A biosensing technique in which biomolecules capable of binding to specific analytes or ligands are first immobilized on one side of a metallic film. Light is then focused on the opposite side of the film to excite the surface plasmons, that is, the oscillations of free electrons propagating along the film's surface. The refractive index of light reflecting off this surface is measured. When the immobilized biomolecules are bound by their ligands, an alteration in surface plasmons on the opposite side of the film is created which is directly proportional to the change in bound, or adsorbed, mass. Binding is measured by changes in the refractive index. The technique is used to study biomolecular interactions, such as antigen-antibody binding.
Any of a variety of procedures which use biomolecular probes to measure the presence or concentration of biological molecules, biological structures, microorganisms, etc., by translating a biochemical interaction at the probe surface into a quantifiable physical signal.
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.
A yellow metallic element with the atomic symbol Au, atomic number 79, and atomic weight 197. It is used in jewelry, goldplating of other metals, as currency, and in dental restoration. Many of its clinical applications, such as ANTIRHEUMATIC AGENTS, are in the form of its salts.
Non-invasive method of demonstrating internal anatomy based on the principle that atomic nuclei in a strong magnetic field absorb pulses of radiofrequency energy and emit them as radiowaves which can be reconstructed into computerized images. The concept includes proton spin tomographic techniques.
Proteins that are chemically bound to a substrate material which renders their location fixed. The immobilization of proteins allows their use in chemical reactions without being diluted by solvent.
The rate dynamics in chemical or physical systems.
Measurement of the index of refraction (the ratio of the velocity of light or other radiation in the first of two media to its velocity in the second as it passes from one into the other).
Nanoparticles produced from metals whose uses include biosensors, optics, and catalysts. In biomedical applications the particles frequently involve the noble metals, especially gold and silver.
The parts of a macromolecule that directly participate in its specific combination with another molecule.
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.
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.
Spectroscopic method of measuring the magnetic moment of elementary particles such as atomic nuclei, protons or electrons. It is employed in clinical applications such as NMR Tomography (MAGNETIC RESONANCE IMAGING).
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.
Models used experimentally or theoretically to study molecular shape, electronic properties, or interactions; includes analogous molecules, computer-generated graphics, and mechanical structures.
Proteins prepared by recombinant DNA technology.
Silver. An element with the atomic symbol Ag, atomic number 47, and atomic weight 107.87. It is a soft metal that is used medically in surgical instruments, dental prostheses, and alloys. Long-continued use of silver salts can lead to a form of poisoning known as ARGYRIA.
Thin strands of transparent material, usually glass, that are used for transmitting light waves over long distances.
Characteristics or attributes of the outer boundaries of objects, including molecules.
A measure of the binding strength between antibody and a simple hapten or antigen determinant. It depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. It includes the concept of "avidity," which refers to the strength of the antigen-antibody bond after formation of reversible complexes.
A molecule that binds to another molecule, used especially to refer to a small molecule that binds specifically to a larger molecule, e.g., an antigen binding to an antibody, a hormone or neurotransmitter binding to a receptor, or a substrate or allosteric effector binding to an enzyme. Ligands are also molecules that donate or accept a pair of electrons to form a coordinate covalent bond with the central metal atom of a coordination complex. (From Dorland, 27th ed)
The measurement of the quantity of heat involved in various processes, such as chemical reactions, changes of state, and formations of solutions, or in the determination of the heat capacities of substances. The fundamental unit of measurement is the joule or the calorie (4.184 joules). (McGraw-Hill Dictionary of Scientific and Technical Terms, 4th ed)
Members of the class of compounds composed of AMINO ACIDS joined together by peptide bonds between adjacent amino acids into linear, branched or cyclical structures. OLIGOPEPTIDES are composed of approximately 2-12 amino acids. Polypeptides are composed of approximately 13 or more amino acids. PROTEINS are linear polypeptides that are normally synthesized on RIBOSOMES.
A 60-kDa extracellular protein of Streptomyces avidinii with four high-affinity biotin binding sites. Unlike AVIDIN, streptavidin has a near neutral isoelectric point and is free of carbohydrate side chains.
The characteristic 3-dimensional shape of a protein, including the secondary, supersecondary (motifs), tertiary (domains) and quaternary structure of the peptide chain. PROTEIN STRUCTURE, QUATERNARY describes the conformation assumed by multimeric proteins (aggregates of more than one polypeptide chain).
Ligand-binding assays that measure protein-protein, protein-small molecule, or protein-nucleic acid interactions using a very large set of capturing molecules, i.e., those attached separately on a solid support, to measure the presence or interaction of target molecules in the sample.
The interaction of two or more substrates or ligands with the same binding site. The displacement of one by the other is used in quantitative and selective affinity measurements.
A change from planar to elliptic polarization when an initially plane-polarized light wave traverses an optically active medium. (McGraw-Hill Dictionary of Scientific and Technical Terms, 4th ed)
A highly acidic mucopolysaccharide formed of equal parts of sulfated D-glucosamine and D-glucuronic acid with sulfaminic bridges. The molecular weight ranges from six to twenty thousand. Heparin occurs in and is obtained from liver, lung, mast cells, etc., of vertebrates. Its function is unknown, but it is used to prevent blood clotting in vivo and vitro, in the form of many different salts.
Partial proteins formed by partial hydrolysis of complete proteins or generated through PROTEIN ENGINEERING techniques.
The development and use of techniques to study physical phenomena and construct structures in the nanoscale size range or smaller.
A family of calcium-binding alpha-globulins that are synthesized in the LIVER and play an essential role in maintaining the solubility of CALCIUM in the BLOOD. In addition the fetuins contain aminoterminal cystatin domains and are classified as type 3 cystatins.
A collection of cloned peptides, or chemically synthesized peptides, frequently consisting of all possible combinations of amino acids making up an n-amino acid peptide.
Incorporation of biotinyl groups into molecules.
The level of protein structure in which regular hydrogen-bond interactions within contiguous stretches of polypeptide chain give rise to alpha helices, beta strands (which align to form beta sheets) or other types of coils. This is the first folding level of protein conformation.
NMR spectroscopy on small- to medium-size biological macromolecules. This is often used for structural investigation of proteins and nucleic acids, and often involves more than one isotope.
The study of crystal structure using X-RAY DIFFRACTION techniques. (McGraw-Hill Dictionary of Scientific and Technical Terms, 4th ed)
The adhesion of gases, liquids, or dissolved solids onto a surface. It includes adsorptive phenomena of bacteria and viruses onto surfaces as well. ABSORPTION into the substance may follow but not necessarily.
Nucleotide sequences, generated by iterative rounds of SELEX APTAMER TECHNIQUE, that bind to a target molecule specifically and with high affinity.
The process by which two molecules of the same chemical composition form a condensation product or polymer.
Antibodies produced by a single clone of cells.
Proteins found in any species of bacterium.
Antibodies that are chemically bound to a substrate material which renders their location fixed.
Enzymes which are immobilized on or in a variety of water-soluble or water-insoluble matrices with little or no loss of their catalytic activity. Since they can be reused continuously, immobilized enzymes have found wide application in the industrial, medical and research fields.
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.
Nanometer-sized tubes composed of various substances including carbon (CARBON NANOTUBES), boron nitride, or nickel vanadate.
A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
A water-soluble, enzyme co-factor present in minute amounts in every living cell. It occurs mainly bound to proteins or polypeptides and is abundant in liver, kidney, pancreas, yeast, and milk.
The use of a quartz crystal microbalance for measuring weights and forces in the micro- to nanogram range. It is used to study the chemical and mechanical properties of thin layers, such as polymer coatings and lipid membranes; and interactions between molecues.
The measurement of the amplitude of the components of a complex waveform throughout the frequency range of the waveform. (McGraw-Hill Dictionary of Scientific and Technical Terms, 6th ed)
A methodology for chemically synthesizing polymer molds of specific molecules or recognition sites of specific molecules. Applications for molecularly imprinted polymers (MIPs) include separations, assays and biosensors, and catalysis.
The degree of similarity between sequences of amino acids. This information is useful for the analyzing genetic relatedness of proteins and species.
A species of gram-negative, facultatively anaerobic, rod-shaped bacteria (GRAM-NEGATIVE FACULTATIVELY ANAEROBIC RODS) commonly found in the lower part of the intestine of warm-blooded animals. It is usually nonpathogenic, but some strains are known to produce DIARRHEA and pyogenic infections. Pathogenic strains (virotypes) are classified by their specific pathogenic mechanisms such as toxins (ENTEROTOXIGENIC ESCHERICHIA COLI), etc.
Proteins that share the common characteristic of binding to carbohydrates. Some ANTIBODIES and carbohydrate-metabolizing proteins (ENZYMES) also bind to carbohydrates, however they are not considered lectins. PLANT LECTINS are carbohydrate-binding proteins that have been primarily identified by their hemagglutinating activity (HEMAGGLUTININS). However, a variety of lectins occur in animal species where they serve diverse array of functions through specific carbohydrate recognition.
Genetically engineered MUTAGENESIS at a specific site in the DNA molecule that introduces a base substitution, or an insertion or deletion.
A type of FLUORESCENCE SPECTROSCOPY using two FLUORESCENT DYES with overlapping emission and absorption spectra, which is used to indicate proximity of labeled molecules. This technique is useful for studying interactions of molecules and PROTEIN FOLDING.
A form of antibodies consisting only of the variable regions of the heavy and light chains (FV FRAGMENTS), connected by a small linker peptide. They are less immunogenic than complete immunoglobulin and thus have potential therapeutic use.
The assembly of the QUATERNARY PROTEIN STRUCTURE of multimeric proteins (MULTIPROTEIN COMPLEXES) from their composite PROTEIN SUBUNITS.
An immunoassay utilizing an antibody labeled with an enzyme marker such as horseradish peroxidase. While either the enzyme or the antibody is bound to an immunosorbent substrate, they both retain their biologic activity; the change in enzyme activity as a result of the enzyme-antibody-antigen reaction is proportional to the concentration of the antigen and can be measured spectrophotometrically or with the naked eye. Many variations of the method have been developed.
Established cell cultures that have the potential to propagate indefinitely.
Artificial, single or multilaminar vesicles (made from lecithins or other lipids) that are used for the delivery of a variety of biological molecules or molecular complexes to cells, for example, drug delivery and gene transfer. They are also used to study membranes and membrane proteins.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
Methods used for studying the interactions of antibodies with specific regions of protein antigens. Important applications of epitope mapping are found within the area of immunochemistry.
A rigorously mathematical analysis of energy relationships (heat, work, temperature, and equilibrium). It describes systems whose states are determined by thermal parameters, such as temperature, in addition to mechanical and electromagnetic parameters. (From Hawley's Condensed Chemical Dictionary, 12th ed)
Artificially produced membranes, such as semipermeable membranes used in artificial kidney dialysis (RENAL DIALYSIS), monomolecular and bimolecular membranes used as models to simulate biological CELL MEMBRANES. These membranes are also used in the process of GUIDED TISSUE REGENERATION.
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.
Process by which unwanted microbial, plant or animal materials or organisms accumulate on man-made surfaces.
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.
Nanometer-sized particles that are nanoscale in three dimensions. They include nanocrystaline materials; NANOCAPSULES; METAL NANOPARTICLES; DENDRIMERS, and QUANTUM DOTS. The uses of nanoparticles include DRUG DELIVERY SYSTEMS and cancer targeting and imaging.
Methods of creating machines and devices.
Polysaccharides are complex carbohydrates consisting of long, often branched chains of repeating monosaccharide units joined together by glycosidic bonds, which serve as energy storage molecules (e.g., glycogen), structural components (e.g., cellulose), and molecular recognition sites in various biological systems.
An anticoccidial agent mainly for poultry.
Partial immunoglobulin molecules resulting from selective cleavage by proteolytic enzymes or generated through PROTEIN ENGINEERING techniques.
Materials which have structured components with at least one dimension in the range of 1 to 100 nanometers. These include NANOCOMPOSITES; NANOPARTICLES; NANOTUBES; and NANOWIRES.
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.
Elements of limited time intervals, contributing to particular results or situations.
LIGHT, it's processes and properties, and the characteristics of materials interacting with it.
CELL LINE derived from the ovary of the Chinese hamster, Cricetulus griseus (CRICETULUS). The species is a favorite for cytogenetic studies because of its small chromosome number. The cell line has provided model systems for the study of genetic alterations in cultured mammalian cells.
Transport proteins that carry specific substances in the blood or across cell membranes.
The relationship between the chemical structure of a compound and its biological or pharmacological activity. Compounds are often classed together because they have structural characteristics in common including shape, size, stereochemical arrangement, and distribution of functional groups.
Methods for determining interaction between PROTEINS.
The characteristic 3-dimensional shape and arrangement of multimeric proteins (aggregates of more than one polypeptide chain).
Nanometer-scale wires made of materials that conduct electricity. They can be coated with molecules such as antibodies that will bind to proteins and other substances.
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.
Protein or glycoprotein substances of plant origin that bind to sugar moieties in cell walls or membranes. Some carbohydrate-metabolizing proteins (ENZYMES) from PLANTS also bind to carbohydrates, however they are not considered lectins. Many plant lectins change the physiology of the membrane of BLOOD CELLS to cause agglutination, mitosis, or other biochemical changes. They may play a role in plant defense mechanisms.
Domesticated bovine animals of the genus Bos, usually kept on a farm or ranch and used for the production of meat or dairy products or for heavy labor.
The lipid- and protein-containing, selectively permeable membrane that surrounds the cytoplasm in prokaryotic and eukaryotic cells.
The location of the atoms, groups or ions relative to one another in a molecule, as well as the number, type and location of covalent bonds.
The evaluation of incidents involving the loss of function of a device. These evaluations are used for a variety of purposes such as to determine the failure rates, the causes of failures, costs of failures, and the reliability and maintainability of devices.
Theoretical representations that simulate the behavior or activity of chemical processes or phenomena; includes the use of mathematical equations, computers, and other electronic equipment.
A subfamily in the family MURIDAE, comprising the hamsters. Four of the more common genera are Cricetus, CRICETULUS; MESOCRICETUS; and PHODOPUS.
Sites on an antigen that interact with specific antibodies.
Layers of lipid molecules which are two molecules thick. Bilayer systems are frequently studied as models of biological membranes.
Polymers made up of a few (2-20) nucleotides. In molecular genetics, they refer to a short sequence synthesized to match a region where a mutation is known to occur, and then used as a probe (OLIGONUCLEOTIDE PROBES). (Dorland, 28th ed)
Substances, usually of biological origin, that cause cells or other organic particles to aggregate and stick to each other. They include those ANTIBODIES which cause aggregation or agglutination of particulate or insoluble ANTIGENS.
Immunoglobulin molecules having a specific amino acid sequence by virtue of which they interact only with the ANTIGEN (or a very similar shape) that induced their synthesis in cells of the lymphoid series (especially PLASMA CELLS).
Local surface sites on antibodies which react with antigen determinant sites on antigens (EPITOPES.) They are formed from parts of the variable regions of FAB FRAGMENTS.
A low affinity interleukin-5 receptor subunit that combines with the CYTOKINE RECEPTOR COMMON BETA SUBUNIT to form a high affinity receptor for INTERLEUKIN-5. Several isoforms of the interleukin-5 receptor alpha subunit exist due to multiple ALTERNATIVE SPLICING.
A heteropolysaccharide that is similar in structure to HEPARIN. It accumulates in individuals with MUCOPOLYSACCHARIDOSIS.
Measurement of the intensity and quality of fluorescence.
The sequence of carbohydrates within POLYSACCHARIDES; GLYCOPROTEINS; and GLYCOLIPIDS.
Behavior of LIGHT and its interactions with itself and materials.

The role of homophilic binding in anti-tumor antibody R24 recognition of molecular surfaces. Demonstration of an intermolecular beta-sheet interaction between vh domains. (1/3140)

The murine antibody R24 and mouse-human Fv-IgG1(kappa) chimeric antibody chR24 are specific for the cell-surface tumor antigen disialoganglioside GD3. X-ray diffraction and surface plasmon resonance experiments have been employed to study the mechanism of "homophilic binding," in which molecules of R24 recognize and bind to other molecules of R24 though their heavy chain variable domains. R24 exhibits strong binding to liposomes containing disialoganglioside GD3; however, the kinetics are unusual in that saturation of binding is not observed. The binding of chR24 to GD3-bearing liposomes is significantly weaker, suggesting that cooperative interactions involving antibody constant regions contribute to R24 binding of membrane-bound GD3. The crystal structures of the Fabs from R24 and chR24 reveal the mechanism for homophilic binding and confirm that the homophilic and antigen-binding idiotopes are distinct. The homophilic binding idiotope is formed largely by an anti-parallel beta-sheet dimerization between the H2 complementarity determining region (CDR) loops of two Fabs, while the antigen-binding idiotope is a pocket formed by the three CDR loops on the heavy chain. The formation of homophilic dimers requires the presence of a canonical conformation for the H2 CDR in conjunction with participation of side chains. The relative positions of the homophilic and antigen-binding sites allows for a lattice of GD3-specific antibodies to be constructed, which is stabilized by the presence of the cell membrane. This model provides for the selective recognition by R24 of cells that overexpress GD3 on the cell surface.  (+info)

A soluble form of the avian hepatitis B virus receptor. Biochemical characterization and functional analysis of the receptor ligand complex. (2/3140)

Avian hepatitis B virus infection is initiated by the specific interaction of the extracellular preS part of the large viral envelope protein with carboxypeptidase D (gp180), the primary cellular receptor. To functionally and biochemically characterize this interaction, we purified a soluble form of duck carboxypeptidase D from a baculovirus expression system, confirmed its receptor function, and investigated the contribution of different preS sequence elements to receptor binding by surface plasmon resonance analysis. We found that preS binds duck carboxypeptidase D with a 1:1 stoichiometry, thereby inducing conformational changes but not oligomerization. The association constant of the complex was determined to be 2.2 x 10(7) M-1 at 37 degreesC, pH 7.4, with an association rate of 4.0 x 10(4) M-1 s-1 and a dissociation rate of 1.9 x 10(-3) s-1, substantiating high affinity interaction of avihepadnaviruses with their receptor carboxypeptidase D. The separately expressed receptor-binding domain, comprising about 50% of preS as defined by mutational analysis, exhibits similar constants. The domain consists of an essential element, probably responsible for the initial receptor contact and a part that contributes to complex stabilization in a conformation sensitive manner. Together with previous results from cell biological studies these data provide new insights into the initial step of hepadnaviral infection.  (+info)

The L1 major capsid protein of human papillomavirus type 11 recombinant virus-like particles interacts with heparin and cell-surface glycosaminoglycans on human keratinocytes. (3/3140)

The L1 major capsid protein of human papillomavirus (HPV) type 11, a 55-kDa polypeptide, forms particulate structures resembling native virus with an average particle diameter of 50-60 nm when expressed in the yeast Saccharomyces cerevisiae. We show in this report that these virus-like particles (VLPs) interact with heparin and with cell-surface glycosaminoglycans (GAGs) resembling heparin on keratinocytes and Chinese hamster ovary cells. The binding of VLPs to heparin is shown to exhibit an affinity comparable to that of other identified heparin-binding proteins. Immobilized heparin chromatography and surface plasmon resonance were used to show that this interaction can be specifically inhibited by free heparin and dextran sulfate and that the effectiveness of the inhibitor is related to its molecular weight and charge density. Sequence comparison of nine human L1 types revealed a conserved region of the carboxyl terminus containing clustered basic amino acids that bear resemblance to proposed heparin-binding motifs in unrelated proteins. Specific enzymatic cleavage of this region eliminated binding to both immobilized heparin and human keratinocyte (HaCaT) cells. Removal of heparan sulfate GAGs on keratinocytes by treatment with heparinase or heparitinase resulted in an 80-90% reduction of VLP binding, whereas treatment of cells with laminin, a substrate for alpha6 integrin receptors, provided minimal inhibition. Cells treated with chlorate or substituted beta-D-xylosides, resulting in undersulfation or secretion of GAG chains, also showed a reduced affinity for VLPs. Similarly, binding of VLPs to a Chinese hamster ovary cell mutant deficient in GAG synthesis was shown to be only 10% that observed for wild type cells. This report establishes for the first time that the carboxyl-terminal portion of HPV L1 interacts with heparin, and that this region appears to be crucial for interaction with the cell surface.  (+info)

Cluster of differentiation antigen 4 (CD4) endocytosis and adaptor complex binding require activation of the CD4 endocytosis signal by serine phosphorylation. (4/3140)

Cluster of differentiation antigen 4 (CD4), the T lymphocyte antigen receptor component and human immunodeficiency virus coreceptor, is down-modulated when cells are activated by antigen or phorbol esters. During down-modulation CD4 dissociates from p56(lck), undergoes endocytosis through clathrin-coated pits, and is then sorted in early endosomes to late endocytic organelles where it is degraded. Previous studies have suggested that phosphorylation and a dileucine sequence are required for down-modulation. Using transfected HeLa cells, in which CD4 endocytosis can be studied in the absence of p56(lck), we show that the dileucine sequence in the cytoplasmic domain is essential for clathrin-mediated CD4 endocytosis. However, this sequence is only functional as an endocytosis signal when neighboring serine residues are phosphorylated. Phosphoserine is required for rapid endocytosis because CD4 molecules in which the cytoplasmic domain serine residues are substituted with glutamic acid residues are not internalized efficiently. Using surface plasmon resonance, we show that CD4 peptides containing the dileucine sequence bind weakly to clathrin adaptor protein complexes 2 and 1. The affinity of this interaction is increased 350- to 700-fold when the peptides also contain phosphoserine residues.  (+info)

Purified meningococcal transferrin-binding protein B interacts with a secondary, strain-specific, binding site in the N-terminal lobe of human transferrin. (5/3140)

Neisseria meningitidis, grown in iron-limited conditions, produces two transferrin-binding proteins (TbpA and TbpB) that independently and specifically bind human serum transferrin (hTF) but not bovine serum transferrin (bTF). We have used surface plasmon resonance to characterize the interaction between individual TbpA and TbpB and a series of full-length human-bovine chimaeric transferrins (hbTFs) under conditions of variable saturation with iron. A comparative analysis of hTF and hbTF chimaera-binding data confirmed that the major features involved in Tbp binding are located in the C-terminal lobe of hTF and that isolated TbpA can recognize distinct sites present in, or conformationally influenced by, residues 598-679. Binding by TbpB was maintained at a significant but decreased level after replacement of the entire hTF C-terminal lobe by the equivalent bovine sequence. The extent of this binding difference was dependent on the meningococcal strain and on the presence of hTF residues 255-350. This indicated that TbpB from strain SD has a secondary, strain-specific, binding site located within this region, whereas TbpB from strain B16B6 does not share this recognition site. Binding of TbpA was influenced primarily by sequence substitutions in the hTF C-terminal lobe, and co-purified TbpA and TbpB (TbpA+B) was functionally distinct from either of its components. The limited divergence between hTF and bTF has been related to observed differences in binding by Tbps and has been used to delineate those regions of hTF that are important for such interactions.  (+info)

Kinetics of the interaction of endotoxin with polymyxin B and its analogs: a surface plasmon resonance analysis. (6/3140)

Lipopolysaccharide, the invariant structural component of Gram-negative bacteria, when present in minute amounts in the circulation in humans elicits 'endotoxic shock' syndrome, which is fatal in 60% of the cases. Polymyxin B (PMB), a cyclic cationic peptide, neutralizes the endotoxin, but also induces many harmful side effects. Many peptide-based drugs mimicking the activity of PMB have been synthesized in an attempt to reduce toxicity while still retaining the anti-endotoxic activity. The study attempts to use the recent technique of surface plasmon resonance (SPR), in determining the kinetics of association and dissociation involved in the interaction of endotoxin with a few selected peptides that have structural features resembling PMB. The results, in conjunction with the thermodynamic data derived using isothermal titration calorimetry (ITC), stress the vital role played by amphiphilicity of the peptides and hydrophobic forces in this biologically important interaction.  (+info)

Interaction of human macrophage C-type lectin with O-linked N-acetylgalactosamine residues on mucin glycopeptides. (7/3140)

A fluorescein-labeled synthetic peptide, PTTTPITTTTK, was converted into O-glycosylated glycopeptides with various numbers of attached N-acetyl-D-galactosamines (GalNAcs) by in vitro glycosylation with UDP-GalNAc and a microsomal fraction of LS174T human colon carcinoma cells. Glycopeptides with 1, 3, 5, and 6 GalNAc residues (G1, G3, G5, and G6) were obtained, and their sizes were confirmed by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Their sequences were determined by a peptide sequencer to be PTTTGalNAcPITTTTK for G1, PTGalNAcTTPITGalNAcTGalNAcTTK for G3, PTTGalNAcTGalNAcPITGalNAcTGalNAcTGalNAcTK for G5, and PTGalNAcTGalNAcTGalNAcPITGalNAcTGalNAcTGalNAcTK for G6. A calcium-type human macrophage lectin (HML) was prepared in a recombinant form, and its interaction with these glycopeptides was investigated by surface plasmon resonance (SPR) spectroscopy and fluorescence polarization. The affinity of recombinant HML (rHML) for immobilized glycopeptides increased, as revealed by SPR, in parallel with the number of GalNAc. The highest affinity was obtained when the G6-peptide was immobilized at high density. Fluorescence polarization equilibrium-binding assays also revealed that the affinity of rHML for soluble gly-copeptides increased, depending on the number of attached GalNAcs. Carbohydrate recognition domain (CRD) fragments of HML were prepared, and their affinity for these four glycopeptides was also determined, this affinity was apparently lower than that of rHML. Affinity constants of rHML for the G3- and G5-peptides were 11- and 38-fold higher, respectively, than for the G1-peptide, whereas those of CRD fragments were only 2- and 6-fold higher, respectively. A chemical cross-linking study revealed that rHML but not recombinant CRD forms trimers in an aqueous solution. Thus, preferential binding of densely glycosylated O-linked glycopeptides should be due to the trimer formation of rHML.  (+info)

Effects of microcystins on phosphorylase-a binding to phosphatase-2A: kinetic analysis by surface plasmon resonance biosensor. (8/3140)

Cyclic heptapeptide microcystins are a group of hepatoxicants which exert the cytotoxic effects by inhibiting the catalytic activities of phosphatase-2A (PP-2A) and phosphatase-1 (PP-1) and thus disrupt the normal signal transduction pathways. Microcystins interact with PP-2A and PP-1 by a two-step mechanism involving rapid binding and inactivation of protein phosphatase catalytic subunit, followed by a slower covalent interaction. It was proposed that inactivation of PP-2A/PP-1 catalytic activity by microcystins precedes covalent adduct formation. In this study, we used a biosensor based on surface plasmon resonance (SPR) to examine the effects of three microcystins, MCLR, MCRR and MCYR, on the binding between PP-2A and its substrate, phosphorylase-a (PL-a), during the first step of the interaction. The SPR biosensor provides real-time information on the association and dissociation kinetics of PL-a with immobilized PP-2A in the absence and presence of microcystins. It was found that the affinity of PL-a to microcystin-bound PP-2A was four times smaller compared to unbound PP-2A, due to 50% decreases in the association rates and two-fold increases in dissociation rates of PL-a binding to PP-2A. The results suggest that the rapid binding of microcystins to the PP-2A catalytic site leads to the formation of a noncovalent microcystin/PP-2A adduct. While the adduct formation fully inhibits the catalytic activity of PP-2A, it only results in partial inhibition of the substrate binding. The similar effects of the three microcystins on PP-2A suggest that the toxins bind to PP-2A at the same site and cause similar conformational changes. The present work also demonstrates the potential application of biosensor technology in environmental toxicological research.  (+info)

Surface Plasmon Resonance (SPR) is a physical phenomenon that occurs at the interface between a metal and a dielectric material, when electromagnetic radiation (usually light) is shone on it. It involves the collective oscillation of free electrons in the metal, known as surface plasmons, which are excited by the incident light. The resonance condition is met when the momentum and energy of the photons match those of the surface plasmons, leading to a strong absorption of light and an evanescent wave that extends into the dielectric material.

In the context of medical diagnostics and research, SPR is often used as a sensitive and label-free detection technique for biomolecular interactions. By immobilizing one binding partner (e.g., a receptor or antibody) onto the metal surface and flowing the other partner (e.g., a ligand or antigen) over it, changes in the refractive index at the interface can be measured in real-time as the plasmons are disturbed by the presence of bound molecules. This allows for the quantification of binding affinities, kinetics, and specificity with high sensitivity and selectivity.

Biosensing techniques refer to the methods and technologies used to detect and measure biological molecules or processes, typically through the use of a physical device or sensor. These techniques often involve the conversion of a biological response into an electrical signal that can be measured and analyzed. Examples of biosensing techniques include electrochemical biosensors, optical biosensors, and piezoelectric biosensors.

Electrochemical biosensors measure the electrical current or potential generated by a biochemical reaction at an electrode surface. This type of biosensor typically consists of a biological recognition element, such as an enzyme or antibody, that is immobilized on the electrode surface and interacts with the target analyte to produce an electrical signal.

Optical biosensors measure changes in light intensity or wavelength that occur when a biochemical reaction takes place. This type of biosensor can be based on various optical principles, such as absorbance, fluorescence, or surface plasmon resonance (SPR).

Piezoelectric biosensors measure changes in mass or frequency that occur when a biomolecule binds to the surface of a piezoelectric crystal. This type of biosensor is based on the principle that piezoelectric materials generate an electrical charge when subjected to mechanical stress, and this charge can be used to detect changes in mass or frequency that are proportional to the amount of biomolecule bound to the surface.

Biosensing techniques have a wide range of applications in fields such as medicine, environmental monitoring, food safety, and biodefense. They can be used to detect and measure a variety of biological molecules, including proteins, nucleic acids, hormones, and small molecules, as well as to monitor biological processes such as cell growth or metabolism.

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.

I believe there may be some confusion in your question. Gold is typically a chemical element with the symbol Au and atomic number 79. It is a dense, soft, malleable, and ductile metal. It is one of the least reactive chemical elements and is solid under standard conditions.

However, if you are referring to "Gold" in the context of medical terminology, it may refer to:

1. Gold salts: These are a group of compounds that contain gold and are used in medicine for their anti-inflammatory properties. They have been used in the treatment of rheumatoid arthritis, although they have largely been replaced by newer drugs with fewer side effects.
2. Gold implants: In some cases, a small amount of gold may be surgically implanted into the eye to treat conditions such as age-related macular degeneration or diabetic retinopathy. The gold helps to hold the retina in place and can improve vision in some patients.
3. Gold thread embedment: This is an alternative therapy used in traditional Chinese medicine, where gold threads are embedded into the skin or acupuncture points for therapeutic purposes. However, there is limited scientific evidence to support its effectiveness.

I hope this information helps! If you have any further questions, please let me know.

Medical Definition:

Magnetic Resonance Imaging (MRI) is a non-invasive diagnostic imaging technique that uses a strong magnetic field and radio waves to create detailed cross-sectional or three-dimensional images of the internal structures of the body. The patient lies within a large, cylindrical magnet, and the scanner detects changes in the direction of the magnetic field caused by protons in the body. These changes are then converted into detailed images that help medical professionals to diagnose and monitor various medical conditions, such as tumors, injuries, or diseases affecting the brain, spinal cord, heart, blood vessels, joints, and other internal organs. MRI does not use radiation like computed tomography (CT) scans.

"Immobilized proteins" refer to proteins that have been fixed or attached to a solid support or matrix, such as beads, resins, membranes, or electrodes. This immobilization can be achieved through various methods including physical adsorption, covalent attachment, cross-linking, or entrapment within the matrix.

Immobilized proteins retain their biological activity and can be used in a variety of applications, such as affinity chromatography, biosensors, enzyme catalysis, and drug delivery systems. The immobilization process allows for the repeated use of the protein, increased stability, and easier separation from reaction mixtures. Additionally, the orientation and density of the immobilized proteins can be controlled to optimize their activity and specificity in these applications.

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

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

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

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

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

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

Refractometry is a medical laboratory technique used to measure the refractive index of a substance, typically a liquid. The refractive index is the ratio of the speed of light in a vacuum to its speed in the substance being measured. In a clinical setting, refractometry is often used to determine the concentration of total solids in a fluid, such as urine or serum, by measuring the angle at which light passes through the sample. This information can be useful in the diagnosis and monitoring of various medical conditions, including dehydration, kidney disease, and diabetes. Refractometry is also used in the field of optometry to measure the refractive error of the eye, or the amount and type of correction needed to provide clear vision.

Medical definitions generally refer to terms and concepts within the medical field. The term "metal nanoparticles" is more commonly used in materials science, chemistry, and physics. However, I can provide a general scientific definition that could be relevant to medical applications:

Metal nanoparticles are tiny particles with at least one dimension ranging from 1 to 100 nanometers (nm), composed of metals or metal compounds. They have unique optical, electronic, and chemical properties due to their small size and high surface-to-volume ratio, making them useful in various fields, including medical research. In medicine, metal nanoparticles can be used in drug delivery systems, diagnostics, and therapeutic applications such as photothermal therapy and radiation therapy. Examples of metals used for nanoparticle synthesis include gold, silver, and iron.

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.

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.

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.

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

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

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

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.

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.

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.

I'm sorry for any confusion, but "Silver" is not a medical term. It is a chemical element with the symbol Ag and atomic number 47. Silver is a soft, white, lustrous transition metal that occurs in its pure, free form (native silver) as an alloy with gold and other metals, and in minerals such as argentite and chlorargyrite.

In the medical field, silver compounds have been used for their antimicrobial properties. For example, silver sulfadiazine is a common topical cream used to prevent or treat wound infections. Colloidal silver, a suspension of silver particles in a liquid, has also been promoted as a dietary supplement and alternative treatment for various conditions, but its effectiveness and safety are not well-established.

Medical Definition of Optical Fibers:

Optical fibers are thin, transparent strands of glass or plastic fiber that are designed to transmit light along their length. In the medical field, optical fibers are used in various applications such as illumination, imaging, and data transmission. For instance, they are used in flexible endoscopes to provide illumination and visualization inside the body during diagnostic or surgical procedures. They are also used in optical communication systems for transmitting information in the form of light signals within medical devices or between medical facilities. The use of optical fibers allows for minimally invasive procedures, improved image quality, and increased data transmission rates.

Surface properties in the context of medical science refer to the characteristics and features of the outermost layer or surface of a biological material or structure, such as cells, tissues, organs, or medical devices. These properties can include physical attributes like roughness, smoothness, hydrophobicity or hydrophilicity, and electrical conductivity, as well as chemical properties like charge, reactivity, and composition.

In the field of biomaterials science, understanding surface properties is crucial for designing medical implants, devices, and drug delivery systems that can interact safely and effectively with biological tissues and fluids. Surface modifications, such as coatings or chemical treatments, can be used to alter surface properties and enhance biocompatibility, improve lubricity, reduce fouling, or promote specific cellular responses like adhesion, proliferation, or differentiation.

Similarly, in the field of cell biology, understanding surface properties is essential for studying cell-cell interactions, cell signaling, and cell behavior. Cells can sense and respond to changes in their environment, including variations in surface properties, which can influence cell shape, motility, and function. Therefore, characterizing and manipulating surface properties can provide valuable insights into the mechanisms of cellular processes and offer new strategies for developing therapies and treatments for various diseases.

Antibody affinity refers to the strength and specificity of the interaction between an antibody and its corresponding antigen at a molecular level. It is a measure of how strongly and selectively an antibody binds to its target antigen. A higher affinity indicates a more stable and specific binding, while a lower affinity suggests weaker and less specific interactions. Affinity is typically measured in terms of the dissociation constant (Kd), which describes the concentration of antigen needed to achieve half-maximal binding to an antibody. Generally, a smaller Kd value corresponds to a higher affinity, indicating a tighter and more selective bond. This parameter is crucial in the development of diagnostic and therapeutic applications, such as immunoassays and targeted therapies, where high-affinity antibodies are preferred for improved sensitivity and specificity.

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.

Calorimetry is the measurement and study of heat transfer, typically using a device called a calorimeter. In the context of medicine and physiology, calorimetry can be used to measure heat production or dissipation in the body, which can provide insight into various bodily functions and metabolic processes.

There are different types of calorimeters used for medical research and clinical applications, including direct and indirect calorimeters. Direct calorimetry measures the heat produced directly by the body, while indirect calorimetry estimates heat production based on oxygen consumption and carbon dioxide production rates. Indirect calorimetry is more commonly used in clinical settings to assess energy expenditure and metabolic rate in patients with various medical conditions or during specific treatments, such as critical illness, surgery, or weight management programs.

In summary, calorimetry in a medical context refers to the measurement of heat exchange within the body or between the body and its environment, which can offer valuable information for understanding metabolic processes and developing personalized treatment plans.

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

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

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

Streptavidin is not a medical term per se, but rather a biochemical term used in the field of medicine and laboratory research. Streptavidin is a protein that is derived from the bacterium Streptomyces avidinii. It has a unique ability to bind very strongly and specifically to another molecule called biotin, with an association constant that is one of the strongest non-covalent interactions known in nature.

This property makes streptavidin a valuable tool in various medical and research applications such as immunoassays, histology, molecular biology, and drug delivery systems. For example, biotinylated molecules (such as antibodies, DNA, or enzymes) can be linked to streptavidin for detection, purification, or targeting purposes.

In summary, streptavidin is a bacterial protein that binds strongly and specifically to biotin, which is used in various medical and research applications as a tool for detection, purification, or targeting purposes.

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

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

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

Protein array analysis is a high-throughput technology used to detect and measure the presence and activity of specific proteins in biological samples. This technique utilizes arrays or chips containing various capture agents, such as antibodies or aptamers, that are designed to bind to specific target proteins. The sample is then added to the array, allowing the target proteins to bind to their corresponding capture agents. After washing away unbound materials, a detection system is used to identify and quantify the bound proteins. This method can be used for various applications, including protein-protein interaction studies, biomarker discovery, and drug development. The results of protein array analysis provide valuable information about the expression levels, post-translational modifications, and functional states of proteins in complex biological systems.

"Competitive binding" is a term used in pharmacology and biochemistry to describe the behavior of two or more molecules (ligands) competing for the same binding site on a target protein or receptor. In this context, "binding" refers to the physical interaction between a ligand and its target.

When a ligand binds to a receptor, it can alter the receptor's function, either activating or inhibiting it. If multiple ligands compete for the same binding site, they will compete to bind to the receptor. The ability of each ligand to bind to the receptor is influenced by its affinity for the receptor, which is a measure of how strongly and specifically the ligand binds to the receptor.

In competitive binding, if one ligand is present in high concentrations, it can prevent other ligands with lower affinity from binding to the receptor. This is because the higher-affinity ligand will have a greater probability of occupying the binding site and blocking access to the other ligands. The competition between ligands can be described mathematically using equations such as the Langmuir isotherm, which describes the relationship between the concentration of ligand and the fraction of receptors that are occupied by the ligand.

Competitive binding is an important concept in drug development, as it can be used to predict how different drugs will interact with their targets and how they may affect each other's activity. By understanding the competitive binding properties of a drug, researchers can optimize its dosage and delivery to maximize its therapeutic effect while minimizing unwanted side effects.

Circular dichroism (CD) is a technique used in physics and chemistry to study the structure of molecules, particularly large biological molecules such as proteins and nucleic acids. It measures the difference in absorption of left-handed and right-handed circularly polarized light by a sample. This difference in absorption can provide information about the three-dimensional structure of the molecule, including its chirality or "handedness."

In more technical terms, CD is a form of spectroscopy that measures the differential absorption of left and right circularly polarized light as a function of wavelength. The CD signal is measured in units of millidegrees (mdeg) and can be positive or negative, depending on the type of chromophore and its orientation within the molecule.

CD spectra can provide valuable information about the secondary and tertiary structure of proteins, as well as the conformation of nucleic acids. For example, alpha-helical proteins typically exhibit a strong positive band near 190 nm and two negative bands at around 208 nm and 222 nm, while beta-sheet proteins show a strong positive band near 195 nm and two negative bands at around 217 nm and 175 nm.

CD spectroscopy is a powerful tool for studying the structural changes that occur in biological molecules under different conditions, such as temperature, pH, or the presence of ligands or other molecules. It can also be used to monitor the folding and unfolding of proteins, as well as the binding of drugs or other small molecules to their targets.

Heparin is defined as a highly sulfated glycosaminoglycan (a type of polysaccharide) that is widely present in many tissues, but is most commonly derived from the mucosal tissues of mammalian lungs or intestinal mucosa. It is an anticoagulant that acts as an inhibitor of several enzymes involved in the blood coagulation cascade, primarily by activating antithrombin III which then neutralizes thrombin and other clotting factors.

Heparin is used medically to prevent and treat thromboembolic disorders such as deep vein thrombosis, pulmonary embolism, and certain types of heart attacks. It can also be used during hemodialysis, cardiac bypass surgery, and other medical procedures to prevent the formation of blood clots.

It's important to note that while heparin is a powerful anticoagulant, it does not have any fibrinolytic activity, meaning it cannot dissolve existing blood clots. Instead, it prevents new clots from forming and stops existing clots from growing larger.

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

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

Nanotechnology is not a medical term per se, but it is a field of study with potential applications in medicine. According to the National Nanotechnology Initiative, nanotechnology is defined as "the understanding and control of matter at the nanoscale, at dimensions between approximately 1 and 100 nanometers, where unique phenomena enable novel applications."

In the context of medicine, nanotechnology has the potential to revolutionize the way we diagnose, treat, and prevent diseases. Nanomedicine involves the use of nanoscale materials, devices, or systems for medical applications. These can include drug delivery systems that target specific cells or tissues, diagnostic tools that detect biomarkers at the molecular level, and tissue engineering strategies that promote regeneration and repair.

While nanotechnology holds great promise for medicine, it is still a relatively new field with many challenges to overcome, including issues related to safety, regulation, and scalability.

Fetuins are a group of proteins that are produced by the liver and found in circulation in the blood. The most well-known fetuin, fetuin-A, is a 64 kDa glycoprotein that is synthesized in the liver and secreted into the bloodstream. Fetuin-A plays a role in several physiological processes, including inhibition of tissue calcification, regulation of insulin sensitivity, and modulation of immune responses.

Fetuin-B is another member of the fetuin family that shares some structural similarities with fetuin-A but has distinct functions. Fetuin-B is also produced by the liver and secreted into the bloodstream, where it plays a role in regulating lipid metabolism and insulin sensitivity.

It's worth noting that while both fetuins have been studied for their roles in various physiological processes, there is still much to be learned about their functions and regulation.

A peptide library is a collection of a large number of peptides, which are short chains of amino acids. Each peptide in the library is typically composed of a defined length and sequence, and may contain a variety of different amino acids. Peptide libraries can be synthesized using automated techniques and are often used in scientific research to identify potential ligands (molecules that bind to specific targets) or to study the interactions between peptides and other molecules.

In a peptide library, each peptide is usually attached to a solid support, such as a resin bead, and the entire library can be created using split-and-pool synthesis techniques. This allows for the rapid and efficient synthesis of a large number of unique peptides, which can then be screened for specific activities or properties.

Peptide libraries are used in various fields such as drug discovery, proteomics, and molecular biology to identify potential therapeutic targets, understand protein-protein interactions, and develop new diagnostic tools.

Biotinyllation is a process of introducing biotin (a vitamin) into a molecule, such as a protein or nucleic acid (DNA or RNA), through chemical reaction. This modification allows the labeled molecule to be easily detected and isolated using streptavidin-biotin interaction, which has one of the strongest non-covalent bonds in nature. Biotinylated molecules are widely used in various research applications such as protein-protein interaction studies, immunohistochemistry, and blotting techniques.

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

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

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

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

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

Adsorption is a process in which atoms, ions, or molecules from a gas, liquid, or dissolved solid accumulate on the surface of a material. This occurs because the particles in the adsorbate (the substance being adsorbed) have forces that attract them to the surface of the adsorbent (the material that the adsorbate is adhering to).

In medical terms, adsorption can refer to the use of materials with adsorptive properties to remove harmful substances from the body. For example, activated charcoal is sometimes used in the treatment of poisoning because it can adsorb a variety of toxic substances and prevent them from being absorbed into the bloodstream.

It's important to note that adsorption is different from absorption, which refers to the process by which a substance is taken up and distributed throughout a material or tissue.

Aptamers are short, single-stranded oligonucleotides (DNA or RNA) that bind to specific target molecules with high affinity and specificity. They are generated through an iterative process called Systematic Evolution of Ligands by EXponential enrichment (SELEX), where large libraries of randomized oligonucleotides are subjected to repeated rounds of selection and amplification until sequences with the desired binding properties are identified. Nucleotide aptamers have potential applications in various fields, including diagnostics, therapeutics, and research tools.

The term "nucleotide" refers to the basic building blocks of nucleic acids (DNA and RNA). A nucleotide consists of a pentose sugar (ribose for RNA and deoxyribose for DNA), a phosphate group, and a nitrogenous base. The nitrogenous bases in nucleotides are adenine, guanine, cytosine, thymine (in DNA) or uracil (in RNA). In aptamers, the nucleotide sequences form specific three-dimensional structures that enable them to recognize and bind to their target molecules.

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

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

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

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

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

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

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

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

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

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

"Immobilized antibodies" refer to antibodies that have been fixed or attached to a solid support or surface. This is often done for use in various diagnostic and research applications, such as immunoassays, biosensors, and affinity chromatography. The immobilization of antibodies allows them to capture and detect specific target molecules (antigens) from complex samples, while remaining stationary and easily recoverable for reuse.

There are several methods for immobilizing antibodies, including physical adsorption, covalent attachment, and non-covalent entrapment. The choice of method depends on the specific application and the desired properties of the immobilized antibodies, such as stability, orientation, and accessibility.

It is important to note that the immobilization process may affect the binding affinity and specificity of the antibodies, and therefore careful optimization and validation are necessary to ensure the performance of the assay or application.

Immobilized enzymes refer to enzymes that have been restricted or fixed in a specific location and are unable to move freely. This is typically achieved through physical or chemical methods that attach the enzyme to a solid support or matrix. The immobilization of enzymes can provide several advantages, including increased stability, reusability, and ease of separation from the reaction mixture.

Immobilized enzymes are widely used in various industrial applications, such as biotransformations, biosensors, and diagnostic kits. They can also be used for the production of pharmaceuticals, food additives, and other fine chemicals. The immobilization techniques include adsorption, covalent binding, entrapment, and cross-linking.

Adsorption involves physically attaching the enzyme to a solid support through weak forces such as van der Waals interactions or hydrogen bonding. Covalent binding involves forming chemical bonds between the enzyme and the support matrix. Entrapment involves encapsulating the enzyme within a porous matrix, while cross-linking involves chemically linking multiple enzyme molecules together to form a stable structure.

Overall, immobilized enzymes offer several advantages over free enzymes, including improved stability, reusability, and ease of separation from the reaction mixture, making them valuable tools in various industrial applications.

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

Nanotubes, in the context of nanotechnology and materials science, refer to hollow cylindrical structures with extremely small diameters, measured in nanometers (nm). They are typically composed of carbon atoms arranged in a hexagonal lattice structure, similar to graphene. The most common types of nanotubes are single-walled nanotubes (SWNTs) and multi-walled nanotubes (MWNTs).

In the field of medicine, nanotubes have been studied for their potential applications in drug delivery, tissue engineering, and medical devices. For example, researchers have explored the use of nanotubes as drug carriers, where drugs can be loaded into the hollow interior of the tube and released in a controlled manner at the target site. Additionally, nanotubes have been used to create conductive scaffolds for tissue engineering, which may help promote nerve regeneration or muscle growth.

However, it's important to note that while nanotubes have shown promise in preclinical studies, their potential use in medical applications is still being researched and developed. There are concerns about the potential toxicity of nanotubes, as well as challenges related to their large-scale production and functionalization for specific medical applications.

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

Biotin is a water-soluble vitamin, also known as Vitamin B7 or Vitamin H. It is a cofactor for several enzymes involved in metabolism, particularly in the synthesis and breakdown of fatty acids, amino acids, and carbohydrates. Biotin plays a crucial role in maintaining healthy skin, hair, nails, nerves, and liver function. It is found in various foods such as nuts, seeds, whole grains, milk, and vegetables. Biotin deficiency is rare but can occur in people with malnutrition, alcoholism, pregnancy, or certain genetic disorders.

A Quartz Crystal Microbalance (QCM) is a type of physical analysis technique that uses the vibrations of a quartz crystal to measure changes in mass at a molecular or nanoscale level. When an alternating electrical field is applied to a quartz crystal, it causes the crystal to vibrate at a specific frequency. This phenomenon is known as the piezoelectric effect.

In QCM techniques, a thin film or material is deposited onto the surface of the quartz crystal, which changes its mass and therefore affects its vibrational frequency. By measuring the change in frequency before and after the deposition of the material, researchers can calculate the mass of the material that was added to the crystal's surface with high precision.

QCM techniques have a wide range of applications in research and industry, including the study of thin films, self-assembled monolayers, biosensors, and environmental monitoring. They are particularly useful for measuring changes in mass that occur on a very small scale, such as those associated with chemical reactions or biological interactions.

I'm sorry for any confusion, but "spectrum analysis" is not a commonly used medical term. Spectrum analysis is a term that is more frequently used in the fields of physics, mathematics, and engineering to describe the process of breaking down a signal or a wave into its different frequencies and amplitudes, creating a visual representation called a spectrum.

If you have any concerns about a medical issue, I would recommend consulting with a healthcare professional for accurate information and guidance.

Molecular imprinting is a technique used in the production of polymer-based materials that have specific recognition sites for target molecules. It is a type of nanotechnology that involves creating a molecular template within a polymer matrix during its synthesis. The template is introduced into the polymer solution, and when the polymer hardens or sets, it takes on the shape and size of the template. After the template is removed, the resulting material has binding sites that are complementary in shape, size, and chemical functionality to the target molecule. These materials can then be used for various applications such as sensors, separations, drug delivery systems, and diagnostics.

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.

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

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

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

Lectins are a type of proteins that bind specifically to carbohydrates and have been found in various plant and animal sources. They play important roles in biological recognition events, such as cell-cell adhesion, and can also be involved in the immune response. Some lectins can agglutinate certain types of cells or precipitate glycoproteins, while others may have a more direct effect on cellular processes. In some cases, lectins from plants can cause adverse effects in humans if ingested, such as digestive discomfort or allergic reactions.

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.

Fluorescence Resonance Energy Transfer (FRET) is not strictly a medical term, but it is a fundamental concept in biophysical and molecular biology research, which can have medical applications. Here's the definition of FRET:

Fluorescence Resonance Energy Transfer (FRET) is a distance-dependent energy transfer process between two fluorophores, often referred to as a donor and an acceptor. The process occurs when the emission spectrum of the donor fluorophore overlaps with the excitation spectrum of the acceptor fluorophore. When the donor fluorophore is excited, it can transfer its energy to the acceptor fluorophore through non-radiative dipole-dipole coupling, resulting in the emission of light from the acceptor at a longer wavelength than that of the donor.

FRET efficiency depends on several factors, including the distance between the two fluorophores, their relative orientation, and the spectral overlap between their excitation and emission spectra. FRET is typically efficient when the distance between the donor and acceptor is less than 10 nm (nanometers), making it a powerful tool for measuring molecular interactions, conformational changes, and distances at the molecular level.

In medical research, FRET has been used to study various biological processes, such as protein-protein interactions, enzyme kinetics, and gene regulation. It can also be used in developing biosensors for detecting specific molecules or analytes in clinical samples, such as blood or tissue.

Single-chain antibodies (scFvs) are small, artificial protein molecules that contain the antigen-binding sites of immunoglobulins. They are formed by linking the variable regions of the heavy and light chains of an antibody via a flexible peptide linker, creating a single polypeptide chain. This design allows scFvs to maintain the specificity of traditional antibodies while being significantly smaller in size, more stable, and easier to produce. They have various applications in research, diagnostics, and therapeutics, including targeted drug delivery, tumor imaging, and the development of novel therapies for cancer and other diseases.

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

An Enzyme-Linked Immunosorbent Assay (ELISA) is a type of analytical biochemistry assay used to detect and quantify the presence of a substance, typically a protein or peptide, in a liquid sample. It takes its name from the enzyme-linked antibodies used in the assay.

In an ELISA, the sample is added to a well containing a surface that has been treated to capture the target substance. If the target substance is present in the sample, it will bind to the surface. Next, an enzyme-linked antibody specific to the target substance is added. This antibody will bind to the captured target substance if it is present. After washing away any unbound material, a substrate for the enzyme is added. If the enzyme is present due to its linkage to the antibody, it will catalyze a reaction that produces a detectable signal, such as a color change or fluorescence. The intensity of this signal is proportional to the amount of target substance present in the sample, allowing for quantification.

ELISAs are widely used in research and clinical settings to detect and measure various substances, including hormones, viruses, and bacteria. They offer high sensitivity, specificity, and reproducibility, making them a reliable choice for many applications.

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.

Liposomes are artificially prepared, small, spherical vesicles composed of one or more lipid bilayers that enclose an aqueous compartment. They can encapsulate both hydrophilic and hydrophobic drugs, making them useful for drug delivery applications in the medical field. The lipid bilayer structure of liposomes is similar to that of biological membranes, which allows them to merge with and deliver their contents into cells. This property makes liposomes a valuable tool in delivering drugs directly to targeted sites within the body, improving drug efficacy while minimizing side effects.

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.

Epitope mapping is a technique used in immunology to identify the specific portion or regions (called epitopes) on an antigen that are recognized and bind to antibodies or T-cell receptors. This process helps to understand the molecular basis of immune responses against various pathogens, allergens, or transplanted tissues.

Epitope mapping can be performed using different methods such as:

1. Peptide scanning: In this method, a series of overlapping peptides spanning the entire length of the antigen are synthesized and tested for their ability to bind to antibodies or T-cell receptors. The peptide that shows binding is considered to contain the epitope.
2. Site-directed mutagenesis: In this approach, specific amino acids within the antigen are altered, and the modified antigens are tested for their ability to bind to antibodies or T-cell receptors. This helps in identifying the critical residues within the epitope.
3. X-ray crystallography and NMR spectroscopy: These techniques provide detailed information about the three-dimensional structure of antigen-antibody complexes, allowing for accurate identification of epitopes at an atomic level.

The results from epitope mapping can be useful in various applications, including vaccine design, diagnostic test development, and understanding the basis of autoimmune diseases.

I'm sorry for any confusion, but "thermodynamics" is not a term that has a specific medical definition. It is a branch of physics that deals with the relationships between heat and other forms of energy. However, the principles of thermodynamics can be applied to biological systems, including those in the human body, such as in the study of metabolism or muscle function. But in a medical context, "thermodynamics" would not be a term used independently as a diagnosis, treatment, or any medical condition.

Artificial membranes are synthetic or man-made materials that possess properties similar to natural biological membranes, such as selective permeability and barrier functions. These membranes can be designed to control the movement of molecules, ions, or cells across them, making them useful in various medical and biotechnological applications.

Examples of artificial membranes include:

1. Dialysis membranes: Used in hemodialysis for patients with renal failure, these semi-permeable membranes filter waste products and excess fluids from the blood while retaining essential proteins and cells.
2. Hemofiltration membranes: Utilized in extracorporeal circuits to remove larger molecules, such as cytokines or inflammatory mediators, from the blood during critical illnesses or sepsis.
3. Drug delivery systems: Artificial membranes can be used to encapsulate drugs, allowing for controlled release and targeted drug delivery in specific tissues or cells.
4. Tissue engineering: Synthetic membranes serve as scaffolds for cell growth and tissue regeneration, guiding the formation of new functional tissues.
5. Biosensors: Artificial membranes can be integrated into biosensing devices to selectively detect and quantify biomolecules, such as proteins or nucleic acids, in diagnostic applications.
6. Microfluidics: Artificial membranes are used in microfluidic systems for lab-on-a-chip applications, enabling the manipulation and analysis of small volumes of fluids for various medical and biological purposes.

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.

Biofouling is the accumulation of microorganisms, algae, plants, and animals on wet surfaces, such as the hulls of ships, pier pilings, and buoys. This growth can have negative impacts on the performance and efficiency of equipment and infrastructure, leading to increased maintenance costs and potential environmental damage. In the medical field, biofouling can also refer to the undesirable accumulation of microorganisms or biomolecules on medical devices, which can lead to infection or device failure.

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.

Nanoparticles are defined in the field of medicine as tiny particles that have at least one dimension between 1 to 100 nanometers (nm). They are increasingly being used in various medical applications such as drug delivery, diagnostics, and therapeutics. Due to their small size, nanoparticles can penetrate cells, tissues, and organs more efficiently than larger particles, making them ideal for targeted drug delivery and imaging.

Nanoparticles can be made from a variety of materials including metals, polymers, lipids, and dendrimers. The physical and chemical properties of nanoparticles, such as size, shape, charge, and surface chemistry, can greatly affect their behavior in biological systems and their potential medical applications.

It is important to note that the use of nanoparticles in medicine is still a relatively new field, and there are ongoing studies to better understand their safety and efficacy.

Equipment design, in the medical context, refers to the process of creating and developing medical equipment and devices, such as surgical instruments, diagnostic machines, or assistive technologies. This process involves several stages, including:

1. Identifying user needs and requirements
2. Concept development and brainstorming
3. Prototyping and testing
4. Design for manufacturing and assembly
5. Safety and regulatory compliance
6. Verification and validation
7. Training and support

The goal of equipment design is to create safe, effective, and efficient medical devices that meet the needs of healthcare providers and patients while complying with relevant regulations and standards. The design process typically involves a multidisciplinary team of engineers, clinicians, designers, and researchers who work together to develop innovative solutions that improve patient care and outcomes.

Polysaccharides are complex carbohydrates consisting of long chains of monosaccharide units (simple sugars) bonded together by glycosidic linkages. They can be classified based on the type of monosaccharides and the nature of the bonds that connect them.

Polysaccharides have various functions in living organisms. For example, starch and glycogen serve as energy storage molecules in plants and animals, respectively. Cellulose provides structural support in plants, while chitin is a key component of fungal cell walls and arthropod exoskeletons.

Some polysaccharides also have important roles in the human body, such as being part of the extracellular matrix (e.g., hyaluronic acid) or acting as blood group antigens (e.g., ABO blood group substances).

Robenidine is not a medication that has been approved for use in humans or animals by regulatory agencies such as the US Food and Drug Administration (FDA) or the European Medicines Agency (EMA). Therefore, there is no established medical definition for this compound.

However, Robenidine is a chemical compound with the molecular formula C10H14ClNO2. It has been studied in laboratory experiments as a potential antiparasitic agent, but its development as a medication has not progressed beyond that stage.

It's important to note that just because a compound has been studied for its potential therapeutic effects does not mean that it is safe or effective for use in humans or animals. All medications must undergo rigorous testing and clinical trials to establish their safety and efficacy before they can be approved for use.

Immunoglobulin fragments refer to the smaller protein units that are formed by the digestion or break-down of an intact immunoglobulin, also known as an antibody. Immunoglobulins are large Y-shaped proteins produced by the immune system to identify and neutralize foreign substances such as pathogens or toxins. They consist of two heavy chains and two light chains, held together by disulfide bonds.

The digestion or break-down of an immunoglobulin can occur through enzymatic cleavage, which results in the formation of distinct fragments. The most common immunoglobulin fragments are:

1. Fab (Fragment, antigen binding) fragments: These are formed by the digestion of an intact immunoglobulin using the enzyme papain. Each Fab fragment contains a single antigen-binding site, consisting of a portion of one heavy chain and one light chain. The Fab fragments retain their ability to bind to specific antigens.
2. Fc (Fragment, crystallizable) fragments: These are formed by the digestion of an intact immunoglobulin using the enzyme pepsin or through the natural breakdown process in the body. The Fc fragment contains the constant region of both heavy chains and is responsible for effector functions such as complement activation, binding to Fc receptors on immune cells, and antibody-dependent cellular cytotoxicity (ADCC).

These immunoglobulin fragments play crucial roles in various immune responses and diagnostic applications. For example, Fab fragments can be used in immunoassays for the detection of specific antigens, while Fc fragments can mediate effector functions that help eliminate pathogens or damaged cells from the body.

Nanostructures, in the context of medical and biomedical research, refer to materials or devices with structural features that have at least one dimension ranging between 1-100 nanometers (nm). At this size scale, the properties of these structures can differ significantly from bulk materials, exhibiting unique phenomena that are often influenced by quantum effects.

Nanostructures have attracted considerable interest in biomedicine due to their potential applications in various areas such as drug delivery, diagnostics, regenerative medicine, and tissue engineering. They can be fabricated from a wide range of materials including metals, polymers, ceramics, and carbon-based materials.

Some examples of nanostructures used in biomedicine include:

1. Nanoparticles: These are tiny particles with at least one dimension in the nanoscale range. They can be made from various materials like metals, polymers, or lipids and have applications in drug delivery, imaging, and diagnostics.
2. Quantum dots: These are semiconductor nanocrystals that exhibit unique optical properties due to quantum confinement effects. They are used as fluorescent labels for bioimaging and biosensing applications.
3. Carbon nanotubes: These are hollow, cylindrical structures made of carbon atoms arranged in a hexagonal lattice. They have exceptional mechanical strength, electrical conductivity, and thermal stability, making them suitable for various biomedical applications such as drug delivery, tissue engineering, and biosensors.
4. Nanofibers: These are elongated nanostructures with high aspect ratios (length much greater than width). They can be fabricated from various materials like polymers, ceramics, or composites and have applications in tissue engineering, wound healing, and drug delivery.
5. Dendrimers: These are highly branched, nanoscale polymers with a well-defined structure and narrow size distribution. They can be used as drug carriers, gene delivery vehicles, and diagnostic agents.
6. Nanoshells: These are hollow, spherical nanoparticles consisting of a dielectric core covered by a thin metallic shell. They exhibit unique optical properties that make them suitable for applications such as photothermal therapy, biosensing, and imaging.

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.

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.

Optical phenomena refer to the various observable patterns and effects that occur due to the interaction of light with the environment or with structures in our eye. These can include natural phenomena such as rainbows, mirages, and halos around the sun or moon, as well as visual artifacts created by the eye itself, such as afterimages, floaters, and flashes of light. Some optical phenomena are caused by the refraction, reflection, or interference of light waves, while others may result from abnormalities in the eye's structure or function. Understanding these phenomena can provide insight into the properties of light and the functioning of the visual system.

CHO cells, or Chinese Hamster Ovary cells, are a type of immortalized cell line that are commonly used in scientific research and biotechnology. They were originally derived from the ovaries of a female Chinese hamster (Cricetulus griseus) in the 1950s.

CHO cells have several characteristics that make them useful for laboratory experiments. They can grow and divide indefinitely under appropriate conditions, which allows researchers to culture large quantities of them for study. Additionally, CHO cells are capable of expressing high levels of recombinant proteins, making them a popular choice for the production of therapeutic drugs, vaccines, and other biologics.

In particular, CHO cells have become a workhorse in the field of biotherapeutics, with many approved monoclonal antibody-based therapies being produced using these cells. The ability to genetically modify CHO cells through various methods has further expanded their utility in research and industrial applications.

It is important to note that while CHO cells are widely used in scientific research, they may not always accurately represent human cell behavior or respond to drugs and other compounds in the same way as human cells do. Therefore, results obtained using CHO cells should be validated in more relevant systems when possible.

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.

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

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

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.

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

I'm sorry for any confusion, but "nanowires" is not a medical term. It is a term used in the field of materials science and nanotechnology. Nanowires are one-dimensional structures with a diameter of 1-100 nanometers (nm) and an aspect ratio (length/diameter) greater than 1000. They have unique electrical, mechanical, and optical properties that make them useful in various applications such as electronics, sensors, energy storage, and biomedical devices.

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.

Plant lectins are proteins or glycoproteins that are abundantly found in various plant parts such as seeds, leaves, stems, and roots. They have the ability to bind specifically to carbohydrate structures present on cell membranes, known as glycoconjugates. This binding property of lectins is reversible and non-catalytic, meaning it does not involve any enzymatic activity.

Lectins play several roles in plants, including defense against predators, pathogens, and herbivores. They can agglutinate red blood cells, stimulate the immune system, and have been implicated in various biological processes such as cell growth, differentiation, and apoptosis (programmed cell death). Some lectins also exhibit mitogenic activity, which means they can stimulate the proliferation of certain types of cells.

In the medical field, plant lectins have gained attention due to their potential therapeutic applications. For instance, some lectins have been shown to possess anti-cancer properties and are being investigated as potential cancer treatments. However, it is important to note that some lectins can be toxic or allergenic to humans and animals, so they must be used with caution.

"Cattle" is a term used in the agricultural and veterinary fields to refer to domesticated animals of the genus *Bos*, primarily *Bos taurus* (European cattle) and *Bos indicus* (Zebu). These animals are often raised for meat, milk, leather, and labor. They are also known as bovines or cows (for females), bulls (intact males), and steers/bullocks (castrated males). However, in a strict medical definition, "cattle" does not apply to humans or other animals.

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.

Molecular structure, in the context of biochemistry and molecular biology, refers to the arrangement and organization of atoms and chemical bonds within a molecule. It describes the three-dimensional layout of the constituent elements, including their spatial relationships, bond lengths, and angles. Understanding molecular structure is crucial for elucidating the functions and reactivities of biological macromolecules such as proteins, nucleic acids, lipids, and carbohydrates. Various experimental techniques, like X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and cryo-electron microscopy (cryo-EM), are employed to determine molecular structures at atomic resolution, providing valuable insights into their biological roles and potential therapeutic targets.

Equipment Failure Analysis is a process of identifying the cause of failure in medical equipment or devices. This involves a systematic examination and evaluation of the equipment, its components, and operational history to determine why it failed. The analysis may include physical inspection, chemical testing, and review of maintenance records, as well as assessment of design, manufacturing, and usage factors that may have contributed to the failure.

The goal of Equipment Failure Analysis is to identify the root cause of the failure, so that corrective actions can be taken to prevent similar failures in the future. This is important in medical settings to ensure patient safety and maintain the reliability and effectiveness of medical equipment.

A chemical model is a simplified representation or description of a chemical system, based on the laws of chemistry and physics. It is used to explain and predict the behavior of chemicals and chemical reactions. Chemical models can take many forms, including mathematical equations, diagrams, and computer simulations. They are often used in research, education, and industry to understand complex chemical processes and develop new products and technologies.

For example, a chemical model might be used to describe the way that atoms and molecules interact in a particular reaction, or to predict the properties of a new material. Chemical models can also be used to study the behavior of chemicals at the molecular level, such as how they bind to each other or how they are affected by changes in temperature or pressure.

It is important to note that chemical models are simplifications of reality and may not always accurately represent every aspect of a chemical system. They should be used with caution and validated against experimental data whenever possible.

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

An epitope is a specific region on the surface of an antigen (a molecule that can trigger an immune response) that is recognized by an antibody, B-cell receptor, or T-cell receptor. It is also commonly referred to as an antigenic determinant. Epitopes are typically composed of linear amino acid sequences or conformational structures made up of discontinuous amino acids in the antigen. They play a crucial role in the immune system's ability to differentiate between self and non-self molecules, leading to the targeted destruction of foreign substances like viruses and bacteria. Understanding epitopes is essential for developing vaccines, diagnostic tests, and immunotherapies.

A lipid bilayer is a thin membrane made up of two layers of lipid molecules, primarily phospholipids. The hydrophilic (water-loving) heads of the lipids face outwards, coming into contact with watery environments on both sides, while the hydrophobic (water-fearing) tails point inward, away from the aqueous surroundings. This unique structure allows lipid bilayers to form a stable barrier that controls the movement of molecules and ions in and out of cells and organelles, thus playing a crucial role in maintaining cellular compartmentalization and homeostasis.

Oligonucleotides are short sequences of nucleotides, the building blocks of DNA and RNA. They typically contain fewer than 100 nucleotides, and can be synthesized chemically to have specific sequences. Oligonucleotides are used in a variety of applications in molecular biology, including as probes for detecting specific DNA or RNA sequences, as inhibitors of gene expression, and as components of diagnostic tests and therapies. They can also be used in the study of protein-nucleic acid interactions and in the development of new drugs.

Agglutinins are antibodies that cause the particles (such as red blood cells, bacteria, or viruses) to clump together. They recognize and bind to specific antigens on the surface of these particles, forming a bridge between them and causing them to agglutinate or clump. Agglutinins are an important part of the immune system's response to infection and help to eliminate pathogens from the body.

There are two main types of agglutinins:

1. Naturally occurring agglutinins: These are present in the blood serum of most individuals, even before exposure to an antigen. They can agglutinate some bacteria and red blood cells without prior sensitization. For example, anti-A and anti-B agglutinins are naturally occurring antibodies found in people with different blood groups (A, B, AB, or O).
2. Immune agglutinins: These are produced by the immune system after exposure to an antigen. They develop as part of the adaptive immune response and target specific antigens that the body has encountered before. Immunization with vaccines often leads to the production of immune agglutinins, which can provide protection against future infections.

Agglutination reactions are widely used in laboratory tests for various diagnostic purposes, such as blood typing, detecting bacterial or viral infections, and monitoring immune responses.

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

A binding site on an antibody refers to the specific region on the surface of the antibody molecule that can recognize and bind to a specific antigen. Antibodies are proteins produced by the immune system in response to the presence of foreign substances called antigens. They have two main functions: to neutralize the harmful effects of antigens and to help eliminate them from the body.

The binding site of an antibody is located at the tips of its Y-shaped structure, formed by the variable regions of the heavy and light chains of the antibody molecule. These regions contain unique amino acid sequences that determine the specificity of the antibody for a particular antigen. The binding site can recognize and bind to a specific epitope or region on the antigen, forming an antigen-antibody complex.

The binding between the antibody and antigen is highly specific and depends on non-covalent interactions such as hydrogen bonds, van der Waals forces, and electrostatic attractions. This interaction plays a crucial role in the immune response, as it allows the immune system to recognize and eliminate pathogens and other foreign substances from the body.

The Interleukin-5 Receptor alpha Subunit (IL-5Rα) is a protein that forms part of the Type I cytokine receptor, specifically for the interleukin-5 (IL-5) cytokine. This receptor is found on the surface of hematopoietic cells, such as eosinophils and basophils. The binding of IL-5 to the IL-5Rα subunit initiates intracellular signaling cascades that regulate the growth, activation, differentiation, and survival of eosinophils and basophils, which are crucial in the immune response against parasitic infections and allergic reactions. Mutations in the gene encoding IL-5Rα can lead to altered immune responses and diseases such as hypereosinophilic syndromes.

Heparin sulfate is not exactly referred to as "heparitin sulfate" in medical terminology. The correct term is heparan sulfate, which is a type of glycosaminoglycan (GAG), a long unbranched chain of repeating disaccharide units composed of a hexuronic acid and a hexosamine.

Heparan sulfate is found on the cell surface and in the extracellular matrix, where it plays crucial roles in various biological processes, including cell signaling, regulation of growth factor activity, and control of blood coagulation. It is also an important component of the proteoglycans, which are complex molecules that help to maintain the structural integrity and function of tissues and organs.

Like heparin, heparan sulfate has a high negative charge due to the presence of sulfate groups, which allows it to bind to and interact with various proteins and growth factors. However, heparan sulfate has a more diverse structure than heparin, with variations in the pattern of sulfation along the chain, which leads to specificity in its interactions with different proteins.

Defects in heparan sulfate biosynthesis or function have been implicated in various human diseases, including certain forms of cancer, developmental disorders, and infectious diseases.

Fluorescence spectrometry is a type of analytical technique used to investigate the fluorescent properties of a sample. It involves the measurement of the intensity of light emitted by a substance when it absorbs light at a specific wavelength and then re-emits it at a longer wavelength. This process, known as fluorescence, occurs because the absorbed energy excites electrons in the molecules of the substance to higher energy states, and when these electrons return to their ground state, they release the excess energy as light.

Fluorescence spectrometry typically measures the emission spectrum of a sample, which is a plot of the intensity of emitted light versus the wavelength of emission. This technique can be used to identify and quantify the presence of specific fluorescent molecules in a sample, as well as to study their photophysical properties.

Fluorescence spectrometry has many applications in fields such as biochemistry, environmental science, and materials science. For example, it can be used to detect and measure the concentration of pollutants in water samples, to analyze the composition of complex biological mixtures, or to study the properties of fluorescent nanomaterials.

A "carbohydrate sequence" refers to the specific arrangement or order of monosaccharides (simple sugars) that make up a carbohydrate molecule, such as a polysaccharide or an oligosaccharide. Carbohydrates are often composed of repeating units of monosaccharides, and the sequence in which these units are arranged can have important implications for the function and properties of the carbohydrate.

For example, in glycoproteins (proteins that contain carbohydrate chains), the specific carbohydrate sequence can affect how the protein is processed and targeted within the cell, as well as its stability and activity. Similarly, in complex carbohydrates like starch or cellulose, the sequence of glucose units can determine whether the molecule is branched or unbranched, which can have implications for its digestibility and other properties.

Therefore, understanding the carbohydrate sequence is an important aspect of studying carbohydrate structure and function in biology and medicine.

"Optical processes" is not a specific medical term, but rather a general term that refers to various phenomena and techniques involving the use of light in physics and engineering, which can have applications in medicine. Here are some examples of optical processes that may be relevant to medical fields:

1. Optical imaging: This refers to the use of light to create images of structures within the body. Examples include endoscopy, microscopy, and ophthalmoscopy.
2. Optical spectroscopy: This involves analyzing the interaction between light and matter to identify the chemical composition or physical properties of a sample. It can be used in medical diagnostics to detect abnormalities in tissues or fluids.
3. Laser therapy: Lasers are highly concentrated beams of light that can be used for a variety of medical applications, including surgery, pain relief, and skin treatments.
4. Optogenetics: This is a technique that involves using light to control the activity of specific cells in living organisms. It has potential applications in neuroscience and other fields of medicine.
5. Photodynamic therapy: This is a treatment that uses light to activate a photosensitizing agent, which then produces a chemical reaction that can destroy abnormal cells or tissues.

Overall, optical processes are an important part of many medical technologies and techniques, enabling doctors and researchers to diagnose and treat diseases with greater precision and effectiveness.

Hydrogen sensor Multi-parametric surface plasmon resonance Nano-optics Plasmon Spinplasmonics Surface plasmon polariton Waves ... Surface plasmon resonance (SPR) is a phenomenon that occurs where electrons in a thin metal sheet become excited by light that ... supporting surface electromagnetic waves with sharper resonances (Bloch surface waves). If the surface is patterned with ... In multi-parametric surface plasmon resonance, two SPR curves are acquired by scanning a range of angles at two different ...
With the emergence of surface plasmon resonance (SPR) arrays, investigators now have high-throughput capabilities to accelerate ... Home Webinars On Demand Accelerating Antibody Screening with Array-Based Surface Plasmon Resonance ... With the emergence of surface plasmon resonance (SPR) arrays, investigators now have high-throughput capabilities to accelerate ...
... the changes of the SPR angle when an oscillating electric potential was applied to a gold film on which surface plasmons were ... sensors allows measurements of small variations in surface potentials to be made. We studied ... The high sensitivity of surface-plasmon resonance (SPR) ... The high sensitivity of surface-plasmon resonance (SPR) sensors ... Effect of varying electric potential on surface-plasmon resonance sensing Appl Opt. 2004 Jun 10;43(17):3426-32. doi: 10.1364/ao ...
Chapter 3. Localized Surface Plasmon Resonance-Active Surfaces Applied to Study Interactions of Biomolecules. Jay K. Bhattarai ... the authors present an overview of the recent research advances in the field of localized surface plasmon resonance (LSPR) ... Surface Plasmon Resonance (SPR): Advances in Research and Applications. ... Chapter 2. Surface Plasmon-Induced Variation in the Properties of Gold Nanoparticle-Doped Nematic Liquid Crystals. Amit ...
Recent advances in surface plasmon resonance biosensors for microRNAs detection. Download Prime PubMed App to iPhone, iPad, or ... Recent advances in surface plasmon resonance biosensors for microRNAs detection.. Biosens Bioelectron. 2020 Dec 01; 169:112599. ... Recent Advances in Surface Plasmon Resonance Biosensors for microRNAs Detection. Biosens Bioelectron. 2020 Dec 1;169:112599. ... A novel surface plasmon resonance biosensor for enzyme-free and highly sensitive detection of microRNA based on multi component ...
Ultra high phase sensitive surface plasmon resonance biosensor Author(s): Ying-Chang Li; Chien Chou ... This study demonstrates a novel phase-sensitive surface plasman resonance biosensor (PS-SPRB) which is able to convert the ...
... the feasibility of a glycopeptide-based biosensor to detect MS specific antibodies in sera using the surface plasmon resonance ... the use of magnetic resonance imaging markers. No alternative simpler assays proven of use, except for cerebrospinal fluid ... Surface Plasmon Resonance (SPR). A stock solution of glycopeptide CSF114(Glc) was prepared in pure water (1 μg/μL) and stored ... Glycopeptide-Based Antibody Detection in Multiple Sclerosis by Surface Plasmon Resonance by Feliciana Real-Fernández ...
Surface Plasmon Resonance (SPR) Biosensor Shared Instrument Facility provides vaccine efficacy tracking, compound, or antibody ... Drexel-SKCC/TJU Biacore S200™ Surface Plasmon Resonance (SPR) Biosensor Shared Instrument Facility. ...
A. Willets et al., Localized Surface Plasmon Resonance Spectroscopy and Sensing, Annu. Rev. Phys. Chem., 2007, 58, 267-297. ... S. Unser et al., Localized Surface Plasmon Resonance Biosensing: Current Challenges and Approaches, Sensors (Basel), 2015, 15, ... More specifically, they are capable of supporting a localised surface plasmon resonance (LSPR), caused by a displacement and ... and optical sensing applications because their optical properties are based on localised surface plasmon resonance (LSPR). ...
535e) Adsorption of Dansylated Amino Acids on Molecularly Imprinted Surfaces: a Surface Plasmon Resonance Study. Conference ... ABSTRACT: Surface plasmon resonance spectroscopy (SPR) was used to measure the adsorption capacities and kinetics of dansylated ... The surface-confined polymer films were grafted from flat gold surfaces using Atom Transfer Radical Polymerization (ATRP). This ... thereby ensuring that the amino acids see a uniform surface during adsorption. N,N-didansyl-L-cystine (DDC) and didansyl-L- ...
I confirm that I would like to be contacted by Bruker to answer my request. I have taken note of the Privacy Notice and I agree to the Terms of Use of the Bruker Website ...
Surface Plasmon Resonance Market. Surface Plasmon Resonance Market - Global Industry, Analysis, Size, Growth, Trends, and ...
Additionally, the availability of versatile products coupled with surface plasmon resonance to enhance the specificity of tests ... Development of new technologies coupled with surface plasmon resonance, such as nanoparticle surface plasmon resonance and ... Key Takeaways of Surface Plasmon Resonance Market Study. *The market share of imaging systems is evaluated to grow at a ... The global surface plasmon resonance market is projected to reach a market value of US$ 770 Mn in 2019, increasing at a decent ...
... conventional surface plasmon resonance (SPR), which suffers from a significant bulk-effect issue, has a plasmon evanescent ... Localized Surface Plasmon Resonance for Bioprocess Development, Monitoring, and Validation. View PDF ... Academic laboratories have embraced localized surface plasmon resonance (LSPR) as the "new wave" of label-free technology (1). ... The lack of bulk effect is due to the localized nature of plasmons in the nanostructured surfaces because the plasmon ...
Characterisation by surface plasmon resonance of monoclonal antibodies that address growth hormone isoforms ... For the characterisation of the antibodies the technique of surface plasmon resonance will be employed. In a straight-forward ... In this study, antibodies were compared using the Surface Plasmon Resonance method and this comparison helped in the selection ... Characterisation by surface plasmon resonance of monoclonal antibodies that address growth hormone isoforms ...
Here we used surface plasmon resonance (SPR) technology to develop an immunosensor for the detection of PB1-F2. The sensor is ... Surface Plasmon Resonance Immunosensor for Detection of PB1-F2 Influenza A Virus Protein in Infected Biological Samples Jasmina ... Surface Plasmon resonance (SPR) analysis was performed at 25°C using the Biacore 3000 (GE Healthcare). To immobilize an anti- ... 2013) Surface Plasmon Resonance Immunosensor for Detection of PB1-F2 Influenza A Virus Protein in Infected Biological Samples. ...
Scuola Scuole Seminario sinu space spazio spending review Stony Brooks University succhi di frutta Surface Plasmon Resonance ( ...
Surface plasmon resonance (SPR) has not been widely used for compound screening and in this study it was used to compare human ... Surface plasmon resonance (SPR) has not been widely used for compound screening and in this study it was used to compare human ... Surface plasmon resonance (SPR) has not been widely used for... (More). Galectin-3 is a beta-galactoside-binding mammalian ... the affinity and kinetics of small molecule glycomimetics for human and mouse galectin-3 using surface plasmon resonance. *Mark ...
Absorption, Nanoparticles, Shells, Solar energy, Surface plasmon resonance, Plasmons (Physics), Resonance, Disperse systems, ... Surface Plasmon Resonance Shifts of a Dispersion of Core-Shell Nanoparticles for Efficient Solar Absorption Wei Lv, Wei Lv ... Lv, W, Otanicar, TP, Phelan, PE, Dai, L, Taylor, RA, & Swaminathan, R. "Surface Plasmon Resonance Shifts of a Dispersion of ... Through our electrostatic model, we achieve a shift in the plasmon resonance peak from a wavelength of about 500 nm to around ...
Analys av haptoglobin i bovint serum med surface plasmon resonance biosensorteknik. SLU, Dept. of Clinical Sciences, Uppsala. ... which results in an optically detectable alteration in mass on the sensor surface. ...
Here, we explore the use of label-free surface-based sensing with surface plasmon resonance (SPR) read-out to determine the ... Determination of exosome concentration in solution using surface plasmon resonance spectroscopy. Analytical Chemistry. Rupert, ... Determination of exosome concentration in solution using surface plasmon resonance spectroscopy. Analytical Chemistry ... Determination of exosome concentration in solution using surface plasmon resonance spectroscopy. Analytical Chemistry ...
Surface plasmon resonance sensors improved with carpet of gold. By R&D Editors , June 5, 2013. ... Incoming light sparks localized plasmon resonances around the nanoparticles that couple to the sensor surface, which causes ... Size dependence of Au NP-enhanced surface plasmon resonance based on differential phase measurement ... has determined the ideal size of nanoparticle to improve surface plasmon resonance (SPR) sensors. ...
Surface plasmon resonance. For binding assay experiments, a Biacore 3000 biosensor platform (GE Biosystems) equipped with a ... 2c-d). In agreement with this, surface plasmon resonance analysis shows that, in solution, sialylated compounds 3SL and 6SL ( ... a-c Three different views of the single-particle cryo-EM from the Nap (beige surface) with the structures of P140 (blue) and ... C-terminal domains act like stalks connecting the large N-terminal domain of each subunit with the outer surface of the cell ...
The SPIRIT is a rugged, portable, multi-analyte, instrument based on surface plasmon resonance (SP... ...
A) Surface plasmon resonance analysis of the interaction of each PfRH5 protein variant (v1.0-v5.0) with basigin. (B) Capture ... Surface plasmon resonance (SPR). The production of recombinant basigin in Origami B (DE3) E. coli has been previously described ... The chip surface was then regenerated with 30 s of 2 M NaCl. Specific binding of the PfRH5 protein was obtained by subtracting ... Position 203 (either C or Y) is exposed on the surface of crystallized constructs of PfRH5, where it lies within the basigin ...
NanoPlasmon - Localized surface plasmon resonance spectrometer. * Realization of real-time biokinetic, immunosensing and ... where it can couple to the surface mode and yield localized surface plasmons. Excitation of localized plasmons is evidenced by ... These nanostructures form the sensitive sensor surface where localized surface plasmons are excited using polarized or ... species present in the probe to the immobilized biosystem affects the effective refractive index in the vicinity of the surface ...
Surface Plasmon Resonance occurs when plane-polarized light hits a metal film at the interface of media with different ... The Biacore T200 system combines surface plasmon resonance with sensor chip technology to monitor the interaction between ... The Biacore T200 system allows surface plasmon resonance spectroscopy for label-free measurement of biomolecular interactions ... At a certain angle of incidence, the light will excite surface plasmons, causing a dip in the intensity of the reflected light ...
When does surface plasmon resonance ( SPR ) occur?. Surface plasmon resonance (SPR) is a phenomenon occuring at metal surfaces ... What is surface plasmon resonance applications?. Surface plasmon resonance (SPR) is an optical effect that can be utilized to ... What causes surface plasmon resonance?. Surface Plasmon Resonance is a phenomenon that occurs when polarized light hits a metal ... When was surface plasmon resonance introduced in BIA?. 1. Introduction Surface plasmon resonance (SPR) was introduced in the ...
Search results: "surface plasmon resonance". Showing 29 search results for the search term "surface plasmon resonance" ...

No FAQ available that match "surface plasmon resonance"

No images available that match "surface plasmon resonance"