional structure, dynamics, and interactions of biomolecules.

NMR spectroscopy is a non-destructive technique that allows for the study of biological molecules in solution, which makes it an important tool for understanding the function and behavior of these molecules in their natural environment. Additionally, NMR can be used to study the effects of drugs, ligands, and other small molecules on biomolecular structure and dynamics, making it a valuable tool in drug discovery and development.

Microfilament proteins are a type of structural protein that form part of the cytoskeleton in eukaryotic cells. They are made up of actin monomers, which polymerize to form long, thin filaments. These filaments are involved in various cellular processes such as muscle contraction, cell division, and cell motility. Microfilament proteins also interact with other cytoskeletal components like intermediate filaments and microtubules to maintain the overall shape and integrity of the cell. Additionally, they play a crucial role in the formation of cell-cell junctions and cell-matrix adhesions, which are essential for tissue structure and function.

Solubility is a fundamental concept in pharmaceutical sciences and medicine, which refers to the maximum amount of a substance (solute) that can be dissolved in a given quantity of solvent (usually water) at a specific temperature and pressure. Solubility is typically expressed as mass of solute per volume or mass of solvent (e.g., grams per liter, milligrams per milliliter). The process of dissolving a solute in a solvent results in a homogeneous solution where the solute particles are dispersed uniformly throughout the solvent.

Understanding the solubility of drugs is crucial for their formulation, administration, and therapeutic effectiveness. Drugs with low solubility may not dissolve sufficiently to produce the desired pharmacological effect, while those with high solubility might lead to rapid absorption and short duration of action. Therefore, optimizing drug solubility through various techniques like particle size reduction, salt formation, or solubilization is an essential aspect of drug development and delivery.

Drug synergism is a pharmacological concept that refers to the interaction between two or more drugs, where the combined effect of the drugs is greater than the sum of their individual effects. This means that when these drugs are administered together, they produce an enhanced therapeutic response compared to when they are given separately.

Drug synergism can occur through various mechanisms, such as:

1. Pharmacodynamic synergism - When two or more drugs interact with the same target site in the body and enhance each other's effects.
2. Pharmacokinetic synergism - When one drug affects the metabolism, absorption, distribution, or excretion of another drug, leading to an increased concentration of the second drug in the body and enhanced therapeutic effect.
3. Physiochemical synergism - When two drugs interact physically, such as when one drug enhances the solubility or permeability of another drug, leading to improved absorption and bioavailability.

It is important to note that while drug synergism can result in enhanced therapeutic effects, it can also increase the risk of adverse reactions and toxicity. Therefore, healthcare providers must carefully consider the potential benefits and risks when prescribing combinations of drugs with known or potential synergistic effects.

Molecular evolution is the process of change in the DNA sequence or protein structure over time, driven by mechanisms such as mutation, genetic drift, gene flow, and natural selection. It refers to the evolutionary study of changes in DNA, RNA, and proteins, and how these changes accumulate and lead to new species and diversity of life. Molecular evolution can be used to understand the history and relationships among different organisms, as well as the functional consequences of genetic changes.

Animal disease models are specialized animals, typically rodents such as mice or rats, that have been genetically engineered or exposed to certain conditions to develop symptoms and physiological changes similar to those seen in human diseases. These models are used in medical research to study the pathophysiology of diseases, identify potential therapeutic targets, test drug efficacy and safety, and understand disease mechanisms.

The genetic modifications can include knockout or knock-in mutations, transgenic expression of specific genes, or RNA interference techniques. The animals may also be exposed to environmental factors such as chemicals, radiation, or infectious agents to induce the disease state.

Examples of animal disease models include:

1. Mouse models of cancer: Genetically engineered mice that develop various types of tumors, allowing researchers to study cancer initiation, progression, and metastasis.
2. Alzheimer's disease models: Transgenic mice expressing mutant human genes associated with Alzheimer's disease, which exhibit amyloid plaque formation and cognitive decline.
3. Diabetes models: Obese and diabetic mouse strains like the NOD (non-obese diabetic) or db/db mice, used to study the development of type 1 and type 2 diabetes, respectively.
4. Cardiovascular disease models: Atherosclerosis-prone mice, such as ApoE-deficient or LDLR-deficient mice, that develop plaque buildup in their arteries when fed a high-fat diet.
5. Inflammatory bowel disease models: Mice with genetic mutations affecting intestinal barrier function and immune response, such as IL-10 knockout or SAMP1/YitFc mice, which develop colitis.

Animal disease models are essential tools in preclinical research, but it is important to recognize their limitations. Differences between species can affect the translatability of results from animal studies to human patients. Therefore, researchers must carefully consider the choice of model and interpret findings cautiously when applying them to human diseases.

Confocal microscopy is a powerful imaging technique used in medical and biological research to obtain high-resolution, contrast-rich images of thick samples. This super-resolution technology provides detailed visualization of cellular structures and processes at various depths within a specimen.

In confocal microscopy, a laser beam focused through a pinhole illuminates a small spot within the sample. The emitted fluorescence or reflected light from this spot is then collected by a detector, passing through a second pinhole that ensures only light from the focal plane reaches the detector. This process eliminates out-of-focus light, resulting in sharp images with improved contrast compared to conventional widefield microscopy.

By scanning the laser beam across the sample in a raster pattern and collecting fluorescence at each point, confocal microscopy generates optical sections of the specimen. These sections can be combined to create three-dimensional reconstructions, allowing researchers to study cellular architecture and interactions within complex tissues.

Confocal microscopy has numerous applications in medical research, including studying protein localization, tracking intracellular dynamics, analyzing cell morphology, and investigating disease mechanisms at the cellular level. Additionally, it is widely used in clinical settings for diagnostic purposes, such as analyzing skin lesions or detecting pathogens in patient samples.

SERPINs are an acronym for "serine protease inhibitors." They are a group of proteins that inhibit serine proteases, which are enzymes that cut other proteins. SERPINs are found in various tissues and body fluids, including blood, and play important roles in regulating biological processes such as inflammation, blood clotting, and cell death. They do this by forming covalent complexes with their target proteases, thereby preventing them from carrying out their proteolytic activities. Mutations in SERPIN genes have been associated with several genetic disorders, including emphysema, cirrhosis, and dementia.

Cysteine is a semi-essential amino acid, which means that it can be produced by the human body under normal circumstances, but may need to be obtained from external sources in certain conditions such as illness or stress. Its chemical formula is HO2CCH(NH2)CH2SH, and it contains a sulfhydryl group (-SH), which allows it to act as a powerful antioxidant and participate in various cellular processes.

Cysteine plays important roles in protein structure and function, detoxification, and the synthesis of other molecules such as glutathione, taurine, and coenzyme A. It is also involved in wound healing, immune response, and the maintenance of healthy skin, hair, and nails.

Cysteine can be found in a variety of foods, including meat, poultry, fish, dairy products, eggs, legumes, nuts, seeds, and some grains. It is also available as a dietary supplement and can be used in the treatment of various medical conditions such as liver disease, bronchitis, and heavy metal toxicity. However, excessive intake of cysteine may have adverse effects on health, including gastrointestinal disturbances, nausea, vomiting, and headaches.

Interferon-gamma (IFN-γ) is a soluble cytokine that is primarily produced by the activation of natural killer (NK) cells and T lymphocytes, especially CD4+ Th1 cells and CD8+ cytotoxic T cells. It plays a crucial role in the regulation of the immune response against viral and intracellular bacterial infections, as well as tumor cells. IFN-γ has several functions, including activating macrophages to enhance their microbicidal activity, increasing the presentation of major histocompatibility complex (MHC) class I and II molecules on antigen-presenting cells, stimulating the proliferation and differentiation of T cells and NK cells, and inducing the production of other cytokines and chemokines. Additionally, IFN-γ has direct antiproliferative effects on certain types of tumor cells and can enhance the cytotoxic activity of immune cells against infected or malignant cells.

Cell cycle proteins are a group of regulatory proteins that control the progression of the cell cycle, which is the series of events that take place in a eukaryotic cell leading to its division and duplication. These proteins can be classified into several categories based on their functions during different stages of the cell cycle.

The major groups of cell cycle proteins include:

1. Cyclin-dependent kinases (CDKs): CDKs are serine/threonine protein kinases that regulate key transitions in the cell cycle. They require binding to a regulatory subunit called cyclin to become active. Different CDK-cyclin complexes are activated at different stages of the cell cycle.
2. Cyclins: Cyclins are a family of regulatory proteins that bind and activate CDKs. Their levels fluctuate throughout the cell cycle, with specific cyclins expressed during particular phases. For example, cyclin D is important for the G1 to S phase transition, while cyclin B is required for the G2 to M phase transition.
3. CDK inhibitors (CKIs): CKIs are regulatory proteins that bind to and inhibit CDKs, thereby preventing their activation. CKIs can be divided into two main families: the INK4 family and the Cip/Kip family. INK4 family members specifically inhibit CDK4 and CDK6, while Cip/Kip family members inhibit a broader range of CDKs.
4. Anaphase-promoting complex/cyclosome (APC/C): APC/C is an E3 ubiquitin ligase that targets specific proteins for degradation by the 26S proteasome. During the cell cycle, APC/C regulates the metaphase to anaphase transition and the exit from mitosis by targeting securin and cyclin B for degradation.
5. Other regulatory proteins: Several other proteins play crucial roles in regulating the cell cycle, such as p53, a transcription factor that responds to DNA damage and arrests the cell cycle, and the polo-like kinases (PLKs), which are involved in various aspects of mitosis.

Overall, cell cycle proteins work together to ensure the proper progression of the cell cycle, maintain genomic stability, and prevent uncontrolled cell growth, which can lead to cancer.

p38 Mitogen-Activated Protein Kinases (p38 MAPKs) are a family of conserved serine-threonine protein kinases that play crucial roles in various cellular processes, including inflammation, immune response, differentiation, apoptosis, and stress responses. They are activated by diverse stimuli such as cytokines, ultraviolet radiation, heat shock, osmotic stress, and lipopolysaccharides (LPS).

Once activated, p38 MAPKs phosphorylate and regulate several downstream targets, including transcription factors and other protein kinases. This regulation leads to the expression of genes involved in inflammation, cell cycle arrest, and apoptosis. Dysregulation of p38 MAPK signaling has been implicated in various diseases, such as cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, p38 MAPKs are considered promising targets for developing new therapeutic strategies to treat these conditions.

Through its death domain, this receptor interacts with TRADD protein, which is known to serve as an adaptor that mediates ... The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor has been shown to activate NF-κB ... a novel death domain-containing TNF receptor" (PDF). FEBS Letters. 431 (3): 351-6. doi:10.1016/S0014-5793(98)00791-1. hdl: ... a novel death domain-containing TNF receptor" (PDF). FEBS Letters. 431 (3): 351-6. doi:10.1016/S0014-5793(98)00791-1. hdl: ...
The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor is activated by tumor necrosis ... "c-E10 is a caspase-recruiting domain-containing protein that interacts with components of death receptors signaling pathway and ... Death receptor 4 (DR4), also known as TRAIL receptor 1 (TRAILR1) and tumor necrosis factor receptor superfamily member 10A ( ... a death domain containing adaptor protein, is required for the apoptosis mediated by this protein. TNFRSF10A has been shown to ...
... is a threonine/serine protein kinase made up of 460 amino acids, which contains both a kinase domain and a death domain. ... The myddosome complex and IRAK1 recruit and activate TNF receptor-associated factor 6 (TRAF6), a ubiquitin protein ligase. ... One of these proteins, MyD88, uses its death domains to recruit, orient, and activate IRAK4. IRAK2 can then be phosphorylated ... and the Toll-like receptor superfamily share an intracytoplasmic Toll-IL-1 receptor (TIR) domain, which mediates recruitment of ...
... typical for the TNF family, but also found in viral capsid proteins. TNF can bind two receptors, TNFR1 (TNF receptor type 1; ... This dissociation enables the adaptor protein TRADD to bind to the death domain, serving as a platform for subsequent protein ... TNF is a member of the TNF superfamily, which consists of various transmembrane proteins with a homologous TNF domain. As an ... Induction of death signaling: Like all death-domain-containing members of the TNFR superfamily, TNFR1 is involved in death ...
"A protein related to a proteasomal subunit binds to the intracellular domain of the p55 TNF receptor upstream to its 'death ... "Two-hybrid cloning of a gene encoding TNF receptor-associated protein 2, a protein that interacts with the intracellular domain ... Hsu H, Huang J, Shu HB, Baichwal V, Goeddel DV (1996). "TNF-dependent recruitment of the protein kinase RIP to the TNF receptor ... "Prevention of constitutive TNF receptor 1 signaling by silencer of death domains". Science. 283 (5401): 543-6. Bibcode:1999Sci ...
"A protein related to a proteasomal subunit binds to the intracellular domain of the p55 TNF receptor upstream to its 'death ... "A protein related to a proteasomal subunit binds to the intracellular domain of the p55 TNF receptor upstream to its 'death ... "Two-hybrid cloning of a gene encoding TNF receptor-associated protein 2, a protein that interacts with the intracellular domain ... "Two-hybrid cloning of a gene encoding TNF receptor-associated protein 2, a protein that interacts with the intracellular domain ...
... and a second DED that is death effector domain in an adaptor protein that is directly associated with activated TNF receptors. ... other recognizable domains in this superfamily are: caspase-recruitment domain (CARD), pyrin domain (PYD) and death domain (DD ... The death-effector domain (DED) is a protein interaction domain found only in eukaryotes that regulates a variety of cellular ... The two tandem DEDs in the pro-domain of caspase induce the protein-protein interactions with other proteins like the FADD. ...
DDs are contained on cytokine receptors in the TNF receptor family. DD proteins function in apoptosis and NF-κB signaling in ... Examples of death fold domains include the death domain (DD), death effector domain (DED), caspase recruitment domain (CARD), ... In these interactions death-folds bind to another death-fold containing domain through the same type of protein interaction ... and pyrin domain (PYD). Death fold domains are an evolutionarily conserved superfamily of domains that mediate apoptotic ...
TNF, and TRAIL) and death receptors. Upon activation, the receptor protein, Fas-associated death domain protein, forms death- ... July 2014). "Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis". Science. 345 (6192 ... The PERK-ATF4-CHOP pathway can induce apoptosis by binding to the death receptors and upregulating the expression of death ... Death receptor-mediated apoptosis occurs via activation of death ligands (Fas, ...
The protein encoded by this gene is a member of the TNF-receptor superfamily, and contains an intracellular death domain. This ... Pan G, Ni J, Wei YF, Yu G, Gentz R, Dixit VM (Aug 1997). "An antagonist decoy receptor and a death domain-containing receptor ... Death receptor 5 (DR5), also known as TRAIL receptor 2 (TRAILR2) and tumor necrosis factor receptor superfamily member 10B ( ... a death domain containing adaptor protein, is required for the apoptosis mediated by this protein. DR5 has been shown to ...
... protein that combines the extracellular and intramembrane domains of the human TNF receptor 1 and the intracellular domain of ... The FasR part of the payload is intended to drive cell death in the endothelium of blood vessels where the chimeric protein is ... the Fas receptor, under the control of a modified version of the murine promoter for endothelin-1, which is called PPE-1. PPE-1 ... expressed; the TNF-R1 part is intended to provide some tumor specificity. Phase I trial results were presented in 2010 and as ...
This protein was found to be associated with the death domain of tumor necrosis factor receptor type 1 (TNF-R1) and death ... Frisch SM (2000). "Evidence for a function of death-receptor-related, death-domain-containing proteins in anoikis". Curr. Biol ... 2003). "Expression of Silencer of Death Domains and Death-Receptor-3 in Normal Human Kidney and in Rejecting Renal Transplants ... 2001). "Isolation of Bcl-2 binding proteins that exhibit homology with BAG-1 and suppressor of death domains protein". Biochem ...
... encoded by this gene is a death domain-containing adaptor protein that interacts with the death domain of TNF-alpha receptor 1 ... death domain protein that interacts with the type 1 tumor necrosis factor receptor and activates mitogen-activated protein ... a TNF receptor binding protein, correlates with neuronal cell death in Alzheimer's disease brain and hippocampal neurons". Proc ... MAP kinase-activating death domain protein is an enzyme that in humans is encoded by the MADD gene. Tumor necrosis factor alpha ...
"Disruption of hsp90 function results in degradation of the death domain kinase, receptor-interacting protein (RIP), and ... but overexpression of the RIP death domain can block NF-kB activation by TNF-R1. The intermediate domain is important for NF-kB ... the intracellular domain of TNFR starts the recruitment of the adaptor TNFR-1-associated death domain protein TRADD, which ... a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death". Cell. 81 (4): ...
... the pathway that leads to caspase activation via the intermediate membrane proteins TNF receptor-associated death domain (TRADD ... The TNF-alpha receptor superfamily also includes death receptors (DRs), such as DR4 and DR5. These receptors bind to the ... For example, the M-T2 protein of myxoma viruses can bind TNF preventing it from binding the TNF receptor and inducing a ... and Fas-associated death domain protein (FADD). cIAP1/2 can inhibit TNF-α signaling by binding to TRAF2. FLIP inhibits the ...
This receptor contains an extracellular TRAIL-binding domain and a transmembrane domain, but no cytoplasmic death domain. This ... is a human cell surface receptor of the TNF-receptor superfamily. The protein encoded by this gene is a member of the TNF- ... 1997). "An antagonist decoy receptor and a death domain-containing receptor for TRAIL". Science. 277 (5327): 815-8. doi:10.1126 ... Decoy receptor 1 (DCR1), also known as TRAIL receptor 3 (TRAILR3) and tumor necrosis factor receptor superfamily member 10C ( ...
This receptor contains an extracellular TRAIL-binding domain, a transmembrane domain, and a truncated cytoplasmic death domain ... is a human cell surface receptor of the TNF-receptor superfamily. The protein encoded by this gene is a member of the TNF- ... "Entrez Gene: TNFRSF10D tumor necrosis factor receptor superfamily, member 10d, decoy with truncated death domain". Kimberley FC ... 2000). "Differential localization and regulation of death and decoy receptors for TNF-related apoptosis-inducing ligand (TRAIL ...
cIAP1 contains baculovirus IAP repeat domains that facilitate binding to caspases and other proteins. cIAP1 is recruited to TNF ... Holcik M, Graber TE (2011). "Distinct roles for the cellular inhibitors of apoptosis proteins 1 and 2". Cell Death & Disease. 2 ... When NOD-like receptors are activated by bacterial peptidoglycans, they oligomerize and recruit cIAP1, cIAP2, TRAF2 and RIP2. ... cIAP1 (also named BIRC2) is the abbreviation for a human protein, cellular inhibitor of apoptosis protein-1. It belongs to the ...
... such as TNFR2 lacking a death domain (DD). The protein encoded by this gene is a member of the tumor necrosis factor receptor ... June 2000). "TTRAP, a novel protein that associates with CD40, tumor necrosis factor (TNF) receptor-75 and TNF receptor- ... This protein and TNF-receptor 1 form a heterocomplex that mediates the recruitment of two anti-apoptotic proteins, c-IAP1 and c ... The function of IAPs in TNF-receptor signalling is unknown, however, c-IAP1 is thought to potentiate TNF-induced apoptosis by ...
The protein encoded by this gene is a member of the TNF receptor associated factor (TRAF) protein family. TRAF proteins ... "Identification of a novel death domain-containing adaptor molecule for ectodysplasin-A receptor that is mutated in crinkled ... This protein directly interacts with TNF receptors, and forms complexes with other TRAF proteins. TRAF2 is required for TNF- ... and functions as a mediator of the anti-apoptotic signals from TNF receptors. The interaction of this protein with TRADD, a TNF ...
SMAC is a mitochondrial protein that promotes cytochrome c- and TNF receptor-dependent activation of apoptosis by inhibiting ... Thus, monomeric SMAC mutants can still bind the BIR3 domain but not the BIR2 domain, which compromises the protein's inhibitory ... Notably, preclinical studies indicate that the use of SMAC mimetics in conjunction with chemotherapy, death receptor ligands ... The homodimer structure also facilitates SMAC-XIAP binding via the BIR2 domain, though it does not form until the protein is ...
TNF)-induced necroptosis, a programmed cell death process, via interaction with receptor-interacting protein 3 (RIP3), which is ... This gene belongs to the protein kinase superfamily. The encoded protein contains a protein kinase-like domain; however, is ... "Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis". Proc. ... "Diverse sequence determinants control human and mouse receptor interacting protein 3 (RIP3) and mixed lineage kinase domain- ...
... a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis". Cell. 81 (4): 505-12 ... Micheau O, Tschopp J (July 2003). "Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes" (PDF ... This allows the adaptor molecule FADD to bind the death domain of Fas through its own death domain. FADD also contains a death ... The Fas receptor is a death receptor on the surface of cells that leads to programmed cell death (apoptosis) if it binds its ...
Most famously the RIG-I CARD domain is phosphorylated by protein kinase C-α (PKC-α) and PKC-β in the resting state to ... PTHR14074: Helicase with Death Domain-Related (filter for human) (Articles with short description, Short description matches ... This binding event is essential to signaling as it causes MAVS to form large functional aggregates in which TRAF3 (TNF receptor ... or proteins such as PP1α and PP1γ that negatively regulate RLR signaling. NOD-like receptor Toll-like receptor This article was ...
Tumor necrosis factor receptor type 1-associated DEATH domain protein is a protein that in humans is encoded by the TRADD gene ... Hsu H, Xiong J, Goeddel DV (June 1995). "The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B ... Hsu H, Huang J, Shu HB, Baichwal V, Goeddel DV (1996). "TNF-dependent recruitment of the protein kinase RIP to the TNF receptor ... TRADD is an adaptor protein. The protein encoded by this gene is a death domain containing adaptor molecule that interacts with ...
The term death receptor refers to those members of the TNF receptor superfamily that contain a death domain, such as TNFR1, Fas ... most TNF receptors require specific adaptor protein such as TRADD, TRAF, RIP and FADD for downstream signalling. TNF receptors ... May 1993). "Crystal structure of the soluble human 55 kd TNF receptor-human TNF beta complex: implications for TNF receptor ... TNF receptor family, Apoptosis, Protein superfamilies, Single-pass transmembrane proteins). ...
... in contrast to the type I death domain found in other TNFR proteins, the type II intracellular death domain of p75NTR does nor ... In the apoptosis pathway, members of the TNF receptor superfamily assemble a death-inducing signaling complex (DISC) in which ... The extracellular domain consists of a stalk domain connecting the transmembrane domain and four cysteine-rich repeat domains, ... The intracellular part is a global-like domain, known as a death domain, which consists of two sets of perpendicular helixes ...
... to dissociate from the MyD88 bound to the DD while maintaining interactions with downstream proteins such as TNF receptor- ... IRAK-1 contains an N-terminal death domain (DD), a ProST domain, a centrally located kinase domain, and a C-terminal domain. ... "Recruitment of IRAK to the interleukin 1 receptor complex requires interleukin 1 receptor accessory protein". Proceedings of ... The IRAK-1 encodes the interleukin-1 receptor-associated kinase 1, which is a serine-threonine protein kinase that is ...
TNFα leads to stimulation of its receptor TNFR1. TNFR1 binding protein TNFR-associated death protein TRADD and TNF receptor- ... First, extrinsic stimulus through the TNF receptor by TNFα signals the recruitment of the TNF receptor-associated death domain ... Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF, Wang FS, Wang X (April 2014). "Mixed lineage kinase domain-like protein MLKL ... The Tumor Necrosis Factor Receptor can signal for both apoptosis and necroptosis. The RIPK1 protein can also signal for both ...
The N-terminal FADD-like death effector domain of this protein suggests that it may interact with Fas-interacting protein FADD ... and TNF receptor-induced cell death". Cell. 85 (6): 803-15. doi:10.1016/s0092-8674(00)81265-9. PMID 8681376. S2CID 7415784. ... a novel death-effector domain-containing protein, interacts with caspases and BclXL and initiates cell death". Proc. Natl. Acad ... "Regulation of Fas-associated death domain interactions by the death effector domain identified by a modified reverse two-hybrid ...
Through its death domain, this receptor interacts with TRADD protein, which is known to serve as an adaptor that mediates ... The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor has been shown to activate NF-κB ... a novel death domain-containing TNF receptor" (PDF). FEBS Letters. 431 (3): 351-6. doi:10.1016/S0014-5793(98)00791-1. hdl: ... a novel death domain-containing TNF receptor" (PDF). FEBS Letters. 431 (3): 351-6. doi:10.1016/S0014-5793(98)00791-1. hdl: ...
TRADD also interacts strongly with another death domain protein; RIP and RIP plays an important role in the TNF signaling ... The interaction between the death domain of TNF. receptor-1 (TNFR1) and TRADD can trigger distinct signaling pathways leading ... "TNF-dependent recruitment of the protein kinase RIP to the TNF receptor-1 signaling complex," Immunity, vol. 4, no. 4, pp. 387- ... receptor expression through interaction with the PDZ domain protein, GIPC," The Journal of Biological Chemistry, vol. 276, no. ...
Fas Associated Death Domain) and TRADD (TNF receptor associated death domain). Together, these molecular pathways induce ... Such prenylation has important roles in the generation of lipidated protein domains that enable protein-protein interactions ... Daniel, P.T.; Wieder, T.; Sturm, I.; Schulze-Osthoff, K. The kiss of death: Promises and failures of death receptors and ... Isolation of proteins that interact specifically with the retinoid X receptor: Two novel orphan receptors. Mol. Endocrinol. ...
The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor contains a death domain. It has ... receptor with its ligand allows the formation of a death-inducing signaling complex that includes Fas-associated death domain ... This receptor has been also shown to activate NF-kappaB, MAPK3/ERK1, and MAPK8/JNK, and is found to be involved in transducing ... The isoforms lacking the transmembrane domain may negatively regulate the apoptosis mediated by the full length isoform. [ ...
... likely through interaction with TNF receptor type 1 (TNFR1)-associated death domain protein (TRADD). Specifically, decreased ... High-efficiency expression/cloning of epidermal growth factor-receptor-binding proteins with Src homology 2 domains. Proc Natl ... 6] ; ADAM-11, a protein implicated in cell-to-cell and cell-to-matrix interactions [7] ; GRB-7, a membrane-bound growth factor ... 4] The genetic defect appears to be a region of the gene encoding the KRT14 nonhelical head (E1/V1) domain located between the ...
Moreover, we discuss the roles of mitochondrial inflammation during cell death and how these processes may potentially be ... In this review, we discuss how mitochondria can initiate inflammation during cell death highlighting parallels with bacterial ... Many of the pro-inflammatory pathways activated during cell death occur upon mitochondrial outer membrane permeabilization ( ... while dispensable for cell death during mitochondrial apoptosis, inhibit activation of pro-inflammatory pathways after MOMP. ...
Death domain-containing protein CRADD (Caspase and RIP adapter with death domain) (RIP-associated protein with a death domain ... Background: Apoptotic adaptor molecule specific for caspase-2 and FASL/TNF receptor-interacting protein RIP. In the presence of ... Caspase and RIP adapter with death domain RIP-associated protein with a death domainGene... ... Recombinant Human Death domain-associated protein 6 (DAXX), partial , CSB-RP012444h Cusabio Human Recombinants ...
TNF receptor-associated factor 1, and cellular Fas-associated death domain protein-like interleukin-1 -converting enzyme ... This was accomplished by down-regulating apoptosis suppressor proteins and other crucial proteins such as the androgen receptor ... death receptor pathway (DR4, DR5), mitochondrial pathways, and protein kinase pathway (JNK, Akt, and AMPK). How curcumin ... The spice suppressed the activation of STAT3, and induced the death receptors (DR)4 and DR5. Turmeric enhanced the production ...
... likely through interaction with TNF receptor type 1 (TNFR1)-associated death domain protein (TRADD). Specifically, decreased ... High-efficiency expression/cloning of epidermal growth factor-receptor-binding proteins with Src homology 2 domains. Proc Natl ... 6] ; ADAM-11, a protein implicated in cell-to-cell and cell-to-matrix interactions [7] ; GRB-7, a membrane-bound growth factor ... 4] The genetic defect appears to be a region of the gene encoding the KRT14 nonhelical head (E1/V1) domain located between the ...
... although both receptors possess death domains with different binding proteins. In this forced induction of death of the ... TNF receptor p55) and introduced it into murine NIH/3T3 cells for their heterotopic expression. Upon stimulation with an ... A small protein model that consists of no more than the functional sites of trypsin1) is designed to detect the active sites ... Both Human Fas and Chimeric Fas/TNFR-1 Mediate Complete Cell Death in Murine NIH/3T3 Cells with Less Distinct Nuclear ...
TNF) receptor-associated factor 2 (TRAF2) [24,25] in the presence of mitogen-activated protein kinase kinase kinase (MAP3K), ... Kim R, Emi M, Tanabe K, Murakami S. Role of the unfolded protein response in cell death. Apoptosis. 2006; 11:5-13. [PMID: ... This 90-kDa protein has a basic leucine zipper domain (bZip) for DNA binding post homo- or heterodimerization. ATF6 binds to ... Protein N-glycosylation, protein folding, and protein quality control. Mol Cells. 2010; 30:497-506. [PMID: 21340671] ...
... the tumor necrosis factor receptor. Nowadays, monoclonal anti-TNF-α antibody is used for the treatment of these diseases ... is implicated in the process of various autoimmune and inflammatory diseases through binding to its receptor, ... because it can neutralize TNF-α to block the relevant signaling responsible for the pathogenesis of these diseases. Currently, ... tumor necrosis factor receptors. TRADD: TNF-receptor-associated death-domain protein. TRAF: TNF receptor associated factor ...
In particular, we will discuss the modifications in intrinsic and extrinsic apoptotic pathways caused by proteins encoded by ... Tumor necrosis factor receptor type 1-associated DEATH domain protein) and FADD (Fas-Associated protein with Death Domain) are ... "death" receptors to the TNF receptor family. TNF-alpha binds to TNF receptor 1 or 2 (TNF R1 and TNF R2), Fas ligand to Fas, and ... poxvirus and herpesvirus both encode proteins with homology to the DED domain, and thus block the binding of death receptors ...
The signal transduction of this receptor is mediated by various death domain containing adaptor proteins. Knockout studies in ... The TNFRSF-1B protein encoded by this gene is a member of the TNF-receptor superfamily. This protein and TNF-receptor 1 form a ... The signal transduction of this receptor is mediated by various death domain containing adaptor proteins. ... The TNF-receptor super-family receptors have been shown to stimulate NF-kappa B activity and regulate cell apoptosis [38]. ...
Fas-associated death domain (FADD), and caspase-8 to the death domain of TNF receptor 1 (TNFR1), thereby establishing the death ... 14.7K did not affect TNF-induced NF-κB activation via recruitment of receptor-interacting protein 1 (RIP-1) and TNF receptor- ... TNF-mediated apoptosis is initiated by ligand-induced recruitment of TNF receptor-associated death domain (TRADD), ... The adenoviral protein E3-14.7K (14.7K) is an inhibitor of TNF-induced apoptosis, but the molecular mechanism underlying this ...
Although dissimilar results have been found, a tissue-specific role of this protein emerges from the data. The objective of ... In the cytosol, Smac/DIABLO interacts and antagonizes inhibitors of apoptosis proteins (IAPs), thus allowing the activation of ... is a proapoptogenic mitochondrial protein that is released to the cytosol in response to diverse apoptotic stimuli, including ... Second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI (Smac/DIABLO) ...
FAS is a type I transmembrane protein and belongs to the TNF receptor superfamily. The extracellular region of FAS possesses ... three cystein-rich domains characteristic of the TNF superfamily, and the intracellular region which includes a death domain ( ... Ligand/Receptor TNFLSF6 (FASL). Cell Type Embryonic Stem Cells Biology Area Apoptosis/Tumor Suppressors/Cell Death, Cell ... Learn more about our Recombinant Proteins.. How does the activity of your recombinant proteins compare to competitors? We ...
... show that TNFR2-mediated apoptosis is blocked by caspase-8 inhibitors and depends on Fas-associated protein with death domain ( ... TNF receptors and then used mutants of human TNF capable of binding either human TNFR1 or human TNFR2 to investigate cell death ... The cytokine tumour necrosis factor (TNF) exerts its many effects through two receptors: TNFR1, the main signalling receptor ... This is a related article to: Induction of apoptosis by TNF receptor 2 in a T-cell hybridoma is FADD dependent and blocked by ...
death domain protein, I B kinase ε, and receptor (TNF receptor superfamily)-interacting serine-threonine kinase ... recep-tors (TLRs)[33], which will recruit adaptor proteins to the receptor, followed by activation of downstream signalling ... and TNF receptor-associated factor-6[35]. Thus, miR-146a functions as a negative feedback regulator of the TLR/ NF-κB pathway. ... Src homology 2 domain-containing inositol-5-phosphatase and CCAAT enhancer-binding protein beta are targeted by miR-155 in B ...
Moreover, ORF111L was verified to directly interact with the zebrafish TNF receptor type 1 associated death domain protein ( ... Proteína de Domínio de Morte Associada a Receptor de TNF/genética , Proteína de Domínio de Morte Associada a Receptor de TNF/ ... especially the death receptor zHDR and its cognate ligand DL1b. Further studies showed that the apoptosis induced by the both ... domain-containing protein, which shows high similarity to the dominant negative form of integrin-linked kinase (ILK); i.e., ...
Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and ... Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and ... TNFα, tumor necrosis factor alpha; TREM2, triggering receptor expressed on myeloid cells-2; TRIF, TIR domain-containing adapter ... complement receptor; DAP12, DNAX-activation protein of 12 KDa; Hsp60, heat-shock protein of 60 KDa; MARCO, macrophage receptor ...
MAVS also activates NF-κB through Fas-associated death domain-containing protein (FADD) and RIP1 interaction. The binding of ... which activates TNF receptor-associated factor 6 (TRAF6), TANK-binding kinase 1/IκB kinase i (TBK1/IKKi), and receptor- ... Activation of TLR3 by viral dsRNA leads to the recruitment of Toll/IL-1R homology domain-containing adaptor-inducing IFN-γ ( ... the endogenous protein PACT/RAX can also activate PKR independently of dsRNA. As suggested by the results of the current study ...
Failure to downregulate NF-kappaB transcriptional activity results in chronic inflammation and cell death, as observed in A20- ... linked ubiquitin chains from receptor interacting protein (RIP), an essential mediator of the proximal TNF receptor 1 (TNFR1) ... We also provide an example of a protein containing separate ubiquitin ligase and DUB domains, both of which participate in ... De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling Nature. 2004 Aug 5;430(7000):694-9. doi ...
We further found that BF-rTK + GCV inhibited the expression of the inflammatory maker of TNF-α. However, BF-rTK + GCV did not ... Furthermore, BF-rTK + GCV showed to repress the inflammation of tumor through downregulating TNF-α expression. Survival ... GCV significantly inhibited TNF-α expression. The results suggested that BF-rTK + GCV induced tumor apoptosis without autophagy ... The analysis of inflammatory markers (TNF-α) in tumor indicated that BF-rTK + ...
... death receptor 5) (7, 46). In response to TRAIL, these receptors recruit the adaptor protein FADD (Fas-associated death domain ... through death domain homophilic interactions (5), and the initiators procaspase-8 and -10, through death effector domain ... TRAIL triggers apoptosis upon engagement of one of its two agonistic receptors, named DR4 (death receptor 4) (33) and DR5 ( ... Tumor necrosis element (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of. Tumor necrosis element (TNF)-related ...
... cells were shown to possess markedly greater basal NF-κB responses and elevated Fas-associated protein with death domain-like ... Sodium salicylate did not affect the intracellular distribution of TNF receptors (TNFRs) but stimulated cell surface TNFR2 ... cells were shown to possess markedly greater basal NF-κB responses and elevated Fas-associated protein with death domain-like ... cells were shown to possess markedly greater basal NF-κB responses and elevated Fas-associated protein with death domain-like ...
TNF-α can bind at least 2 receptors on colonic epithelial cells. Tnfrsf1a receptors possess a cytoplasmic death domain that ... with protein content normalized using recombinant mouse TNF-α (Cell Signaling Technology). NF-κB was irreversibly blocked using ... TNF-α increases Tnfrsf1b receptors. A, expression of Tnfrsf1b receptors increases in YAMC cells at 100 ng/mL TNF-α at 24, 48, ... TNF-α increases Tnfrsf1b receptors. A, expression of Tnfrsf1b receptors increases in YAMC cells at 100 ng/mL TNF-α at 24, 48, ...
Through its death domain, this receptor interacts with TRADD protein, which is known to serve as an adaptor that mediates ... The protein encoded by this gene is a member of the TNF-receptor superfamily. This receptor has been shown to activate NF-κB ... Receptor/signaling modulators. Signaling peptide/protein receptor modulators. Growth factor receptor modulators. ... a novel death domain-containing TNF receptor" (PDF). FEBS Letters. 431 (3): 351-6. doi:10.1016/S0014-5793(98)00791-1. PMID ...
Western blotting was used to analyze protein extracts, and IL-8 release into cell culture supernatants was assessed by ELISA. ... TNF) receptor-associated factors (TRAF)2/6, and TNF receptor type 1-associated death domain protein (TRADD). ... Other important components of the macromolecular complex associated with TNF receptor superfamily, whose stimulation leads to ... TNF) receptor-associated factor 6 (TRAF6). Similarly to MyD88, the opposite actions of H2O2 and simvastatin translated into ...
cIAPs and XIAP negatively regulate cytokine production, including TNF to disrupt myeloid lineage differentiation.IAPs prevent ... Death domain-containing members of the TNFR superfamily and pattern recognition receptor family members (TLR) can not only ... A-induced TNF protein production and secretion (Figure 5F), did not reduce induction of TNF mRNA. On the contrary, TNF mRNA ... The death domain kinase RIP mediates the TNF-induced NF-kappaB signal. ...
  • TNF signaling controls immune cell activation via regulation of gene expression of chemokines, cytokines, and toxic molecules, including the reactive oxygen species, transcription factor nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK). (xiahepublishing.com)
  • Activation of TLR3 by viral dsRNA leads to the recruitment of Toll/IL-1R homology domain-containing adaptor-inducing IFN-γ (TRIF), which activates TNF receptor-associated factor 6 (TRAF6), TANK-binding kinase 1/IκB kinase i (TBK1/IKKi), and receptor-interacting protein 1 (RIP1). (jci.org)
  • In particular, the ERK1/2 subgroup of MAPK is activated by a MAPK kinase kinase named Raf (most commonly Raf-1), whose activation in turn requires the GTP-bound form of Ras family proteins [ 4 ]. (biomedcentral.com)
  • Loss of inhibitor of apoptosis proteins (IAPs), particularly cIAP1, can promote production of tumor necrosis factor (TNF) and sensitize cancer cell lines to TNF-induced necroptosis by promoting formation of a death-inducing signaling complex containing receptor-interacting serine/threonine-protein kinase (RIPK) 1 and 3. (ox.ac.uk)
  • The loss of RIPK1 or RIPK3, but not the RIPK3 substrate mixed lineage kinase domain-like protein, attenuated TNF secretion and thereby prevented apoptotic cell death and not necrosis. (ox.ac.uk)
  • Cell Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. (emisoft.cn)
  • TRADD recruits additional proteins that indirectly activate IκB kinase (IKK). (oncotarget.com)
  • Activation of DR3 by TL1A induced the formation of a signaling complex containing TRADD, TRAF2, and RIP and activated the NF-κB and the ERK, JNK, and p38 mitogen-activated protein kinase pathways. (enzolifesciences.com)
  • Here, we display that a kinase-independent function of RIP1 dampens the consequences of innate immune cell death. (5htreceptor.com)
  • herpesviruseceptor interacting protein (RIP) kinase RIP1 (RIPK1) functions as an vital adapter in a quantity of innate immune TrkA supplier signal transduction pathways, together with people initiated by Toll-like receptor (TLR)three, TLR4, and retinoic acid-inducible gene one (RIGI)-like receptors, on top of that to death receptors (1). (5htreceptor.com)
  • Furthermore, pathway analysis showed that YQRG treatment downregulated the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase/Akt (PI3K/AKT) signalling pathways and upregulated other signalling pathways, including those related to peroxisome proliferator-activated receptors(PPAR) and AMP-activated protein kinase(AMPK), with these findings subsequently verified experimentally. (biomedcentral.com)
  • Additionally, the direct binding of the core protein towards the cytoplasmic domains with the CD95 and kinase inhibitor Fingolimod TNF receptors has become reported to induce a pro apoptotic effect by altering mitochondrial function. (pkc-inhibitors.com)
  • Src kinase family is a family of non-receptor tyrosine kinases. (srcsignaling.com)
  • The TNF-α/TNFR1 complicated also contains the TNF-α receptor-associated protein with loss of life domains (TRADD) TNF-α receptor-associated protein 2 receptor-associated protein kinase 1 (RIPK1) and mobile inhibitor of apoptosis protein 1 (cIAP1) and cIAP2. (healthdisparitiesks.org)
  • Its activation is tightly controlled by numerous other signaling proteins including protein kinase C (PKC), Sab/SH3BP, and caveolin-1. (medscape.com)
  • It is a transmembrane protein with 212 amino acid sequence, existing in a homotrimer stable form that binds with two receptors, TNFR1 and TNFR2. (xiahepublishing.com)
  • TNF-mediated apoptosis is initiated by ligand-induced recruitment of TNF receptor-associated death domain (TRADD), Fas-associated death domain (FADD), and caspase-8 to the death domain of TNF receptor 1 (TNFR1), thereby establishing the death-inducing signaling complex (DISC). (jci.org)
  • Here we report that adenovirus 14.7K protein inhibits ligand-induced TNFR1 internalization. (jci.org)
  • Furthermore, we found that the same TNF defense mechanisms were instrumental in protecting wild-type adenovirus-infected human cells expressing 14.7K. This study describes a new molecular mechanism implemented by a virus to escape immunosurveillance by selectively targeting TNFR1 endocytosis to prevent TNF-induced DISC formation. (jci.org)
  • The authors engineered PC60 cells to express both human TNF receptors and then used mutants of human TNF capable of binding either human TNFR1 or human TNFR2 to investigate cell death induction in the T cell hybridoma. (biologists.com)
  • Among other results, they show that TNFR2-mediated apoptosis is blocked by caspase-8 inhibitors and depends on Fas-associated protein with death domain (FADD) but does not require simultaneous activation of TNFR1. (biologists.com)
  • The amino-terminal domain of A20, which is a de-ubiquitinating (DUB) enzyme of the OTU (ovarian tumour) family, removes lysine-63 (K63)-linked ubiquitin chains from receptor interacting protein (RIP), an essential mediator of the proximal TNF receptor 1 (TNFR1) signalling complex. (nih.gov)
  • TNF-α binds to TNF Receptor 1 (TNFR1), which causes the receptor to oligomerize and recruit TNF receptor associated death domain (TRADD). (oncotarget.com)
  • It looks likely that dysregulation of extra signaling pathways contributes to this phenotype, provided that deficiency in TNF receptor one (TNFR1) only modestly extends the lifespan of RIP1-null mice and deficiency in TNFR2 only rescues thymocytes from death (7). (5htreceptor.com)
  • The results of the present study suggest that P. vivax infection induces apoptosis of CD4 + T cells mediated by two types of signaling: by activation of the TNFR1 death receptor (extrinsic pathway), which is amplified by Bid, and by decreased expression of the anti-apoptotic protein Bcl-2 (intrinsic pathway). (biomedcentral.com)
  • TNF-α is normally a continuing feature of cyst liquids sampled in the Tmem33 kidneys of ADPKD sufferers.9 TNF-α binds to receptor I (TNFR1) to initiate the forming of a multimeric signaling complex that regulates cell survival and cell death. (healthdisparitiesks.org)
  • Through its death domain , this receptor interacts with TRADD protein, which is known to serve as an adaptor that mediates signal transduction of TNF-receptors. (wikidoc.org)
  • ROS may interfere with signal transduction pathways regulating the functions of transcription factors such as nuclear factor κB (NF-κB) and activator protein-1 (AP-1) [ 3 ]. (biomedcentral.com)
  • These proteins are involved in signal transduction from ectoderm to mesenchyme during development of the fetus and are indispensable for the differentiation of ectoderm-derived structures such as eccrine sweat glands, teeth, hair, skin, and/or nails. (springer.com)
  • Within the last 10 years significant progress has been made in understanding the pathogenesis of HED and this is mainly due to the discovery of unknown proteins and the elucidation of their function in signal transduction via the TNFα-related pathway. (springer.com)
  • Death receptor 4 (DR4), also known as TRAIL receptor 1 (TRAILR1) and tumor necrosis factor receptor superfamily member 10A (TNFRSF10A), is a cell surface receptor of the TNF-receptor superfamily that binds TRAIL and mediates apoptosis. (wikipedia.org)
  • Death receptor 6 ( DR6 ), also known as tumor necrosis factor receptor superfamily member 21 ( TNFRSF21 ), is a cell surface receptor of the tumor necrosis factor receptor superfamily which activates the JNK and NF-κB pathways. (wikidoc.org)
  • This in turn induces the recruitment of adapter proteins (FADD, TRADD o RAIDD) to form the so-called death-inducing signal complex (DISC), which activates caspase-8. (biomedcentral.com)
  • The aggregated death domains provide a novel molecular interface that interacts specifically with the death domain of tradd. (lu.se)
  • Various tradd-interacting proteins such as trafs, rip and possibly fadd, are recruited to the complex by their association with tradd. (lu.se)
  • Studies with FADD-deficient mice suggested that FADD, a death domain containing adaptor protein, is required for the apoptosis mediated by this protein. (wikipedia.org)
  • The interaction of this receptor with its ligand allows the formation of a death-inducing signaling complex that includes Fas-associated death domain protein (FADD), caspase 8, and caspase 10. (cancerindex.org)
  • The binding of FASL to FAS induces conformational changes in FAS, leading to the recruitment of FADD and procaspase-8 and the assembly of the death-inducing signaling complex (DISC). (biolegend.com)
  • MAVS also activates NF-κB through Fas-associated death domain-containing protein (FADD) and RIP1 interaction. (jci.org)
  • In response to TRAIL, these receptors recruit the adaptor protein FADD (Fas-associated death domain), through death domain homophilic interactions (5), and the initiators procaspase-8 and -10, through death effector domain interactions with FADD, hence forming the macromolecular complex called DISC (death-inducing signaling complex). (opioid-receptors.com)
  • On top of that, it's been postulated that this protein can bind to death domains, such as FADD and to the c FLIP inhibitor, resulting in an anti apoptotic result. (pkc-inhibitors.com)
  • The adapter molecule fadd recruits caspase-8 to the activated receptor. (lu.se)
  • This receptor is activated by tumor necrosis factor-related apoptosis inducing ligand (TNFSF10/TRAIL), and thus transduces cell death signal and induces cell apoptosis. (wikipedia.org)
  • The results suggest that exposure to GLA, even in low doses, during infantile period in the rat induces alterations in the kainic acid receptor in the brain. (go.jp)
  • Tumor necrosis element (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family that induces cancer cell death by apoptosis with some selectivity. (opioid-receptors.com)
  • [7] DR6 induces apoptosis when it is over expressed, however the manner in which the death signal is intracellularly transduced is currently unknown. (wikidoc.org)
  • Tumor necrosis factor (TNF) is a multifunctional cytokine, which induces proliferation or death in a cell type-dependent manner. (elsevierpure.com)
  • TNF-α induces its transcription in mutant renal epithelial cells recommending that. (healthdisparitiesks.org)
  • α-TEA induces apoptosis via activation of extrinsic death receptors Fas (CD95) and DR5, JNK/p73/Noxa pathways, and suppression of anti-apoptotic mediators Akt, ERK, c-FLIP and survivin in breast, ovarian and prostate cancer cells. (biomedcentral.com)
  • Many of the pro-inflammatory pathways activated during cell death occur upon mitochondrial outer membrane permeabilization (MOMP), the pivotal commitment point to cell death during mitochondrial apoptosis. (nature.com)
  • Caspases, while dispensable for cell death during mitochondrial apoptosis, inhibit activation of pro-inflammatory pathways after MOMP. (nature.com)
  • In particular, we will discuss the modifications in intrinsic and extrinsic apoptotic pathways caused by proteins encoded by HPV early genes. (mdpi.com)
  • Apoptosis can be activated through two pathways: The extrinsic pathway (mediated by death receptors) or the intrinsic pathway (mediated by mitochondria). (biomedcentral.com)
  • In this context, a new molecule, B-cell activating factor (BAFF), has emerged as a positive regulator of B cell survival and differentiation functioning through various signaling pathways and potentiating the activity of various receptor complexes through pleiotropic means. (medsci.org)
  • Signaling by means of these pathways bifurcates in the amount of RIP1 to produce opposing outcomes, a prosurvival inflammatory response counterbalanced by extrinsic cell death signaling that drives both apoptosis or necroptosis. (5htreceptor.com)
  • The company's pipeline of immunotherapy drug candidates targets different tumor necrosis factor (TNF) superfamily-dependent signaling pathways in order to restore the anti-tumor immune response in cancer patients and reduce lymphopenia and inflammatory cell death in patients with viral infections. (pharma-industry-review.com)
  • For instance, CSC maintenance in glioblastoma, colon cancer, gastric cancer, and prostate cancer is regulated by CD133-mediated AKT, leucine-rich G-protein-coupled receptor 5 (LGR5)-mediated Wnt/β-catenin and speckle-type POZ protein (SPOP)-mediated Nanog pathways [ 4 - 8 ] . (oaepublish.com)
  • In contrast, regulation of breast cancer CSCs (BCSCs) occurs by CD44 standard splice isoform (CD44s)-activated platelet-derived growth factor receptor b (PDGFRb)/signal transducer and activator of transcription 3 (STAT3), forkhead box C1 (FOXC1)-activated sonic hedgehog (SHH), and sphingosine-1-phosphate (S1P)/S1PR3-activated NOTCH pathways [ 10 - 13 ] . (oaepublish.com)
  • The binding with the ligands to the death receptors final results within the activation of caspase eight which in flip, activates two signaling pathways. (pkc-inhibitors.com)
  • Apoptotic pathways of cell death. (cdc.gov)
  • Communication between the pathways exists through cleavage of Bcl-2 interacting domain (Bid) by active caspase-8 to form truncated Bid (tBid). (cdc.gov)
  • The protein encoded by this gene is a member of the TNF-receptor superfamily. (wikipedia.org)
  • FAS is a type I transmembrane protein and belongs to the TNF receptor superfamily. (biolegend.com)
  • The extracellular region of FAS possesses three cystein-rich domains characteristic of the TNF superfamily, and the intracellular region which includes a death domain (DD). (biolegend.com)
  • The secreted decoy receptor 3 (DcR3), a soluble protein of the tumor necrosis factor receptor (TNFR) superfamily blocks the action of TL1A. (enzolifesciences.com)
  • DR6) is an orphan TNF receptor superfamily member and belongs to a subgroup of receptors called death receptors. (elabscience.com)
  • The extrinsic pathway is mediated by a variety of death receptor ligands, including tumor necrosis factor (TNF) and Fas ligand (FaSL), that trigger apoptosis by binding to cell surface receptors. (cdc.gov)
  • Furthermore, we identified key ligand-receptor pairs between crucial subtypes after MONs treatment that are known to promote liver fibrosis. (bvsalud.org)
  • [8] APP ( amyloid precursor protein ) is the natural ligand of DR6 and is first cleaved into Aβ and N-APP. (wikidoc.org)
  • TL1A (TNFSF15), a ligand belonging to the tumor necrosis factor (TNF) family, is expressed predominantly by endothelial cells and monocytes. (enzolifesciences.com)
  • ED1 , encoding a ligand-ectodysplasinA-A1 (EDA-A1), EDAR , coding for ectodysplasinA-A1 receptor, EDARADD , programming the structure of EDAR-associated death domain protein and NEMO whose protein product, NFκB essential modulator (NEMO), is necessary for an indirect activation of nuclear factor κB (NFκB). (springer.com)
  • The former is activated in response to the engagement of ligands such as CD95 or TNF-α with their receptors. (biomedcentral.com)
  • Also, this system may be mediated from the receptors and ligands on the Tumor Necrosis Factor family members, particularly, the TNF one recep tors, CD95 CD95 ligands, and TRAIL receptors one and two. (pkc-inhibitors.com)
  • This protein can inhibit CD95 receptors and TNF in duced apoptosis by inhibiting the liberation of cyto chrome c and, thus, by activating caspases 9, three and 7. (pkc-inhibitors.com)
  • The p38α further activates CCAAT/enhancer-binding protein β (C/EBPβ) resulting in cancer-related consequences, i.e. protein translation, cell differentiation and cell growth. (xiahepublishing.com)
  • Stress not only affects the neuroendocrine system but also immediately activates the white blood cells to produce proinflammatory interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF alpha). (entokey.com)
  • BSAC is a nuclear protein, and overexpression of BSAC potently activates promoters containing A + T-rich sequences named CArG boxes. (elsevierpure.com)
  • Tumor necrosis factor-alpha (TNF-α) is implicated in the process of various autoimmune and inflammatory diseases through binding to its receptor, the tumor necrosis factor receptor. (xiahepublishing.com)
  • The proinflammatory cytokine, tumor necrosis factor (TNF-α) makes a great contribution to immune system regulation, being involved in cell growth and proliferation upon binding with its receptor. (xiahepublishing.com)
  • As its name suggests, tumor necrosis factor (TNF) is known to induce cytotoxicity in a wide variety of tumor cells and cell lines. (elsevierpure.com)
  • Recent advances in understanding the molecular events underlying hypohidrotic ectodermal dysplasia (HED) caused by mutations of the genes encoding proteins of the tumor necrosis factor α (TNFα)-related signaling pathway have been presented. (springer.com)
  • HED embraces a genetically heterogeneous group of diseases and is due to mutations of several genes that encode components of the tumor necrosis factor α (TNFα)-related signaling pathway (Cluzeau et al. (springer.com)
  • Identification of a novel transcriptional activator, BSAC, by a functional cloning to inhibit tumor necrosis factor-induced cell death. (elsevierpure.com)
  • Extracellular Domain Of The 55Kda Tumor Necrosis Factor Receptor. (lu.se)
  • Crystallographic evidence for dimerization of unliganded tumor necrosis factor receptor. (lu.se)
  • Intracellular signaling peptides and proteins that bind directly or indirectly to the cytoplasmic portion of TUMOR NECROSIS FACTOR RECEPTORS. (bvsalud.org)
  • A family of genetically conserved signal transducing proteins that bind directly to TUMOR NECROSIS FACTOR RECEPTORS. (bvsalud.org)
  • Some drugs (eg, hydralazine , procainamide , isoniazid , anti-tumor necrosis factor [TNF] drugs) cause a reversible lupus-like syndrome. (msdmanuals.com)
  • Second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI (Smac/DIABLO) is a proapoptogenic mitochondrial protein that is released to the cytosol in response to diverse apoptotic stimuli, including commonly used chemotherapeutic drugs. (biomedcentral.com)
  • Smac (Second mitochondria-derived activator of caspase) protein, also known as DIABLO (Direct Inhibitor of Apoptosis-Binding protein with LOw pI), is codified by a nuclear gene. (biomedcentral.com)
  • The core protein It's been demonstrated the core protein of HCV has both professional apoptotic and anti apoptotic functions. (pkc-inhibitors.com)
  • CDBPA exerted a strong apoptotic cell death. (longdom.org)
  • Other biological effects not leading to apoptotic cell death have also been described. (biolegend.com)
  • ED 50 = 10 - 50 ng/ml, corresponding to a specific activity of 0.02 - 0.1 x 10 6 units/mg, as determined by inhibition of apoptotic cell death induced by FASL on Jurkat cells. (biolegend.com)
  • This type I transmembrane receptor possesses four extracellular cysteine-rich motifs and a cytoplasmic death domain. (elabscience.com)
  • Fas-activated DCs increase the expression of MHC II, B7 co-stimulatory molecules, and DC-lysosome-associated membrane proteins (DC-LAMP), and secrete proinflammatory cytokines such as IL-1β and TNF-α. (biolegend.com)
  • NF-κB is activated by TNF-α and other proinflammatory cytokines. (oncotarget.com)
  • TL1a is upregulated by the proinflammatory cytokines TNF and IL-1 and also by immune complexes (IC). (enzolifesciences.com)
  • On activated T cells, TL1A functions specifically via its surface-bound receptor DR3, (a member of the death-domain containing TNF receptor family) to promote cell survival and secretion of proinflammatory cytokines. (enzolifesciences.com)
  • In the cytosol, Smac/DIABLO interacts and antagonizes inhibitors of apoptosis proteins (IAPs), thus allowing the activation of caspases and apoptosis. (biomedcentral.com)
  • Apoptotic adaptor molecule specific for caspase-2 and FASL/TNF receptor-interacting protein RIP. (joplink.net)
  • The linker region between the BIR1 and BIR2 is implicated in the inhibition of caspase-3 and -7 whereas the BIR2 domain inhibits caspase-7 in a non-competitive manner [ 9 ]. (biomedcentral.com)
  • Inhibition of NF-κB or FLIP levels in human erythroleukemia cells by pharmacological or molecular-biological means also resulted in switching the character of these cells from a TNF-responsive proliferative phenotype into an apoptotic one. (elsevierpure.com)
  • [2] The DR6 is an alpha-helical integral membrane receptor protein that shows evidence that it has something to do with the inhibition of blood vessels forming on tumors which would allow them to grow larger. (wikidoc.org)
  • The cytokine production and consequent cell death caused by IAP depletion was attenuated by loss or inhibition of TNF or TNF receptor 1. (ox.ac.uk)
  • Interestingly, inhibition of TNF-induced cell death was not observed in DKO MEFs transfected with either N-terminal or C-terminal deletion mutant of BSAC, revealing an intimate correlation between transcriptional activity and antiapoptotic function. (elsevierpure.com)
  • DcR1 is characterized by the presence of a glycosylphosphatidylinositol membrane anchor and therefore has no intracellular domain, whereas the death domain of DcR2 is truncated. (opioid-receptors.com)
  • Sodium salicylate did not affect the intracellular distribution of TNF receptors (TNFRs) but stimulated cell surface TNFR2 shedding. (elsevierpure.com)
  • Cytoplasmic dsRNA is also recognized by the RLH system, which interacts with MAVS via caspase recruitment domains (CARDs). (jci.org)
  • Interacts with hcv core protein. (lu.se)
  • Death receptor 6 gets a chemical message and starts a signaling pathway that causes apoptosis, also known as cell death, to occur. (wikidoc.org)
  • In addition to its role in glucose metabolism, this pathway also regulates the redirection of free amino acids to protein synthesis via the mTOR-signaling pathway. (hindawi.com)
  • Novel data were reviewed and discussed on the structure and functions of the components of TNFα-related signaling pathway, the consequences of mutations of the genes encoding these proteins, and the prospect for further investigations, which might elucidate the origin of HED. (springer.com)
  • With the exception of EDA1 and NEMO , both localized on the X chromosome, all other genes encoding components of the TNFα-related signaling pathway involved in differentiation of skin appendages, are localized on the autosomes (Table 1 ). (springer.com)
  • The purpose of this report is to review current literature on the structure and function of components of the TNFα-related signaling pathway, to present a novel approach to their contribution in the differentiation of skin appendages and to discuss the role of mutations of genes encoding components of this pathway in the origin of HED. (springer.com)
  • Apoptotic caspase activity inhibits cell death-associated inflammation. (nature.com)
  • CusabioAlternative Name(s): Caspase and RIP adapter with death domain RIP-associated protein with a death domainGene. (joplink.net)
  • The resulting death-inducing signaling complex (disc) performs caspase-8 proteolytic activation which initiates the subsequent cascade of caspases (aspartate-specific cysteine proteases) mediating apoptosis. (lu.se)
  • These NFPs are a chemoattractant for host phagocytes and can be recognised by formyl peptide receptors on the plasma membrane [ 8 , 9 ]. (nature.com)
  • Type I membrane protein and secreted. (lu.se)
  • Failure to downregulate NF-kappaB transcriptional activity results in chronic inflammation and cell death, as observed in A20-deficient mice. (nih.gov)
  • Extending these observations, colon biopsies from E. faecalis -colonized IL-10 −/− mice exhibited crypt hyperplasia and increased staining for macrophages, TNF-α, netrin-1, NF-κB, Tnfrsf1b, and the proliferation marker proliferating cell nuclear antigen while also displaying a reduction in epithelial cell apoptosis. (aacrjournals.org)
  • Despite the typical improvement of quite a few organs and neuromuscular architecture, RIP1-null mice die inside some days of birth with indicators of edema at the same time as major amounts of cell death inside of lymphoid tissues, especially immature thymocytes (five). (5htreceptor.com)
  • While TNF-signaling contributes to this perinatal death (six) and implicates the prosurvival purpose of RIP1 in activating nuclear aspect B (NF-B) (5), the exact mechanism accountable for developmental failure of RIP1-deficient mice stays unresolved. (5htreceptor.com)
  • The signaling via the TNFα-related pathway, which is critical for differentiation of skin appendages, has been elucidated mainly due to the investigations performed in mice (Mikkola 2009 ). (springer.com)
  • We previously showed that murine embryonic fibroblasts (MEFs) from TNF receptor-associated factor 2 (Traf2) and Traf5 double-deficient (double knockout (DKO)) mice were highly susceptible to TNF-induced cell death. (elsevierpure.com)
  • DR6 is an extensively posttranslationally modified transmembrane protein and that N- and O-glycosylations of amino acids in its extracellular part. (elabscience.com)
  • Binding of TNF to the extracellular domain leads to homotrimerization. (lu.se)
  • Here, we report that the central proinflammatory cytokine TNF-α acts as a diffusible mediator of the bystander effects induced by macrophages, an effect caused by a proliferation of macrophages that trigger epithelial cell production of Netrin-1, a neuronal guidance molecule. (aacrjournals.org)
  • deletion of an exon encoding the transmembrane domain results in a soluble FAS molecule that blocks apoptosis. (biolegend.com)
  • Crystal structure of the soluble human 55 kd TNF receptor-human TNF complex: implications for TNF receptor activation. (lu.se)
  • A skewed preference for BV7 genes and sequential "G" amino acids motif is observed in complementarity-determining region-3 of T cell receptor-β. (nature.com)
  • TNF-α was declared, in 1975, as a circulating antitumorigenic cytokine produced from such immune cells as activated macrophages and lymphocytes. (xiahepublishing.com)
  • TNF-α-mediated bystander assays used a murine coculture system of primary colonic epithelial cells and E. faecalis -infected macrophages ( in vitro ), with an interleukin 10 (IL-10)-deficient mouse model of colon cancer that involves long-term colonization with E. faecalis ( in vivo ). (aacrjournals.org)
  • In vitro differentiation of macrophages from bone marrow in the absence of cIAPs and XIAP led to detectable levels of TNF and resulted in reduced numbers of mature macrophages. (ox.ac.uk)
  • The immune response is mediated mainly by macrophages and nat ural killer cells, which can right lead to the death of your contaminated cells. (pkc-inhibitors.com)
  • 1 , 2 Improper TNF-α production has been seen in a variety of human diseases, including Alzheimer's disease, 3 cancer, 4 major depression, 5 psoriasis, 6 and inflammatory bowel disease (IBD). (xiahepublishing.com)
  • The analysis of inflammatory markers (TNF-α) in tumor indicated that BF-rTK + GCV significantly inhibited TNF-α expression. (biomedcentral.com)
  • We further found that BF-rTK + GCV inhibited the expression of the inflammatory maker of TNF-α. (biomedcentral.com)
  • The evolutionary process positively selected individuals that upon being wounded (which was connected with emotional stress, imminent infection, and a consequent danger of serious illness and death), quickly responded with an inflammatory reaction. (entokey.com)
  • In addition to the agonistic TRAIL receptors DR4 and DR5, TRAIL can bind to related but antagonistic receptors, including TRID or TRAIL-R3 (11, 27, 32) and TRUNDD or TRAIL-R4 (10), also coined DcR1 (decoy receptor 1) and DcR2 (decoy receptor 2), respectively. (opioid-receptors.com)
  • cIAPs and XIAP negatively regulate cytokine production, including TNF to disrupt myeloid lineage differentiation. (ashpublications.org)
  • It has been shown to play a central role in the physiological regulation of programmed cell death, and has been implicated in the pathogenesis of various malignancies and diseases of the immune system. (cancerindex.org)
  • Mitochondria have recently emerged as key drivers of inflammation associated with cell death. (nature.com)
  • In this review, we discuss how mitochondria can initiate inflammation during cell death highlighting parallels with bacterial activation of inflammation. (nature.com)
  • Moreover, we discuss the roles of mitochondrial inflammation during cell death and how these processes may potentially be harnessed therapeutically, for instance to improve cancer treatment. (nature.com)
  • Mitochondrial-driven inflammation can enhance the immunogenicity of cell death. (nature.com)
  • Is mitochondrial-driven inflammation during cell death driven by bacterial-like DAMPs? (nature.com)
  • What physiological functions has mitochondrial inflammation during cell death? (nature.com)
  • TAK1 adaptor proteins, TAB2 and TAB3, link the signalosome to B-cell receptor-induced IKK activation. (xiahepublishing.com)
  • This type of cell death is fundamental for the maintenance of tissue homeostasis and immune system development [ 2 ]. (biomedcentral.com)
  • 5 ), cigarette smoke may (as reflected by dashed arrows) affect lung cell responses to RNA viruses by enhancing MAVS-PKR signaling and therefore trigger alveolar cell death. (jci.org)
  • Transient overexpression of DcR1 or DcR2 in TRAIL-sensitive Sav1 tumor cells prevents cell death triggering by TRAIL (10, 11), and recent evidence indicates that tumor and normal cells can acquire resistance to TRAIL-induced killing by up-regulating TRAIL antagonistic receptors (6, 8, 9, 34). (opioid-receptors.com)
  • In cell cocultures, we observed increased expression of the TNF-α receptor Tnfrsf1b and Netrin-1. (aacrjournals.org)
  • RNAi-mediated attenuation of Tnfrsf1b decreased TNF-α-induced netrin-1 production and augmented epithelial cell apoptosis in culture. (aacrjournals.org)
  • This receptor has been shown to activate NF-κB and MAPK8 / JNK , and induce cell apoptosis . (wikidoc.org)
  • Tumor cells can induce, through exposition of amyloid precursor protein (APP), DR6-mediated endothelial cell necroptosis allowing tumors metastasis. (wikidoc.org)
  • The free serum levels of this DR6 are heightened with anti-cell death factors in patients that have later stage ovarian cancer. (wikidoc.org)
  • Western blotting was used to analyze protein extracts, and IL-8 release into cell culture supernatants was assessed by ELISA. (biomedcentral.com)
  • Mes as broad as cytokine activation and cell death. (5htreceptor.com)
  • Increased binding scores are observed in T-cell clones with complementarity-determining region-3 of T cell receptor-β with a "(G)GG" motif. (nature.com)
  • An individual T-cell has a specific complementarity-determining region 3 (CDR3) of T cell receptor (TCR)-α and -β, which is a result of the recombination of somatic TCR V-(D)-J genes and junction diversity. (nature.com)
  • Overexpression of BSAC in DKO MEFs partially inhibited TNF-induced cell death by suppressing activation of caspases. (elsevierpure.com)
  • Vaccine-infected B cells were characterized by a higher percentage of cells positive for viral protein, a higher level of viral transcription and reduced cell death compared to wild-type infected cells, regardless of B cell subtype. (cdc.gov)
  • [ 4 ] The genetic defect appears to be a region of the gene encoding the KRT14 nonhelical head (E1/V1) domain located between the microsatellite markers D17S798 and D17S957, which are separated by approximately 26.97 cM. (medscape.com)
  • Protein level and Gene expression were tested by Western blotting analysis and by the Applied Biosystems® Tagman® Array Plates, respectively. (longdom.org)
  • A gene expression profile analysis has been performed and the results indicated that CBPDA is significantly increased the expression of five genes: CARD9, BNIP, TNFRSF-1B, TNFRSF25 and TNF-alpha, that have an important role in induction of apoptosis. (longdom.org)
  • The more studied member is XIAP, formed by three BIR (Baculoviral IAP Repeat) domains located in the NH 2 -terminus and one RING (Really Interesting New Gene) domain in the CO 2 H-terminus. (biomedcentral.com)
  • The binding of type I IFNs to their receptor (IFNR) causes JAK/STAT-mediated synthesis of IFN-stimulated gene (ISG) products, including PKR. (jci.org)
  • Additionally, we performed transcriptome analysis using RNA-sequencing (RNA-seq) technology, including cluster, Gene Ontology (GO), and pathway analyses, to identify differentially expressed genes (DEGs), and protein and gene expression were detected by immunofluorescence (IFC), western blot and real-time quantitative PCR. (biomedcentral.com)
  • Mutations of these genes are responsible for systemic tooth agenesis, in addition to the defects of other ectodermal structures, and this review is limited to description of the function of their protein products. (springer.com)
  • The molecular chaperone glucose-regulated protein 78 ( GRP78 ) or binding immunoglobulin protein (BiP), in addition to Ca 2+ binding and protein processing functions, possesses one more key role: master initiator of early UPR signaling. (molvis.org)
  • The adenoviral protein E3-14.7K (14.7K) is an inhibitor of TNF-induced apoptosis, but the molecular mechanism underlying this protective effect has not yet been explained exhaustively. (jci.org)
  • The 397 amino acid recombinant protein has a predicted molecular mass of approximately 44.7 kD. (biolegend.com)
  • Classical NF-κB is a cytoplasmic inactive heterodimer of p65 and p50 bound to Inhibitor of NF-κB (IκB) proteins [ 9 ]. (oncotarget.com)
  • Although it is unclear whether PKR may be directly stimulated by the interaction of MAVS and dsRNA (as indicated by the dashed arrow), the endogenous protein PACT/RAX can also activate PKR independently of dsRNA. (jci.org)
  • RIP1 orchestrates assembly of distinct signaling platforms by way of two C-terminal protein rotein AChE Inhibitor Purity & Documentation binding domains: a death domain as well as a RIP homotypic interaction motif (RHIM) (three, 4). (5htreceptor.com)
  • Prevention of raft reorganization into larger platforms by inactivation of acid sphingomyelinase (ASMase), renders cells resistant to Fas clustering and apoptosis [ 9 ], indicating that ASMase/ceramide plays an important role in death receptor-mediated apoptosis. (biomedcentral.com)
  • Contributes to the induction of noncytocidal TNF effects including anti-viral state and activation of the acid sphingomyelinase. (lu.se)
  • In contrast, 14.7K did not affect TNF-induced NF-κB activation via recruitment of receptor-interacting protein 1 (RIP-1) and TNF receptor-associated factor 2 (TRAF-2). (jci.org)
  • The appearance of TNF-α is normally controlled through its receptor-mediated activation of NF-κB.29 Quantitative RT-PCR demonstrated that TNF-α mRNA was increased in null mouse embryonic kidney (MEK) cells (Amount 1A) and postnatal homozygous PN24 cells (Amount 1B) aswell as the kidneys from and wild-type MEK cells heterozygous PH2 cells and wild-type kidneys respectively. (healthdisparitiesks.org)
  • Due to its bacterial ancestry, mitochondrial formylation of methionine is required for translation initiation of mtDNA-derived mRNA [ 7 ] and will therefore be recognised by the same formyl peptide receptors. (nature.com)
  • TNF-α mRNA was further XL184 free base (Cabozantinib) elevated in response to exterior TNF-α arousal in null MEK cells and PN24 cells (Amount 1 A and B). This response is normally mediated through canonical NF-κB signaling because adding an NF-κB inhibitor SN50 avoided the upsurge in TNF-α mRNA in mutant renal epithelial cells treated with TNF-α (Amount 1A). (healthdisparitiesks.org)
  • The isoforms lacking the transmembrane domain may negatively regulate the apoptosis mediated by the full length isoform. (cancerindex.org)
  • 8 , 9 On binding with receptors, it controls the expression of inflammation and injury. (xiahepublishing.com)
  • Furthermore, BF-rTK + GCV showed to repress the inflammation of tumor through downregulating TNF-α expression. (biomedcentral.com)
  • STAT5 protein, involved in cytosolic signaling, is activated by TRAF2 through PI3K. (xiahepublishing.com)
  • Within this context a key role is played by mitogen-activated protein kinases (MAPK), whose targets are mainly represented by nuclear transcription factors, also including those involved in oxidative stress. (biomedcentral.com)
  • Asunercept is expected to prevent the death of immune and lung cells leading to lymphopenia and acute respiratory distress syndrome, thus reducing the number of COVID-19 patients who require intensive care or even die from this disease. (pharma-industry-review.com)