Antibody

From WikiProjectMed
(Redirected from Antibodies)
Jump to navigation Jump to search

Each antibody binds to a specific antigen in a highly specific interaction analogous to a lock and key.

An antibody (Ab) is the secreted form of a B cell receptor; the term immunoglobulin can refer to either the membrane-bound form or the secreted form of the B cell receptor, but they are, broadly speaking, the same protein, and so the terms are often treated as synonymous.[1] Antibodies are large, Y-shaped proteins belonging to the immunoglobulin superfamily which are used by the immune system to identify and neutralize foreign objects such as bacteria and viruses, including those that cause disease. Antibodies can recognize virtually any size antigen with diverse chemical compositions from molecules.[2] Each antibody recognizes one or more specific antigens.[3][4] This term literally means "antibody generator", as it is the presence of an antigen that drives the formation of an antigen-specific antibody. Each tip of the "Y" of an antibody contains a paratope that specifically binds to one particular epitope on an antigen, allowing the two molecules to bind together with precision. Using this mechanism, antibodies can effectively "tag" a microbe or an infected cell for attack by other parts of the immune system, or can neutralize it directly (for example, by blocking a part of a virus that is essential for its invasion).

To allow the immune system to recognize millions of different antigens, the antigen-binding sites at both tips of the antibody come in an equally wide variety. The rest of the antibody structure is relatively generic. In humans, antibodies occur in five classes, sometimes called isotypes: IgA, IgD, IgE, IgG, and IgM. Human IgG and IgA antibodies are also divided into discrete subclasses (IgG1, IgG2, IgG3, IgG4; IgA1 and IgA2). The class refers to the functions triggered by the antibody (also known as effector functions), in addition to some other structural features. Antibodies from different classes also differ in where they are released in the body and at what stage of an immune response. Importantly, while classes and subclasses of antibodies may be shared between species (at least in name), their functions and distribution throughout the body may be different. For example, mouse IgG1 is closer to human IgG2 than human IgG1 in terms of its function.

The term humoral immunity is often treated as synonymous with the antibody response, describing the function of the immune system that exists in the body's humors (fluids) in the form of soluble proteins, as distinct from cell-mediated immunity, which generally describes the responses of T cells (especially cytotoxic T cells). In general, antibodies are considered part of the adaptive immune system, though this classification can become complicated. For example, natural IgM,[5] which are made by B-1 lineage cells that have properties more similar to innate immune cells than adaptive, refers to IgM antibodies made independently of an immune response that demonstrate polyreactivity- they recognize multiple distinct (unrelated) antigens. These can work with the complement system in the earliest phases of an immune response to help facilitate clearance of the offending antigen and delivery of the resulting immune complexes to the lymph nodes or spleen for initiation of an immune response. Hence in this capacity, the function of antibodies is more akin to that of innate immunity than adaptive. Nonetheless, in general antibodies are regarded as part of the adaptive immune system because they demonstrate exceptional specificity (with some exception), are produced through genetic rearrangements (rather than being encoded directly in germline), and are a manifestation of immunological memory.

In the course of an immune response, B cells can progressively differentiate into antibody-secreting cells (B cells themselves do not secrete antibody; B cells do, however, express B cell receptors, the membrane-bound form of the antibody, on their surface) or memory B cells.[6] Antibody-secreting cells comprise plasmablasts and plasma cells, which differ mainly in the degree to which they secrete antibody, their lifespan, metabolic adaptations, and surface markers.[7] Plasmablasts are rapidly proliferating, short-lived cells produced in the early phases of the immune response (classically described as arising extrafollicularly rather than from the germinal center) which have the potential to differentiate further into plasma cells.[8] The literature is sloppy at times and often describes plasmablasts as just short-lived plasma cells- formally this is incorrect. Plasma cells, in contrast, do not divide (they are terminally differentiated), and rely on survival niches comprising specific cell types and cytokines to persist.[9] Plasma cells will secrete huge quantities of antibody regardless of whether or not their cognate antigen is present, ensuring that antibody levels to the antigen in question do not fall to 0, provided the plasma cell stays alive. The rate of antibody secretion, however, can be regulated, for example, by the presence of adjuvant molecules that stimulate the immune response such as TLR ligands.[10] Long-lived plasma cells can live for potentially the entire lifetime of the organism.[11] Classically, the survival niches that house long-lived plasma cells reside in the bone marrow,[12] though it cannot be assumed that any given plasma cell in the bone marrow will be long-lived. However, other work indicates that survival niches can readily be established within the mucosal tissues- though the classes of antibodies involved show a different hierarchy from those in the bone marrow.[13][14] B cells can also differentiate into memory B cells which can persist for decades similarly to long-lived plasma cells. These cells can be rapidly recalled in a secondary immune response, undergoing class switching, affinity maturation, and differentiating into antibody-secreting cells.

Antibodies are central to the immune protection elicited by most vaccines and infections (although other components of the immune system certainly participate and for some diseases are considerably more important than antibodies in generating an immune response, e.g. herpes zoster).[15] Durable protection from infections caused by a given microbe – that is, the ability of the microbe to enter the body and begin to replicate (not necessarily to cause disease) – depends on sustained production of large quantities of antibodies, meaning that effective vaccines ideally elicit persistent high levels of antibody, which relies on long-lived plasma cells. At the same time, many microbes of medical importance have the ability to mutate to escape antibodies elicited by prior infections, and long-lived plasma cells cannot undergo affinity maturation or class switching. This is compensated for through memory B cells: novel variants of a microbe that still retain structural features of previously encountered antigens can elicit memory B cell responses that adapt to those changes. It has been suggested that long-lived plasma cells secrete B cell receptors with higher affinity than those on the surfaces of memory B cells, but findings are not entirely consistent on this point.[16]

Structure

Schematic structure of an antibody: two heavy chains (blue, yellow) and the two light chains (green, pink). The antigen binding site is circled.
Model of an antibody showing beta strands
Surface model of an antibody at the molecular level
A more accurate depiction of an antibody (3D structure at RCSB PDB). Glycans in the Fc region are shown in black.

Antibodies are heavy (~150 kDa) proteins of about 10 nm in size,[17] arranged in three globular regions that roughly form a Y shape.

In humans and most other mammals, an antibody unit consists of four polypeptide chains; two identical heavy chains and two identical light chains connected by disulfide bonds.[18] Each chain is a series of domains: somewhat similar sequences of about 110 amino acids each. These domains are usually represented in simplified schematics as rectangles. Light chains consist of one variable domain VL and one constant domain CL, while heavy chains contain one variable domain VH and three to four constant domains CH1, CH2, ...[19]

Structurally an antibody is also partitioned into two antigen-binding fragments (Fab), containing one VL, VH, CL, and CH1 domain each, as well as the crystallisable fragment (Fc), forming the trunk of the Y shape.[20] In between them is a hinge region of the heavy chains, whose flexibility allows antibodies to bind to pairs of epitopes at various distances, to form complexes (dimers, trimers, etc.), and to bind effector molecules more easily.[21]

In an electrophoresis test of blood proteins, antibodies mostly migrate to the last, gamma globulin fraction. Conversely, most gamma-globulins are antibodies, which is why the two terms were historically used as synonyms, as were the symbols Ig and γ. This variant terminology fell out of use due to the correspondence being inexact and due to confusion with γ (gamma) heavy chains which characterize the IgG class of antibodies.[22][23]

Antigen-binding site

The variable domains can also be referred to as the FV region. It is the subregion of Fab that binds to an antigen. More specifically, each variable domain contains three hypervariable regions – the amino acids seen there vary the most from antibody to antibody. When the protein folds, these regions give rise to three loops of β-strands, localized near one another on the surface of the antibody. These loops are referred to as the complementarity-determining regions (CDRs), since their shape complements that of an antigen. Three CDRs from each of the heavy and light chains together form an antibody-binding site whose shape can be anything from a pocket to which a smaller antigen binds, to a larger surface, to a protrusion that sticks out into a groove in an antigen. Typically however only a few residues contribute to most of the binding energy.[3]

The existence of two identical antibody-binding sites allows antibody molecules to bind strongly to multivalent antigen (repeating sites such as polysaccharides in bacterial cell walls, or other sites at some distance apart), as well as to form antibody complexes and larger antigen-antibody complexes.[3] The resulting cross-linking plays a role in activating other parts of the immune system.[citation needed]

The structures of CDRs have been clustered and classified by Chothia et al.[24] and more recently by North et al.[25] and Nikoloudis et al.[26] However, describing an antibody's binding site using only one single static structure limits the understanding and characterization of the antibody's function and properties. To improve antibody structure prediction and to take the strongly correlated CDR loop and interface movements into account, antibody paratopes should be described as interconverting states in solution with varying probabilities.[27]

In the framework of the immune network theory, CDRs are also called idiotypes. According to immune network theory, the adaptive immune system is regulated by interactions between idiotypes.

Fc region

The Fc region (the trunk of the Y shape) is composed of constant domains from the heavy chains. Its role is in modulating immune cell activity: it is where effector molecules bind to, triggering various effects after the antibody Fab region binds to an antigen.[3][21] Effector cells (such as macrophages or natural killer cells) bind via their Fc receptors (FcR) to the Fc region of an antibody, while the complement system is activated by binding the C1q protein complex. IgG or IgM can bind to C1q, but IgA cannot, therefore IgA does not activate the classical complement pathway.[28]

Another role of the Fc region is to selectively distribute different antibody classes across the body. In particular, the neonatal Fc receptor (FcRn) binds to the Fc region of IgG antibodies to transport it across the placenta, from the mother to the fetus. In addition to this, binding to FcRn endows IgG with an exceptionally long half-life relative to other plasma proteins of 3-4 weeks. IgG3 in most cases (depending on allotype) has mutations at the FcRn binding site which lower affinity for FcRn, which are thought to have evolved to limit the highly inflammatory effects of this subclass.[29]

Antibodies are glycoproteins,[30] that is, they have carbohydrates (glycans) added to conserved amino acid residues.[30][31] These conserved glycosylation sites occur in the Fc region and influence interactions with effector molecules.[30][32]

Protein structure

The N-terminus of each chain is situated at the tip. Each immunoglobulin domain has a similar structure, characteristic of all the members of the immunoglobulin superfamily: it is composed of between 7 (for constant domains) and 9 (for variable domains) β-strands, forming two beta sheets in a Greek key motif. The sheets create a "sandwich" shape, the immunoglobulin fold, held together by a disulfide bond.

Antibody complexes

Some antibodies form complexes that bind to multiple antigen molecules.

Secreted antibodies can occur as a single Y-shaped unit, a monomer. However, some antibody classes also form dimers with two Ig units (as with IgA), tetramers with four Ig units (like teleost fish IgM), or pentamers with five Ig units (like shark IgW or mammalian IgM, which occasionally forms hexamers as well, with six units).[33] IgG can also form hexamers, though no J chain is required.[34] IgA tetramers and pentamers have also been reported.[35]

Antibodies also form complexes by binding to antigen: this is called an antigen-antibody complex or immune complex. Small antigens can cross-link two antibodies, also leading to the formation of antibody dimers, trimers, tetramers, etc. Multivalent antigens (e.g., cells with multiple epitopes) can form larger complexes with antibodies. An extreme example is the clumping, or agglutination, of red blood cells with antibodies in the Coombs test to determine blood groups: the large clumps become insoluble, leading to visually apparent precipitation.

B cell receptors

The membrane-bound form of an antibody may be called a surface immunoglobulin (sIg) or a membrane immunoglobulin (mIg). It is part of the B cell receptor (BCR), which allows a B cell to detect when a specific antigen is present in the body and triggers B cell activation.[36] The BCR is composed of surface-bound IgD or IgM antibodies and associated Ig-α and Ig-β heterodimers, which are capable of signal transduction.[37] A typical human B cell will have 50,000 to 100,000 antibodies bound to its surface.[37] Upon antigen binding, they cluster in large patches, which can exceed 1 micrometer in diameter, on lipid rafts that isolate the BCRs from most other cell signaling receptors.[37] These patches may improve the efficiency of the cellular immune response.[38] In humans, the cell surface is bare around the B cell receptors for several hundred nanometers,[37] which further isolates the BCRs from competing influences.

Classes

Antibodies can come in different varieties known as isotypes or classes. In humans there are five antibody classes known as IgA, IgD, IgE, IgG, and IgM, which are further subdivided into subclasses such as IgA1, IgA2. The prefix "Ig" stands for immunoglobulin, while the suffix denotes the type of heavy chain the antibody contains: the heavy chain types α (alpha), γ (gamma), δ (delta), ε (epsilon), μ (mu) give rise to IgA, IgG, IgD, IgE, IgM, respectively. The distinctive features of each class are determined by the part of the heavy chain within the hinge and Fc region.[3]

The classes differ in their biological properties, functional locations and ability to deal with different antigens, as depicted in the table.[18] For example, IgE antibodies are responsible for an allergic response consisting of histamine release from mast cells, often a sole contributor to asthma (though other pathways exist as do exist symptoms very similar to yet not technically asthma). The antibody's variable region binds to allergic antigen, for example house dust mite particles, while its Fc region (in the ε heavy chains) binds to Fc receptor ε on a mast cell, triggering its degranulation: the release of molecules stored in its granules.[39]

Antibody isotypes of humans
Class Subclasses Description
IgA 2 Found in mucosal areas, such as the gut, respiratory tract and urogenital tract, and prevents colonization by pathogens.[40] Also found in saliva, tears, and breast milk.
IgD 1 Functions mainly as an antigen receptor on B cells that have not been exposed to antigens.[41] It has been shown to activate basophils and mast cells to produce antimicrobial factors.[42]
IgE 1 Binds to allergens and triggers histamine release from mast cells and basophils, and is involved in allergy. Humans and other animals evolved IgE to protect against parasitic worms, though in the present, IgE is primarily related to allergies and asthma.[43]
IgG 4 In its four forms, provides the majority of antibody-based immunity against invading pathogens.[43] The only antibody capable of crossing the placenta to give passive immunity to the fetus.
IgM 1 Expressed on the surface of B cells (monomer) and in a secreted form (pentamer) with very high avidity. Eliminates pathogens in the early stages of B cell-mediated (humoral) immunity before there is sufficient IgG.[43][41]

The antibody isotype of a B cell changes during cell development and activation. Immature B cells, which have never been exposed to an antigen, express only the IgM isotype in a cell surface bound form. The B lymphocyte, in this ready-to-respond form, is known as a "naive B lymphocyte." The naive B lymphocyte expresses both surface IgM and IgD. The co-expression of both of these immunoglobulin isotypes renders the B cell ready to respond to antigen.[44] B cell activation follows engagement of the cell-bound antibody molecule with an antigen, causing the cell to divide and differentiate into an antibody-producing cell called a plasma cell. In this activated form, the B cell starts to produce antibody in a secreted form rather than a membrane-bound form. Some daughter cells of the activated B cells undergo isotype switching, a mechanism that causes the production of antibodies to change from IgM or IgD to the other antibody isotypes, IgE, IgA, or IgG, that have defined roles in the immune system.

Light chain types

In mammals there are two types of immunoglobulin light chain, which are called lambda (λ) and kappa (κ). However, there is no known functional difference between them, and both can occur with any of the five major types of heavy chains.[3] Each antibody contains two identical light chains: both κ or both λ. Proportions of κ and λ types vary by species and can be used to detect abnormal proliferation of B cell clones. Other types of light chains, such as the iota (ι) chain, are found in other vertebrates like sharks (Chondrichthyes) and bony fishes (Teleostei).

In non-mammalian animals

In most placental mammals, the structure of antibodies is generally the same. Jawed fish appear to be the most primitive animals that are able to make antibodies similar to those of mammals, although many features of their adaptive immunity appeared somewhat earlier.[45]

Cartilaginous fish (such as sharks) produce heavy-chain-only antibodies (i.e., lacking light chains) which moreover feature longer chain pentamers (with five constant units per molecule). Camelids (such as camels, llamas, alpacas) are also notable for producing heavy-chain-only antibodies.[3][46]

Antibody classes not found in mammals
Class Types Description
IgY Found in birds and reptiles; related to mammalian IgG.[47]
IgW Found in sharks and skates; related to mammalian IgD.[48]
IgT/Z Found in teleost fish[49]

Antibody–antigen interactions

The antibody's paratope interacts with the antigen's epitope. An antigen usually contains different epitopes along its surface arranged discontinuously, and dominant epitopes on a given antigen are called determinants.

Antibody and antigen interact by spatial complementarity (lock and key). The molecular forces involved in the Fab-epitope interaction are weak and non-specific – for example electrostatic forces, hydrogen bonds, hydrophobic interactions, and van der Waals forces. This means binding between antibody and antigen is reversible, and the antibody's affinity towards an antigen is relative rather than absolute. Relatively weak binding also means it is possible for an antibody to cross-react with different antigens of different relative affinities.

Function

  1. Antibodies (A) and pathogens (B) free roam in the blood.
  2. The antibodies bind to pathogens, and can do so in different formations such as:
    1. opsonization,
    2. neutralisation, and
    3. agglutination.
  3. A phagocyte (C) approaches the pathogen, and the Fc region (D) of the antibody binds to one of the Fc receptors (E) of the phagocyte.
  4. Phagocytosis occurs as the pathogen is ingested.

The main categories of antibody action include the following:

More indirectly, an antibody can signal immune cells to present antibody fragments to T cells, or downregulate other immune cells to avoid autoimmunity.

Activated B cells differentiate into either antibody-producing cells called plasma cells that secrete soluble antibody or memory cells that survive in the body for years afterward in order to allow the immune system to remember an antigen and respond faster upon future exposures.[50]

At the prenatal and neonatal stages of life, the presence of antibodies is provided by passive immunization from the mother. Early endogenous antibody production varies for different kinds of antibodies, and usually appear within the first years of life. Since antibodies exist freely in the bloodstream, they are said to be part of the humoral immune system. Circulating antibodies are produced by clonal B cells that specifically respond to only one antigen (an example is a virus capsid protein fragment). Antibodies contribute to immunity in three ways: They prevent pathogens from entering or damaging cells by binding to them; they stimulate removal of pathogens by macrophages and other cells by coating the pathogen; and they trigger destruction of pathogens by stimulating other immune responses such as the complement pathway.[51] Antibodies will also trigger vasoactive amine degranulation to contribute to immunity against certain types of antigens (helminths, allergens).

The secreted mammalian IgM has five Ig units. Each Ig unit (labeled 1) has two epitope binding Fab regions, so IgM is capable of binding up to 10 epitopes.

Activation of complement

Antibodies that bind to surface antigens (for example, on bacteria) will attract the first component of the complement cascade with their Fc region and initiate activation of the "classical" complement system.[51] This results in the killing of bacteria in two ways.[43] First, the binding of the antibody and complement molecules marks the microbe for ingestion by phagocytes in a process called opsonization; these phagocytes are attracted by certain complement molecules generated in the complement cascade. Second, some complement system components form a membrane attack complex to assist antibodies to kill the bacterium directly (bacteriolysis).[52]

Activation of effector cells

To combat pathogens that replicate outside cells, antibodies bind to pathogens to link them together, causing them to agglutinate. Since an antibody has at least two paratopes, it can bind more than one antigen by binding identical epitopes carried on the surfaces of these antigens. By coating the pathogen, antibodies stimulate effector functions against the pathogen in cells that recognize their Fc region.[43]

Those cells that recognize coated pathogens have Fc receptors, which, as the name suggests, interact with the Fc region of IgA, IgG, and IgE antibodies. The engagement of a particular antibody with the Fc receptor on a particular cell triggers an effector function of that cell; phagocytes will phagocytose, mast cells and neutrophils will degranulate, natural killer cells will release cytokines and cytotoxic molecules; that will ultimately result in destruction of the invading microbe. The activation of natural killer cells by antibodies initiates a cytotoxic mechanism known as antibody-dependent cell-mediated cytotoxicity (ADCC) – this process may explain the efficacy of monoclonal antibodies used in biological therapies against cancer. The Fc receptors are isotype-specific, which gives greater flexibility to the immune system, invoking only the appropriate immune mechanisms for distinct pathogens.[3]

Natural antibodies

Humans and higher primates also produce "natural antibodies" that are present in serum before viral infection. Natural antibodies have been defined as antibodies that are produced without any previous infection, vaccination, other foreign antigen exposure or passive immunization. These antibodies can activate the classical complement pathway leading to lysis of enveloped virus particles long before the adaptive immune response is activated. Many natural antibodies are directed against the disaccharide galactose α(1,3)-galactose (α-Gal), which is found as a terminal sugar on glycosylated cell surface proteins, and generated in response to production of this sugar by bacteria contained in the human gut.[53] Rejection of xenotransplantated organs is thought to be, in part, the result of natural antibodies circulating in the serum of the recipient binding to α-Gal antigens expressed on the donor tissue.[54]

Immunoglobulin diversity

Virtually all microbes can trigger an antibody response. Successful recognition and eradication of many different types of microbes requires diversity among antibodies; their amino acid composition varies allowing them to interact with many different antigens.[55] It has been estimated that humans generate about 10 billion different antibodies, each capable of binding a distinct epitope of an antigen.[56] Although a huge repertoire of different antibodies is generated in a single individual, the number of genes available to make these proteins is limited by the size of the human genome. Several complex genetic mechanisms have evolved that allow vertebrate B cells to generate a diverse pool of antibodies from a relatively small number of antibody genes.[57]

Domain variability

The complementarity determining regions of the heavy chain are shown in red (PDB: 1IGT​)

The chromosomal region that encodes an antibody is large and contains several distinct gene loci for each domain of the antibody—the chromosome region containing heavy chain genes (IGH@) is found on chromosome 14, and the loci containing lambda and kappa light chain genes (IGL@ and IGK@) are found on chromosomes 22 and 2 in humans. One of these domains is called the variable domain, which is present in each heavy and light chain of every antibody, but can differ in different antibodies generated from distinct B cells. Differences between the variable domains are located on three loops known as hypervariable regions (HV-1, HV-2 and HV-3) or complementarity-determining regions (CDR1, CDR2 and CDR3). CDRs are supported within the variable domains by conserved framework regions. The heavy chain locus contains about 65 different variable domain genes that all differ in their CDRs. Combining these genes with an array of genes for other domains of the antibody generates a large cavalry of antibodies with a high degree of variability. This combination is called V(D)J recombination discussed below.[58]

V(D)J recombination

Simplified overview of V(D)J recombination of immunoglobulin heavy chains

Somatic recombination of immunoglobulins, also known as V(D)J recombination, involves the generation of a unique immunoglobulin variable region. The variable region of each immunoglobulin heavy or light chain is encoded in several pieces—known as gene segments (subgenes). These segments are called variable (V), diversity (D) and joining (J) segments.[57] V, D and J segments are found in Ig heavy chains, but only V and J segments are found in Ig light chains. Multiple copies of the V, D and J gene segments exist, and are tandemly arranged in the genomes of mammals. In the bone marrow, each developing B cell will assemble an immunoglobulin variable region by randomly selecting and combining one V, one D and one J gene segment (or one V and one J segment in the light chain). As there are multiple copies of each type of gene segment, and different combinations of gene segments can be used to generate each immunoglobulin variable region, this process generates a huge number of antibodies, each with different paratopes, and thus different antigen specificities.[59] The rearrangement of several subgenes (i.e. V2 family) for lambda light chain immunoglobulin is coupled with the activation of microRNA miR-650, which further influences biology of B-cells.

RAG proteins play an important role with V(D)J recombination in cutting DNA at a particular region.[59] Without the presence of these proteins, V(D)J recombination would not occur.[59]

After a B cell produces a functional immunoglobulin gene during V(D)J recombination, it cannot express any other variable region (a process known as allelic exclusion) thus each B cell can produce antibodies containing only one kind of variable chain.[3][60]

Somatic hypermutation and affinity maturation

Following activation with antigen, B cells begin to proliferate rapidly. In these rapidly dividing cells, the genes encoding the variable domains of the heavy and light chains undergo a high rate of point mutation, by a process called somatic hypermutation (SHM). SHM results in approximately one nucleotide change per variable gene, per cell division.[61] As a consequence, any daughter B cells will acquire slight amino acid differences in the variable domains of their antibody chains.

This serves to increase the diversity of the antibody pool and impacts the antibody's antigen-binding affinity.[62] Some point mutations will result in the production of antibodies that have a weaker interaction (low affinity) with their antigen than the original antibody, and some mutations will generate antibodies with a stronger interaction (high affinity).[63] B cells that express high affinity antibodies on their surface will receive a strong survival signal during interactions with other cells, whereas those with low affinity antibodies will not, and will die by apoptosis.[63] Thus, B cells expressing antibodies with a higher affinity for the antigen will outcompete those with weaker affinities for function and survival allowing the average affinity of antibodies to increase over time. The process of generating antibodies with increased binding affinities is called affinity maturation. Affinity maturation occurs in mature B cells after V(D)J recombination, and is dependent on help from helper T cells.[64]

Class switching

Mechanism of class switch recombination that allows isotype switching in activated B cells

Isotype or class switching is a biological process occurring after activation of the B cell, which allows the cell to produce different classes of antibody (IgA, IgE, or IgG).[59] The different classes of antibody, and thus effector functions, are defined by the constant (C) regions of the immunoglobulin heavy chain. Initially, naive B cells express only cell-surface IgM and IgD with identical antigen binding regions. Each isotype is adapted for a distinct function; therefore, after activation, an antibody with an IgG, IgA, or IgE effector function might be required to effectively eliminate an antigen. Class switching allows different daughter cells from the same activated B cell to produce antibodies of different isotypes. Only the constant region of the antibody heavy chain changes during class switching; the variable regions, and therefore antigen specificity, remain unchanged. Thus the progeny of a single B cell can produce antibodies, all specific for the same antigen, but with the ability to produce the effector function appropriate for each antigenic challenge. Class switching is triggered by cytokines; the isotype generated depends on which cytokines are present in the B cell environment.[65]

Class switching occurs in the heavy chain gene locus by a mechanism called class switch recombination (CSR). This mechanism relies on conserved nucleotide motifs, called switch (S) regions, found in DNA upstream of each constant region gene (except in the δ-chain). The DNA strand is broken by the activity of a series of enzymes at two selected S-regions.[66][67] The variable domain exon is rejoined through a process called non-homologous end joining (NHEJ) to the desired constant region (γ, α or ε). This process results in an immunoglobulin gene that encodes an antibody of a different isotype.[68]

Specificity designations

An antibody can be called monospecific if it has specificity for a single antigen or epitope,[69] or bispecific if it has affinity for two different antigens or two different epitopes on the same antigen.[70] A group of antibodies can be called polyvalent (or unspecific) if they have affinity for various antigens[71] or microorganisms.[71] Intravenous immunoglobulin, if not otherwise noted, consists of a variety of different IgG (polyclonal IgG). In contrast, monoclonal antibodies are identical antibodies produced by a single B cell.

Asymmetrical antibodies

Heterodimeric antibodies, which are also asymmetrical antibodies, allow for greater flexibility and new formats for attaching a variety of drugs to the antibody arms. One of the general formats for a heterodimeric antibody is the "knobs-into-holes" format. This format is specific to the heavy chain part of the constant region in antibodies. The "knobs" part is engineered by replacing a small amino acid with a larger one. It fits into the "hole", which is engineered by replacing a large amino acid with a smaller one. What connects the "knobs" to the "holes" are the disulfide bonds between each chain. The "knobs-into-holes" shape facilitates antibody dependent cell mediated cytotoxicity. Single-chain variable fragments (scFv) are connected to the variable domain of the heavy and light chain via a short linker peptide. The linker is rich in glycine, which gives it more flexibility, and serine/threonine, which gives it specificity. Two different scFv fragments can be connected together, via a hinge region, to the constant domain of the heavy chain or the constant domain of the light chain.[72] This gives the antibody bispecificity, allowing for the binding specificities of two different antigens.[73] The "knobs-into-holes" format enhances heterodimer formation but does not suppress homodimer formation.

To further improve the function of heterodimeric antibodies, many scientists are looking towards artificial constructs. Artificial antibodies are largely diverse protein motifs that use the functional strategy of the antibody molecule, but are not limited by the loop and framework structural constraints of the natural antibody.[74] Being able to control the combinational design of the sequence and three-dimensional space could transcend the natural design and allow for the attachment of different combinations of drugs to the arms.

Heterodimeric antibodies have a greater range in shapes they can take and the drugs that are attached to the arms do not have to be the same on each arm, allowing for different combinations of drugs to be used in cancer treatment. Pharmaceuticals are able to produce highly functional bispecific, and even multispecific, antibodies. The degree to which they can function is impressive given that such a change of shape from the natural form should lead to decreased functionality.

Interchromosomal DNA Transposition

Antibody diversification typically occurs through somatic hypermutation, class switching, and affinity maturation targeting the BCR gene loci, but on occasion more unconventional forms of diversification have been documented.[75] For example, in the case of malaria caused by Plasmodium falciparum, some antibodies from those who had been infected demonstrated an insertion from chromosome 19 containing a 98-amino acid stretch from leukocyte-associated immunoglobulin-like receptor 1, LAIR1, in the elbow joint. This represents a form of interchromosomal transposition. LAIR1 normally binds collagen, but can recognize repetitive interspersed families of polypeptides (RIFIN) family members that are highly expressed on the surface of P. falciparum-infected red blood cells. In fact, these antibodies underwent affinity maturation that enhanced affinity for RIFIN but abolished affinity for collagen. These "LAIR1-containing" antibodies have been found in 5-10% of donors from Tanzania and Mali, though not in European donors.[76] European donors did show 100-1000 nucleotide stretches inside the elbow joints as well, however. This particular phenomenon may be specific to malaria, as infection is known to induce genomic instability.[77]

History

The first use of the term "antibody" occurred in a text by Paul Ehrlich. The term Antikörper (the German word for antibody) appears in the conclusion of his article "Experimental Studies on Immunity", published in October 1891, which states that, "if two substances give rise to two different Antikörper, then they themselves must be different".[78] However, the term was not accepted immediately and several other terms for antibody were proposed; these included Immunkörper, Amboceptor, Zwischenkörper, substance sensibilisatrice, copula, Desmon, philocytase, fixateur, and Immunisin.[78] The word antibody has formal analogy to the word antitoxin and a similar concept to Immunkörper (immune body in English).[78] As such, the original construction of the word contains a logical flaw; the antitoxin is something directed against a toxin, while the antibody is a body directed against something.[78]

Angel of the West (2008) by Julian Voss-Andreae is a sculpture based on the antibody structure published by E. Padlan.[79] Created for the Florida campus of the Scripps Research Institute,[80] the antibody is placed into a ring referencing Leonardo da Vinci's Vitruvian Man thus highlighting the similarity of the antibody and the human body.[81]

The study of antibodies began in 1890 when Emil von Behring and Kitasato Shibasaburō described antibody activity against diphtheria and tetanus toxins. Von Behring and Kitasato put forward the theory of humoral immunity, proposing that a mediator in serum could react with a foreign antigen.[82][83] His idea prompted Paul Ehrlich to propose the side-chain theory for antibody and antigen interaction in 1897, when he hypothesized that receptors (described as "side-chains") on the surface of cells could bind specifically to toxins – in a "lock-and-key" interaction – and that this binding reaction is the trigger for the production of antibodies.[84] Other researchers believed that antibodies existed freely in the blood and, in 1904, Almroth Wright suggested that soluble antibodies coated bacteria to label them for phagocytosis and killing; a process that he named opsoninization.[85]

Michael Heidelberger

In the 1920s, Michael Heidelberger and Oswald Avery observed that antigens could be precipitated by antibodies and went on to show that antibodies are made of protein.[86] The biochemical properties of antigen-antibody-binding interactions were examined in more detail in the late 1930s by John Marrack.[87] The next major advance was in the 1940s, when Linus Pauling confirmed the lock-and-key theory proposed by Ehrlich by showing that the interactions between antibodies and antigens depend more on their shape than their chemical composition.[88] In 1948, Astrid Fagraeus discovered that B cells, in the form of plasma cells, were responsible for generating antibodies.[89]

Further work concentrated on characterizing the structures of the antibody proteins. A major advance in these structural studies was the discovery in the early 1960s by Gerald Edelman and Joseph Gally of the antibody light chain,[90] and their realization that this protein is the same as the Bence-Jones protein described in 1845 by Henry Bence Jones.[91] Edelman went on to discover that antibodies are composed of disulfide bond-linked heavy and light chains. Around the same time, antibody-binding (Fab) and antibody tail (Fc) regions of IgG were characterized by Rodney Porter.[92] Together, these scientists deduced the structure and complete amino acid sequence of IgG, a feat for which they were jointly awarded the 1972 Nobel Prize in Physiology or Medicine.[92] The Fv fragment was prepared and characterized by David Givol.[93] While most of these early studies focused on IgM and IgG, other immunoglobulin isotypes were identified in the 1960s: Thomas Tomasi discovered secretory antibody (IgA);[94] David S. Rowe and John L. Fahey discovered IgD;[95] and Kimishige Ishizaka and Teruko Ishizaka discovered IgE and showed it was a class of antibodies involved in allergic reactions.[96] In a landmark series of experiments beginning in 1976, Susumu Tonegawa showed that genetic material can rearrange itself to form the vast array of available antibodies.[97]

Medical applications

Disease diagnosis

Detection of particular antibodies is a very common form of medical diagnostics, and applications such as serology depend on these methods.[98] For example, in biochemical assays for disease diagnosis,[99] a titer of antibodies directed against Epstein-Barr virus or Lyme disease is estimated from the blood. If those antibodies are not present, either the person is not infected or the infection occurred a very long time ago, and the B cells generating these specific antibodies have naturally decayed.

In clinical immunology, levels of individual classes of immunoglobulins are measured by nephelometry (or turbidimetry) to characterize the antibody profile of patient.[100] Elevations in different classes of immunoglobulins are sometimes useful in determining the cause of liver damage in patients for whom the diagnosis is unclear.[1] For example, elevated IgA indicates alcoholic cirrhosis, elevated IgM indicates viral hepatitis and primary biliary cirrhosis, while IgG is elevated in viral hepatitis, autoimmune hepatitis and cirrhosis.

Autoimmune disorders can often be traced to antibodies that bind the body's own epitopes; many can be detected through blood tests. Antibodies directed against red blood cell surface antigens in immune mediated hemolytic anemia are detected with the Coombs test.[101] The Coombs test is also used for antibody screening in blood transfusion preparation and also for antibody screening in antenatal women.[101]

Practically, several immunodiagnostic methods based on detection of complex antigen-antibody are used to diagnose infectious diseases, for example ELISA, immunofluorescence, Western blot, immunodiffusion, immunoelectrophoresis, and magnetic immunoassay.[102] Antibodies raised against human chorionic gonadotropin are used in over the counter pregnancy tests.

New dioxaborolane chemistry enables radioactive fluoride (18F) labeling of antibodies, which allows for positron emission tomography (PET) imaging of cancer.[103]

Disease therapy

Targeted monoclonal antibody therapy is employed to treat diseases such as rheumatoid arthritis,[104] multiple sclerosis,[105] psoriasis,[106] and many forms of cancer including non-Hodgkin's lymphoma,[107] colorectal cancer, head and neck cancer and breast cancer.[108]

Some immune deficiencies, such as X-linked agammaglobulinemia and hypogammaglobulinemia, result in partial or complete lack of antibodies.[109] These diseases are often treated by inducing a short-term form of immunity called passive immunity. Passive immunity is achieved through the transfer of ready-made antibodies in the form of human or animal serum, pooled immunoglobulin or monoclonal antibodies, into the affected individual.[110]

Prenatal therapy

Rh factor, also known as Rh D antigen, is an antigen found on red blood cells; individuals that are Rh-positive (Rh+) have this antigen on their red blood cells and individuals that are Rh-negative (Rh–) do not. During normal childbirth, delivery trauma or complications during pregnancy, blood from a fetus can enter the mother's system. In the case of an Rh-incompatible mother and child, consequential blood mixing may sensitize an Rh- mother to the Rh antigen on the blood cells of the Rh+ child, putting the remainder of the pregnancy, and any subsequent pregnancies, at risk for hemolytic disease of the newborn.[111]

Rho(D) immune globulin antibodies are specific for human RhD antigen.[112] Anti-RhD antibodies are administered as part of a prenatal treatment regimen to prevent sensitization that may occur when a Rh-negative mother has a Rh-positive fetus. Treatment of a mother with Anti-RhD antibodies prior to and immediately after trauma and delivery destroys Rh antigen in the mother's system from the fetus. This occurs before the antigen can stimulate maternal B cells to "remember" Rh antigen by generating memory B cells. Therefore, her humoral immune system will not make anti-Rh antibodies, and will not attack the Rh antigens of the current or subsequent babies. Rho(D) Immune Globulin treatment prevents sensitization that can lead to Rh disease, but does not prevent or treat the underlying disease itself.[112]

Research applications

Immunofluorescence image of the eukaryotic cytoskeleton. Microtubules as shown in green, are marked by an antibody conjugated to a green fluorescing molecule, FITC.

Specific antibodies are produced by injecting an antigen into a mammal, such as a mouse, rat, rabbit, goat, sheep, or horse for large quantities of antibody. Blood isolated from these animals contains polyclonal antibodies—multiple antibodies that bind to the same antigen—in the serum, which can now be called antiserum. Antigens are also injected into chickens for generation of polyclonal antibodies in egg yolk.[113] To obtain antibody that is specific for a single epitope of an antigen, antibody-secreting lymphocytes are isolated from the animal and immortalized by fusing them with a cancer cell line. The fused cells are called hybridomas, and will continually grow and secrete antibody in culture. Single hybridoma cells are isolated by dilution cloning to generate cell clones that all produce the same antibody; these antibodies are called monoclonal antibodies.[114] Polyclonal and monoclonal antibodies are often purified using Protein A/G or antigen-affinity chromatography.[115]

In research, purified antibodies are used in many applications. Antibodies for research applications can be found directly from antibody suppliers, or through use of a specialist search engine. Research antibodies are most commonly used to identify and locate intracellular and extracellular proteins. Antibodies are used in flow cytometry to differentiate cell types by the proteins they express; different types of cells express different combinations of cluster of differentiation molecules on their surface, and produce different intracellular and secretable proteins.[116] They are also used in immunoprecipitation to separate proteins and anything bound to them (co-immunoprecipitation) from other molecules in a cell lysate,[117] in Western blot analyses to identify proteins separated by electrophoresis,[118] and in immunohistochemistry or immunofluorescence to examine protein expression in tissue sections or to locate proteins within cells with the assistance of a microscope.[116][119] Proteins can also be detected and quantified with antibodies, using ELISA and ELISpot techniques.[120][121]

Antibodies used in research are some of the most powerful, yet most problematic reagents with a tremendous number of factors that must be controlled in any experiment including cross reactivity, or the antibody recognizing multiple epitopes and affinity, which can vary widely depending on experimental conditions such as pH, solvent, state of tissue etc. Multiple attempts have been made to improve both the way that researchers validate antibodies[122][123] and ways in which they report on antibodies. Researchers using antibodies in their work need to record them correctly in order to allow their research to be reproducible (and therefore tested, and qualified by other researchers). Less than half of research antibodies referenced in academic papers can be easily identified.[124] Papers published in F1000 in 2014 and 2015 provide researchers with a guide for reporting research antibody use.[125][126] The RRID paper, is co-published in 4 journals that implemented the RRIDs Standard for research resource citation, which draws data from the antibodyregistry.org as the source of antibody identifiers[127] (see also group at Force11[128]).

Antibody regions can be used to further biomedical research by acting as a guide for drugs to reach their target. Several application involve using bacterial plasmids to tag plasmids with the Fc region of the antibody such as pFUSE-Fc plasmid.

Regulations

Production and testing

There are several ways to obtain antibodies, including in vivo techniques like animal immunization and various in vitro approaches, such as the phage display method.[129] Traditionally, most antibodies are produced by hybridoma cell lines through immortalization of antibody-producing cells by chemically induced fusion with myeloma cells. In some cases, additional fusions with other lines have created "triomas" and "quadromas". The manufacturing process should be appropriately described and validated. Validation studies should at least include:

  • The demonstration that the process is able to produce in good quality (the process should be validated)
  • The efficiency of the antibody purification (all impurities and virus must be eliminated)
  • The characterization of purified antibody (physicochemical characterization, immunological properties, biological activities, contaminants, ...)
  • Determination of the virus clearance studies

Before clinical trials

  • Product safety testing: Sterility (bacteria and fungi), in vitro and in vivo testing for adventitious viruses, murine retrovirus testing..., product safety data needed before the initiation of feasibility trials in serious or immediately life-threatening conditions, it serves to evaluate dangerous potential of the product.
  • Feasibility testing: These are pilot studies whose objectives include, among others, early characterization of safety and initial proof of concept in a small specific patient population (in vitro or in vivo testing).

Preclinical studies

  • Testing cross-reactivity of antibody: to highlight unwanted interactions (toxicity) of antibodies with previously characterized tissues. This study can be performed in vitro (reactivity of the antibody or immunoconjugate should be determined with a quick-frozen adult tissues) or in vivo (with appropriates animal models).
  • Preclinical pharmacology and toxicity testing: preclinical safety testing of antibody is designed to identify possible toxicity in humans, to estimate the likelihood and severity of potential adverse events in humans, and to identify a safe starting dose and dose escalation, when possible.
  • Animal toxicity studies: Acute toxicity testing, repeat-dose toxicity testing, long-term toxicity testing
  • Pharmacokinetics and pharmacodynamics testing: Use for determinate clinical dosages, antibody activities, evaluation of the potential clinical effects

Structure prediction and computational antibody design

The importance of antibodies in health care and the biotechnology industry demands knowledge of their structures at high resolution. This information is used for protein engineering, modifying the antigen binding affinity, and identifying an epitope, of a given antibody. X-ray crystallography is one commonly used method for determining antibody structures. However, crystallizing an antibody is often laborious and time-consuming. Computational approaches provide a cheaper and faster alternative to crystallography, but their results are more equivocal, since they do not produce empirical structures. Online web servers such as Web Antibody Modeling (WAM)[130] and Prediction of Immunoglobulin Structure (PIGS)[131] enables computational modeling of antibody variable regions. Rosetta Antibody is a novel antibody FV region structure prediction server, which incorporates sophisticated techniques to minimize CDR loops and optimize the relative orientation of the light and heavy chains, as well as homology models that predict successful docking of antibodies with their unique antigen.[132] However, describing an antibody's binding site using only one single static structure limits the understanding and characterization of the antibody's function and properties. To improve antibody structure prediction and to take the strongly correlated CDR loop and interface movements into account, antibody paratopes should be described as interconverting states in solution with varying probabilities.[27]

The ability to describe the antibody through binding affinity to the antigen is supplemented by information on antibody structure and amino acid sequences for the purpose of patent claims.[133] Several methods have been presented for computational design of antibodies based on the structural bioinformatics studies of antibody CDRs.[134][135][136]

There are a variety of methods used to sequence an antibody including Edman degradation, cDNA, etc.; albeit one of the most common modern uses for peptide/protein identification is liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS).[137] High volume antibody sequencing methods require computational approaches for the data analysis, including de novo sequencing directly from tandem mass spectra[138] and database search methods that use existing protein sequence databases.[139][140] Many versions of shotgun protein sequencing are able to increase the coverage by utilizing CID/HCD/ETD[141] fragmentation methods and other techniques, and they have achieved substantial progress in attempt to fully sequence proteins, especially antibodies. Other methods have assumed the existence of similar proteins,[142] a known genome sequence,[143] or combined top-down and bottom up approaches.[144] Current technologies have the ability to assemble protein sequences with high accuracy by integrating de novo sequencing peptides, intensity, and positional confidence scores from database and homology searches.[145]

Antibody mimetic

Antibody mimetics are organic compounds, like antibodies, that can specifically bind antigens. They consist of artificial peptides or proteins, or aptamer-based nucleic acid molecules with a molar mass of about 3 to 20 kDa. Antibody fragments, such as Fab and nanobodies are not considered as antibody mimetics. Common advantages over antibodies are better solubility, tissue penetration, stability towards heat and enzymes, and comparatively low production costs. Antibody mimetics have being developed and commercialized as research, diagnostic and therapeutic agents.[146]

Binding antibody unit

BAU (binding antibody unit, often as BAU/mL) is a measurement unit defined by the WHO for the comparison of assays detecting the same class of immunoglobulins with the same specificity.[147][148][149]

See also

References

  1. ^ a b Rhoades RA, Pflanzer RG (2002). Human Physiology (5th ed.). Thomson Learning. p. 584. ISBN 978-0-534-42174-8.
  2. ^ Wilson IA, Stanfield RL (3 May 2021). "50 Years of structural immunology". The Journal of Biological Chemistry. 296: 100745. doi:10.1016/j.jbc.2021.100745. ISSN 0021-9258. PMC 8163984. PMID 33957119. Antibodies (A–D) can recognize virtually any antigen whether large or small, and which can have diverse chemical compositions from small molecules (A) to carbohydrates to lipids to peptides (B) to proteins (C and D) and combinations thereof.
  3. ^ a b c d e f g h i Janeway C (2001). Immunobiology (5th ed.). Garland Publishing. ISBN 978-0-8153-3642-6.
  4. ^ Litman GW, Rast JP, Shamblott MJ, Haire RN, Hulst M, Roess W, et al. (January 1993). "Phylogenetic diversification of immunoglobulin genes and the antibody repertoire". Molecular Biology and Evolution. 10 (1): 60–72. doi:10.1093/oxfordjournals.molbev.a040000. PMID 8450761.
  5. ^ Ehrenstein MR, Notley CA (15 October 2010). "The importance of natural IgM: scavenger, protector and regulator". Nature Reviews Immunology. 10 (11): 778–786. doi:10.1038/nri2849. ISSN 1474-1733. PMID 20948548. S2CID 35784099.
  6. ^ Akkaya M, Kwak K, Pierce SK (April 2020). "B cell memory: building two walls of protection against pathogens". Nature Reviews Immunology. 20 (4): 229–238. doi:10.1038/s41577-019-0244-2. ISSN 1474-1741. PMC 7223087. PMID 31836872.
  7. ^ Tellier J, Nutt SL (15 October 2018). "Plasma cells: The programming of an antibody-secreting machine". European Journal of Immunology. 49 (1): 30–37. doi:10.1002/eji.201847517. hdl:11343/284565. ISSN 0014-2980. PMID 30273443.
  8. ^ "B Cell Memory and Plasma Cell Development", Molecular Biology of B Cells, Elsevier, pp. 227–249, 2015, doi:10.1016/b978-0-12-397933-9.00014-x, ISBN 978-0-12-397933-9, retrieved 24 January 2024
  9. ^ Chu VT, Berek C (19 December 2012). "The establishment of the plasma cell survival niche in the bone marrow". Immunological Reviews. 251 (1): 177–188. doi:10.1111/imr.12011. ISSN 0105-2896. PMID 23278749. S2CID 205212187.
  10. ^ Dorner M, Brandt S, Tinguely M, Zucol F, Bourquin J, Zauner L, et al. (6 November 2009). "Plasma cell toll-like receptor (TLR) expression differs from that of B cells, and plasma cell TLR triggering enhances immunoglobulin production". Immunology. 128 (4): 573–579. doi:10.1111/j.1365-2567.2009.03143.x. ISSN 0019-2805. PMC 2792141. PMID 19950420.
  11. ^ Joyner CJ, Ley AM, Nguyen DC, Ali M, Corrado A, Tipton C, et al. (March 2022). "Generation of human long-lived plasma cells by developmentally regulated epigenetic imprinting". Life Science Alliance. 5 (3): e202101285. doi:10.26508/lsa.202101285. ISSN 2575-1077. PMC 8739272. PMID 34952892.
  12. ^ Halliley JL, Tipton CM, Liesveld J, Rosenberg AF, Darce J, Gregoretti IV, et al. (July 2015). "Long-Lived Plasma Cells Are Contained within the CD19−CD38hiCD138+ Subset in Human Bone Marrow". Immunity. 43 (1): 132–145. doi:10.1016/j.immuni.2015.06.016. PMC 4680845. PMID 26187412.
  13. ^ Tellier J, Tarasova I, Nie J, Smillie CS, Fedele PL, Cao WH, et al. (3 January 2024). "Unraveling the diversity and functions of tissue-resident plasma cells". Nature Immunology. 25 (2): 330–342. doi:10.1038/s41590-023-01712-w. ISSN 1529-2908. PMID 38172260. S2CID 266752931.
  14. ^ Landsverk OJ, Snir O, Casado RB, Richter L, Mold JE, Réu P, et al. (February 2017). "Antibody-secreting plasma cells persist for decades in human intestine". The Journal of Experimental Medicine. 214 (2): 309–317. doi:10.1084/jem.20161590. ISSN 1540-9538. PMC 5294861. PMID 28104812.
  15. ^ Plotkin SA (2022). "Recent updates on correlates of vaccine-induced protection". Frontiers in Immunology. 13: 1081107. doi:10.3389/fimmu.2022.1081107. ISSN 1664-3224. PMC 9912984. PMID 36776392.
  16. ^ Sutton HJ, Gao X, Kelly HG, Parker BJ, Lofgren M, Dacon C, et al. (12 January 2024). "Lack of affinity signature for germinal center cells that have initiated plasma cell differentiation". Immunity. 57 (2): S1074–7613(23)00541–1. doi:10.1016/j.immuni.2023.12.010. ISSN 1097-4180. PMC 10922795. PMID 38228150.
  17. ^ Reth M (August 2013). "Matching cellular dimensions with molecular sizes" (PDF). Nature Immunology. 14 (8): 765–7. doi:10.1038/ni.2621. PMID 23867923. S2CID 24333875. Archived from the original (PDF) on 2 May 2018. Retrieved 1 May 2018.
  18. ^ a b Woof JM, Burton DR (February 2004). "Human antibody-Fc receptor interactions illuminated by crystal structures". Nature Reviews. Immunology. 4 (2): 89–99. doi:10.1038/nri1266. PMID 15040582. S2CID 30584218.
  19. ^ Barclay AN (August 2003). "Membrane proteins with immunoglobulin-like domains—a master superfamily of interaction molecules". Seminars in Immunology. 15 (4): 215–23. doi:10.1016/S1044-5323(03)00047-2. PMID 14690046.
  20. ^ Putnam FW, Liu YS, Low TL (April 1979). "Primary structure of a human IgA1 immunoglobulin. IV. Streptococcal IgA1 protease, digestion, Fab and Fc fragments, and the complete amino acid sequence of the alpha 1 heavy chain". The Journal of Biological Chemistry. 254 (8): 2865–74. doi:10.1016/S0021-9258(17)30153-9. PMID 107164.
  21. ^ a b Delves PJ, Martin SJ, Burton DR, Roitt IM (2017). Roitt's essential immunology (13th ed.). Chichester, West Sussex. ISBN 978-1-118-41577-1. OCLC 949912256.{{cite book}}: CS1 maint: location missing publisher (link)
  22. ^ "MeSH Browser – gamma-Globulins". meshb.nlm.nih.gov. Retrieved 18 October 2020.
  23. ^ "Recommendations for the nomenclature of human immunoglobulins". Journal of Immunology. 108 (6): 1733–4. June 1972. doi:10.4049/jimmunol.108.6.1733. PMID 5031329.
  24. ^ Al-Lazikani B, Lesk AM, Chothia C (November 1997). "Standard conformations for the canonical structures of immunoglobulins". Journal of Molecular Biology. 273 (4): 927–48. doi:10.1006/jmbi.1997.1354. PMID 9367782.
  25. ^ North B, Lehmann A, Dunbrack RL (February 2011). "A new clustering of antibody CDR loop conformations". Journal of Molecular Biology. 406 (2): 228–56. doi:10.1016/j.jmb.2010.10.030. PMC 3065967. PMID 21035459.
  26. ^ Nikoloudis D, Pitts JE, Saldanha JW (2014). "A complete, multi-level conformational clustering of antibody complementarity-determining regions". PeerJ. 2 (e456): e456. doi:10.7717/peerj.456. PMC 4103072. PMID 25071986.
  27. ^ a b Fernández-Quintero ML, Georges G, Varga JM, Liedl KR (2021). "Ensembles in solution as a new paradigm for antibody structure prediction and design". mAbs. 13 (1): 1923122. doi:10.1080/19420862.2021.1923122. PMC 8158028. PMID 34030577.
  28. ^ Woof JM, Russell RW (2011). "Structure and function relationships in IgA". Mucosal Immunology. 4 (6): 590–597. doi:10.1038/mi.2011.39. PMID 21937984.
  29. ^ Damelang T, Rogerson SJ, Kent SJ, Chung AW (March 2019). "Role of IgG3 in Infectious Diseases". Trends in Immunology. 40 (3): 197–211. doi:10.1016/j.it.2019.01.005. hdl:11343/284299. ISSN 1471-4906. PMID 30745265. S2CID 73419807.
  30. ^ a b c Maverakis E, Kim K, Shimoda M, Gershwin ME, Patel F, Wilken R, et al. (February 2015). "Glycans in the immune system and The Altered Glycan Theory of Autoimmunity: a critical review". Journal of Autoimmunity. 57 (6): 1–13. doi:10.1016/j.jaut.2014.12.002. PMC 4340844. PMID 25578468.
  31. ^ Mattu TS, Pleass RJ, Willis AC, Kilian M, Wormald MR, Lellouch AC, et al. (January 1998). "The glycosylation and structure of human serum IgA1, Fab, and Fc regions and the role of N-glycosylation on Fcα receptor interactions". The Journal of Biological Chemistry. 273 (4): 2260–72. doi:10.1074/jbc.273.4.2260. PMID 9442070.
  32. ^ Cobb BA (March 2020). "The history of IgG glycosylation and where we are now". Glycobiology. 30 (4): 202–213. doi:10.1093/glycob/cwz065. PMC 7109348. PMID 31504525.
  33. ^ Roux KH (October 1999). "Immunoglobulin structure and function as revealed by electron microscopy". International Archives of Allergy and Immunology. 120 (2): 85–99. doi:10.1159/000024226. PMID 10545762. S2CID 12187510.
  34. ^ Diebolder CA, Beurskens FJ, de Jong RN, Koning RI, Strumane K, Lindorfer MA, et al. (14 March 2014). "Complement Is Activated by IgG Hexamers Assembled at the Cell Surface". Science. 343 (6176): 1260–1263. Bibcode:2014Sci...343.1260D. doi:10.1126/science.1248943. ISSN 0036-8075. PMC 4250092. PMID 24626930.
  35. ^ Kumar N, Arthur CP, Ciferri C, Matsumoto ML (28 February 2020). "Structure of the secretory immunoglobulin A core". Science. 367 (6481): 1008–1014. Bibcode:2020Sci...367.1008K. doi:10.1126/science.aaz5807. ISSN 0036-8075. PMID 32029686.
  36. ^ Parker DC (1993). "T cell-dependent B cell activation". Annual Review of Immunology. 11 (1): 331–60. doi:10.1146/annurev.iy.11.040193.001555. PMID 8476565.
  37. ^ a b c d Wintrobe MM (2004). Greer JG, Foerster F, Lukens JN, Rodgers GM, Paraskevas F (eds.). Wintrobe's clinical hematology (11 ed.). Hagerstown, MD: Lippincott Williams & Wilkins. pp. 453–456. ISBN 978-0-7817-3650-3.
  38. ^ Tolar P, Sohn HW, Pierce SK (February 2008). "Viewing the antigen-induced initiation of B-cell activation in living cells". Immunological Reviews. 221 (1): 64–76. doi:10.1111/j.1600-065X.2008.00583.x. PMID 18275475. S2CID 38464264.
  39. ^ Williams CM, Galli SJ (May 2000). "The diverse potential effector and immunoregulatory roles of mast cells in allergic disease". The Journal of Allergy and Clinical Immunology. 105 (5): 847–59. doi:10.1067/mai.2000.106485. PMID 10808163.
  40. ^ Underdown BJ, Schiff JM (1986). "Immunoglobulin A: strategic defense initiative at the mucosal surface". Annual Review of Immunology. 4 (1): 389–417. doi:10.1146/annurev.iy.04.040186.002133. PMID 3518747.
  41. ^ a b Geisberger R, Lamers M, Achatz G (August 2006). "The riddle of the dual expression of IgM and IgD". Immunology. 118 (4): 429–37. doi:10.1111/j.1365-2567.2006.02386.x. PMC 1782314. PMID 16895553.
  42. ^ Chen K, Xu W, Wilson M, He B, Miller NW, Bengtén E, et al. (August 2009). "Immunoglobulin D enhances immune surveillance by activating antimicrobial, proinflammatory and B cell-stimulating programs in basophils". Nature Immunology. 10 (8): 889–98. doi:10.1038/ni.1748. PMC 2785232. PMID 19561614.
  43. ^ a b c d e Pier GB, Lyczak JB, Wetzler LM (2004). Immunology, Infection, and Immunity. ASM Press. ISBN 978-1-55581-246-1.
  44. ^ Goding JW (1978). "Allotypes of IgM and IgD Receptors in the Mouse: A Probe for Lymphocyte Differentiation". Contemporary Topics in Immunobiology. Vol. 8. pp. 203–43. doi:10.1007/978-1-4684-0922-2_7. ISBN 978-1-4684-0924-6. PMID 357078.
  45. ^ Litman GW, Rast JP, Fugmann SD (August 2010). "The origins of vertebrate adaptive immunity". Nature Reviews. Immunology. 10 (8): 543–53. doi:10.1038/nri2807. PMC 2919748. PMID 20651744.
  46. ^ Litman GW, Rast JP, Fugmann SD (August 2010). "The origins of vertebrate adaptive immunity". Nature Reviews. Immunology. John Wiley & Sons, Ltd. 10 (8): 543–53. doi:10.1002/9783527699124.ch4. ISBN 978-3-527-69912-4. PMC 2919748. PMID 20651744.
  47. ^ Lundqvist ML, Middleton DL, Radford C, Warr GW, Magor KE (2006). "Immunoglobulins of the non-galliform birds: antibody expression and repertoire in the duck". Developmental and Comparative Immunology. 30 (1–2): 93–100. doi:10.1016/j.dci.2005.06.019. PMC 1317265. PMID 16150486.
  48. ^ Berstein RM, Schluter SF, Shen S, Marchalonis JJ (April 1996). "A new high molecular weight immunoglobulin class from the carcharhine shark: implications for the properties of the primordial immunoglobulin". Proceedings of the National Academy of Sciences of the United States of America. 93 (8): 3289–93. Bibcode:1996PNAS...93.3289B. doi:10.1073/pnas.93.8.3289. PMC 39599. PMID 8622930.
  49. ^ Salinas, I., & Parra, D. (2015). Fish mucosal immunity: Intestine. In Mucosal Health in Aquaculture. Elsevier Inc. https://doi.org/10.1016/B978-0-12-417186-2.00006-6
  50. ^ Borghesi L, Milcarek C (2006). "From B cell to plasma cell: regulation of V(D)J recombination and antibody secretion". Immunologic Research. 36 (1–3): 27–32. doi:10.1385/IR:36:1:27. PMID 17337763. S2CID 27041937.
  51. ^ a b Ravetch JV, Bolland S (2001). "IgG Fc receptors". Annual Review of Immunology. 19 (1): 275–90. doi:10.1146/annurev.immunol.19.1.275. PMID 11244038.
  52. ^ Rus H, Cudrici C, Niculescu F (2005). "The role of the complement system in innate immunity". Immunologic Research. 33 (2): 103–12. doi:10.1385/IR:33:2:103. PMID 16234578. S2CID 46096567.
  53. ^ Racaniello, Vincent (6 October 2009). "Natural antibody protects against viral infection". Virology Blog. Archived from the original on 20 February 2010. Retrieved 22 January 2010.
  54. ^ Milland J, Sandrin MS (December 2006). "ABO blood group and related antigens, natural antibodies and transplantation". Tissue Antigens. 68 (6): 459–66. doi:10.1111/j.1399-0039.2006.00721.x. PMID 17176435.
  55. ^ Mian IS, Bradwell AR, Olson AJ (January 1991). "Structure, function and properties of antibody binding sites". Journal of Molecular Biology. 217 (1): 133–51. doi:10.1016/0022-2836(91)90617-F. PMID 1988675.
  56. ^ Fanning LJ, Connor AM, Wu GE (April 1996). "Development of the immunoglobulin repertoire". Clinical Immunology and Immunopathology. 79 (1): 1–14. doi:10.1006/clin.1996.0044. PMID 8612345.
  57. ^ a b Nemazee D (October 2006). "Receptor editing in lymphocyte development and central tolerance". Nature Reviews. Immunology. 6 (10): 728–40. doi:10.1038/nri1939. PMID 16998507. S2CID 2234228.
  58. ^ Peter Parham. The Immune System. 2nd ed. Garland Science: New York, 2005. pg.47–62
  59. ^ a b c d Market E, Papavasiliou FN (October 2003). "V(D)J recombination and the evolution of the adaptive immune system". PLOS Biology. 1 (1): E16. doi:10.1371/journal.pbio.0000016. PMC 212695. PMID 14551913.
  60. ^ Bergman Y, Cedar H (October 2004). "A stepwise epigenetic process controls immunoglobulin allelic exclusion". Nature Reviews. Immunology. 4 (10): 753–61. doi:10.1038/nri1458. PMID 15459667. S2CID 8579156.
  61. ^ Diaz M, Casali P (April 2002). "Somatic immunoglobulin hypermutation". Current Opinion in Immunology. 14 (2): 235–40. doi:10.1016/S0952-7915(02)00327-8. PMC 4621002. PMID 11869898.
  62. ^ Honjo T, Habu S (1985). "Origin of immune diversity: genetic variation and selection". Annual Review of Biochemistry. 54 (1): 803–30. doi:10.1146/annurev.bi.54.070185.004103. PMID 3927822.
  63. ^ a b Or-Guil M, Wittenbrink N, Weiser AA, Schuchhardt J (April 2007). "Recirculation of germinal center B cells: a multilevel selection strategy for antibody maturation". Immunological Reviews. 216: 130–41. doi:10.1111/j.1600-065X.2007.00507.x. PMID 17367339. S2CID 37636392.
  64. ^ Neuberger MS, Ehrenstein MR, Rada C, Sale J, Batista FD, Williams G, et al. (March 2000). "Memory in the B-cell compartment: antibody affinity maturation". Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 355 (1395): 357–60. doi:10.1098/rstb.2000.0573. PMC 1692737. PMID 10794054.
  65. ^ Stavnezer J, Amemiya CT (August 2004). "Evolution of isotype switching". Seminars in Immunology. 16 (4): 257–75. doi:10.1016/j.smim.2004.08.005. PMID 15522624.
  66. ^ Durandy A (August 2003). "Activation-induced cytidine deaminase: a dual role in class-switch recombination and somatic hypermutation". European Journal of Immunology. 33 (8): 2069–73. doi:10.1002/eji.200324133. PMID 12884279. S2CID 32059768.
  67. ^ Casali P, Zan H (November 2004). "Class switching and Myc translocation: how does DNA break?". Nature Immunology. 5 (11): 1101–3. doi:10.1038/ni1104-1101. PMC 4625794. PMID 15496946.
  68. ^ Lieber MR, Yu K, Raghavan SC (September 2006). "Roles of nonhomologous DNA end joining, V(D)J recombination, and class switch recombination in chromosomal translocations". DNA Repair. 5 (9–10): 1234–45. doi:10.1016/j.dnarep.2006.05.013. PMID 16793349.
  69. ^ p. 22 in: Shoenfeld Y, Meroni PL, Gershwin ME (2007). Autoantibodie. Amsterdam; Boston: Elsevier. ISBN 978-0-444-52763-9.
  70. ^ Spiess C, Zhai Q, Carter PJ (October 2015). "Alternative molecular formats and therapeutic applications for bispecific antibodies". Molecular Immunology. 67 (2 Pt A): 95–106. doi:10.1016/j.molimm.2015.01.003. PMID 25637431.
  71. ^ a b Farlex dictionary > polyvalent Citing: The American Heritage Medical Dictionary. 2004
  72. ^ Gunasekaran K, Pentony M, Shen M, Garrett L, Forte C, Woodward A, et al. (June 2010). "Enhancing antibody Fc heterodimer formation through electrostatic steering effects: applications to bispecific molecules and monovalent IgG". The Journal of Biological Chemistry. 285 (25): 19637–46. doi:10.1074/jbc.M110.117382. PMC 2885242. PMID 20400508.
  73. ^ Muller KM (1998). "The first constant domain (CH1 and CL) of an antibody used as heterodimerization domain for bispecific miniantibodies". FEBS Letters. 422 (2): 259–264. doi:10.1016/s0014-5793(98)00021-0. PMID 9490020. S2CID 35243494.
  74. ^ Gao C, Mao S, Lo CH, Wirsching P, Lerner RA, Janda KD (May 1999). "Making artificial antibodies: a format for phage display of combinatorial heterodimeric arrays". Proceedings of the National Academy of Sciences of the United States of America. 96 (11): 6025–30. Bibcode:1999PNAS...96.6025G. doi:10.1073/pnas.96.11.6025. PMC 26829. PMID 10339535.
  75. ^ Kanyavuz A, Marey-Jarossay A, Lacroix-Desmazes S, Dimitrov JD (June 2019). "Breaking the law: unconventional strategies for antibody diversification". Nature Reviews Immunology. 19 (6): 355–368. doi:10.1038/s41577-019-0126-7. ISSN 1474-1741. PMID 30718829. S2CID 59603663.
  76. ^ Pieper K, Tan J, Piccoli L, Foglierini M, Barbieri S, Chen Y, et al. (August 2017). "Public antibodies to malaria antigens generated by two LAIR1 insertion modalities". Nature. 548 (7669): 597–601. Bibcode:2017Natur.548..597P. doi:10.1038/nature23670. ISSN 0028-0836. PMC 5635981. PMID 28847005.
  77. ^ Robbiani DF, Deroubaix S, Feldhahn N, Oliveira TY, Callen E, Wang Q, et al. (August 2015). "Plasmodium Infection Promotes Genomic Instability and AID-Dependent B Cell Lymphoma". Cell. 162 (4): 727–737. doi:10.1016/j.cell.2015.07.019. PMC 4538708. PMID 26276629.
  78. ^ a b c d Lindenmann J (April 1984). "Origin of the terms 'antibody' and 'antigen'". Scandinavian Journal of Immunology. 19 (4): 281–5. doi:10.1111/j.1365-3083.1984.tb00931.x. PMID 6374880. S2CID 222200504.
  79. ^ Padlan EA (February 1994). "Anatomy of the antibody molecule". Molecular Immunology. 31 (3): 169–217. doi:10.1016/0161-5890(94)90001-9. PMID 8114766.
  80. ^ Sauter E (10 November 2018). "New Sculpture Portraying Human Antibody as Protective Angel Installed on Scripps Florida Campus". News & Views. Vol. 8, no. 34. The Scripps Research Institute. Archived from the original on 10 January 2011. Retrieved 12 December 2008.
  81. ^ Pescovitz D (22 October 2008). "Protein sculpture inspired by Vitruvian Man". boingboing (Blog). Archived from the original on 4 November 2010. Retrieved 12 December 2008.
  82. ^ Emil von Behring – Biographical. NobelPrize.org. Nobel Media AB 2020. Mon. 20 January 2020. <https://www.nobelprize.org/prizes/medicine/1901/behring/biographical/>
  83. ^ AGN (August 1931). "The Late Baron Shibasaburo Kitasato". Canadian Medical Association Journal. 25 (2): 206. PMC 382621. PMID 20318414.
  84. ^ Winau F, Westphal O, Winau R (July 2004). "Paul Ehrlich—in search of the magic bullet". Microbes and Infection. 6 (8): 786–9. doi:10.1016/j.micinf.2004.04.003. PMID 15207826.
  85. ^ Silverstein AM (May 2003). "Cellular versus humoral immunology: a century-long dispute". Nature Immunology. 4 (5): 425–8. doi:10.1038/ni0503-425. PMID 12719732. S2CID 31571243.
  86. ^ Van Epps HL (January 2006). "Michael Heidelberger and the demystification of antibodies". The Journal of Experimental Medicine. 203 (1): 5. doi:10.1084/jem.2031fta. PMC 2118068. PMID 16523537.
  87. ^ Marrack JR (1938). Chemistry of antigens and antibodies (2nd ed.). London: His Majesty's Stationery Office. OCLC 3220539.
  88. ^ "The Linus Pauling Papers: How Antibodies and Enzymes Work". Archived from the original on 5 December 2010. Retrieved 5 June 2007.
  89. ^ Silverstein AM (December 2004). "Labeled antigens and antibodies: the evolution of magic markers and magic bullets" (PDF). Nature Immunology. 5 (12): 1211–7. doi:10.1038/ni1140. PMID 15549122. S2CID 40595920. Archived from the original (PDF) on 25 March 2009.
  90. ^ Edelman GM, Gally JA (August 1962). "The nature of Bence-Jones proteins. Chemical similarities to polypetide chains of myeloma globulins and normal gamma-globulins". The Journal of Experimental Medicine. 116 (2): 207–27. doi:10.1084/jem.116.2.207. PMC 2137388. PMID 13889153.
  91. ^ Stevens FJ, Solomon A, Schiffer M (July 1991). "Bence Jones proteins: a powerful tool for the fundamental study of protein chemistry and pathophysiology". Biochemistry. 30 (28): 6803–5. doi:10.1021/bi00242a001. PMID 2069946.
  92. ^ a b Raju TN (September 1999). "The Nobel chronicles. 1972: Gerald M Edelman (b 1929) and Rodney R Porter (1917–85)". Lancet. 354 (9183): 1040. doi:10.1016/S0140-6736(05)76658-7. PMID 10501404. S2CID 54380536.
  93. ^ Hochman J, Inbar D, Givol D (March 1973). "An active antibody fragment (Fv) composed of the variable portions of heavy and light chains". Biochemistry. 12 (6): 1130–5. doi:10.1021/bi00730a018. PMID 4569769.
  94. ^ Tomasi TB (October 1992). "The discovery of secretory IgA and the mucosal immune system". Immunology Today. 13 (10): 416–8. doi:10.1016/0167-5699(92)90093-M. PMID 1343085.
  95. ^ Preud'homme JL, Petit I, Barra A, Morel F, Lecron JC, Lelièvre E (October 2000). "Structural and functional properties of membrane and secreted IgD". Molecular Immunology. 37 (15): 871–87. doi:10.1016/S0161-5890(01)00006-2. PMID 11282392.
  96. ^ Johansson SG (2006). "The discovery of immunoglobulin E". Allergy and Asthma Proceedings. 27 (2 Suppl 1): S3–6. PMID 16722325.
  97. ^ Hozumi N, Tonegawa S (October 1976). "Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions". Proceedings of the National Academy of Sciences of the United States of America. 73 (10): 3628–32. Bibcode:1976PNAS...73.3628H. doi:10.1073/pnas.73.10.3628. PMC 431171. PMID 824647.
  98. ^ "Animated depictions of how antibodies are used in ELISA assays". Cellular Technology Ltd.—Europe. Archived from the original on 14 June 2011. Retrieved 8 May 2007.
  99. ^ "Animated depictions of how antibodies are used in ELISPOT assays". Cellular Technology Ltd.—Europe. Archived from the original on 16 May 2011. Retrieved 8 May 2007.
  100. ^ Stern P (2006). "Current possibilities of turbidimetry and nephelometry" (PDF). Klin Biochem Metab. 14 (3): 146–151. Archived from the original (PDF) on 10 April 2008.
  101. ^ a b Dean L (2005). "Chapter 4: Hemolytic disease of the newborn". Blood Groups and Red Cell Antigens. NCBI Bethesda (MD): National Library of Medicine (US).
  102. ^ Sullivan MV, Stockburn WJ, Hawes PC, Mercer T, Reddy SM (26 February 2021). "Green synthesis as a simple and rapid route to protein modified magnetic nanoparticles for use in the development of a fluorometric molecularly imprinted polymer-based assay for detection of myoglobin". Nanotechnology. 32 (9): 095502. Bibcode:2021Nanot..32i5502S. doi:10.1088/1361-6528/abce2d. PMC 8314874. PMID 33242844.
  103. ^ Rodriguez EA, Wang Y, Crisp JL, Vera DR, Tsien RY, Ting R (May 2016). "New Dioxaborolane Chemistry Enables [(18)F]-Positron-Emitting, Fluorescent [(18)F]-Multimodality Biomolecule Generation from the Solid Phase". Bioconjugate Chemistry. 27 (5): 1390–1399. doi:10.1021/acs.bioconjchem.6b00164. PMC 4916912. PMID 27064381.
  104. ^ Feldmann M, Maini RN (2001). "Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned?". Annual Review of Immunology. 19 (1): 163–96. doi:10.1146/annurev.immunol.19.1.163. PMID 11244034.
  105. ^ Doggrell SA (June 2003). "Is natalizumab a breakthrough in the treatment of multiple sclerosis?". Expert Opinion on Pharmacotherapy. 4 (6): 999–1001. doi:10.1517/14656566.4.6.999. PMID 12783595. S2CID 16104816.
  106. ^ Krueger GG, Langley RG, Leonardi C, Yeilding N, Guzzo C, Wang Y, et al. (February 2007). "A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis". The New England Journal of Medicine. 356 (6): 580–92. doi:10.1056/NEJMoa062382. PMID 17287478.
  107. ^ Plosker GL, Figgitt DP (2003). "Rituximab: a review of its use in non-Hodgkin's lymphoma and chronic lymphocytic leukaemia". Drugs. 63 (8): 803–43. doi:10.2165/00003495-200363080-00005. PMID 12662126.
  108. ^ Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, et al. (2001). "First-line Herceptin monotherapy in metastatic breast cancer". Oncology. 61. 61 (Suppl. 2): 37–42. doi:10.1159/000055400. PMID 11694786. S2CID 24924864.
  109. ^ LeBien TW (July 2000). "Fates of human B-cell precursors". Blood. 96 (1): 9–23. doi:10.1182/blood.V96.1.9. PMID 10891425. Archived from the original on 29 April 2010. Retrieved 31 March 2007.
  110. ^ Ghaffer A (26 March 2006). "Immunization". Immunology — Chapter 14. University of South Carolina School of Medicine. Archived from the original on 18 October 2010. Retrieved 6 June 2007.
  111. ^ Urbaniak SJ, Greiss MA (March 2000). "RhD haemolytic disease of the fetus and the newborn". Blood Reviews. 14 (1): 44–61. doi:10.1054/blre.1999.0123. PMID 10805260.
  112. ^ a b Fung Kee Fung K, Eason E, Crane J, Armson A, De La Ronde S, Farine D, et al. (September 2003). "Prevention of Rh alloimmunization". Journal of Obstetrics and Gynaecology Canada. 25 (9): 765–73. doi:10.1016/S1701-2163(16)31006-4. PMID 12970812.
  113. ^ Tini M, Jewell UR, Camenisch G, Chilov D, Gassmann M (March 2002). "Generation and application of chicken egg-yolk antibodies". Comparative Biochemistry and Physiology. Part A, Molecular & Integrative Physiology. 131 (3): 569–74. doi:10.1016/S1095-6433(01)00508-6. PMID 11867282.
  114. ^ Cole SP, Campling BG, Atlaw T, Kozbor D, Roder JC (June 1984). "Human monoclonal antibodies". Molecular and Cellular Biochemistry. 62 (2): 109–20. doi:10.1007/BF00223301. PMID 6087121. S2CID 12616168.
  115. ^ Kabir S (2002). "Immunoglobulin purification by affinity chromatography using protein A mimetic ligands prepared by combinatorial chemical synthesis". Immunological Investigations. 31 (3–4): 263–78. doi:10.1081/IMM-120016245. PMID 12472184. S2CID 12785078.
  116. ^ a b Brehm-Stecher BF, Johnson EA (September 2004). "Single-cell microbiology: tools, technologies, and applications". Microbiology and Molecular Biology Reviews. 68 (3): 538–59, table of contents. doi:10.1128/MMBR.68.3.538-559.2004. PMC 515252. PMID 15353569.
  117. ^ Williams NE (2000). "Chapter 23 Immunoprecipitation Procedures". Methods in Cell Biology Volume 62. Vol. 62. San Diego, CA : Academic Press. pp. 449–53. doi:10.1016/S0091-679X(08)61549-6. ISBN 978-0-12-544164-3. PMID 10503210.
  118. ^ Kurien BT, Scofield RH (April 2006). "Western blotting". Methods. 38 (4): 283–93. doi:10.1016/j.ymeth.2005.11.007. PMID 16483794.
  119. ^ Scanziani E (1998). "Immunohistochemical staining of fixed tissues". Mycoplasma Protocols. Methods in Molecular Biology. Vol. 104. Totowa, N.J. : Humana Press. pp. 133–40. doi:10.1385/0-89603-525-5:133. ISBN 978-0-89603-525-6. PMID 9711649.
  120. ^ Reen DJ (1994). "Enzyme-linked immunosorbent assay (ELISA)". Basic Protein and Peptide Protocols. Methods in Molecular Biology. Vol. 32. pp. 461–6. doi:10.1385/0-89603-268-X:461. ISBN 978-0-89603-268-2. PMC 2366430. PMID 7951745.
  121. ^ Kalyuzhny AE (2005). "Chemistry and biology of the ELISPOT assay". Handbook of ELISPOT. Methods in Molecular Biology. Vol. 302. pp. 15–31. doi:10.1385/1-59259-903-6:015. ISBN 978-1-59259-903-5. PMID 15937343.
  122. ^ Saper CB (December 2005). "An open letter to our readers on the use of antibodies". The Journal of Comparative Neurology. 493 (4): 477–8. doi:10.1002/cne.20839. PMID 16304632. S2CID 14082678.
  123. ^ "NOT-OD-16-011: Implementing Rigor and Transparency in NIH & AHRQ Research Grant Applications". grants.nih.gov.
  124. ^ Vasilevsky NA, Brush MH, Paddock H, Ponting L, Tripathy SJ, Larocca GM, et al. (2 September 2013). "On the reproducibility of science: unique identification of research resources in the biomedical literature". PeerJ. 1: e148. doi:10.7717/peerj.148. PMC 3771067. PMID 24032093.
  125. ^ Bandrowski A, Brush M, Grethe JS, Haendel MA, Kennedy DN, Hill S, et al. (2015). "The Resource Identification Initiative: A cultural shift in publishing". F1000Research. 4: 134. doi:10.12688/f1000research.6555.2. PMC 4648211. PMID 26594330.
  126. ^ Helsby MA, Fenn JR, Chalmers AD (23 August 2013). "Reporting research antibody use: how to increase experimental reproducibility". F1000Research. 2: 153. doi:10.12688/f1000research.2-153.v2. PMC 3829129. PMID 24358895.
  127. ^ "The Antibody Registry". antibodyregistry.org.
  128. ^ "Resource Identification Initiative". FORCE11. 14 August 2013. Retrieved 18 April 2016.
  129. ^ Eberle C (20 February 2023). "Antibody Production simply explained". Retrieved 7 December 2023.
  130. ^ Archived 17 July 2011 at the Wayback Machine
    WAM
  131. ^ Marcatili P, Rosi A, Tramontano A (September 2008). "PIGS: automatic prediction of antibody structures". Bioinformatics. 24 (17): 1953–4. doi:10.1093/bioinformatics/btn341. PMID 18641403. Archived from the original on 26 November 2010.
    Prediction of Immunoglobulin Structure (PIGS) Archived 26 November 2010 at the Wayback Machine
  132. ^ Archived 19 July 2011 at the Wayback Machine
    RosettaAntibody
  133. ^ Park H. "Written Description Problems of the Monoclonal Antibody Patents after Centocor v. Abbott". jolt.law.harvard.edu. Archived from the original on 13 December 2014. Retrieved 12 December 2014.
  134. ^ Adolf-Bryfogle J, Kalyuzhniy O, Kubitz M, Weitzner BD, Hu X, Adachi Y, et al. (April 2018). "RosettaAntibodyDesign (RAbD): A general framework for computational antibody design". PLOS Computational Biology. 14 (4): e1006112. Bibcode:2018PLSCB..14E6112A. doi:10.1371/journal.pcbi.1006112. PMC 5942852. PMID 29702641.
  135. ^ Lapidoth GD, Baran D, Pszolla GM, Norn C, Alon A, Tyka MD, et al. (August 2015). "AbDesign: An algorithm for combinatorial backbone design guided by natural conformations and sequences". Proteins. 83 (8): 1385–406. doi:10.1002/prot.24779. PMC 4881815. PMID 25670500.
  136. ^ Li T, Pantazes RJ, Maranas CD (2014). "OptMAVEn--a new framework for the de novo design of antibody variable region models targeting specific antigen epitopes". PLOS ONE. 9 (8): e105954. Bibcode:2014PLoSO...9j5954L. doi:10.1371/journal.pone.0105954. PMC 4143332. PMID 25153121.
  137. ^ Pham V, Henzel WJ, Arnott D, Hymowitz S, Sandoval WN, Truong BT, et al. (May 2006). "De novo proteomic sequencing of a monoclonal antibody raised against OX40 ligand". Analytical Biochemistry. 352 (1): 77–86. doi:10.1016/j.ab.2006.02.001. PMID 16545334.
  138. ^ Ma B, Zhang K, Hendrie C, Liang C, Li M, Doherty-Kirby A, et al. (2003). "PEAKS: powerful software for peptide de novo sequencing by tandem mass spectrometry". Rapid Communications in Mass Spectrometry. 17 (20): 2337–42. Bibcode:2003RCMS...17.2337M. doi:10.1002/rcm.1196. PMID 14558135.
  139. ^ Zhang J, Xin L, Shan B, Chen W, Xie M, Yuen D, et al. (April 2012). "PEAKS DB: de novo sequencing assisted database search for sensitive and accurate peptide identification". Molecular & Cellular Proteomics. 11 (4): M111.010587. doi:10.1074/mcp.M111.010587. PMC 3322562. PMID 22186715.
  140. ^ Perkins DN, Pappin DJ, Creasy DM, Cottrell JS (December 1999). "Probability-based protein identification by searching sequence databases using mass spectrometry data". Electrophoresis. 20 (18): 3551–67. doi:10.1002/(SICI)1522-2683(19991201)20:18<3551::AID-ELPS3551>3.0.CO;2-2. PMID 10612281. S2CID 42423655.
  141. ^ Bandeira N, Tang H, Bafna V, Pevzner P (December 2004). "Shotgun protein sequencing by tandem mass spectra assembly". Analytical Chemistry. 76 (24): 7221–33. doi:10.1021/ac0489162. PMID 15595863.
  142. ^ Liu X, Han Y, Yuen D, Ma B (September 2009). "Automated protein (re)sequencing with MS/MS and a homologous database yields almost full coverage and accuracy". Bioinformatics. 25 (17): 2174–80. doi:10.1093/bioinformatics/btp366. PMID 19535534.
  143. ^ Castellana NE, Pham V, Arnott D, Lill JR, Bafna V (June 2010). "Template proteogenomics: sequencing whole proteins using an imperfect database". Molecular & Cellular Proteomics. 9 (6): 1260–70. doi:10.1074/mcp.M900504-MCP200. PMC 2877985. PMID 20164058.
  144. ^ Liu X, Dekker LJ, Wu S, Vanduijn MM, Luider TM, Tolić N, et al. (July 2014). "De novo protein sequencing by combining top-down and bottom-up tandem mass spectra". Journal of Proteome Research. 13 (7): 3241–8. doi:10.1021/pr401300m. PMID 24874765.
  145. ^ Tran NH, Rahman MZ, He L, Xin L, Shan B, Li M (August 2016). "Complete De Novo Assembly of Monoclonal Antibody Sequences". Scientific Reports. 6: 31730. Bibcode:2016NatSR...631730T. doi:10.1038/srep31730. PMC 4999880. PMID 27562653.
  146. ^ Gebauer M, Skerra A (June 2009). "Engineered protein scaffolds as next-generation antibody therapeutics". Current Opinion in Chemical Biology. 13 (3): 245–55. doi:10.1016/j.cbpa.2009.04.627. PMID 19501012.
  147. ^ Kristiansen PA, Page M, Bernasconi V, Mattiuzzo G, Dull P, Makar K, et al. (10 April 2021) [2021-02-23]. "WHO International Standard for anti-SARS-CoV-2 immunoglobulin". The Lancet. World Health Organization / Elsevier. 397 (10282): 1347–1348. doi:10.1016/S0140-6736(21)00527-4. PMC 7987302. PMID 33770519.
  148. ^ Knezevic I, Mattiuzzo G, Page M, Minor P, Griffiths E, Nuebling M, et al. (26 October 2021). "WHO International Standard for evaluation of the antibody response to COVID-19 vaccines: call for urgent action by the scientific community". The Lancet Microbe. World Health Organization / Elsevier. 3 (3): e235–e240. doi:10.1016/S2666-5247(21)00266-4. PMC 8547804. PMID 34723229.
  149. ^ Knezevic I (10 November 2021). "Training webinar for the calibration of quantitative serology assays using the WHO International Standard for anti-SARS-CoV-2 immunoglobulin" (PDF). Archived (PDF) from the original on 18 February 2022. Retrieved 5 March 2022. (68 pages)

External links