The act of ligating UBIQUITINS to PROTEINS to form ubiquitin-protein ligase complexes to label proteins for transport to the PROTEASOME ENDOPEPTIDASE COMPLEX where proteolysis occurs.
A highly conserved 76-amino acid peptide universally found in eukaryotic cells that functions as a marker for intracellular PROTEIN TRANSPORT and degradation. Ubiquitin becomes activated through a series of complicated steps and forms an isopeptide bond to lysine residues of specific proteins within the cell. These "ubiquitinated" proteins can be recognized and degraded by proteosomes or be transported to specific compartments within the cell.
A diverse class of enzymes that interact with UBIQUITIN-CONJUGATING ENZYMES and ubiquitination-specific protein substrates. Each member of this enzyme group has its own distinct specificity for a substrate and ubiquitin-conjugating enzyme. Ubiquitin-protein ligases exist as both monomeric proteins multiprotein complexes.
A family of proteins that are structurally-related to Ubiquitin. Ubiquitins and ubiquitin-like proteins participate in diverse cellular functions, such as protein degradation and HEAT-SHOCK RESPONSE, by conjugation to other proteins.
A large multisubunit complex that plays an important role in the degradation of most of the cytosolic and nuclear proteins in eukaryotic cells. It contains a 700-kDa catalytic sub-complex and two 700-kDa regulatory sub-complexes. The complex digests ubiquitinated proteins and protein activated via ornithine decarboxylase antizyme.
A class of enzymes that form a thioester bond to UBIQUITIN with the assistance of UBIQUITIN-ACTIVATING ENZYMES. They transfer ubiquitin to the LYSINE of a substrate protein with the assistance of UBIQUITIN-PROTEIN LIGASES.
A set of protein subcomplexes involved in PROTEIN SORTING of UBIQUITINATED PROTEINS into intraluminal vesicles of MULTIVESICULAR BODIES and in membrane scission during formation of intraluminal vesicles, during the final step of CYTOKINESIS, and during the budding of enveloped viruses. The ESCRT machinery is comprised of the protein products of Class E vacuolar protein sorting genes.
A class of enzymes that catalyze the formation of a bond between two substrate molecules, coupled with the hydrolysis of a pyrophosphate bond in ATP or a similar energy donor. (Dorland, 28th ed) EC 6.
An essential amino acid. It is often added to animal feed.
Cleavage of proteins into smaller peptides or amino acids either by PROTEASES or non-enzymatically (e.g., Hydrolysis). It does not include Protein Processing, Post-Translational.
A family of structurally related proteins that were originally discovered for their role in cell-cycle regulation in CAENORHABDITIS ELEGANS. They play important roles in regulation of the CELL CYCLE and as components of UBIQUITIN-PROTEIN LIGASES.
Proteins covalently modified with UBIQUITINS or UBIQUITIN-LIKE PROTEINS.
An oligomer formed from the repetitive linking of the C-terminal glycine of one UBIQUITIN molecule via an isopeptide bond to a lysine residue on a second ubiquitin molecule. It is structurally distinct from UBIQUITIN C, which is a single protein containing a tandemly arrayed ubiquitin peptide sequence.
Complexes of enzymes that catalyze the covalent attachment of UBIQUITIN to other proteins by forming a peptide bond between the C-terminal GLYCINE of UBIQUITIN and the alpha-amino groups of LYSINE residues in the protein. The complexes play an important role in mediating the selective-degradation of short-lived and abnormal proteins. The complex of enzymes can be broken down into three components that involve activation of ubiquitin (UBIQUITIN-ACTIVATING ENZYMES), conjugation of ubiquitin to the ligase complex (UBIQUITIN-CONJUGATING ENZYMES), and ligation of ubiquitin to the substrate protein (UBIQUITIN-PROTEIN LIGASES).
An E3 UBIQUITIN LIGASE that interacts with and inhibits TUMOR SUPPRESSOR PROTEIN P53. Its ability to ubiquitinate p53 is regulated by TUMOR SUPPRESSOR PROTEIN P14ARF.
A thioester hydrolase which acts on esters formed between thiols such as DITHIOTHREITOL or GLUTATHIONE and the C-terminal glycine residue of UBIQUITIN.
A subset of ubiquitin protein ligases that are formed by the association of a SKP DOMAIN PROTEIN, a CULLIN DOMAIN PROTEIN and a F-BOX DOMAIN PROTEIN.
A family of F-box domain proteins that contain sequences that are homologous to the beta subunit of transducin (BETA-TRANSDUCIN). They play an important role in the protein degradation pathway by becoming components of SKP CULLIN F-BOX PROTEIN LIGASES, which selectively act on a subset of proteins including beta-catenin and IkappaBbeta.
A family of proteins that share the F-BOX MOTIF and are involved in protein-protein interactions. They play an important role in process of protein ubiquition by associating with a variety of substrates and then associating into SCF UBIQUITIN LIGASE complexes. They are held in the ubiquitin-ligase complex via binding to SKP DOMAIN PROTEINS.
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.
Any of various enzymatically catalyzed post-translational modifications of PEPTIDES or PROTEINS in the cell of origin. These modifications include carboxylation; HYDROXYLATION; ACETYLATION; PHOSPHORYLATION; METHYLATION; GLYCOSYLATION; ubiquitination; oxidation; proteolysis; and crosslinking and result in changes in molecular weight and electrophoretic motility.
Proto-oncogene proteins that negatively regulate RECEPTOR PROTEIN-TYROSINE KINASE signaling. It is a UBIQUITIN-PROTEIN LIGASE and the cellular homologue of ONCOGENE PROTEIN V-CBL.
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.
Established cell cultures that have the potential to propagate indefinitely.
Systems of enzymes which function sequentially by catalyzing consecutive reactions linked by common metabolic intermediates. They may involve simply a transfer of water molecules or hydrogen atoms and may be associated with large supramolecular structures such as MITOCHONDRIA or RIBOSOMES.
A cell line generated from human embryonic kidney cells that were transformed with human adenovirus type 5.
A group of acylated oligopeptides produced by Actinomycetes that function as protease inhibitors. They have been known to inhibit to varying degrees trypsin, plasmin, KALLIKREINS, papain and the cathepsins.
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.
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.
A family of structurally-related proteins that were originally identified by their ability to complex with cyclin proteins (CYCLINS). They share a common domain that binds specifically to F-BOX MOTIFS. They take part in SKP CULLIN F-BOX PROTEIN LIGASES, where they can bind to a variety of F-BOX PROTEINS.
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.
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.
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.
The ability of a protein to retain its structural conformation or its activity when subjected to physical or chemical manipulations.
The introduction of a phosphoryl group into a compound through the formation of an ester bond between the compound and a phosphorus moiety.
Compounds that inhibit the function or proteolytic action of the PROTEASOME.
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.
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.
A class of enzymes that catalyzes the ATP-dependent formation of a thioester bond between itself and UBIQUITIN. It then transfers the activated ubiquitin to one of the UBIQUITIN-PROTEIN LIGASES.
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.
An E3 ubiquitin ligase primarily involved in regulation of the metaphase-to-anaphase transition during MITOSIS through ubiquitination of specific CELL CYCLE PROTEINS. Enzyme activity is tightly regulated through subunits and cofactors, which modulate activation, inhibition, and substrate specificity. The anaphase-promoting complex, or APC-C, is also involved in tissue differentiation in the PLACENTA, CRYSTALLINE LENS, and SKELETAL MUSCLE, and in regulation of postmitotic NEURONAL PLASTICITY and excitability.
Cellular uptake of extracellular materials within membrane-limited vacuoles or microvesicles. ENDOSOMES play a central role in endocytosis.
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.
Cell surface receptors for AUTOCRINE MOTILITY FACTOR, which is the secreted form of GLUCOSE-6-PHOSPHATE ISOMERASE. The receptor has an unusual composition in that it shares some structural similarities with G-PROTEIN-COUPLED RECEPTORS and functions as an ubiquitin protein ligase when internalized.
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.
Proteins obtained from the species SACCHAROMYCES CEREVISIAE. The function of specific proteins from this organism are the subject of intense scientific interest and have been used to derive basic understanding of the functioning similar proteins in higher eukaryotes.
Proteins that control the CELL DIVISION CYCLE. This family of proteins includes a wide variety of classes, including CYCLIN-DEPENDENT KINASES, mitogen-activated kinases, CYCLINS, and PHOSPHOPROTEIN PHOSPHATASES as well as their putative substrates such as chromatin-associated proteins, CYTOSKELETAL PROTEINS, and TRANSCRIPTION FACTORS.
Endosomes containing intraluminal vesicles which are formed by the inward budding of the endosome membrane. Multivesicular bodies (MVBs) may fuse with other organelles such as LYSOSOMES or fuse back with the PLASMA MEMBRANE releasing their contents by EXOCYTOSIS. The MVB intraluminal vesicles released into the extracellular environment are known as EXOSOMES.
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 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.
Nuclear phosphoprotein encoded by the p53 gene (GENES, P53) whose normal function is to control CELL PROLIFERATION and APOPTOSIS. A mutant or absent p53 protein has been found in LEUKEMIA; OSTEOSARCOMA; LUNG CANCER; and COLORECTAL CANCER.
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.
Transport proteins that carry specific substances in the blood or across cell membranes.
Members of the peptidase C19 family which regulate signal transduction by removing UBIQUITIN from specific protein substrates via a process known as deubiquitination or deubiquitylation.
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.
A cell line derived from cultured tumor cells.
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.
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.
A type of POST-TRANSLATIONAL PROTEIN MODIFICATION by SMALL UBIQUITIN-RELATED MODIFIER PROTEINS (also known as SUMO proteins).
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 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.
Exogenous and endogenous compounds which inhibit CYSTEINE ENDOPEPTIDASES.
A class of morphologically heterogeneous cytoplasmic particles in animal and plant tissues characterized by their content of hydrolytic enzymes and the structure-linked latency of these enzymes. The intracellular functions of lysosomes depend on their lytic potential. The single unit membrane of the lysosome acts as a barrier between the enzymes enclosed in the lysosome and the external substrate. The activity of the enzymes contained in lysosomes is limited or nil unless the vesicle in which they are enclosed is ruptured. Such rupture is supposed to be under metabolic (hormonal) control. (From Rieger et al., Glossary of Genetics: Classical and Molecular, 5th ed)
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.
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.
A gene silencing phenomenon whereby specific dsRNAs (RNA, DOUBLE-STRANDED) trigger the degradation of homologous mRNA (RNA, MESSENGER). The specific dsRNAs are processed into SMALL INTERFERING RNA (siRNA) which serves as a guide for cleavage of the homologous mRNA in the RNA-INDUCED SILENCING COMPLEX. DNA METHYLATION may also be triggered during this process.
CELL LINES derived from the CV-1 cell line by transformation with a replication origin defective mutant of SV40 VIRUS, which codes for wild type large T antigen (ANTIGENS, POLYOMAVIRUS TRANSFORMING). They are used for transfection and cloning. (The CV-1 cell line was derived from the kidney of an adult male African green monkey (CERCOPITHECUS AETHIOPS).)
A subclass of PEPTIDE HYDROLASES that catalyze the internal cleavage of PEPTIDES or PROTEINS.
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.
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.
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 zinc-binding domain defined by the sequence Cysteine-X2-Cysteine-X(9-39)-Cysteine-X(l-3)-His-X(2-3)-Cysteine-X2-Cysteine -X(4-48)-Cysteine-X2-Cysteine, where X is any amino acid. The RING finger motif binds two atoms of zinc, with each zinc atom ligated tetrahedrally by either four cysteines or three cysteines and a histidine. The motif also forms into a unitary structure with a central cross-brace region and is found in many proteins that are involved in protein-protein interactions. The acronym RING stands for Really Interesting New Gene.
A 1.5-kDa small ubiquitin-related modifier protein that can covalently bind via an isopeptide link to a number of cellular proteins. It may play a role in intracellular protein transport and a number of other cellular processes.
Proteins involved in the transport of NUCLEOTIDES across cellular membranes.
Cytoplasmic vesicles formed when COATED VESICLES shed their CLATHRIN coat. Endosomes internalize macromolecules bound by receptors on the cell surface.
Endogenous substances, usually proteins, which are effective in the initiation, stimulation, or termination of the genetic transcription process.
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.
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.
Proteins that are normally involved in holding cellular growth in check. Deficiencies or abnormalities in these proteins may lead to unregulated cell growth and tumor development.
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.
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.
The degree of similarity between sequences of amino acids. This information is useful for the analyzing genetic relatedness of proteins and species.
Macromolecular complexes formed from the association of defined protein subunits.
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.
Injuries to DNA that introduce deviations from its normal, intact structure and which may, if left unrepaired, result in a MUTATION or a block of DNA REPLICATION. These deviations may be caused by physical or chemical agents and occur by natural or unnatural, introduced circumstances. They include the introduction of illegitimate bases during replication or by deamination or other modification of bases; the loss of a base from the DNA backbone leaving an abasic site; single-strand breaks; double strand breaks; and intrastrand (PYRIMIDINE DIMERS) or interstrand crosslinking. Damage can often be repaired (DNA REPAIR). If the damage is extensive, it can induce APOPTOSIS.
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.
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)
Small chromosomal proteins (approx 12-20 kD) possessing an open, unfolded structure and attached to the DNA in cell nuclei by ionic linkages. Classification into the various types (designated histone I, histone II, etc.) is based on the relative amounts of arginine and lysine in each.
Ligases that catalyze the joining of adjacent AMINO ACIDS by the formation of carbon-nitrogen bonds between their carboxylic acid groups and amine groups.
A system of cisternae in the CYTOPLASM of many cells. In places the endoplasmic reticulum is continuous with the plasma membrane (CELL MEMBRANE) or outer membrane of the nuclear envelope. If the outer surfaces of the endoplasmic reticulum membranes are coated with ribosomes, the endoplasmic reticulum is said to be rough-surfaced (ENDOPLASMIC RETICULUM, ROUGH); otherwise it is said to be smooth-surfaced (ENDOPLASMIC RETICULUM, SMOOTH). (King & Stansfield, A Dictionary of Genetics, 4th ed)
A non-essential amino acid occurring in natural form as the L-isomer. It is synthesized from GLYCINE or THREONINE. It is involved in the biosynthesis of PURINES; PYRIMIDINES; and other amino acids.
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.
A family of inhibitory proteins which bind to the REL PROTO-ONCOGENE PROTEINS and modulate their activity. In the CYTOPLASM, I-kappa B proteins bind to the transcription factor NF-KAPPA B. Cell stimulation causes its dissociation and translocation of active NF-kappa B to the nucleus.
The parts of a macromolecule that directly participate in its specific combination with another molecule.
A group of enzymes that catalyzes the phosphorylation of serine or threonine residues in proteins, with ATP or other nucleotides as phosphate donors.
A class of structurally related proteins of 12-20 kDa in size. They covalently modify specific proteins in a manner analogous to UBIQUITIN.
Proteins prepared by recombinant DNA technology.
A family of pheromone receptors that were initially discovered in SACCHAROMYCES CEREVISIAE as proteins necessary for fungal conjugation. Each mating factor receptor is expressed in HAPLOID CELLS of a single mating type.
A species of CERCOPITHECUS containing three subspecies: C. tantalus, C. pygerythrus, and C. sabeus. They are found in the forests and savannah of Africa. The African green monkey (C. pygerythrus) is the natural host of SIMIAN IMMUNODEFICIENCY VIRUS and is used in AIDS research.
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.
An oncoprotein from the Cas NS-1 murine retrovirus that induces pre- B-CELL LYMPHOMA and MYELOID LEUKEMIAS. v-cbl protein is a tyrosine-phosphorylated, truncated form of its cellular homologue, PROTO-ONCOGENE PROTEIN C-CBL.
Genetically engineered MUTAGENESIS at a specific site in the DNA molecule that introduces a base substitution, or an insertion or deletion.
The artificial induction of GENE SILENCING by the use of RNA INTERFERENCE to reduce the expression of a specific gene. It includes the use of DOUBLE-STRANDED RNA, such as SMALL INTERFERING RNA and RNA containing HAIRPIN LOOP SEQUENCE, and ANTI-SENSE OLIGONUCLEOTIDES.
A characteristic feature of enzyme activity in relation to the kind of substrate on which the enzyme or catalytic molecule reacts.
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.
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)
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.
A continuous cell line of high contact-inhibition established from NIH Swiss mouse embryo cultures. The cells are useful for DNA transfection and transformation studies. (From ATCC [Internet]. Virginia: American Type Culture Collection; c2002 [cited 2002 Sept 26]. Available from http://www.atcc.org/)
A ubiquitin-protein ligase that mediates OXYGEN-dependent polyubiquitination of HYPOXIA-INDUCIBLE FACTOR 1, ALPHA SUBUNIT. It is inactivated in VON HIPPEL-LINDAU SYNDROME.
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 degradation process whereby incorrectly folded proteins are selectively transported out of the ENDOPLASMIC RETICULUM and into the CYTOSOL. The misfolded proteins are subsequently ubiquitinated and degraded by the PROTEASOME.
Proteins produced from GENES that have acquired MUTATIONS.
The biosynthesis of RNA carried out on a template of DNA. The biosynthesis of DNA from an RNA template is called REVERSE TRANSCRIPTION.
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.
The complex series of phenomena, occurring between the end of one CELL DIVISION and the end of the next, by which cellular material is duplicated and then divided between two daughter cells. The cell cycle includes INTERPHASE, which includes G0 PHASE; G1 PHASE; S PHASE; and G2 PHASE, and CELL DIVISION PHASE.
Hydrolases that specifically cleave the peptide bonds found in PROTEINS and PEPTIDES. Examples of sub-subclasses for this group include EXOPEPTIDASES and ENDOPEPTIDASES.
Cell surface receptors that respond to PHEROMONES.
Nuclear antigen with a role in DNA synthesis, DNA repair, and cell cycle progression. PCNA is required for the coordinated synthesis of both leading and lagging strands at the replication fork during DNA replication. PCNA expression correlates with the proliferation activity of several malignant and non-malignant cell types.
Together with the Apc2 subunit, forms the catalytic core of the E3 ubiquitin ligase, anaphase-promoting complex-cyclosome. It has a RING H2 domain which interacts with the cullin domain of Apc2. Apc11 also interacts with the E2 ubiquitin ligases involved in APC-C ubiquitination reactions.
A conserved class of proteins that control APOPTOSIS in both VERTEBRATES and INVERTEBRATES. IAP proteins interact with and inhibit CASPASES, and they function as ANTI-APOPTOTIC PROTEINS. The protein class is defined by an approximately 80-amino acid motif called the baculoviral inhibitor of apoptosis repeat.
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.
The lipid- and protein-containing, selectively permeable membrane that surrounds the cytoplasm in prokaryotic and eukaryotic cells.
A class of MOLECULAR CHAPERONES whose members act in the mechanism of SIGNAL TRANSDUCTION by STEROID RECEPTORS.
Gated transport mechanisms by which proteins or RNA are moved across the NUCLEAR MEMBRANE.
Connective tissue cells which secrete an extracellular matrix rich in collagen and other macromolecules.
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.
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.
Regulatory proteins that down-regulate phosphorylated G-protein membrane receptors, including rod and cone photoreceptors and adrenergic receptors.
Proteins found in any species of fungus.
A casein kinase that was originally described as a monomeric enzyme with a molecular weight of 30-40 kDa. Several ISOENZYMES of casein kinase I have been found which are encoded by separate genes. Many of the casein kinase I isoenzymes have been shown to play distinctive roles in intracellular SIGNAL TRANSDUCTION.
Cdh1 is an activator of the anaphase-promoting complex-cyclosome, and is involved in substrate recognition. It associates with the complex in late MITOSIS from anaphase through G1 to regulate activity of CYCLIN-DEPENDENT KINASES and to prevent premature DNA replication.
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.
Mitogen-activated protein kinase kinase kinases (MAPKKKs) are serine-threonine protein kinases that initiate protein kinase signaling cascades. They phosphorylate MITOGEN-ACTIVATED PROTEIN KINASE KINASES; (MAPKKs) which in turn phosphorylate MITOGEN-ACTIVATED PROTEIN KINASES; (MAPKs).
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.
The largest subunit of the anaphase-promoting complex. It acts primarily as a scaffold for the proper organization and arrangement of subunits. The C-terminal region of Apc1 contains a series of tandem amino acid repeats that are also seen in the 26S proteasome regulatory particle, and may assist with forming and stabilizing protein-protein interactions.
The N-acetyl derivative of CYSTEINE. It is used as a mucolytic agent to reduce the viscosity of mucous secretions. It has also been shown to have antiviral effects in patients with HIV due to inhibition of viral stimulation by reactive oxygen intermediates.
Interruption or suppression of the expression of a gene at transcriptional or translational levels.
Highly conserved proteins that specifically bind to and activate the anaphase-promoting complex-cyclosome, promoting ubiquitination and proteolysis of cell-cycle-regulatory proteins. Cdc20 is essential for anaphase-promoting complex activity, initiation of anaphase, and cyclin proteolysis during mitosis.
Methods for determining interaction between PROTEINS.
Formation of an acetyl derivative. (Stedman, 25th ed)
Diffusible gene products that act on homologous or heterologous molecules of viral or cellular DNA to regulate the expression of proteins.
Elements of limited time intervals, contributing to particular results or situations.
A class of MOLECULAR CHAPERONES found in both prokaryotes and in several compartments of eukaryotic cells. These proteins can interact with polypeptides during a variety of assembly processes in such a way as to prevent the formation of nonfunctional structures.
Proteolytic breakdown of the MITOCHONDRIA.
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.
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)
A glycogen synthase kinase that was originally described as a key enzyme involved in glycogen metabolism. It regulates a diverse array of functions such as CELL DIVISION, microtubule function and APOPTOSIS.
The time it takes for a substance (drug, radioactive nuclide, or other) to lose half of its pharmacologic, physiologic, or radiologic activity.
Models used experimentally or theoretically to study molecular shape, electronic properties, or interactions; includes analogous molecules, computer-generated graphics, and mechanical structures.
An inhibitor of apoptosis protein that is translated by a rare cap-independent mechanism. It blocks caspase-mediated cellular destruction by inhibiting CASPASE 3; CASPASE 7; and CASPASE 9.
The simplest of all peptides. It functions as a gamma-glutamyl acceptor.
Processes that stimulate the GENETIC TRANSCRIPTION of a gene or set of genes.
A cyclin-dependent kinase inhibitor that coordinates the activation of CYCLIN and CYCLIN-DEPENDENT KINASES during the CELL CYCLE. It interacts with active CYCLIN D complexed to CYCLIN-DEPENDENT KINASE 4 in proliferating cells, while in arrested cells it binds and inhibits CYCLIN E complexed to CYCLIN-DEPENDENT KINASE 2.
Human COLORECTAL CARCINOMA cell line.
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.
The main structural coat protein of COATED VESICLES which play a key role in the intracellular transport between membranous organelles. Each molecule of clathrin consists of three light chains (CLATHRIN LIGHT CHAINS) and three heavy chains (CLATHRIN HEAVY CHAINS) that form a structure called a triskelion. Clathrin also interacts with cytoskeletal proteins.
A transferase that catalyzes the addition of aliphatic, aromatic, or heterocyclic FREE RADICALS as well as EPOXIDES and arene oxides to GLUTATHIONE. Addition takes place at the SULFUR. It also catalyzes the reduction of polyol nitrate by glutathione to polyol and nitrite.
'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.
A family of intracellular signaling adaptor proteins that contain caspase activation and recruitment domains. Proteins that contain this domain play a role in APOPTOSIS-related signal transduction by associating with other CARD domain-containing members and in activating INITIATOR CASPASES that contain CARD domains within their N-terminal pro-domain region.
A type of CELL NUCLEUS division by means of which the two daughter nuclei normally receive identical complements of the number of CHROMOSOMES of the somatic cells of the species.
A multi-functional catenin that participates in CELL ADHESION and nuclear signaling. Beta catenin binds CADHERINS and helps link their cytoplasmic tails to the ACTIN in the CYTOSKELETON via ALPHA CATENIN. It also serves as a transcriptional co-activator and downstream component of WNT PROTEIN-mediated SIGNAL TRANSDUCTION PATHWAYS.
A mutation in which a codon is mutated to one directing the incorporation of a different amino acid. This substitution may result in an inactive or unstable product. (From A Dictionary of Genetics, King & Stansfield, 5th ed)
The process of cleaving a chemical compound by the addition of a molecule of water.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
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.
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.
That portion of the electromagnetic spectrum immediately below the visible range and extending into the x-ray frequencies. The longer wavelengths (near-UV or biotic or vital rays) are necessary for the endogenous synthesis of vitamin D and are also called antirachitic rays; the shorter, ionizing wavelengths (far-UV or abiotic or extravital rays) are viricidal, bactericidal, mutagenic, and carcinogenic and are used as disinfectants.
The phase of cell nucleus division following METAPHASE, in which the CHROMATIDS separate and migrate to opposite poles of the spindle.
Any spaces or cavities within a cell. They may function in digestion, storage, secretion, or excretion.
All of the processes involved in increasing CELL NUMBER including CELL DIVISION.
A family of histone acetyltransferases that is structurally-related to CREB-BINDING PROTEIN and to E1A-ASSOCIATED P300 PROTEIN. They function as transcriptional coactivators by bridging between DNA-binding TRANSCRIPTION FACTORS and the basal transcription machinery. They also modify transcription factors and CHROMATIN through ACETYLATION.
An essential amino acid occurring naturally in the L-form, which is the active form. It is found in eggs, milk, gelatin, and other proteins.
A family of structurally related proteins that are constitutively expressed and that negatively regulate cytokine-mediated SIGNAL TRANSDUCTION PATHWAYS. PIAS proteins inhibit the activity of signal transducers and activators of transcription.
A multisubunit polycomb protein complex with affinity for CHROMATIN that contains methylated HISTONE H3. It contains an E3 ubiquitin ligase activity that is specific for HISTONE H2A and works in conjunction with POLYCOMB REPRESSIVE COMPLEX 2 to effect EPIGENETIC REPRESSION.

Influence of TRP53 status on FAS membrane localization, CFLAR (c-FLIP) ubiquitinylation, and sensitivity of GC-2spd (ts) cells to undergo FAS-mediated apoptosis. (1/3688)

Previously we reported that testicular germ cells undergo FAS-mediated apoptosis after exposure of mice to the Sertoli cell toxicant mono-(2-ethylhexyl) phthalate (MEHP) and that this process is partially dependent on the TRP53 protein (p53). Recent reports have suggested that TRP53 may influence the ubiquitinylation and consequent proteosomal degradation of a negative regulator of FAS, CFLAR (L) (c-FLIP [L]), in human colon cancer cells. To further characterize the relationship between CFLAR and TRP53, we used the transformed germ cell line GC-2spd (ts), which harbors a temperature-sensitive Trp53 mutation that allows for TRP53 activation at 32 degrees C. We report here that GC-2 cells expressed a 10-fold increase in basal cell membrane FAS levels and an increased sensitivity to FAS agonistic antibody (JO2)-triggered apoptosis only when they were maintained at the permissive TRP53 temperature. After JO2 exposure, CFLAR (L) protein levels were enhanced only at the nonpermissive TRP53 temperature (37 degrees C) while real-time PCR results indicated an absence of Cflar (L) mRNA changes in GC-2 cells regardless of the temperature. Furthermore, transfection of GC-2 cells at 37 degrees C with siRNA against Cflar resulted in reduction of CFLAR (L) protein levels and increased sensitivity to JO2-mediated apoptosis. The CFLAR (L) protein was also more strongly ubiquitinylated in response to JO2 treatment at the permissive TRP53 temperature. Taken together, these data suggest that the TRP53 protein influences the sensitivity of GC-2 cells to undergo FAS-mediated apoptosis by modulating the expression of FAS on their cell membranes and subsequently influencing the degradation of the antiapoptotic protein CFLAR (L).  (+info)

Mutations within the membrane domain of HMG-CoA reductase confer resistance to sterol-accelerated degradation. (2/3688)

The pivotal event for sterol-induced degradation of the cholesterol biosynthetic enzyme HMG-CoA reductase is binding of its membrane domain to Insig proteins in the endoplasmic reticulum. Insigs are carriers for gp78, an E3 ubiquitin ligase that marks reductase for proteasomal degradation. We report here the isolation of mutant Chinese hamster ovary cell lines, designated SRD-16, -17, and -18, in which sterol-induced ubiquitination and degradation of reductase are severely impaired. These cells were produced by chemical mutagenesis and selection with SR-12813, a compound that mimics sterols in stimulating ubiquitination and degradation of reductase. Each SRD cell line was found to contain a point mutation in one reductase allele, resulting in substitutions of aspartate for serine-60 (SRD-16), arginine for glycine-87 (SRD-17), and proline for alanine-333 (SRD-18). Sterols failed to promote ubiquitination and degradation of these reductase mutants, owing to their decreased affinity for Insigs. Thus, three different point mutations in reductase, all of which localize to the membrane domain, disrupt Insig binding and abolish sterol-accelerated degradation of the enzyme.  (+info)

The dark side of EGFP: defective polyubiquitination. (3/3688)

Enhanced Green Fluorescent Protein (EGFP) is the most commonly used live cell reporter despite a number of conflicting reports that it can affect cell physiology. Thus far, the precise mechanism of GFP-associated defects remained unclear. Here we demonstrate that EGFP and EGFP fusion proteins inhibit polyubiquitination, a posttranslational modification that controls a wide variety of cellular processes, like activation of kinase signalling or protein degradation by the proteasome. As a consequence, the NF-kappaB and JNK signalling pathways are less responsive to activation, and the stability of the p53 tumour suppressor is enhanced in cell lines and in vivo. In view of the emerging role of polyubiquitination in the regulation of numerous cellular processes, the use of EGFP as a live cell reporter should be carefully considered.  (+info)

Gene-specific selection against experimental fanconi anemia gene inactivation in human cancer. (4/3688)

The Fanconi anemia (FA) gene family comprises at least 12 genes interacting in a common pathway involved in DNA repair. To gain insight into the role of FA gene inactivation occurring in tumors among the general population, we endogenously targeted in cancer cells four FA genes that act at different stages of the FA pathway. After successful mono-allelic deletion of all genes, the sequential homozygous deletion was achieved only for FANCC and FANCG, acting upstream, but not for BRCA2 or FANCD2, acting downstream in the FA pathway. Targeting of the second allele in in BRCA2 and FANCD2 heterozygote clones resulted in redeletion exclusively of the already defective allele in multiple instances (13x concerning BRCA2, 25x concerning FANCD2), strongly suggesting a detrimental phenotype. Unlike complete FANCD2 disruption, the mere reduction of FANCD2 protein levels had no discernible effect. In addition, we confirmed that human cancer cells harboring the Seckel ATR mutation display impaired FANCD2 monoubiquitination and FANCD2 nuclear focus formation, as well as an increased sensitivity to DNA interstrand-crosslinking agents. Nevertheless, these cells were viable, indicating an ATR-independent function of FANCD2, distinct from its major known functions, to be responsible for the detrimental effects of FANCD2 loss. In conclusion, we established the downstream FA genes FANCD2 and BRCA2 to represent particularly vulnerable parts of the FA pathway, providing direct evidence for the paradoxical assumption that their inactivation could be predominantly selected against in cancer cells. This would explain why certain FA gene defects, despite an apparent selection for FA pathway inactivation in cancer, are rarely observed in tumors among the general population.  (+info)

Role of ubiquitination in IGF-1 receptor signaling and degradation. (5/3688)

BACKGROUND: The insulin-like growth factor 1 receptor (IGF-1R) plays numerous crucial roles in cancer biology. The majority of knowledge on IGF-1R signaling is concerned with its role in the activation of the canonical phosphatidyl inositol-3 kinase (PI3K)/Akt and MAPK/ERK pathways. However, the role of IGF-1R ubiquitination in modulating IGF-1R function is an area of current research. In light of this we sought to determine the relationship between IGF-1R phosphorylation, ubiquitination, and modulation of growth signals. METHODOLOGY: Wild type and mutant constructs of IGF-1R were transfected into IGF-1R null fibroblasts. IGF-1R autophosphorylation and ubiquitination were determined by immunoprecipitation and western blotting. IGF-1R degradation and stability was determined by cyclohexamide-chase assay in combination with lysosome and proteasome inhibitors. PRINCIPAL FINDINGS: IGF-1R autophosphorylation was found to be an absolute requirement for receptor ubiquitination. Deletion of C-terminal domain had minimal effect on IGF-1 induced receptor autophosphorylation, however, ubiquitination and ERK activation were completely abolished. Cells expressing kinase impaired IGF-1R, exhibited both receptor ubiquitination and ERK phosphorylation, however failed to activate Akt. While IGF-1R mutants with impaired PI3K/Akt signaling were degraded mainly by the proteasomes, the C-terminal truncated one was exclusively degraded through the lysosomal pathway. CONCLUSIONS: Our data suggest important roles of ubiquitination in mediating IGF-1R signaling and degradation. Ubiquitination of IGF-1R requires receptor tyrosine kinase activity, but is not involved in Akt activation. In addition we show that the C-terminal domain of IGF-1R is a necessary requisite for ubiquitination and ERK phosphorylation as well as for proteasomal degradation of the receptor.  (+info)

ElaD, a Deubiquitinating protease expressed by E. coli. (6/3688)

BACKGROUND: Ubiquitin and ubiquitin-like proteins (Ubl) are designed to modify polypeptides in eukaryotes. Covalent binding of ubiquitin or Ubls to substrate proteins can be reversed by specific hydrolases. One particular set of cysteine proteases, the CE clan, which targets ubiquitin and Ubls, has homologs in eukaryotes, prokaryotes, and viruses. FINDINGS: We have cloned and analyzed the E. coli protein elaD, which is distantly related to eukaryotic CE clan members of the ULP/SENP protease family that are specific for SUMO and Nedd8. Previously misannotated as a putative sulfatase/phosphatase, elaD is an efficient and specific deubiquitinating enzyme in vitro. Interestingly, elaD is present in all intestinal pathogenic E. coli strains, but conspicuously absent from extraintestinal pathogenic strains (ExPECs). Further homologs of this protease can be found in Acanthamoeba Polyphaga Mimivirus, and in Alpha-, Beta-and Gammaproteobacteria. CONCLUSION: The expression of ULP/SENP-related hydrolases in bacteria therefore extends to plant pathogens and medically relevant strains of Escherichia coli, Legionella pneumophila, Rickettsiae, Chlamydiae, and Salmonellae, in which the elaD ortholog sseL has recently been identified as a virulence factor with deubiquitinating activity. As a counterpoint, our phylogenetic and functional examination reveals that ancient eukaryotic ULP/SENP proteases also have the potential of ubiquitin-specific hydrolysis, suggesting an early common origin of this peptidase clan.  (+info)

Biogenesis and function of multivesicular bodies. (7/3688)

The two major cellular sites for membrane protein degradation are the proteasome and the lysosome. Ubiquitin attachment is a sorting signal for both degradation routes. For lysosomal degradation, ubiquitination triggers the sorting of cargo proteins into the lumen of late endosomal multivesicular bodies (MVBs)/endosomes. MVB formation occurs when a portion of the limiting membrane of an endosome invaginates and buds into its own lumen. Intralumenal vesicles are degraded when MVBs fuse to lysosomes. The proper delivery of proteins to the MVB interior relies on specific ubiquitination of cargo, recognition and sorting of ubiquitinated cargo to endosomal subdomains, and the formation and scission of cargo-filled intralumenal vesicles. Over the past five years, a number of proteins that may directly participate in these aspects of MVB function and biogenesis have been identified. However, major questions remain as to exactly what these proteins do at the molecular level and how they may accomplish these tasks.  (+info)

Ubiquitin-mediated degradation of Rpn4 is controlled by a phosphorylation-dependent ubiquitylation signal. (8/3688)

A ubiquitylation signal of a protein substrate is defined as a short primary sequence or a structural feature recognized by a specific E3. Our previous work has mapped the ubiquitylation signal of Rpn4, the transcription activator for the Saccharomyces cerevisiae proteasome genes, to an N-terminal acidic domain (NAD) consisting of amino acids 211-229. However, the molecular mechanism by which Ubr2, the cognate E3, recognizes NAD remains unclear. Here we show that phosphorylation of either Ser-214 or Ser-220 enhances the binding of NAD to Ubr2. However, phosphorylation of Ser-220 but not Ser-214 plays a predominant role in Rpn4 ubiquitylation and degradation. Interestingly, NAD does not constitute the major Ubr2-binding site of Rpn4 even though it serves as the ubiquitylation signal essential for Rpn4 degradation. By contrast, the stable binding with Ubr2 conferred by other domains of Rpn4 is not required for Rpn4 degradation. Our results indicate that ubiquitin-mediated degradation of Rpn4 is controlled by a phosphorylation-dependent ubiquitylation signal. This study also suggests that binding to E3 may be only a part of the function of a ubiquitylation signal.  (+info)

Ubiquitination is a post-translational modification process in which a ubiquitin protein is covalently attached to a target protein. This process plays a crucial role in regulating various cellular functions, including protein degradation, DNA repair, and signal transduction. The addition of ubiquitin can lead to different outcomes depending on the number and location of ubiquitin molecules attached to the target protein. Monoubiquitination (the attachment of a single ubiquitin molecule) or multiubiquitination (the attachment of multiple ubiquitin molecules) can mark proteins for degradation by the 26S proteasome, while specific types of ubiquitination (e.g., K63-linked polyubiquitination) can serve as a signal for nonproteolytic functions such as endocytosis, autophagy, or DNA repair. Ubiquitination is a highly regulated process that involves the coordinated action of three enzymes: E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme, and E3 ubiquitin ligase. Dysregulation of ubiquitination has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

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.

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.

Ubiquitin is a small protein that is present in most tissues in the body. It plays a critical role in regulating many important cellular processes, such as protein degradation and DNA repair. Ubiquitin can attach to other proteins in a process called ubiquitination, which can target the protein for degradation or modify its function.

Ubiquitination involves a series of enzymatic reactions that ultimately result in the attachment of ubiquitin molecules to specific lysine residues on the target protein. The addition of a single ubiquitin molecule is called monoubiquitination, while the addition of multiple ubiquitin molecules is called polyubiquitination.

Polyubiquitination can serve as a signal for proteasomal degradation, where the target protein is broken down into its component amino acids by the 26S proteasome complex. Monoubiquitination and other forms of ubiquitination can also regulate various cellular processes, such as endocytosis, DNA repair, and gene expression.

Dysregulation of ubiquitin-mediated protein degradation has been implicated in a variety of diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

The proteasome endopeptidase complex is a large protein complex found in the cells of eukaryotic organisms, as well as in archaea and some bacteria. It plays a crucial role in the degradation of damaged or unneeded proteins through a process called proteolysis. The proteasome complex contains multiple subunits, including both regulatory and catalytic particles.

The catalytic core of the proteasome is composed of four stacked rings, each containing seven subunits, forming a structure known as the 20S core particle. Three of these rings are made up of beta-subunits that contain the proteolytic active sites, while the fourth ring consists of alpha-subunits that control access to the interior of the complex.

The regulatory particles, called 19S or 11S regulators, cap the ends of the 20S core particle and are responsible for recognizing, unfolding, and translocating targeted proteins into the catalytic chamber. The proteasome endopeptidase complex can cleave peptide bonds in various ways, including hydrolysis of ubiquitinated proteins, which is an essential mechanism for maintaining protein quality control and regulating numerous cellular processes, such as cell cycle progression, signal transduction, and stress response.

In summary, the proteasome endopeptidase complex is a crucial intracellular machinery responsible for targeted protein degradation through proteolysis, contributing to various essential regulatory functions in cells.

Ubiquitin-conjugating enzymes (UBCs or E2 enzymes) are a family of enzymes that play a crucial role in the ubiquitination process, which is a post-translational modification of proteins. This process involves the covalent attachment of the protein ubiquitin to specific lysine residues on target proteins, ultimately leading to their degradation by the 26S proteasome.

Ubiquitination is a multi-step process that requires the coordinated action of three types of enzymes: E1 (ubiquitin-activating), E2 (ubiquitin-conjugating), and E3 (ubiquitin ligases). Ubiquitin-conjugating enzymes are responsible for transferring ubiquitin from the E1 enzyme to the target protein, which is facilitated by an E3 ubiquitin ligase. The human genome encodes around 40 different UBCs, each with unique substrate specificities and functions in various cellular processes, such as protein degradation, DNA repair, and signal transduction.

Ubiquitination is a highly regulated process that can be reversed by the action of deubiquitinating enzymes (DUBs), which remove ubiquitin molecules from target proteins. Dysregulation of the ubiquitination pathway has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

Endosomal Sorting Complexes Required for Transport (ESCRT) are a set of protein complexes found in the endosomal membrane of eukaryotic cells. They play a crucial role in the sorting and trafficking of proteins and lipids between various cellular compartments, particularly in the formation of vesicles and the budding of viruses.

The ESCRT system is composed of several distinct complexes (ESCRT-0, -I, -II, and -III) that work together in a coordinated manner to carry out their functions. ESCRT-0 recognizes and binds to ubiquitinated proteins on the endosomal membrane, initiating the sorting process. ESCRT-I and -II then help to deform the membrane and recruit ESCRT-III, which forms a tight spiral around the neck of the budding vesicle. Finally, the AAA+ ATPase Vps4 disassembles the ESCRT-III complex, allowing for the release of the vesicle into the lumen of the endosome or extracellular space.

Defects in the ESCRT system have been linked to a variety of human diseases, including neurological disorders, cancer, and viral infections.

Ligases are a group of enzymes that catalyze the formation of a covalent bond between two molecules, usually involving the joining of two nucleotides in a DNA or RNA strand. They play a crucial role in various biological processes such as DNA replication, repair, and recombination. In DNA ligases, the enzyme seals nicks or breaks in the phosphodiester backbone of the DNA molecule by catalyzing the formation of an ester bond between the 3'-hydroxyl group and the 5'-phosphate group of adjacent nucleotides. This process is essential for maintaining genomic integrity and stability.

Lysine is an essential amino acid, which means that it cannot be synthesized by the human body and must be obtained through the diet. Its chemical formula is (2S)-2,6-diaminohexanoic acid. Lysine is necessary for the growth and maintenance of tissues in the body, and it plays a crucial role in the production of enzymes, hormones, and antibodies. It is also essential for the absorption of calcium and the formation of collagen, which is an important component of bones and connective tissue. Foods that are good sources of lysine include meat, poultry, fish, eggs, and dairy products.

Proteolysis is the biological process of breaking down proteins into smaller polypeptides or individual amino acids by the action of enzymes called proteases. This process is essential for various physiological functions, including digestion, protein catabolism, cell signaling, and regulation of numerous biological activities. Dysregulation of proteolysis can contribute to several pathological conditions, such as cancer, neurodegenerative diseases, and inflammatory disorders.

Cullin proteins are a family of structurally related proteins that play a crucial role in the function of E3 ubiquitin ligase complexes. These complexes are responsible for targeting specific cellular proteins for degradation by the proteasome, which is a key process in maintaining protein homeostasis within cells.

Cullin proteins act as scaffolds that bring together different components of the E3 ubiquitin ligase complex, including RING finger proteins and substrate receptors. There are several different cullin proteins identified in humans (CUL1, CUL2, CUL3, CUL4A, CUL4B, CUL5, and CUL7), each of which can form distinct E3 ubiquitin ligase complexes with unique substrate specificities.

The regulation of cullin proteins is critical for normal cellular function, and dysregulation of these proteins has been implicated in various diseases, including cancer. For example, mutations in CUL1 have been found in certain types of breast and ovarian cancers, while alterations in CUL3 have been linked to neurodegenerative disorders such as Parkinson's disease.

Overall, cullin proteins are essential components of the ubiquitin-proteasome system, which plays a critical role in regulating protein turnover and maintaining cellular homeostasis.

Ubiquitinated proteins are proteins that have been marked for degradation through the covalent attachment of ubiquitin molecules. Ubiquitination is a post-translational modification process that involves the activation of ubiquitin by an E1 activating enzyme, its transfer to an E2 conjugating enzyme, and finally its attachment to a lysine residue on the target protein by an E3 ligase enzyme.

The addition of a single ubiquitin molecule can alter the function or localization of a protein, while the attachment of a polyubiquitin chain (a chain of multiple ubiquitin molecules) typically targets the protein for degradation by the 26S proteasome. Ubiquitination plays a crucial role in regulating various cellular processes, including protein quality control, DNA repair, and cell signaling.

Abnormalities in ubiquitination have been implicated in several diseases, including neurodegenerative disorders, cancer, and inflammatory conditions. Therefore, understanding the mechanisms of ubiquitination and developing strategies to modulate this process has important therapeutic potential.

Polyubiquitin refers to the formation of chains of ubiquitin molecules that are attached to a protein substrate. Ubiquitination is a post-translational modification where ubiquitin, a small regulatory protein, is covalently attached to lysine residues on target proteins. When multiple ubiquitin molecules are linked together through their C-terminal glycine residue to one of the seven lysine residues (K6, K11, K27, K29, K33, K48, or K63) on another ubiquitin molecule, it results in the formation of polyubiquitin chains.

Different types of polyubiquitination chains have distinct functions within the cell. For instance, K48-linked polyubiquitin chains typically target proteins for proteasomal degradation, while K63-linked polyubiquitin chains are involved in various signaling pathways, including DNA damage response, endocytosis, and inflammation.

Polyubiquitination is a dynamic process that can be reversed by the action of deubiquitinating enzymes (DUBs), which cleave ubiquitin chains from substrate proteins or disassemble polyubiquitin chains into individual ubiquitin molecules. Dysregulation of polyubiquitination and deubiquitination processes has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

Ubiquitin-Protein Ligase Complexes, 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 altering their function, localization, or interaction with other proteins.

The ubiquitination process involves three main steps:

1. Ubiquitin activation: Ubiquitin is activated by an E1 ubiquitin-activating enzyme in an ATP-dependent reaction.
2. Ubiquitin conjugation: The activated ubiquitin is then transferred to an E2 ubiquitin-conjugating enzyme.
3. Ubiquitin ligation: Finally, the E2 ubiquitin-conjugating enzyme interacts with a specific E3 ubiquitin ligase complex, which facilitates the transfer and ligation of ubiquitin to the target protein.

Ubiquitin-Protein Ligase Complexes are responsible for recognizing and binding to specific substrate proteins, ensuring that ubiquitination occurs on the correct targets. They can be divided into three main categories based on their structural features and mechanisms of action:

1. Really Interesting New Gene (RING) finger E3 ligases: These E3 ligases contain a RING finger domain, which directly interacts with both the E2 ubiquitin-conjugating enzyme and the substrate protein. They facilitate the transfer of ubiquitin from the E2 to the target protein by bringing them into close proximity.
2. Homologous to E6-AP C terminus (HECT) E3 ligases: These E3 ligases contain a HECT domain, which interacts with the E2 ubiquitin-conjugating enzyme and forms a thioester bond with ubiquitin before transferring it to the substrate protein.
3. RING-between-RING (RBR) E3 ligases: These E3 ligases contain both RING finger and HECT-like domains, which allow them to function similarly to both RING finger and HECT E3 ligases. They first form a thioester bond with ubiquitin using their RING1 domain before transferring it to the substrate protein via their RING2 domain.

Dysregulation of Ubiquitin-Protein Ligase Complexes has been implicated in various diseases, including cancer and neurodegenerative disorders. Understanding their mechanisms and functions can provide valuable insights into disease pathogenesis and potential therapeutic strategies.

Proto-oncogene proteins, such as c-MDM2, are normal cellular proteins that play crucial roles in regulating various cellular processes, including cell growth, differentiation, and apoptosis (programmed cell death). When these genes undergo mutations or are overexpressed, they can become oncogenes, which contribute to the development of cancer.

The c-MDM2 protein is a key regulator of the cell cycle and is involved in the negative regulation of the tumor suppressor protein p53. Under normal conditions, p53 helps prevent the formation of tumors by inducing cell cycle arrest or apoptosis in response to DNA damage or other stress signals. However, when c-MDM2 is overexpressed or mutated, it can bind and inhibit p53, leading to uncontrolled cell growth and increased risk of cancer development.

In summary, proto-oncogene proteins like c-MDM2 are important regulators of normal cellular processes, but when they become dysregulated through mutations or overexpression, they can contribute to the formation of tumors and cancer progression.

I'm sorry for any confusion, but "Ubiquitin Thiolesterase" is not a widely recognized medical term or a well-defined concept in the field of medicine. Ubiquitination, however, is a post-translational modification that plays a crucial role in various cellular processes, including protein degradation and regulation of signaling pathways.

Ubiquitin Thiolesterase could potentially refer to an enzyme that catalyzes the hydrolysis of a thioester bond between ubiquitin and a target protein. This process would be part of the ubiquitination cascade, where ubiquitin is transferred from one protein to another through various intermediates, including thioester bonds. However, I would recommend consulting primary literature or speaking with an expert in the field for more precise information on this topic.

SKP (S-phase kinase associated protein) Cullin F-box protein ligases, also known as SCF complexes, are a type of E3 ubiquitin ligase that play a crucial role in the ubiquitination and subsequent degradation of proteins. These complexes are composed of several subunits: SKP1, Cul1 (Cullin 1), Rbx1 (Ring-box 1), and an F-box protein. The F-box protein is a variable component that determines the substrate specificity of the SCF complex.

The ubiquitination process mediated by SCF complexes involves the sequential transfer of ubiquitin molecules to a target protein, leading to its degradation by the 26S proteasome. This pathway is essential for various cellular processes, including cell cycle regulation, signal transduction, and DNA damage response.

Dysregulation of SCF complexes has been implicated in several diseases, such as cancer and neurodegenerative disorders, making them potential targets for therapeutic intervention.

Beta-transducin repeat-containing proteins (β-TrCP) are a group of proteins that are involved in the regulation of various cellular processes, including protein degradation and signal transduction. They are named after their structural similarity to the beta subunit of transducin, a G protein that plays a role in visual signaling.

β-TrCP proteins contain multiple repeats of a specific motif known as a WD40 domain, which is involved in protein-protein interactions. They function as substrate recognition components of an E3 ubiquitin ligase complex, which targets specific proteins for degradation by the proteasome.

One well-studied function of β-TrCP is its role in the regulation of the cell cycle and DNA damage response. It recognizes and binds to phosphorylated forms of certain proteins, leading to their ubiquitination and subsequent degradation. This helps to ensure proper progression through the cell cycle and prevents the accumulation of damaged or mutated proteins that could lead to cancer or other diseases.

Other functions of β-TrCP include regulating gene transcription, modulating immune responses, and controlling cell survival and death pathways. Dysregulation of β-TrCP has been implicated in various human diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

F-box proteins are a family of proteins that are characterized by the presence of an F-box domain, which is a motif of about 40-50 amino acids. This domain is responsible for binding to Skp1, a component of the SCF (Skp1-Cul1-F-box protein) E3 ubiquitin ligase complex. The F-box proteins serve as the substrate recognition subunit of this complex and are involved in targeting specific proteins for ubiquitination and subsequent degradation by the 26S proteasome.

There are multiple types of F-box proteins, including FBXW (also known as β-TrCP), FBXL, and FBLX, each with different substrate specificities. These proteins play important roles in various cellular processes such as cell cycle regulation, signal transduction, and DNA damage response by controlling the stability of key regulatory proteins.

Abnormal regulation of F-box proteins has been implicated in several human diseases, including cancer, developmental disorders, and neurodegenerative diseases.

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.

Post-translational protein processing refers to the modifications and changes that proteins undergo after their synthesis on ribosomes, which are complex molecular machines responsible for protein synthesis. These modifications occur through various biochemical processes and play a crucial role in determining the final structure, function, and stability of the protein.

The process begins with the translation of messenger RNA (mRNA) into a linear polypeptide chain, which is then subjected to several post-translational modifications. These modifications can include:

1. Proteolytic cleavage: The removal of specific segments or domains from the polypeptide chain by proteases, resulting in the formation of mature, functional protein subunits.
2. Chemical modifications: Addition or modification of chemical groups to the side chains of amino acids, such as phosphorylation (addition of a phosphate group), glycosylation (addition of sugar moieties), methylation (addition of a methyl group), acetylation (addition of an acetyl group), and ubiquitination (addition of a ubiquitin protein).
3. Disulfide bond formation: The oxidation of specific cysteine residues within the polypeptide chain, leading to the formation of disulfide bonds between them. This process helps stabilize the three-dimensional structure of proteins, particularly in extracellular environments.
4. Folding and assembly: The acquisition of a specific three-dimensional conformation by the polypeptide chain, which is essential for its function. Chaperone proteins assist in this process to ensure proper folding and prevent aggregation.
5. Protein targeting: The directed transport of proteins to their appropriate cellular locations, such as the nucleus, mitochondria, endoplasmic reticulum, or plasma membrane. This is often facilitated by specific signal sequences within the protein that are recognized and bound by transport machinery.

Collectively, these post-translational modifications contribute to the functional diversity of proteins in living organisms, allowing them to perform a wide range of cellular processes, including signaling, catalysis, regulation, and structural support.

Proto-oncogene proteins c-cbl are a group of E3 ubiquitin ligases that play crucial roles in regulating various cellular processes, including cell survival, proliferation, differentiation, and migration. The c-cbl gene encodes for the c-Cbl protein, which is a member of the Cbl family of proteins that also includes Cbl-b and Cbl-c.

The c-Cbl protein contains several functional domains, including an N-terminal tyrosine kinase binding domain, a RING finger domain, a proline-rich region, and a C-terminal ubiquitin association domain. These domains enable c-Cbl to interact with various signaling molecules, such as receptor tyrosine kinases (RTKs), G protein-coupled receptors (GPCRs), and growth factor receptors, and regulate their activity through ubiquitination.

Ubiquitination is a post-translational modification that involves the addition of ubiquitin molecules to proteins, leading to their degradation or altered function. c-Cbl functions as an E3 ubiquitin ligase, which catalyzes the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to a specific target protein.

Proto-oncogene proteins c-cbl can act as tumor suppressors by negatively regulating signaling pathways that promote cell growth and survival. Mutations in the c-cbl gene or dysregulation of c-Cbl function have been implicated in various types of cancer, including leukemia, lymphoma, and solid tumors. These mutations can lead to increased RTK signaling, enhanced cell proliferation, and decreased apoptosis, contributing to tumor development and progression.

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.

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.

Multienzyme complexes are specialized protein structures that consist of multiple enzymes closely associated or bound together, often with other cofactors and regulatory subunits. These complexes facilitate the sequential transfer of substrates along a series of enzymatic reactions, also known as a metabolic pathway. By keeping the enzymes in close proximity, multienzyme complexes enhance reaction efficiency, improve substrate specificity, and maintain proper stoichiometry between different enzymes involved in the pathway. Examples of multienzyme complexes include the pyruvate dehydrogenase complex, the citrate synthase complex, and the fatty acid synthetase complex.

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.

Leupeptins are a type of protease inhibitors, which are substances that can inhibit the activity of enzymes called proteases. Proteases play a crucial role in breaking down proteins into smaller peptides or individual amino acids. Leupeptins are naturally occurring compounds found in some types of bacteria and are often used in laboratory research to study various cellular processes that involve protease activity.

Leupeptins can inhibit several different types of proteases, including serine proteases, cysteine proteases, and some metalloproteinases. They work by binding to the active site of these enzymes and preventing them from cleaving their protein substrates. Leupeptins have been used in various research applications, such as studying protein degradation, signal transduction pathways, and cell death mechanisms.

It is important to note that leupeptins are not typically used as therapeutic agents in clinical medicine due to their potential toxicity and lack of specificity for individual proteases. Instead, they are primarily used as research tools in basic science investigations.

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.

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.

S-phase kinase-associated proteins (Skp2) are a group of proteins that are associated with the S-phase kinase, which is a type of enzyme that helps to regulate the cell cycle. Specifically, Skp2 is involved in the ubiquitination and degradation of certain proteins that play a role in controlling the progression of the cell cycle.

Skp2 is a member of the F-box protein family, which are components of the Skp1-Cul1-F-box (SCF) complex, a type of E3 ubiquitin ligase. The SCF complex recognizes and binds to specific proteins, tagging them for ubiquitination and subsequent degradation by the proteasome.

One of the key targets of Skp2 is the tumor suppressor protein p27, which inhibits the activity of cyclin-dependent kinases (CDKs) and helps to regulate the transition from the G1 phase to the S phase of the cell cycle. By targeting p27 for degradation, Skp2 promotes the progression of the cell cycle and has been implicated in the development of various types of cancer.

Overall, Skp2 plays a critical role in regulating the cell cycle and has important implications for the development and treatment of various diseases, including cancer.

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.

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.

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.

Protein stability refers to the ability of a protein to maintain its native structure and function under various physiological conditions. It is determined by the balance between forces that promote a stable conformation, such as intramolecular interactions (hydrogen bonds, van der Waals forces, and hydrophobic effects), and those that destabilize it, such as thermal motion, chemical denaturation, and environmental factors like pH and salt concentration. A protein with high stability is more resistant to changes in its structure and function, even under harsh conditions, while a protein with low stability is more prone to unfolding or aggregation, which can lead to loss of function or disease states, such as protein misfolding 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.

Proteasome inhibitors are a class of medications that work by blocking the action of proteasomes, which are protein complexes that play a critical role in the breakdown and recycling of damaged or unwanted proteins within cells. By inhibiting the activity of these proteasomes, proteasome inhibitors can cause an accumulation of abnormal proteins within cells, leading to cell death.

This effect is particularly useful in the treatment of certain types of cancer, such as multiple myeloma and mantle cell lymphoma, where malignant cells often have an overproduction of abnormal proteins that can be targeted by proteasome inhibitors. The three main proteasome inhibitors currently approved for use in cancer therapy are bortezomib (Velcade), carfilzomib (Kyprolis), and ixazomib (Ninlaro). These medications have been shown to improve outcomes and extend survival in patients with these types of cancers.

It's important to note that proteasome inhibitors can also have off-target effects on other cells in the body, leading to side effects such as neurotoxicity, gastrointestinal symptoms, and hematologic toxicities. Therefore, careful monitoring and management of these side effects is necessary during treatment with proteasome inhibitors.

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.

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.

Ubiquitin-activating enzymes, also known as E1 enzymes, are a class of enzymes that play a crucial role in the ubiquitination pathway. Ubiquitination is a post-translational modification process that targets proteins for degradation or regulates their function by attaching a small protein called ubiquitin to them.

E1 enzymes initiate the ubiquitination process by activating ubiquitin through a two-step reaction. First, they catalyze the adenylation of ubiquitin's carboxyl terminus using ATP as an energy source, forming an adenylated ubiquitin intermediate. Then, the E1 enzyme transfers the activated ubiquitin to a cysteine residue on its own active site, forming a thioester bond between the ubiquitin and the E1 enzyme.

After activation, ubiquitin is transferred from the E1 enzyme to an E2 ubiquitin-conjugating enzyme, which then works with an E3 ubiquitin ligase to transfer ubiquitin to a specific lysine residue on the target protein. The addition of multiple ubiquitin molecules can create a polyubiquitin chain, leading to proteasomal degradation or other functional changes in the targeted protein.

There are two main families of E1 enzymes: UBA1 and UBA6. Dysregulation of ubiquitination pathways has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions. Therefore, understanding the function and regulation of E1 enzymes is essential for developing potential therapeutic strategies targeting these pathways.

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.

The Anaphase-Promoting Complex/Cyclosome (APC/C) is a large E3 ubiquitin ligase complex that plays a crucial role in the regulation of the cell cycle. It is responsible for targeting specific proteins for degradation by the proteasome, which is a multi-subunit protein complex that mediates the controlled breakdown of ubiquitinated proteins.

During anaphase, the final stage of mitosis, the APC/C becomes active and triggers the degradation of several key regulatory proteins, including securin and cyclin B. The destruction of these proteins allows for the separation of chromosomes and the completion of cell division.

The APC/C is composed of multiple subunits, including a catalytic core that binds to ubiquitin-conjugating enzymes (E2s) and several coactivators that regulate its activity. The activation of the APC/C requires the binding of one of two coactivators, Cdc20 or CDH1, which recognize specific substrates for degradation.

Dysregulation of the APC/C has been implicated in various human diseases, including cancer and neurodegenerative disorders. Therefore, understanding the mechanisms that regulate its activity is an important area of research with potential therapeutic implications.

Endocytosis is the process by which cells absorb substances from their external environment by engulfing them in membrane-bound structures, resulting in the formation of intracellular vesicles. This mechanism allows cells to take up large molecules, such as proteins and lipids, as well as small particles, like bacteria and viruses. There are two main types of endocytosis: phagocytosis (cell eating) and pinocytosis (cell drinking). Phagocytosis involves the engulfment of solid particles, while pinocytosis deals with the uptake of fluids and dissolved substances. Other specialized forms of endocytosis include receptor-mediated endocytosis and caveolae-mediated endocytosis, which allow for the specific internalization of molecules through the interaction with cell surface receptors.

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.

Autocrine motility factor (AMF) receptors are cell surface proteins that bind to autocrine motility factor, a cytokine produced and released by certain types of cancer cells. The binding of AMF to its receptor activates signaling pathways within the same cell that produces it, leading to changes in cell behavior such as increased motility and invasiveness. This process is known as autocrine signaling and plays a role in tumor progression and metastasis.

The AMF receptor has been identified as the product of the gene for the insulin-like growth factor I receptor (IGF1R), which is a tyrosine kinase receptor that regulates cell growth, differentiation, and survival. The activation of the IGF1R by AMF leads to the activation of downstream signaling pathways such as the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/Akt pathways, which promote cell motility and invasion.

In summary, Autocrine Motility Factor (AMF) receptors are a type of cell surface proteins that bind to AMF, leading to the activation of signaling pathways within the same cell that produces it, promoting changes in cell behavior such as increased motility and invasiveness, which play a role in tumor progression and metastasis.

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.

Saccharomyces cerevisiae proteins are the proteins that are produced by the budding yeast, Saccharomyces cerevisiae. This organism is a single-celled eukaryote that has been widely used as a model organism in scientific research for many years due to its relatively simple genetic makeup and its similarity to higher eukaryotic cells.

The genome of Saccharomyces cerevisiae has been fully sequenced, and it is estimated to contain approximately 6,000 genes that encode proteins. These proteins play a wide variety of roles in the cell, including catalyzing metabolic reactions, regulating gene expression, maintaining the structure of the cell, and responding to environmental stimuli.

Many Saccharomyces cerevisiae proteins have human homologs and are involved in similar biological processes, making this organism a valuable tool for studying human disease. For example, many of the proteins involved in DNA replication, repair, and recombination in yeast have human counterparts that are associated with cancer and other diseases. By studying these proteins in yeast, researchers can gain insights into their function and regulation in humans, which may lead to new treatments for disease.

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.

Multivesicular bodies (MVBs) are membrane-bound organelles found within eukaryotic cells, including animal and human cells. They are involved in the transport and disposal of cellular components, such as proteins and lipids. MVBs are characterized by the presence of multiple intraluminal vesicles (ILVs) contained within a larger compartment. These ILVs form through the inward budding of the limiting membrane, creating a complex internal structure.

MVBs play a crucial role in the process of autophagy, where they help to degrade damaged organelles and protein aggregates by fusing with lysosomes. Additionally, MVBs are essential for the downregulation of cell surface receptors through a process called endocytosis. In this pathway, activated receptors on the plasma membrane are internalized into early endosomes, which then mature into late endosomes or multivesicular bodies. The ILVs within MVBs contain these receptors along with other cellular components, and upon fusion of MVBs with lysosomes, the contents are degraded by hydrolytic enzymes.

In summary, multivesicular bodies (MVBs) are membrane-bound organelles containing multiple intraluminal vesicles that participate in autophagy and endocytosis for the disposal of cellular components and downregulation of surface receptors.

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.

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.

Tumor suppressor protein p53, also known as p53 or tumor protein p53, is a nuclear phosphoprotein that plays a crucial role in preventing cancer development and maintaining genomic stability. It does so by regulating the cell cycle and acting as a transcription factor for various genes involved in apoptosis (programmed cell death), DNA repair, and cell senescence (permanent cell growth arrest).

In response to cellular stress, such as DNA damage or oncogene activation, p53 becomes activated and accumulates in the nucleus. Activated p53 can then bind to specific DNA sequences and promote the transcription of target genes that help prevent the proliferation of potentially cancerous cells. These targets include genes involved in cell cycle arrest (e.g., CDKN1A/p21), apoptosis (e.g., BAX, PUMA), and DNA repair (e.g., GADD45).

Mutations in the TP53 gene, which encodes p53, are among the most common genetic alterations found in human cancers. These mutations often lead to a loss or reduction of p53's tumor suppressive functions, allowing cancer cells to proliferate uncontrollably and evade apoptosis. As a result, p53 has been referred to as "the guardian of the genome" due to its essential role in preventing tumorigenesis.

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.

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.

Ubiquitin-specific proteases (USPs) are a type of deubiquitinating enzymes (DUBs) that specifically cleave ubiquitin from proteins. Ubiquitination is a post-translational modification in which ubiquitin molecules are attached to proteins, targeting them for degradation by the proteasome. USPs reverse this process by removing ubiquitin molecules from proteins, thereby regulating protein stability, localization, and activity.

USPs contain a conserved catalytic domain that is responsible for the deubiquitinating activity. They are involved in various cellular processes, including DNA damage repair, gene expression regulation, inflammation, and immune response. Dysregulation of USP function has been implicated in several diseases, such as cancer, neurodegenerative disorders, and viral infections. Therefore, USPs are considered potential therapeutic targets for the development of drugs to treat these conditions.

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.

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.

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.

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.

Sumoylation is a post-translational modification process in which a small ubiquitin-like modifier (SUMO) protein is covalently attached to specific lysine residues on target proteins. This conjugation is facilitated by an enzymatic cascade involving E1 activating enzyme, E2 conjugating enzyme, and E3 ligase. Sumoylation can regulate various cellular functions such as protein stability, subcellular localization, activity, and interaction with other proteins. It plays crucial roles in numerous biological processes including DNA replication, repair, transcription, and chromatin remodeling, as well as stress response and regulation of the cell cycle. Dysregulation of sumoylation has been implicated in various human diseases, such as cancer, neurodegenerative disorders, and viral infections.

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.

"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.

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.

Lysosomes are membrane-bound organelles found in the cytoplasm of eukaryotic cells. They are responsible for breaking down and recycling various materials, such as waste products, foreign substances, and damaged cellular components, through a process called autophagy or phagocytosis. Lysosomes contain hydrolytic enzymes that can break down biomolecules like proteins, nucleic acids, lipids, and carbohydrates into their basic building blocks, which can then be reused by the cell. They play a crucial role in maintaining cellular homeostasis and are often referred to as the "garbage disposal system" of the cell.

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.

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

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.

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.

Endopeptidases are a type of enzyme that breaks down proteins by cleaving peptide bonds inside the polypeptide chain. They are also known as proteinases or endoproteinases. These enzymes work within the interior of the protein molecule, cutting it at specific points along its length, as opposed to exopeptidases, which remove individual amino acids from the ends of the protein chain.

Endopeptidases play a crucial role in various biological processes, such as digestion, blood coagulation, and programmed cell death (apoptosis). They are classified based on their catalytic mechanism and the structure of their active site. Some examples of endopeptidase families include serine proteases, cysteine proteases, aspartic proteases, and metalloproteases.

It is important to note that while endopeptidases are essential for normal physiological functions, they can also contribute to disease processes when their activity is unregulated or misdirected. For instance, excessive endopeptidase activity has been implicated in the pathogenesis of neurodegenerative disorders, cancer, and inflammatory conditions.

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.

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.

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.

Ring finger domains (RFIDs) are a type of protein domain that contain a characteristic cysteine-rich motif. They were initially identified in the RAS-associated proteins called Ras GTPase-activating proteins (GAPs), where they are involved in mediating protein-protein interactions.

The name "ring finger" comes from the fact that these domains contain a series of cysteine and histidine residues that coordinate a central zinc ion, forming a structural ring. This ring is thought to play a role in stabilizing the overall structure of the domain and facilitating its interactions with other proteins.

RFIDs are found in a wide variety of proteins, including transcription factors, chromatin modifiers, and signaling molecules. They have been implicated in a range of cellular processes, including transcriptional regulation, DNA repair, and signal transduction. Mutations in RFID-containing proteins have been linked to various human diseases, including cancer and neurological disorders.

SUMO-1 (Small Ubiquitin-like Modifier 1) protein is a member of the SUMO family of post-translational modifiers, which are involved in the regulation of various cellular processes such as nuclear-cytoplasmic transport, transcriptional regulation, and DNA repair. The SUMO-1 protein is covalently attached to specific lysine residues on target proteins through a process called sumoylation, which can alter the activity, localization, or stability of the modified protein. Sumoylation plays a crucial role in maintaining cellular homeostasis and has been implicated in several human diseases, including cancer and neurodegenerative disorders.

Nucleotide transport proteins are specialized membrane-bound proteins that facilitate the passive or active transport of nucleotides, such as adenosine triphosphate (ATP), guanosine triphosphate (GTP), and their precursors, across biological membranes. These proteins play a crucial role in maintaining the intracellular concentration of nucleotides, which are essential for various cellular processes, including energy metabolism, biosynthesis, and signal transduction.

There are two main types of nucleotide transport proteins: equilibrative nucleoside transporters (ENTs) and concentrative nucleoside transporters (CNTs). ENTs facilitate the passive diffusion of nucleosides and some nucleotides down their concentration gradient, while CNTs actively transport these molecules against their concentration gradient using energy derived from sodium or proton gradients.

These proteins are vital for cellular homeostasis and have been implicated in several diseases, including cancer and neurological disorders. Understanding the structure, function, and regulation of nucleotide transport proteins can provide valuable insights into their role in health and disease, potentially leading to the development of novel therapeutic strategies.

Endosomes are membrane-bound compartments within eukaryotic cells that play a critical role in intracellular trafficking and sorting of various cargoes, including proteins and lipids. They are formed by the invagination of the plasma membrane during endocytosis, resulting in the internalization of extracellular material and cell surface receptors.

Endosomes can be classified into early endosomes, late endosomes, and recycling endosomes based on their morphology, molecular markers, and functional properties. Early endosomes are the initial sorting stations for internalized cargoes, where they undergo sorting and processing before being directed to their final destinations. Late endosomes are more acidic compartments that mature from early endosomes and are responsible for the transport of cargoes to lysosomes for degradation.

Recycling endosomes, on the other hand, are involved in the recycling of internalized cargoes back to the plasma membrane or to other cellular compartments. Endosomal sorting and trafficking are regulated by a complex network of molecular interactions involving various proteins, lipids, and intracellular signaling pathways.

Defects in endosomal function have been implicated in various human diseases, including neurodegenerative disorders, developmental abnormalities, and cancer. Therefore, understanding the mechanisms underlying endosomal trafficking and sorting is of great importance for developing therapeutic strategies to treat these conditions.

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.

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.

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.

Tumor suppressor proteins are a type of regulatory protein that helps control the cell cycle and prevent cells from dividing and growing in an uncontrolled manner. They work to inhibit tumor growth by preventing the formation of tumors or slowing down their progression. These proteins can repair damaged DNA, regulate gene expression, and initiate programmed cell death (apoptosis) if the damage is too severe for repair.

Mutations in tumor suppressor genes, which provide the code for these proteins, can lead to a decrease or loss of function in the resulting protein. This can result in uncontrolled cell growth and division, leading to the formation of tumors and cancer. Examples of tumor suppressor proteins include p53, Rb (retinoblastoma), and BRCA1/2.

"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.

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.

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.

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.

'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.

DNA damage refers to any alteration in the structure or composition of deoxyribonucleic acid (DNA), which is the genetic material present in cells. DNA damage can result from various internal and external factors, including environmental exposures such as ultraviolet radiation, tobacco smoke, and certain chemicals, as well as normal cellular processes such as replication and oxidative metabolism.

Examples of DNA damage include base modifications, base deletions or insertions, single-strand breaks, double-strand breaks, and crosslinks between the two strands of the DNA helix. These types of damage can lead to mutations, genomic instability, and chromosomal aberrations, which can contribute to the development of diseases such as cancer, neurodegenerative disorders, and aging-related conditions.

The body has several mechanisms for repairing DNA damage, including base excision repair, nucleotide excision repair, mismatch repair, and double-strand break repair. However, if the damage is too extensive or the repair mechanisms are impaired, the cell may undergo apoptosis (programmed cell death) to prevent the propagation of potentially harmful mutations.

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.

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.

Histones are highly alkaline proteins found in the chromatin of eukaryotic cells. They are rich in basic amino acid residues, such as arginine and lysine, which give them their positive charge. Histones play a crucial role in packaging DNA into a more compact structure within the nucleus by forming a complex with it called a nucleosome. Each nucleosome contains about 146 base pairs of DNA wrapped around an octamer of eight histone proteins (two each of H2A, H2B, H3, and H4). The N-terminal tails of these histones are subject to various post-translational modifications, such as methylation, acetylation, and phosphorylation, which can influence chromatin structure and gene expression. Histone variants also exist, which can contribute to the regulation of specific genes and other nuclear processes.

Peptide synthases are a group of enzymes that catalyze the formation of peptide bonds between specific amino acids to produce peptides or proteins. They are responsible for the biosynthesis of many natural products, including antibiotics, bacterial toxins, and immunomodulatory peptides.

Peptide synthases are large, complex enzymes that consist of multiple domains and modules, each of which is responsible for activating and condensing specific amino acids. The activation of amino acids involves the formation of an aminoacyl-adenylate intermediate, followed by transfer of the activated amino acid to a thiol group on the enzyme. The condensation of two activated amino acids results in the formation of a peptide bond and release of adenosine monophosphate (AMP) and pyrophosphate.

Peptide synthases are found in all three domains of life, but are most commonly associated with bacteria and fungi. They play important roles in the biosynthesis of many natural products that have therapeutic potential, making them targets for drug discovery and development.

The endoplasmic reticulum (ER) is a network of interconnected tubules and sacs that are present in the cytoplasm of eukaryotic cells. It is a continuous membranous organelle that plays a crucial role in the synthesis, folding, modification, and transport of proteins and lipids.

The ER has two main types: rough endoplasmic reticulum (RER) and smooth endoplasmic reticulum (SER). RER is covered with ribosomes, which give it a rough appearance, and is responsible for protein synthesis. On the other hand, SER lacks ribosomes and is involved in lipid synthesis, drug detoxification, calcium homeostasis, and steroid hormone production.

In summary, the endoplasmic reticulum is a vital organelle that functions in various cellular processes, including protein and lipid metabolism, calcium regulation, and detoxification.

Serine is an amino acid, which is a building block of proteins. More specifically, it is a non-essential amino acid, meaning that the body can produce it from other compounds, and it does not need to be obtained through diet. Serine plays important roles in the body, such as contributing to the formation of the protective covering of nerve fibers (myelin sheath), helping to synthesize another amino acid called tryptophan, and taking part in the metabolism of fatty acids. It is also involved in the production of muscle tissues, the immune system, and the forming of cell structures. Serine can be found in various foods such as soy, eggs, cheese, meat, peanuts, lentils, and many others.

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).

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.

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.

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.

Small Ubiquitin-Related Modifier (SUMO) proteins are a type of post-translational modifier, similar to ubiquitin, that can be covalently attached to other proteins in a process called sumoylation. This modification plays a crucial role in regulating various cellular processes such as nuclear transport, transcriptional regulation, DNA repair, and protein stability. Sumoylation is a dynamic and reversible process, which allows for precise control of these functions under different physiological conditions.

The human genome encodes four SUMO paralogs (SUMO1-4), among which SUMO2 and SUMO3 share 97% sequence identity and are often referred to as a single entity, SUMO2/3. The fourth member, SUMO4, is less conserved and has a more restricted expression pattern compared to the other three paralogs.

Similar to ubiquitination, sumoylation involves an enzymatic cascade consisting of an E1 activating enzyme (SAE1/UBA2 heterodimer), an E2 conjugating enzyme (UBC9), and an E3 ligase that facilitates the transfer of SUMO from the E2 to the target protein. The process can be reversed by SUMO-specific proteases, which cleave the isopeptide bond between the modified lysine residue on the target protein and the C-terminal glycine of the SUMO molecule.

Dysregulation of sumoylation has been implicated in various human diseases, including cancer, neurodegenerative disorders, and viral infections. Therefore, understanding the molecular mechanisms governing this post-translational modification is essential for developing novel therapeutic strategies targeting these conditions.

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.

"Mating factor receptors" are a type of cell surface receptor found in certain species of fungi that play a crucial role in the process of mating and sexual reproduction. These receptors are responsible for recognizing and binding to specific signaling molecules, known as "mating factors," which are released by potential mating partners.

In the fungus Saccharomyces cerevisiae (yeast), for example, there are two types of mating factors: a-factor and α-factor. The a-factor is produced by cells with the mating type "a," while the α-factor is produced by cells with the mating type "α." When these factors come into contact with receptor proteins on the surface of cells with the opposite mating type, they trigger a series of intracellular signaling events that ultimately lead to the fusion of the two cells and the formation of a diploid zygote.

The receptors for mating factors are typically transmembrane proteins with an extracellular domain that binds to the mating factor, and an intracellular domain that activates downstream signaling pathways. In S. cerevisiae, the a-factor receptor is called "Ste3," while the α-factor receptor is called "Ste2."

It's worth noting that while mating factor receptors are essential for sexual reproduction in fungi, they have also been implicated in other important cellular processes, such as cell growth, differentiation, and pathogenesis.

'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.

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.

v-Cbl is a type of oncogene protein that is derived from the cellular c-Cbl protein. Oncogenes are genes that have the potential to cause cancer, and they can do this by promoting cell growth and division when they should not. The v-Cbl protein is created when a virus called the avian reticuloendotheliosis virus infects a host cell and inserts its own version of the c-Cbl gene into the host's DNA. This results in the production of the abnormal v-Cbl protein, which can contribute to the development of cancer by disrupting the normal regulation of cell growth and division.

The c-Cbl protein is a type of E3 ubiquitin ligase, which is an enzyme that helps to tag other proteins for degradation. The v-Cbl protein retains this function, but it also has additional activities that allow it to promote cell growth and division. For example, v-Cbl can activate signaling pathways that lead to the activation of transcription factors, which are proteins that control the expression of genes involved in cell growth and division.

In addition to its role in cancer, v-Cbl has also been implicated in the development of other diseases, including immune disorders and neurological conditions. However, more research is needed to fully understand the various functions of this oncogene protein and how it contributes to disease.

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.

Gene knockdown techniques are methods used to reduce the expression or function of specific genes in order to study their role in biological processes. These techniques typically involve the use of small RNA molecules, such as siRNAs (small interfering RNAs) or shRNAs (short hairpin RNAs), which bind to and promote the degradation of complementary mRNA transcripts. This results in a decrease in the production of the protein encoded by the targeted gene.

Gene knockdown techniques are often used as an alternative to traditional gene knockout methods, which involve completely removing or disrupting the function of a gene. Knockdown techniques allow for more subtle and reversible manipulation of gene expression, making them useful for studying genes that are essential for cell survival or have redundant functions.

These techniques are widely used in molecular biology research to investigate gene function, genetic interactions, and disease mechanisms. However, it is important to note that gene knockdown can have off-target effects and may not completely eliminate the expression of the targeted gene, so results should be interpreted with caution.

Substrate specificity in the context of medical biochemistry and enzymology refers to the ability of an enzyme to selectively bind and catalyze a chemical reaction with a particular substrate (or a group of similar substrates) while discriminating against other molecules that are not substrates. This specificity arises from the three-dimensional structure of the enzyme, which has evolved to match the shape, charge distribution, and functional groups of its physiological substrate(s).

Substrate specificity is a fundamental property of enzymes that enables them to carry out highly selective chemical transformations in the complex cellular environment. The active site of an enzyme, where the catalysis takes place, has a unique conformation that complements the shape and charge distribution of its substrate(s). This ensures efficient recognition, binding, and conversion of the substrate into the desired product while minimizing unwanted side reactions with other molecules.

Substrate specificity can be categorized as:

1. Absolute specificity: An enzyme that can only act on a single substrate or a very narrow group of structurally related substrates, showing no activity towards any other molecule.
2. Group specificity: An enzyme that prefers to act on a particular functional group or class of compounds but can still accommodate minor structural variations within the substrate.
3. Broad or promiscuous specificity: An enzyme that can act on a wide range of structurally diverse substrates, albeit with varying catalytic efficiencies.

Understanding substrate specificity is crucial for elucidating enzymatic mechanisms, designing drugs that target specific enzymes or pathways, and developing biotechnological applications that rely on the controlled manipulation of enzyme activities.

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).

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.

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.

NIH 3T3 cells are a type of mouse fibroblast cell line that was developed by the National Institutes of Health (NIH). The "3T3" designation refers to the fact that these cells were derived from embryonic Swiss mouse tissue and were able to be passaged (i.e., subcultured) more than three times in tissue culture.

NIH 3T3 cells are widely used in scientific research, particularly in studies involving cell growth and differentiation, signal transduction, and gene expression. They have also been used as a model system for studying the effects of various chemicals and drugs on cell behavior. NIH 3T3 cells are known to be relatively easy to culture and maintain, and they have a stable, flat morphology that makes them well-suited for use in microscopy studies.

It is important to note that, as with any cell line, it is essential to verify the identity and authenticity of NIH 3T3 cells before using them in research, as contamination or misidentification can lead to erroneous results.

The Von Hippel-Lindau (VHL) tumor suppressor protein is a crucial component in the regulation of cellular growth and division, specifically through its role in oxygen sensing and the ubiquitination of hypoxia-inducible factors (HIFs). The VHL protein forms part of an E3 ubiquitin ligase complex that targets HIFs for degradation under normoxic conditions. In the absence of functional VHL protein or in hypoxic environments, HIFs accumulate and induce the transcription of genes involved in angiogenesis, cell proliferation, and metabolism.

Mutations in the VHL gene can lead to the development of Von Hippel-Lindau syndrome, a rare inherited disorder characterized by the growth of tumors and cysts in various organs, including the central nervous system, retina, kidneys, adrenal glands, and pancreas. These tumors often arise from the overactivation of HIF-mediated signaling pathways due to the absence or dysfunction of VHL protein.

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).

Endoplasmic reticulum-associated degradation (ERAD) is a cellular process that targets and degrades misfolded or damaged proteins located in the endoplasmic reticulum (ER). The ER is a network of membrane-bound tubules and sacs within eukaryotic cells where proteins are synthesized, folded, and modified before being transported to their final destinations.

When proteins fail to fold correctly or become damaged in the ER, they can be recognized and tagged for degradation through the ERAD pathway. This process involves several steps:

1. Recognition: Misfolded or damaged proteins are recognized by specific chaperone proteins and ubiquitin ligases in the ER. Chaperones help proteins fold correctly, while ubiquitin ligases tag misfolded proteins with ubiquitin molecules, marking them for degradation.
2. Retrotranslocation: The marked proteins are then retrotranslocated (or "pulled back") across the ER membrane into the cytosol by a protein complex called the ERAD machinery.
3. Ubiquitination: Once in the cytosol, the ubiquitin molecules attached to the misfolded proteins are recognized by another set of ubiquitin ligases, which add more ubiquitin molecules, creating a polyubiquitin chain.
4. Degradation: The polyubiquitinated protein is then targeted to and degraded by the 26S proteasome, a large protein complex responsible for breaking down unwanted or damaged proteins in the cell.

ERAD plays a crucial role in maintaining protein quality control and ensuring proper cellular function. Dysregulation of ERAD has been implicated in various diseases, including neurodegenerative disorders, cancer, and viral infections.

A mutant protein is a protein that has undergone a genetic mutation, resulting in an altered amino acid sequence and potentially changed structure and function. These changes can occur due to various reasons such as errors during DNA replication, exposure to mutagenic substances, or inherited genetic disorders. The alterations in the protein's structure and function may have no significant effects, lead to benign phenotypic variations, or cause diseases, depending on the type and location of the mutation. Some well-known examples of diseases caused by mutant proteins include cystic fibrosis, sickle cell anemia, and certain types of cancer.

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.

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.

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.

Peptide hydrolases, also known as proteases or peptidases, are a group of enzymes that catalyze the hydrolysis of peptide bonds in proteins and peptides. They play a crucial role in various biological processes such as protein degradation, digestion, cell signaling, and regulation of various physiological functions. Based on their catalytic mechanism and the specificity for the peptide bond, they are classified into several types, including serine proteases, cysteine proteases, aspartic proteases, and metalloproteases. These enzymes have important clinical applications in the diagnosis and treatment of various diseases, such as cancer, viral infections, and inflammatory disorders.

Pheromone receptors are a specific type of sensory receptor found in many animals, including insects and mammals, that detect and respond to pheromones. Pheromones are chemical signals that are released by an individual and received by another individual of the same species, which can elicit various behavioral or physiological responses.

Pheromone receptors are located in the sensory organs responsible for detecting chemical stimuli, such as the antennae of insects or the vomeronasal organ (VNO) in mammals. These receptors contain specialized proteins called G protein-coupled receptors (GPCRs) that bind to specific pheromone molecules and trigger a cascade of intracellular signaling events, ultimately leading to the activation of downstream effector pathways.

In insects, pheromone receptors are typically found in olfactory sensory neurons located on the antennae or other peripheral organs. These receptors can detect a wide range of pheromones, including sex pheromones that play a critical role in mating behavior, as well as aggregation pheromones that help to coordinate group behaviors such as feeding or nesting.

In mammals, pheromone receptors are found in the vomeronasal organ (VNO), which is located in the nasal cavity and contains specialized sensory neurons called vomeronasal sensory neurons (VSNs). VSNs express a variety of pheromone receptors that can detect different types of pheromones, including those involved in social recognition, mating behavior, and aggression.

Overall, the activation of pheromone receptors plays a critical role in mediating various aspects of animal behavior and physiology, highlighting their importance in chemical communication and social interaction.

Proliferating Cell Nuclear Antigen (PCNA) is a protein that plays an essential role in the process of DNA replication and repair in eukaryotic cells. It functions as a cofactor for DNA polymerase delta, enhancing its activity during DNA synthesis. PCNA forms a sliding clamp around DNA, allowing it to move along the template and coordinate the actions of various enzymes involved in DNA metabolism.

PCNA is often used as a marker for cell proliferation because its levels increase in cells that are actively dividing or have been stimulated to enter the cell cycle. Immunostaining techniques can be used to detect PCNA and determine the proliferative status of tissues or cultures. In this context, 'proliferating' refers to the rapid multiplication of cells through cell division.

APC11 subunit is a component of the Anaphase-Promoting Complex/Cyclosome (APC/C), which is an E3 ubiquitin ligase complex that plays a critical role in regulating the cell cycle. The APC/C complex is responsible for targeting specific proteins for degradation by the proteasome, thereby controlling various stages of mitosis and the transition to G1 phase of the cell cycle.

APC11 is one of the essential subunits of the APC/C complex, and it functions as a part of the catalytic core that mediates the transfer of ubiquitin molecules to the target proteins. Specifically, APC11 interacts with another subunit, APC2, to form the catalytic site of the APC/C complex. Together, they facilitate the formation of a polyubiquitin chain on the target protein, marking it for degradation by the 26S proteasome.

APC11 has been shown to be critical for the proper functioning of the APC/C complex and is required for the ubiquitination and subsequent degradation of several key cell cycle regulators, including securin, cyclin A, and cyclin B. Mutations in the APC11 gene have been associated with various types of cancer, highlighting its importance in maintaining genomic stability.

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.

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.

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.

HSP90 (Heat Shock Protein 90) refers to a family of highly conserved molecular chaperones that are expressed in all eukaryotic cells. They play a crucial role in protein folding, assembly, and transport, thereby assisting in the maintenance of proper protein function and cellular homeostasis. HSP90 proteins are named for their increased expression during heat shock and other stress conditions, which helps protect cells by facilitating the refolding or degradation of misfolded proteins that can accumulate under these circumstances.

HSP90 chaperones are ATP-dependent and consist of multiple domains: a N-terminal nucleotide binding domain (NBD), a middle domain, and a C-terminal dimerization domain. They exist as homodimers and interact with a wide range of client proteins, including transcription factors, kinases, and steroid hormone receptors. By regulating the activity and stability of these client proteins, HSP90 chaperones contribute to various cellular processes such as signal transduction, cell cycle progression, and stress response. Dysregulation of HSP90 function has been implicated in numerous diseases, including cancer, neurodegenerative disorders, and infectious diseases, making it an attractive target for therapeutic intervention.

I'm sorry for any confusion, but "Active Transport, Cell Nucleus" is not a widely recognized or established medical term. Active transport typically refers to the energy-dependent process by which cells move molecules across their membranes against their concentration gradient. This process is facilitated by transport proteins and requires ATP as an energy source. However, this process primarily occurs in the cell membrane and not in the cell nucleus.

The cell nucleus, on the other hand, contains genetic material (DNA) and is responsible for controlling various cellular activities such as gene expression, replication, and repair. While there are transport processes that occur within the nucleus, they do not typically involve active transport in the same way that it occurs at the cell membrane.

Therefore, a medical definition of "Active Transport, Cell Nucleus" would not be applicable or informative in this context.

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).

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.

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.

Arrestins are a family of proteins that play a crucial role in regulating G protein-coupled receptor (GPCR) signaling. There are four main types of arrestins: visual arrestin (also known as arr1 or S-arrestin), β-arrestin1 (also known as arr2 or Kon/Vec), β-arrestin2 (also known as arr3 or hTHT), and arrestin-domain containing protein 1 (ARRDC1).

Arrestins bind to the intracellular domains of activated GPCRs, which leads to several outcomes:

1. They prevent further activation of G proteins by the receptor, effectively "arresting" the signal transduction process.
2. They promote the internalization (endocytosis) of the receptor from the cell membrane into endosomes, where it can be either degraded or recycled back to the cell surface.
3. They act as scaffolds for various signaling complexes and mediate interactions between GPCRs and other intracellular signaling proteins, leading to the activation of different signaling pathways.

Overall, arrestins play a critical role in fine-tuning GPCR signaling, ensuring appropriate cellular responses to hormones, neurotransmitters, and other extracellular signals.

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.

Casein Kinase 1 (CK1) is a type of serine/threonine protein kinase that plays a crucial role in various cellular processes, including the regulation of circadian rhythms, signal transduction, and DNA damage response. CK1 phosphorylates specific serine or threonine residues on its target proteins, thereby modulating their activity, localization, or stability.

There are several isoforms of CK1, including CK1α, CK1δ, CK1ε, and CK1γ, which exhibit distinct subcellular distributions and functions. Dysregulation of CK1 has been implicated in several human diseases, such as cancer, neurodegenerative disorders, and metabolic syndromes. Therefore, understanding the molecular mechanisms underlying CK1 function is essential for developing novel therapeutic strategies to treat these conditions.

Cdh1 proteins are part of the anaphase-promoting complex/cyclosome (APC/C), which is a multi-subunit E3 ubiquitin ligase that plays a critical role in regulating the cell cycle. Cdh1, specifically, is a regulatory subunit of the APC/C and is essential for the proper progression through the cell cycle.

Cdh1 binds to and activates the APC/C in late mitosis and early G1 phase, targeting specific proteins for ubiquitination and subsequent degradation by the proteasome. This helps to ensure that key events of the cell cycle, such as chromosome segregation and mitotic exit, occur in a timely and orderly fashion.

Cdh1 has been shown to regulate the degradation of several important cell cycle regulators, including cyclins A and B, securin, and aurora kinase A. By targeting these proteins for destruction, Cdh1 helps to prevent premature entry into mitosis and ensures that cells do not exit mitosis until all chromosomes have been properly aligned and segregated.

Mutations in the genes encoding Cdh1 and other components of the APC/C have been implicated in a variety of human cancers, highlighting the importance of this complex in maintaining genomic stability.

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.

MAP (Mitogen-Activated Protein) Kinase Kinase Kinases (MAP3K or MAPKKK) are a group of protein kinases that play a crucial role in intracellular signal transduction pathways, which regulate various cellular processes such as proliferation, differentiation, survival, and apoptosis. They are called "kinases" because they catalyze the transfer of a phosphate group from ATP to specific serine or threonine residues on their target proteins.

MAP3Ks function upstream of MAP Kinase Kinases (MKKs or MAP2K) and MAP Kinases (MPKs or MAPK) in the MAP kinase cascade. Upon activation by various extracellular signals, such as growth factors, cytokines, stress, and hormones, MAP3Ks phosphorylate and activate MKKs, which subsequently phosphorylate and activate MPKs. Activated MPKs then regulate the activity of downstream transcription factors and other target proteins to elicit appropriate cellular responses.

There are several subfamilies of MAP3Ks, including ASK, DLK, TAK, MEKK, MLK, and ZAK, among others. Each subfamily has distinct structural features and functions in different signaling pathways. Dysregulation of MAP kinase cascades, including MAP3Ks, has been implicated in various human diseases, such as cancer, inflammation, and neurodegenerative disorders.

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.

APC1 (Anaphase-Promoting Complex-Cyclosome) subunit is a component of the multi-subunit E3 ubiquitin ligase complex known as the Anaphase-Promoting Complex/Cyclosome (APC/C). The APC/C plays a crucial role in regulating the cell cycle, specifically during mitosis and meiosis.

The APC/C is responsible for targeting specific proteins for degradation by the ubiquitin-proteasome system. This degradation leads to the regulation of various cell cycle events, such as sister chromatid separation during anaphase and the exit from mitosis.

APC1 is one of the several subunits that make up the APC/C complex. It serves as a scaffold protein, helping to assemble and maintain the structural integrity of the complex. Additionally, APC1 has been shown to play a role in substrate recognition by the APC/C, contributing to the specificity of ubiquitination and subsequent degradation of target proteins.

The medical relevance of understanding the APC/C and its subunits, including APC1, lies in their essential roles in cell cycle regulation. Dysregulation of these processes can lead to various diseases, such as cancer, where uncontrolled cell division is a hallmark feature. Studying the APC/C and its components may provide insights into potential therapeutic targets for treating such conditions.

Acetylcysteine is a medication that is used for its antioxidant effects and to help loosen thick mucus in the lungs. It is commonly used to treat conditions such as chronic bronchitis, emphysema, and cystic fibrosis. Acetylcysteine is also known by the brand names Mucomyst and Accolate. It works by thinning and breaking down mucus in the airways, making it easier to cough up and clear the airways. Additionally, acetylcysteine is an antioxidant that helps to protect cells from damage caused by free radicals. It is available as a oral tablet, liquid, or inhaled medication.

Gene silencing is a process by which the expression of a gene is blocked or inhibited, preventing the production of its corresponding protein. This can occur naturally through various mechanisms such as RNA interference (RNAi), where small RNAs bind to and degrade specific mRNAs, or DNA methylation, where methyl groups are added to the DNA molecule, preventing transcription. Gene silencing can also be induced artificially using techniques such as RNAi-based therapies, antisense oligonucleotides, or CRISPR-Cas9 systems, which allow for targeted suppression of gene expression in research and therapeutic applications.

CDC20 proteins are a type of regulatory protein that play a crucial role in the cell cycle, which is the process by which cells grow and divide. Specifically, CDC20 proteins are involved in the transition from metaphase to anaphase during mitosis, the phase of the cell cycle where chromosomes are separated and distributed to two daughter cells.

CDC20 proteins function as part of a larger complex called the anaphase-promoting complex/cyclosome (APC/C), which targets specific proteins for degradation by the proteasome. During metaphase, CDC20 binds to the APC/C and helps to activate it, leading to the degradation of securin and cyclin B, two proteins that are essential for maintaining the proper attachment of chromosomes to the spindle apparatus.

Once these proteins are degraded, the sister chromatids can be separated and moved to opposite poles of the cell, allowing for the completion of mitosis and the formation of two genetically identical daughter cells. In addition to their role in mitosis, CDC20 proteins have also been implicated in other cellular processes, including meiosis, DNA damage repair, and apoptosis.

Protein interaction mapping is a research approach used to identify and characterize the physical interactions between different proteins within a cell or organism. This process often involves the use of high-throughput experimental techniques, such as yeast two-hybrid screening, mass spectrometry-based approaches, or protein fragment complementation assays, to detect and quantify the binding affinities of protein pairs. The resulting data is then used to construct a protein interaction network, which can provide insights into functional relationships between proteins, help elucidate cellular pathways, and inform our understanding of biological processes in health and disease.

Acetylation is a chemical process that involves the addition of an acetyl group (-COCH3) to a molecule. In the context of medical biochemistry, acetylation often refers to the post-translational modification of proteins, where an acetyl group is added to the amino group of a lysine residue in a protein by an enzyme called acetyltransferase. This modification can alter the function or stability of the protein and plays a crucial role in regulating various cellular processes such as gene expression, DNA repair, and cell signaling. Acetylation can also occur on other types of molecules, including lipids and carbohydrates, and has important implications for drug metabolism and toxicity.

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.

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.

HSP70 heat-shock proteins are a family of highly conserved molecular chaperones that play a crucial role in protein folding and protection against stress-induced damage. They are named after the fact that they were first discovered in response to heat shock, but they are now known to be produced in response to various stressors, such as oxidative stress, inflammation, and exposure to toxins.

HSP70 proteins bind to exposed hydrophobic regions of unfolded or misfolded proteins, preventing their aggregation and assisting in their proper folding. They also help target irreversibly damaged proteins for degradation by the proteasome. In addition to their role in protein homeostasis, HSP70 proteins have been shown to have anti-inflammatory and immunomodulatory effects, making them a subject of interest in various therapeutic contexts.

Mitochondrial degradation, also known as mitophagy, is a process by which damaged or dysfunctional mitochondria are eliminated from the cell. Mitochondria are essential organelles that generate energy for the cell through a process called oxidative phosphorylation. However, they can become damaged due to various factors such as mutations in mitochondrial DNA, oxidative stress, or protein misfolding.

Mitophagy is a selective form of autophagy, which is the process by which cells break down and recycle their own components. During mitophagy, damaged mitochondria are tagged with ubiquitin molecules, which serve as a signal for their recognition and engulfment by autophagosomes. Autophagosomes are double-membraned vesicles that enclose cellular components and fuse with lysosomes, where the contents are broken down and recycled.

Mitophagy plays an important role in maintaining mitochondrial quality control and preventing the accumulation of damaged mitochondria, which can lead to cellular dysfunction and disease. Defects in mitophagy have been implicated in various pathologies, including neurodegenerative disorders, cardiovascular diseases, and aging-related conditions.

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.

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.

Glycogen Synthase Kinase 3 (GSK-3) is a serine/threonine protein kinase that plays a crucial role in the regulation of several cellular processes, including glycogen metabolism, cell signaling, gene transcription, and apoptosis. It was initially discovered as a key enzyme involved in glycogen metabolism due to its ability to phosphorylate and inhibit glycogen synthase, an enzyme responsible for the synthesis of glycogen from glucose.

GSK-3 exists in two isoforms, GSK-3α and GSK-3β, which share a high degree of sequence similarity and are widely expressed in various tissues. Both isoforms are constitutively active under normal conditions and are regulated through inhibitory phosphorylation by several upstream signaling pathways, such as insulin, Wnt, and Hedgehog signaling.

Dysregulation of GSK-3 has been implicated in the pathogenesis of various diseases, including diabetes, neurodegenerative disorders, and cancer. In recent years, GSK-3 has emerged as an attractive therapeutic target for the development of novel drugs to treat these conditions.

In the context of pharmacology, "half-life" refers to the time it takes for the concentration or amount of a drug in the body to be reduced by half during its elimination phase. This is typically influenced by factors such as metabolism and excretion rates of the drug. It's a key factor in determining dosage intervals and therapeutic effectiveness of medications, as well as potential side effects or toxicity risks.

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.

The X-linked inhibitor of apoptosis protein (XIAP) is a member of the inhibitor of apoptosis (IAP) family, which are proteins that play a crucial role in regulating programmed cell death, also known as apoptosis. XIAP is located on the X chromosome and functions by binding to and inhibiting certain caspases, which are enzymes that play an essential role in initiating and executing the apoptotic process. By inhibiting these caspases, XIAP promotes cell survival and prevents excessive cell death, which can contribute to cancer development and resistance to therapy. Additionally, XIAP has been implicated in the regulation of inflammation and immune responses, making it a target for therapeutic intervention in various diseases.

Glycylglycine is not a medical condition or term, but rather it is a chemical compound. It is a dipeptide, which means it is composed of two amino acids linked together. Specifically, glycylglycine consists of two glycine molecules joined by an amide bond (also known as a peptide bond) between the carboxyl group of one glycine and the amino group of the other glycine.

Glycylglycine is often used in laboratory research as a buffer, a substance that helps maintain a stable pH level in a solution. It has a relatively simple structure and is not naturally found in significant amounts in living organisms.

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.

Cyclin-Dependent Kinase Inhibitor p27, also known as CDKN1B or p27Kip1, is a protein that regulates the cell cycle. It inhibits the activity of certain cyclin-dependent kinases (CDKs), which are enzymes that play key roles in regulating the progression of the cell cycle.

The cell cycle is a series of events that cells undergo as they grow and divide. Cyclins and CDKs help to control the different stages of the cell cycle by activating and deactivating various proteins at specific times. The p27 protein acts as a brake on the cell cycle, preventing cells from dividing too quickly or abnormally.

When p27 binds to a CDK-cyclin complex, it prevents the complex from phosphorylating its target proteins, which are necessary for the progression of the cell cycle. By inhibiting CDK activity, p27 helps to ensure that cells divide only when the proper conditions are met.

Mutations in the CDKN1B gene, which encodes p27, have been associated with several types of cancer, including breast, lung, and prostate cancer. These mutations can lead to decreased levels of p27 or impaired function, allowing cells to divide uncontrollably and form tumors.

HCT116 cells are a type of human colon cancer cell line that is widely used in scientific research. They were originally established in the early 1980s from a primary colon tumor that had metastasized to the liver. HCT116 cells are known for their stability, robust growth, and susceptibility to various genetic manipulations, making them a popular choice for studying cancer biology, drug discovery, and gene function.

These cells have several important features that make them useful in research. For example, they harbor mutations in key genes involved in colorectal cancer development, such as the adenomatous polyposis coli (APC) gene and the KRAS oncogene. Additionally, HCT116 cells can be easily cultured in the lab and are amenable to a variety of experimental techniques, including genetic modification, drug screening, and protein analysis.

It is important to note that while HCT116 cells provide valuable insights into colon cancer biology, they represent only one type of cancer cell line, and their behavior may not necessarily reflect the complexity of human tumors in vivo. Therefore, researchers must exercise caution when interpreting results obtained from these cells and consider other complementary approaches to validate their findings.

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.

Clathrin is a type of protein that plays a crucial role in the formation of coated vesicles within cells. These vesicles are responsible for transporting materials between different cellular compartments, such as from the plasma membrane to the endoplasmic reticulum or Golgi apparatus. Clathrin molecules form a lattice-like structure that curves around the vesicle, providing stability and shape to the coated vesicle. This process is known as clathrin-mediated endocytosis.

The formation of clathrin-coated vesicles begins with the recruitment of clathrin proteins to specific sites on the membrane, where they assemble into a polygonal lattice structure. As more clathrin molecules join the assembly, the lattice curves and eventually pinches off from the membrane, forming a closed vesicle. The clathrin coat then disassembles, releasing the vesicle to continue with its intracellular transport mission.

Disruptions in clathrin-mediated endocytosis can lead to various cellular dysfunctions and diseases, including neurodegenerative disorders and certain types of cancer.

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.

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.

CARD (caspase recruitment domain) signaling adaptor proteins are a group of intracellular signaling molecules that play a crucial role in the regulation of various cellular processes, including inflammation, immunity, and programmed cell death or apoptosis. These proteins contain a CARD domain, which is a protein-protein interaction module that enables them to bind to other CARD-containing proteins and form large signaling complexes.

CARD signaling adaptor proteins function as molecular scaffolds that help bring together various signaling components in response to different stimuli, such as pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). By doing so, they facilitate the activation of downstream signaling cascades and the initiation of appropriate cellular responses.

Some examples of CARD signaling adaptor proteins include:

1. Myeloid differentiation factor 88 (MyD88): This protein is involved in the signaling pathways of most Toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R) family members, which are critical for the detection of microbial components and the initiation of innate immune responses.
2. CARD9: This protein is involved in the signaling pathways of several C-type lectin receptors (CLRs), which recognize fungal and other pathogens, and plays a key role in antifungal immunity.
3. ASC (apoptosis-associated speck-like protein containing a CARD): This protein is involved in the formation of inflammasomes, which are large cytosolic complexes that activate caspase-1 and promote the maturation and secretion of proinflammatory cytokines.
4. RIPK2 (receptor-interacting serine/threonine-protein kinase 2): This protein is involved in the signaling pathways of NOD1 and NOD2, which are intracellular sensors of bacterial peptidoglycan, and plays a role in the regulation of inflammation and apoptosis.

Overall, CARD-containing proteins play crucial roles in various immune signaling pathways by mediating protein-protein interactions and downstream signal transduction events, ultimately leading to the activation of innate immunity and inflammatory responses.

Mitosis is a type of cell division in which the genetic material of a single cell, called the mother cell, is equally distributed into two identical daughter cells. It's a fundamental process that occurs in multicellular organisms for growth, maintenance, and repair, as well as in unicellular organisms for reproduction.

The process of mitosis can be broken down into several stages: prophase, prometaphase, metaphase, anaphase, and telophase. During prophase, the chromosomes condense and become visible, and the nuclear envelope breaks down. In prometaphase, the nuclear membrane is completely disassembled, and the mitotic spindle fibers attach to the chromosomes at their centromeres.

During metaphase, the chromosomes align at the metaphase plate, an imaginary line equidistant from the two spindle poles. In anaphase, sister chromatids are pulled apart by the spindle fibers and move toward opposite poles of the cell. Finally, in telophase, new nuclear envelopes form around each set of chromosomes, and the chromosomes decondense and become less visible.

Mitosis is followed by cytokinesis, a process that divides the cytoplasm of the mother cell into two separate daughter cells. The result of mitosis and cytokinesis is two genetically identical cells, each with the same number and kind of chromosomes as the original parent cell.

Beta-catenin is a protein that plays a crucial role in gene transcription and cell-cell adhesion. It is a key component of the Wnt signaling pathway, which regulates various processes such as cell proliferation, differentiation, and migration during embryonic development and tissue homeostasis in adults.

In the absence of Wnt signals, beta-catenin forms a complex with other proteins, including adenomatous polyposis coli (APC) and axin, which targets it for degradation by the proteasome. When Wnt ligands bind to their receptors, this complex is disrupted, allowing beta-catenin to accumulate in the cytoplasm and translocate to the nucleus. In the nucleus, beta-catenin interacts with T cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcription factors to activate the transcription of target genes involved in cell fate determination, survival, and proliferation.

Mutations in the genes encoding components of the Wnt signaling pathway, including beta-catenin, have been implicated in various human diseases, such as cancer, developmental disorders, and degenerative conditions.

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.

Hydrolysis is a chemical process, not a medical one. However, it is relevant to medicine and biology.

Hydrolysis is the breakdown of a chemical compound due to its reaction with water, often resulting in the formation of two or more simpler compounds. In the context of physiology and medicine, hydrolysis is a crucial process in various biological reactions, such as the digestion of food molecules like proteins, carbohydrates, and fats. Enzymes called hydrolases catalyze these hydrolysis reactions to speed up the breakdown process in the body.

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.

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.

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.

According to the medical definition, ultraviolet (UV) rays are invisible radiations that fall in the range of the electromagnetic spectrum between 100-400 nanometers. UV rays are further divided into three categories: UVA (320-400 nm), UVB (280-320 nm), and UVC (100-280 nm).

UV rays have various sources, including the sun and artificial sources like tanning beds. Prolonged exposure to UV rays can cause damage to the skin, leading to premature aging, eye damage, and an increased risk of skin cancer. UVA rays penetrate deeper into the skin and are associated with skin aging, while UVB rays primarily affect the outer layer of the skin and are linked to sunburns and skin cancer. UVC rays are the most harmful but fortunately, they are absorbed by the Earth's atmosphere and do not reach the surface.

Healthcare professionals recommend limiting exposure to UV rays, wearing protective clothing, using broad-spectrum sunscreen with an SPF of at least 30, and avoiding tanning beds to reduce the risk of UV-related health problems.

Anaphase is a stage in the cell division process called mitosis, where sister chromatids (the two copies of each chromosome formed during DNA replication) separate at the centromeres and move toward opposite poles of the cell. This separation is facilitated by the attachment of microtubules from the spindle apparatus to the kinetochores, protein structures located on the centromeres of each sister chromatid. Anaphase is followed by telophase, during which the nuclear membrane reforms around each set of separated chromosomes, and cytokinesis, the division of the cytoplasm to form two separate daughter cells.

Vacuoles are membrane-bound organelles found in the cells of most eukaryotic organisms. They are essentially fluid-filled sacs that store various substances, such as enzymes, waste products, and nutrients. In plants, vacuoles often contain water, ions, and various organic compounds, while in fungi, they may store lipids or pigments. Vacuoles can also play a role in maintaining the turgor pressure of cells, which is critical for cell shape and function.

In animal cells, vacuoles are typically smaller and less numerous than in plant cells. Animal cells have lysosomes, which are membrane-bound organelles that contain digestive enzymes and break down waste materials, cellular debris, and foreign substances. Lysosomes can be considered a type of vacuole, but they are more specialized in their function.

Overall, vacuoles are essential for maintaining the health and functioning of cells by providing a means to store and dispose of various substances.

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.

P300 and CREB binding protein (CBP) are both transcriptional coactivators that play crucial roles in regulating gene expression. They function by binding to various transcription factors and modifying the chromatin structure to allow for the recruitment of the transcriptional machinery. The P300-CBP complex is essential for many cellular processes, including development, differentiation, and oncogenesis.

P300-CBP transcription factors refer to a family of proteins that include both p300 and CBP, as well as their various isoforms and splice variants. These proteins share structural and functional similarities and are often referred to together due to their overlapping roles in transcriptional regulation.

The P300-CBP complex plays a key role in the P300-CBP-mediated signal integration, which allows for the coordinated regulation of gene expression in response to various signals and stimuli. Dysregulation of P300-CBP transcription factors has been implicated in several diseases, including cancer, neurodevelopmental disorders, and inflammatory diseases.

In summary, P300-CBP transcription factors are a family of proteins that play crucial roles in regulating gene expression through their ability to bind to various transcription factors and modify the chromatin structure. Dysregulation of these proteins has been implicated in several diseases, making them important targets for therapeutic intervention.

Threonine is an essential amino acid, meaning it cannot be synthesized by the human body and must be obtained through the diet. Its chemical formula is HO2CCH(NH2)CH(OH)CH3. Threonine plays a crucial role in various biological processes, including protein synthesis, immune function, and fat metabolism. It is particularly important for maintaining the structural integrity of proteins, as it is often found in their hydroxyl-containing regions. Foods rich in threonine include animal proteins such as meat, dairy products, and eggs, as well as plant-based sources like lentils and soybeans.

Protein Inhibitors of Activated STAT (PIAS) are a family of proteins that regulate the activity of signal transducer and activator of transcription (STAT) proteins, which are involved in various cellular processes such as differentiation, proliferation, and apoptosis. PIAS proteins function as E3 ubiquitin ligases and SUMO (small ubiquitin-like modifier) ligases, modifying STAT proteins and other transcription factors by adding SUMO molecules to them. This modification can alter the activity, localization, or stability of the target protein, thereby regulating its function in the cell. PIAS proteins have been shown to play a role in various physiological and pathological processes, including inflammation, cancer, and neurodegenerative diseases. Inhibiting PIAS proteins has emerged as a potential therapeutic strategy for the treatment of certain diseases associated with aberrant STAT activation.

Polycomb Repressive Complex 1 (PRC1) is a protein complex that plays a crucial role in the epigenetic regulation of gene expression, primarily through the process of histone modification. It is associated with the maintenance of gene repression during development and differentiation. PRC1 facilitates the monoubiquitination of histone H2A at lysine 119 (H2AK119ub1), leading to chromatin compaction and transcriptional silencing. This complex is composed of several core subunits, including BMI1, RING1A/B, and one of the six PCGF proteins, which define different PRC1 variants. Dysregulation of PRC1 has been implicated in various human diseases, such as cancers and developmental disorders.

Ubiquitination involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s, ubiquitin-conjugating enzymes ... Pickart CM (2001). "Mechanisms underlying ubiquitination". Annu. Rev. Biochem. 70: 503-33. doi:10.1146/annurev.biochem.70.1.503 ... mediate the ubiquitination of RING-finger proteins, ARA54 and RNF8". Eur. J. Biochem. 268 (9): 2725-32. doi:10.1046/j.1432- ...
Ben-Neriah Y (Jan 2002). "Regulatory functions of ubiquitination in the immune system". Nature Immunology. 3 (1): 20-6. doi: ... Pickart CM (2001). "Mechanisms underlying ubiquitination". Annual Review of Biochemistry. 70: 503-33. doi:10.1146/annurev. ... Protein ubiquitination and subsequent proteolysis and degradation by the proteasome are important mechanisms in the regulation ...
Phosphorylation and ubiquitination of Bim dependent on ERK caused Bim to be degraded in proteasomes. However, presence of ... Pickart, Cecile M. (June 2001). "Mechanisms Underlying Ubiquitination". Annual Review of Biochemistry. 70 (1): 503-533. doi: ... due to its previous ubiquitination. However, in cancer cells and CAFs, Snail1 degradation is prevented. One of the mechanisms ... the reversal process of ubiquitination, regulating the fate of ubiquitin-conjugated proteins. Based on its conserved catalytic ...
Busch H (1984). "[23] Ubiquitination of Proteins". Posttranslational Modifications Part A. pp. 238-262. doi:10.1016/0076-6879( ...
Either by autoubiquitination or by ubiquitination of a cofactor, it is then responsible for directing the virions to the ... Fukuda-Kamitani T, Kamitani T (July 2002). "Ubiquitination of Ro52 autoantigen". Biochemical and Biophysical Research ...
Additionally, histone ubiquitination is related to genomic maintenance, as ubiquitination of histone H2AX is involved in DNA ... Ubiquitination of the H2A core histone typically represses gene expression as it prevents methylation at H3K4, while H2B ... Ubiquitination is a post-translational modification involving the addition of ubiquitin proteins onto target proteins. Histones ... "Histone Ubiquitination - an overview , ScienceDirect Topics". www.sciencedirect.com. Retrieved 2021-12-02. Ikura T, Tashiro S, ...
Ubiquitination plays a central role in cell signaling that regulates processes including protein degradation and immunological ... Ubiquitination is a component of several immune responses. Without ubiquitin's proper functioning, the invasion of pathogens ... Trujillo, Marco; Shirasu, Ken (1 August 2010). "Ubiquitination in plant immunity". Current Opinion in Plant Biology. 13 (4): ... "Role of Ubiquitination in Plant Innate Immunity and Pathogen Virulence". Journal of Plant Biology. 53 (1): 10-18. doi:10.1007/ ...
STUB1 mediates the ubiquitination of the chaperone-bound client, which induces the recruitment of the autophagic ubiquitin ... Shaid S, Brandts CH, Serve H, Dikic I (January 2013). "Ubiquitination and selective autophagy". Cell Death Differ. 20 (1): 21- ...
... ubiquitination, triggered by K411-m1. Effects on biological progress - cell growth, inhibited, triggered by K411-m1. The effect ...
Ubiquitination occurs via the BRCA1 fusion protein and is abolished by zinc chelation. The enzyme activity of the fusion ... BARD1 is thought to be involved in the recognition and binding of protein targets for ubiquitination. It attaches to proteins ... Vandenberg CJ, Gergely F, Ong CY, Pace P, Mallery DL, Hiom K, Patel KJ (July 2003). "BRCA1-independent ubiquitination of FANCD2 ... The ring domain is an important element of ubiquitin E3 ligases, which catalyze protein ubiquitination. Ubiquitin is a small ...
Mono-ubiquitination is also required for interaction with the nuclease FAN1. FAN1 recruitment and its consequent activity ... Vandenberg CJ, Gergely F, Ong CY, Pace P, Mallery DL, Hiom K, Patel KJ (July 2003). "BRCA1-independent ubiquitination of FANCD2 ... Vandenberg CJ, Gergely F, Ong CY, Pace P, Mallery DL, Hiom K, Patel KJ (July 2003). "BRCA1-independent ubiquitination of FANCD2 ...
Ubiquitination has been observed to be most involved in the final stages of the connexin protein's lifecycle, regulating both ... The distinct effects of ubiquitination tend to vary widely, depending on the tissues and subcellular location where it occurs ... Leithe, Edward; Rivedal, Edgar (2007-11-14). "Ubiquitination of Gap Junction Proteins". Journal of Membrane Biology. 217 (1-3 ... Ubiquitination of Cx43 that is at the plasma membrane and organized into gap junctions will result in internalization, or ...
Ubiquitination involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s, ubiquitin-conjugating enzymes ... Vandenberg CJ, Gergely F, Ong CY, Pace P, Mallery DL, Hiom K, Patel KJ (July 2003). "BRCA1-independent ubiquitination of FANCD2 ... Kamura T, Sato S, Iwai K, Czyzyk-Krzeska M, Conaway RC, Conaway JW (2000). "Activation of HIF1α ubiquitination by a ... It also functions in the ubiquitination of the tumor-suppressor protein p53 and the hypoxia-inducible transcription factor ...
It is responsible for the mono-ubiquitination of histone H2A at lysine 119, a modification commonly associated with ... Weake VM, Workman JL (March 2008). "Histone ubiquitination: triggering gene activity". Molecular Cell. 29 (6): 653-63. doi: ...
Vandenberg CJ, Gergely F, Ong CY, Pace P, Mallery DL, Hiom K, Patel KJ (Jul 2003). "BRCA1-independent ubiquitination of FANCD2 ...
Ubiquitination--more than two to tango". Science. 289 (5487): 2061-2. doi:10.1126/science.289.5487.2061. PMID 11032556. S2CID ... Ubiquitination involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s, ubiquitin-conjugating enzymes ... 1999). "Structure of an E6AP-UbcH7 complex: insights into ubiquitination by the E2-E3 enzyme cascade". Science. 286 (5443): ...
Their activity is modified by many post translational activities, such as phosphorylation, ubiquitination, and acetylation. ... "Mdm2 induces mono-ubiquitination of FOXO4". PLOS ONE. 3 (7): e2819. Bibcode:2008PLoSO...3.2819B. doi:10.1371/journal.pone. ...
2003). "BRCA1-independent ubiquitination of FANCD2". Mol. Cell. 12 (1): 247-254. doi:10.1016/S1097-2765(03)00281-8. PMID ... "Differential functions of FANCI and FANCD2 ubiquitination stabilize ID2 complex on DNA". EMBO Reports. 21 (7): e50133. doi: ... "Structural and biochemical basis of interdependent FANCI-FANCD2 ubiquitination". The EMBO Journal. 42 (3): e111898. doi: ...
2009). Ubiquitination and Death-receptor signaling. Death Receptors and Cognate Ligands in Cancer. Results Probl Cell Differ, ...
Song MS, Song SJ, Kim SY, Oh HJ, Lim DS (July 2008). "The tumour suppressor RASSF1A promotes MDM2 self-ubiquitination by ... Brenkman AB, de Keizer PL, van den Broek NJ, Jochemsen AG, Burgering BM (2008). "Mdm2 induces mono-ubiquitination of FOXO4". ... and the ubiquitination of histone 2A at lysine 119 (H2AK119) at its target genes. Removing MDM2 simultaneously with the H2AK119 ... Several lysine residues in p53 C-terminus have been identified as the sites of ubiquitination, and it has been shown that p53 ...
Ubiquitination involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s, ubiquin-conjugating enzymes, ... Ubiquitination--more than two to tango". Science. 289 (5487): 2061-2. doi:10.1126/science.289.5487.2061. PMID 11032556. S2CID ... 1999). "Structure of an E6AP-UbcH7 complex: insights into ubiquitination by the E2-E3 enzyme cascade". Science. 286 (5443): ... Webster JM, Tiwari S, Weissman AM, Wojcikiewicz RJ (2003). "Inositol 1,4,5-trisphosphate receptor ubiquitination is mediated by ...
Ubiquitination of histone H2B in response to DNA damage is important for timely initiation of DNA repair. Ubiquitinase RNF20/ ... Histone ubiquitination in the DNA damage response. DNA Repair (Amst). 2017 Aug;56:92-101. doi: 10.1016/j.dnarep.2017.06.011. ... Acetylation and ubiquitination are examples of two post-translational modifications that affect the function of histone H2B in ... Acetylation, phosphorylation, and ubiquitination are the most common and most studied modifications of histone H2B. Histone H2B ...
2002). "Histone ubiquitination: a tagging tail unfolds?". BioEssays. 24 (2): 166-74. doi:10.1002/bies.10038. PMID 11835281. ...
Site specific ubiquitination of histone H2A has a role in the recruitment of DNA repair proteins to DNA double strand breaks ... Histone ubiquitination in the DNA damage response. DNA Repair (Amst). 2017 Aug;56:92-101. doi: 10.1016/j.dnarep.2017.06.011. ... In the DNA damage response, it is thought that ubiquitination of H2A by the BRCA1/BARD1 heterodimer promotes homologous ... recombination, and that ubiquitination of H2A by RNF168 protein promotes non-homologous end joining. H2A is coded by many genes ...
Yang WL, Zhang X, Lin HK (August 2010). "Emerging role of Lys-63 ubiquitination in protein kinase and phosphatase activation ... The regulation could be related to ubiquitin function; proteins with ubiquitination type function were found to interact with ... Furthermore, multiple proteins were involved in ubiquitination. Some of the interacting yeast proteins with the higher ... ubiquitination, and ribosomes in the cytoplasm. The basis of these arguments is on the interaction data of human CXorf26 as ...
... promotes the skipping of exon 9 of the NUMB transcript, producing a NUMB isoform that causes ubiquitination followed by ... June 2014). "A COFRADIC protocol to study protein ubiquitination". Journal of Proteome Research. 13 (6): 3107-13. doi:10.1021/ ... July 2018). "UbiSite approach for comprehensive mapping of lysine and N-terminal ubiquitination sites". Nature Structural & ...
In COS-1 monkey kidney cells transfected with mouse HAS2 and HAS3 plasmids, one site of ubiquitination is seen on the lysine at ... "The Activity of Hyaluronan Synthase 2 Is Regulated by Dimerization and Ubiquitination". Journal of Biological Chemistry. 285 ( ... HAS2 is regulated by dimerization and ubiquitination. ...
"Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI)". MBL International. Retrieved 2019-12-12. ScienceDirect - Cell ...
Song MS, Song SJ, Kim SY, Oh HJ, Lim DS (July 2008). "The tumour suppressor RASSF1A promotes MDM2 self-ubiquitination by ... Carpentier I, Coornaert B, Beyaert R (October 2008). "Smurf2 is a TRAF2 binding protein that triggers TNF-R2 ubiquitination and ... Ott DE, Coren LV, Chertova EN, Gagliardi TD, Schubert U (December 2000). "Ubiquitination of HIV-1 and MuLV Gag". Virology. 278 ... "cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination". Molecular Cell ...
HO-1 enzymes are degraded via ubiquitination. The enzyme has been the subject of extensive investigation into its regulatory ...
UBE4A catalyzes NRF1 ubiquitination and facilitates DDI2-mediated NRF1 cleavage. Title: UBE4A catalyzes NRF1 ubiquitination and ... UBE4A ubiquitination factor E4A [Homo sapiens] UBE4A ubiquitination factor E4A [Homo sapiens]. Gene ID:9354 ... ubiquitination factor E4Aprovided by HGNC. Primary source. HGNC:HGNC:12499 See related. Ensembl:ENSG00000110344 MIM:603753; ... UBE4A catalyzes NRF1 ubiquitination and facilitates DDI2-mediated NRF1 cleavage. Hu X, et al. Biochim Biophys Acta Gene Regul ...
... : Use of mouse models for analyzing ubiquitination processes ... The role of the ubiquitination-proteasome pathway in breast cancer: Applying drugs that affect the ubiquitin-proteasome pathway ... The role of the ubiquitination-proteasome pathway in breast cancer: Ubiquitin mediated degradation of growth factor receptors ... The role of the ubiquitination-proteasome pathway in breast cancer. Nancy Hynes ...
Study Rationale: Parkinsons disease (PD) involves accumulation of toxic proteins in cells in specific areas of the brain. These cells normally have protective mechanisms to remove these toxic proteins and dysfunction of these mechanisms may play a role in the development of PD. A major goal of this grant is to explore novel ways to allow these cells to restore their ability to remove these toxic proteins.Hypothesis:Key members of the deubiquitinase (DUB) family of enzymes are thought to play a central role in the development and progression of PD by allowing the build-up of toxic proteins. This study will determine if inhibitors of these enzymes can prevent or halt the progression of PD. Study Design:The grant recipients have expertise in the identification of drugs that block the action of DUBs. The team will apply this expertise to DUB family members that are thought to play a central role in the development and progression of PD to find new drugs. Once candidate drugs are identified, the team will
... Nature. 2004 Aug 5;430(7000):694-9. doi ...
Additionally, MG-132 is a broad-spectrum ubiquitination inhibitor that generally blocks all ubiquitination pathways, making it ... 2003) Ubiquitination regulates PSD-95 degradation and AMPA receptor surface expression. Neuron 40:595-607, doi:10.1016/S0896- ... 2010) Ubiquitination acutely regulates presynaptic neurotransmitter release in mammalian neurons. J Neurosci 30:3157-3166, doi: ... 2007) SCRAPPER-dependent ubiquitination of active zone protein RIM1 regulates synaptic vesicle release. Cell 130:943-957, doi: ...
Background: Cholangiocarcinoma (CCA) represents the epithelial cell cancer with high aggressiveness whose five-year survival rate is poor with standard treatment. Calcyclin-binding protein (CACYBP) shows aberrant expression ... | Find, read and cite all the research you need on Tech Science Press
title = "Structural basis of specific H2A K13/K15 ubiquitination by RNF168",. abstract = "Ubiquitination of chromatin by ... Ubiquitination of chromatin by modification of histone H2A is a critical step in both regulation of DNA repair and regulation ... Structural basis of specific H2A K13/K15 ubiquitination by RNF168. / Horn, Velten ; Uckelmann, Michael; Zhang, Heyi et al. In: ... N2 - Ubiquitination of chromatin by modification of histone H2A is a critical step in both regulation of DNA repair and ...
RNF112-mediated FOXM1 ubiquitination suppresses the proliferation and invasion of gastric cancer. ... RNF112-mediated FOXM1 ubiquitination suppresses the proliferation and invasion of gastric cancer. ... which further promoted FOXM1 ubiquitination and subsequently exerted promising anticancer effects in vitro and in vivo. ...
TRAF6-mediated ubiquitination of MST1/STK4 attenuates the TLR4-NF-κB signaling pathway in macrophages.. Sep 28, 2020 ... In addition, we found that TRAF6 mediates the LPS-induced activation of MST1/STK4 by catalyzing its ubiquitination, resulting ...
Ubiquitination and Degradation of Neuronal Nitric-Oxide Synthase in Vitro: Dimer Stabilization Protects the Enzyme from ... Ubiquitination and Degradation of Neuronal Nitric-Oxide Synthase in Vitro: Dimer Stabilization Protects the Enzyme from ... Ubiquitination and Degradation of Neuronal Nitric-Oxide Synthase in Vitro: Dimer Stabilization Protects the Enzyme from ... Ubiquitination and Degradation of Neuronal Nitric-Oxide Synthase in Vitro: Dimer Stabilization Protects the Enzyme from ...
... deficiency il 17 district il 17 drugs il17a il17d cd93 il17d receptor il17f il17ra il17rb il17rd kif4a hbv kif4a ubiquitination ...
These modifications did not affect any biochemical properties of pol but prevented its ubiquitination and its subsequent ... These modifications did not affect any biochemical properties of pol but prevented its ubiquitination and its subsequent ... These modifications did not affect any biochemical properties of pol but prevented its ubiquitination and its subsequent ...
This review outlines the current understandings of the regulatory mechanisms and assay capabilities of ubiquitination, ... It will further highlight the role of ubiquitination, SUMOylation, and NEDDylation in tumorigenesis. ... into ubiquitin and ubiquitin-like pathways involved in cancer biology reveal a potential interplay between ubiquitination, ... The Next Frontier: Translational Development of Ubiquitination, SUMOylation, and NEDDylation in Cancer. ...
Role of IkappaBalpha ubiquitination in signal-induced activation of NFkappaB in vivo. ... Thus signal-induced activation of NF-kappaB involves phosphorylation-dependent ubiquitination of IkappaBalpha, which targets ... that is neither phosphorylated nor degraded in response to signal induction fails to undergo inducible ubiquitination in vivo. ...
WWP2 interacts with transcription factor IRF7, promoting its non-degradative mono-ubiquitination, nuclear translocation and ... In addition to their physical interaction, we observed that WWP2 promotes the ubiquitination of IRF7. Ubiquitination assay ... 6: WWP2 regulates IRF7 transcriptional activity in macrophages through non-degradative ubiquitination.. ... In addition to having relatively higher ubiquitination, WT BMDMs showed higher levels of IRF7 (Fig. 5h, i) and p-IRF7 (Fig. 6d ...
BARD1 reads H2A lysine 15 ubiquitination to direct homologous recombination. Research output: Contribution to journal › Journal ...
... www.aklectures.com/lecture/ubiquitination-of-proteins Facebook link: https://www.facebook.com/aklectures Website link: http:// ... Ubiquitination of Proteins Donate here: http://www.aklectures.com/donate.php Buy shirt: https://www.etsy.com/listing/458728846/ ... Website video: http://www.aklectures.com/lecture/ubiquitination-of-proteins. Facebook link: https://www.facebook.com/aklectures ... short-sleeve-t-shirt-white?ref=shop_home_active_1 Website video: http://www.aklectures.com/lecture/ubiquitination-of-proteins ...
Keywords: Ubiquitination Pathway, Ubiquitination Autophagy, Ubiquitin Signaling. 1. Ubiquitin-mediated Autophagy Pathway. ... Currently, ubiquitination is involved in most of cellular processes, signaling pathway and regulation of diseases.. ... Ubiquitination refers to the ubiquitin-protein conjugation process. Ubiquitin usually conjugates with proteins by different ...
Ubiquitination. Another highly dynamic PTM that is implicated in signaling pathways is ubiquitination. Ub (ubiquitin) is a ... Ubiquitination involves one or more covalent additions of Ub to the lysine residues of target proteins (i.e., mono- or poly- ... 2002). SEL-10 interacts with presenilin 1, facilitates its ubiquitination, and alters A-beta peptide production. J. Neurochem. ... 2016). Rapid identification of ubiquitination and SUMOylation target sites by microfluidic peptide array. Biochem. Biophys. Rep ...
We here report that APE1 is also modified by ubiquitination. APE1 ubiquitination occurred specifically at Lys residues near the ... We here report that APE1 is also modified by ubiquitination. APE1 ubiquitination occurred specifically at Lys residues near the ... We here report that APE1 is also modified by ubiquitination. APE1 ubiquitination occurred specifically at Lys residues near the ... We here report that APE1 is also modified by ubiquitination. APE1 ubiquitination occurred specifically at Lys residues near the ...
BARD1 reads H2A lysine 15 ubiquitination to direct homologous recombination Becker JR., Clifford G., Bonnet C., Groth A., ...
Histone Ubiquitination : An Integrative Signaling Platform in Genome Stability. In: Trends in Genetics. 2021 ; Vol. 37, Nr. 6. ... Ubiquitination of chromatin plays a central role in these mechanisms. The ever-growing complexity of the ubiquitin (Ub) code ... Ubiquitination of chromatin plays a central role in these mechanisms. The ever-growing complexity of the ubiquitin (Ub) code ... Ubiquitination of chromatin plays a central role in these mechanisms. The ever-growing complexity of the ubiquitin (Ub) code ...
A retrospective graph review was done using the computerized individual record system in the Memphis VA INFIRMARY for 614 individuals with type 2 diabetes and previously collected supplement B12 levels. Individuals were excluded if indeed they were older than 60 years; on the vegetarian diet; have been identified as having pernicious anemia, recorded with a… Continue reading A retrospective graph review was done using the computerized individual record. ...
... Gerlach, Björn;Cordier, Stefanie M.;Schmukle, ... We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced ... We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced ... Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating ...
Mechanisms of ubiquitination. Even though ubiquitination participates in most, if not all cellular processes, many questions in ... These include questions regarding the basis of the ubiquitination process. The final step is carried out by the ubiquitin ...
Ubiquitin E3 ligases are present at the outer membrane of both organelles where they mediate ubiquitination and turnover of ... Ubiquitin E3 ligases are present at the outer membrane of both organelles where they mediate ubiquitination and turnover of ...
Beyond K48 and K63: non-canonical protein ubiquitination. Overview of attention for article published in Cellular & Molecular ... RNF144A promotes antiviral responses by modulating STING ubiquitination. Article in EMBO Reports (November 2023) ...
For postnatal development and regeneration of skeletal muscle mass satellite television cells Mirtazapine a self-renewing pool of muscle mass stem cells give rise to child myogenic precursor cells that contribute to the formation of fresh muscle mass fibers. This review outlines recent findings concerning hematopoietic stem/progenitor cell populations residing in adult skeletal muscle mass and… Continue reading For postnatal development and regeneration of skeletal muscle mass satellite television. ...
... MANDILI, GIORGIA;KHADJAVI, AMINA;GALLO, ... Here we show that nanomolar Doxorubicin treatment of neuroblastoma cells caused: (a) dose-dependent over-ubiquitination of a ... Here we show that nanomolar Doxorubicin treatment of neuroblastoma cells caused: (a) dose-dependent over-ubiquitination of a ...
  • Turnover of several regulatory proteins results from targeted destruction via ubiquitination and subsequent degradation through the proteosome. (biomedcentral.com)
  • Nevertheless, the mechanism for ubiquitination-regulated turnover of presynaptic active zone proteins in the progression of spinal plasticity-associated neuropathic pain remains unclear. (jneurosci.org)
  • Studies investigating pain pathology have demonstrated that ubiquitination contributes to chronic pain by regulating the turnover of synaptic proteins. (jneurosci.org)
  • Ubiquitin E3 ligases are present at the outer membrane of both organelles where they mediate ubiquitination and turnover of other organellar proteins. (ox.ac.uk)
  • In addition, SARS-CoV-2 proteins were found to be ubiquitinated during infection despite the fact that SARS-CoV-2 itself didn't code any E3 ligase, and that ubiquitination at three sites on the Spike protein could significantly enhance viral infection. (gob.pe)
  • Here we show that ubiquitination can also regulate proteins by transiently inactivating enzymatic function through conformational change in a dimeric enzyme, which can be reversed upon deubiquitination. (deiodinase.org)
  • The protein produced from the RNF216 gene is involved in a cellular process, called ubiquitination, by which unneeded proteins are tagged with a molecule called ubiquitin. (medlineplus.gov)
  • That is, RIM1α-facilitated Ca V 2.2 expression plays a role in the downstream signaling of Fbxo3-dependent Fbxl2 ubiquitination/degradation to promote spinal plasticity underlying the progression of nociceptive hypersensitivity following neuropathic injury. (jneurosci.org)
  • SIGNIFICANCE STATEMENT Ubiquitination is a well known process required for protein degradation. (jneurosci.org)
  • The mechanism of this selective proteolytic degradation, or in essence how the nNOS becomes labilized and recognized for ubiquitination and subsequent degradation, has not been determined. (aspetjournals.org)
  • Downmodulation of MDM2 increased APE1 level, suggesting that MDM2-mediated ubiquitination can be a signal for APE1 degradation. (uky.edu)
  • Among the diverse signaling outcomes associated with ubiquitination, the most well-established is the targeted degradation of substrates via the proteasome. (intechopen.com)
  • Critical cysteine residues of Keap1 in suppression of Nrf2 basal activity and arsenic-sensing by regulating the ubiquitination-proteasomal degradation of Nrf2 protein. (cdc.gov)
  • Activation of Nrf2 involves modulation of Keap1/Cul3-mediated ubiquitination-proteasomal degradation of Nrf2. (cdc.gov)
  • Prolactin stimulates ubiquitination, initial internalization, and degradation of its receptor via catalytic activation of Janus kinase 2. (medscape.com)
  • We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced cell death and, thereby, prevents inflammation. (unicatt.it)
  • Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating immune signalling. (unicatt.it)
  • Until now, there's little information about linear ubiquitination or SHARPIN in mouse platelets. (wearecellix.com)
  • Platelets from cpdm/cpdm mice do not express SHARPIN, have reduced linear ubiquitination and show reduced accumulation on collagen under shear. (wearecellix.com)
  • Platelets from cpdm/cpdm mice don't express SHARPIN and have reduced linear ubiquitination. (wearecellix.com)
  • 01J09219) Investigating the effects of regulatory phosphorylation and linear ubiquitination on Pyrin-mediated auto-inflammatory disorders. (ugent.be)
  • In this project, we will investigate the mechanisms by which Pyrin signaling is regulated by phosphorylation and linear ubiquitination, and how this modulates Pyrin-mediated auto-inflammation in FMF and ORAS mouse models. (ugent.be)
  • Here we review the current knowledge on specific, low-abundant histone ubiquitination events that are highly regulated within the cellular DNA damage response (DDR), with particular emphasis on the latest discovery of Ub phosphorylation as a novel regulator of the DDR signaling pathway. (knaw.nl)
  • Ubiquitination and phosphorylation are reversible posttranslational protein modifications regulating physiological and pathological processes. (life-science-alliance.org)
  • DSBs elicit a signalling cascade that modifies the chromatin surrounding the break, first by ATM-dependent phosphorylation and then by RNF8-, RNF168- and BRCA1-dependent regulatory ubiquitination. (elsevierpure.com)
  • In addition, Fbxo3 modifies this pathway by inhibiting Fbxl2-mediated RIM1α ubiquitination, suggesting that presynaptic protein ubiquitination makes a crucial contribution to the development of neuropathic pain. (jneurosci.org)
  • TRAF6-mediated ubiquitination of MST1/STK4 attenuates the TLR4-NF-κB signaling pathway in macrophages. (physiciansweekly.com)
  • In addition, we found that TRAF6 mediates the LPS-induced activation of MST1/STK4 by catalyzing its ubiquitination, resulting in negative feedback regulation by MST1/STK4 of the LPS-induced pathway leading to cytokine production in macrophages. (physiciansweekly.com)
  • Currently, ubiquitination is involved in most of cellular processes, signaling pathway and regulation of diseases. (fn-test.com)
  • The ever-growing complexity of the ubiquitin (Ub) code and of chromatin modifications and dynamics challenges our ability to fully understand how histone ubiquitination regulates genome stability. (knaw.nl)
  • WWP2 interacts with transcription factor IRF7, promoting its non-degradative mono-ubiquitination, nuclear translocation and transcriptional activity, leading to upregulation of Ccl5 at transcriptional level. (nature.com)
  • The structural model highlights the role of E3 and nucleosome in promoting ubiquitination and provides a basis for understanding and engineering of chromatin ubiquitination specificity. (monash.edu)
  • Here we report that OTUB1, a deubiquitinating enzyme, is an inhibitor of DSB-induced chromatin ubiquitination. (elsevierpure.com)
  • Even though ubiquitination participates in most, if not all cellular processes, many questions in the field remain unanswered. (trujillolab.com)
  • Ubiquitination of chromatin by modification of histone H2A is a critical step in both regulation of DNA repair and regulation of cell fate. (monash.edu)
  • These results reveal a novel regulation of APE1 through ubiquitination. (uky.edu)
  • Dynamic regulation of endosymbiotic organelles by ubiquitination. (ox.ac.uk)
  • Scholars@Duke publication: {beta}-Arrestin is crucial for ubiquitination and down-regulation of the insulin-like growth factor-1 receptor by acting as adaptor for the MDM2 E3 ligase. (duke.edu)
  • In this way, beta-arrestin acts as a crucial component in the ubiquitination and down-regulation of the receptor. (duke.edu)
  • Here, we provide the first insight into the regulation of BCL2A1 ubiquitination. (edu.au)
  • One area of particular interest is the role of ubiquitination in directing receptor translocation to intralumenal vesicles and its requirement or otherwise for trafficking to the lysosome. (abdn.ac.uk)
  • B) Detection of linear (Met1-linked) ubiquitination by LUB9 antibody in basal and stimulated wild-type and cpdm/cpdm platelets. (wearecellix.com)
  • We after that immunoprecipated lysates with anti-Nrf2 antibody and blotted with anti-HA antibody to identify ubiquitination. (rawveronica.com)
  • This review outlines the current understandings of the regulatory mechanisms and assay capabilities of ubiquitination, SUMOylation, and NEDDylation. (ox.ac.uk)
  • Ubiquitination of chromatin plays a central role in these mechanisms. (knaw.nl)
  • APE1 ubiquitination occurred specifically at Lys residues near the N-terminus, and was markedly enhanced by mouse double minute 2 (MDM2), the major intracellular p53 inhibitor. (uky.edu)
  • Moreover, DNA-damaging reagents and nutlin-3, an inhibitor of MDM2-p53 interaction, increased APE1 ubiquitination in the presence of p53. (uky.edu)
  • Spinal plasticity, a key process mediating neuropathic pain development, requires ubiquitination-dependent protein turnover. (jneurosci.org)
  • These results confirm the prediction that spinal presynaptic Fbxo3-dependent Fbxl2 ubiquitination promotes the subsequent RIM1α/Ca V 2.2 cascade in SNL-induced neuropathic pain. (jneurosci.org)
  • This multiomics combined with high-throughput screening study reveals that SARS-CoV-2 not only modulates innate immunity, but also promotes viral infection, by hijacking ubiquitination-specific processes, highlighting potential antiviral and anti-inflammation targets. (gob.pe)
  • Surprisingly, we found that OTUB1 suppresses RNF168-dependent poly-ubiquitination independently of its catalytic activity. (elsevierpure.com)
  • Ubiquitination is a critical posttranslational regulator of protein stability and/or subcellular localization. (deiodinase.org)
  • Data now suggest that delocalization, accumulation, and ubiquitination of TDP-43 in the cytoplasm of motor neurons are early dysfunctions in the cascade of the events leading to motor neuron degeneration in ALS. (medscape.com)
  • To verify that Arkadia was actually mediating the Glyparamide forming of the HA-Ub-Nrf2 types, HepG2 cells had been transfected with either Arkadia WT, an Arkadia*Band mutant (a mutation that makes Arkadia not capable of catalyzing ubiquitination), or with an Arkadia*SIM mutant plasmid (a mutation where all three SIMs of Arkadia Glyparamide are mutated to disrupt identification of sumoylated substrates) [32] accompanied by treatment with arsenic trioxide (As203). (rawveronica.com)
  • Our outcomes present a rise in the known degrees of Nrf2 ubiquitination in cells expressing WT Arkadia and needlessly to say, a reduction in the known degrees of Nrf2 ubiquitination in cells expressing both Band and SIM mutants. (rawveronica.com)
  • This shows that Nrf2 ubiquitination was mediated by expressed Arkadia exogenously. (rawveronica.com)
  • Overexpression of Arkadia stabilizes Nrf2 To see whether the Arkadia-mediated ubiquitination of polysumoylated Nrf2 happened within PML-NB domains, we isolated PML-NBs from Simply because2O3-treated HepG2 cells and blotted for portrayed Arkadia exogenously. (rawveronica.com)
  • This shows that the ID1 consequences of Arkadia-mediated ubiquitination of Nrf2 may occur in PML-NBs. (rawveronica.com)
  • Numerous E3 ubiquitin ligases, which facilitate the ubiquitination of specific substrates, have been shown to control G1/S. In this chapter, we will discuss components in the ubiquitin proteasome system that are implicated in G1/S control, how these enzymes are interconnected, gaps in our current knowledge, and the potential role of these pathways in the cancer cycle and disease proliferation. (intechopen.com)
  • We discuss players involved and potential implications of histone (phospho)ubiquitination on chromatin structure, and we highlight exciting open questions for future research. (knaw.nl)
  • Role of NOXA and its ubiquitination in proteasome inhibitor-induced apoptosis in chronic lymphocytic leukemia cells. (le.ac.uk)
  • Novel insights into ubiquitin and ubiquitin-like pathways involved in cancer biology reveal a potential interplay between ubiquitination, SUMOylation, and NEDDylation. (ox.ac.uk)
  • This interplay is regulated by the ubiquitination of non-phosphorylated RUNX2-S118, which is evident in the dominant mutant RUNX2-S118D. (uohyd.ac.in)
  • Interestingly, the well-established small-molecule compound RCM-1 significantly enhanced the interaction between RNF112 and FOXM1, which further promoted FOXM1 ubiquitination and subsequently exerted promising anticancer effects in vitro and in vivo. (jci.org)
  • This can involve ubiquitin regulated sorting by the ESCRT complex (2), initial ubiquitin-indendent sorting (possibly by GASP) before ubiquitin and ESCRT mediated transfer to Intralumenal vesicles (3) or direct interaction with the ESCRT III component, independent of ubiquitination (4) mediated by ALIX. (abdn.ac.uk)
  • Peli1 causes nondegradative ubiquitination of TSC1 and promotes TSC1-TSC2 dimerization and TSC2 stabilization. (medscape.com)
  • A role for SHARPIN in platelet linear protein ubiquitination and function - By Moore et al. (wearecellix.com)
  • The molecular origin of ubiquitination site specificity by these related E3 enzymes is one of the open questions in the field. (monash.edu)
  • Among them are enzymes involved in altering the host-ubiquitination system. (elsevierpure.com)
  • Ubiquitination involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s, ubiquitin-conjugating enzymes, or E2s, and ubiquitin-protein ligases, or E3s. (wikipedia.org)
  • It will further highlight the role of ubiquitination, SUMOylation, and NEDDylation in tumorigenesis. (ox.ac.uk)
  • Ubiquitination and NfKB play a role in platelet function. (wearecellix.com)
  • Indeed, endogenous TRIM28 and BCL2A1 bind to each other at the mitochondria and TRIM28 knock-down decreases BCL2A1 ubiquitination. (edu.au)
  • Ubiquitination-Induced Conformational Change within the Deiodinase Dimer Is a Switch Regulating Enzyme Activity. (deiodinase.org)
  • In mouse P6 cells (overexpressing human IGF-1R) absence of beta-arrestin 1, but not of beta-arrestin 2, blocked ubiquitination of IGF-1R. (duke.edu)
  • Ubiquitination refers to the ubiquitin-protein conjugation process. (fn-test.com)
  • These include questions regarding the basis of the ubiquitination process. (trujillolab.com)

No images available that match "ubiquitination"