Antibody

Source: Wikipedia, the free encyclopedia.
(Redirected from
Fv region
)

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

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:

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

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.
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

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

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)
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

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]
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

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
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.

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,

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
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

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

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]

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

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

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

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).

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

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

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,

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

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,

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.

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]

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

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

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

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

primary biliary cirrhosis, while IgG is elevated in viral hepatitis, autoimmune hepatitis
and cirrhosis.

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

Some immune deficiencies, such as

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

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

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

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:

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
    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] enable 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

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

WHO for the comparison of assays detecting the same class of immunoglobulins with the same specificity.[147][148][149]

See also

References

  1. ^ .
  2. . 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. ^ .
  4. .
  5. .
  6. .
  7. .
  8. , retrieved 24 January 2024
  9. .
  10. .
  11. .
  12. .
  13. .
  14. .
  15. .
  16. .
  17. S2CID 24333875. Archived from the original
    (PDF) on 2 May 2018. Retrieved 1 May 2018.
  18. ^ .
  19. .
  20. .
  21. ^
    OCLC 949912256.{{cite book}}: CS1 maint: location missing publisher (link
    )
  22. ^ "MeSH Browser – gamma-Globulins". meshb.nlm.nih.gov. Retrieved 18 October 2020.
  23. 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.
    PMID 9367782
    .
  25. .
  26. .
  27. ^ .
  28. .
  29. .
  30. ^ .
  31. .
  32. .
  33. .
  34. .
  35. .
  36. .
  37. ^ .
  38. .
  39. .
  40. .
  41. ^ .
  42. .
  43. ^ .
  44. .
  45. .
  46. .
  47. .
  48. .
  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. S2CID 27041937
    .
  51. ^ .
  52. .
  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. PMID 17176435
    .
  55. .
  56. .
  57. ^ .
  58. ^ Peter Parham. The Immune System. 2nd ed. Garland Science: New York, 2005. pg.47–62
  59. ^
    PMID 14551913
    .
  60. .
  61. .
  62. .
  63. ^ .
  64. .
  65. .
  66. .
  67. .
  68. .
  69. .
  70. .
  71. ^ a b Farlex dictionary > polyvalent Citing: The American Heritage Medical Dictionary. 2004
  72. PMID 20400508
    .
  73. .
  74. .
  75. .
  76. .
  77. .
  78. ^ .
  79. .
  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. PMID 20318414
    .
  84. .
  85. .
  86. .
  87. .
  88. ^ "The Linus Pauling Papers: How Antibodies and Enzymes Work". Archived from the original on 5 December 2010. Retrieved 5 June 2007.
  89. S2CID 40595920. Archived from the original
    (PDF) on 25 March 2009.
  90. .
  91. .
  92. ^ .
  93. .
  94. .
  95. .
  96. .
  97. .
  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. PMID 33242844
    .
  103. .
  104. .
  105. .
  106. .
  107. .
  108. .
  109. 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. PMID 10805260
    .
  112. ^ .
  113. .
  114. .
  115. .
  116. ^ .
  117. .
  118. .
  119. .
  120. .
  121. .
  122. .
  123. ^ "NOT-OD-16-011: Implementing Rigor and Transparency in NIH & AHRQ Research Grant Applications". grants.nih.gov.
  124. PMID 24032093
    .
  125. .
  126. .
  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. 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. PMID 29702641
    .
  135. .
  136. .
  137. .
  138. .
  139. .
  140. .
  141. .
  142. .
  143. .
  144. .
  145. .
  146. .
  147. .
  148. .
  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