Fact-checked by Grok 2 weeks ago

Adaptive immune system

The adaptive immune system is a subsystem of the that mounts targeted defenses against specific pathogens and foreign substances known as antigens, providing long-lasting protection through its capacity for immunological memory. Unlike the , which delivers rapid but non-specific responses to a broad range of threats, the adaptive immune system develops over several days upon first exposure and exhibits high specificity, diversity, and the ability to "remember" previous encounters for quicker subsequent reactions. This antigen-dependent mechanism is essential for eliminating infections that evade initial innate defenses and forms the foundation of strategies. Key components of the adaptive immune system include lymphocytes—primarily B cells and T cells—that circulate in the blood, , and lymphoid organs such as the , lymph nodes, and . B cells differentiate into plasma cells that secrete antibodies to neutralize extracellular pathogens, while T cells include helper T cells that coordinate responses, cytotoxic T cells that destroy infected cells, and regulatory T cells that maintain tolerance to self-antigens. Antigen-presenting cells, like dendritic cells and macrophages, bridge the innate and adaptive systems by processing and displaying antigens on (MHC) molecules to activate lymphocytes. The system's specificity arises from and hypermutation processes that generate billions of unique receptors on B and T cells, allowing recognition of virtually any foreign molecule while avoiding self-reactivity. Upon , clonal occurs, where antigen-specific lymphocytes proliferate rapidly, leading to effector functions such as production () or direct cell killing (). Memory cells persist long-term, enabling lifelong immunity to certain diseases, though dysregulation can contribute to autoimmune disorders or immunodeficiencies.

Overview

Definition and characteristics

The adaptive immune system is a subsystem of the that provides targeted defense against pathogens by mounting specific responses to antigens, primarily through the actions of lymphocytes. Unlike the , which offers immediate but non-specific protection, the adaptive system develops tailored reactions that improve with exposure. This subsystem relies on the of specific antigens to initiate protective mechanisms. Its defining characteristics include antigen specificity, which enables precise identification of invading agents such as viruses or ; diversity, allowing the generation of receptors capable of recognizing millions of different ; immunological memory, which confers long-term immunity and accelerates subsequent responses to the same ; and self/non-self recognition, which prevents attacks on the host's own tissues while targeting foreign entities. These features collectively ensure a highly effective, adaptable defense that evolves during an individual's lifetime. The terminology has evolved historically; early descriptions referred to it as "specific" or "acquired" immunity to highlight its pathogen-targeted nature and development through exposure, but the term "adaptive" gained prominence starting in , when Robert A. Good and colleagues used it to describe the system's developmental and phylogenetic adaptability in vertebrates like frogs. This modern usage underscores its capacity for learning and refinement beyond mere acquisition.

Functions and importance

The adaptive immune system plays a central role in defending against specific pathogens by recognizing and eliminating them through targeted responses, contrasting with the broader, faster action of the . It achieves this by detecting specific antigens on pathogens, leading to their precise neutralization or destruction, which amplifies the initial innate defenses to clear infections more effectively. This specificity allows the system to mount responses that are tailored to individual threats, such as viruses or , rather than relying on generalized mechanisms. A key function is ongoing for abnormal cells, including those infected by viruses or transformed into cancerous states, where the system identifies and eliminates threats that evade innate detection. For instance, it recognizes mutated proteins on tumor cells as foreign, enabling targeted elimination to prevent progression. Additionally, the establishment of immunological ensures long-term protection, with memory cells persisting after initial exposure to enable rapid reactivation upon re-encounter with the same . This memory formation is crucial for preventing reinfections, as seen in diseases like , where prior exposure confers lifelong immunity. The importance of the adaptive immune system extends to its role in vaccine efficacy, where mimics to generate responses without causing , thereby protecting populations from outbreaks. Quantitatively, while the innate response activates within hours, the requires 4–7 days for a primary response, involving clonal expansion of lymphocytes to amplify effector cells by orders of magnitude. Secondary responses, however, occur within 1–3 days due to pre-existing , highlighting its in sustained defense. Dysfunction in the adaptive immune system underscores its critical role in health; for example, targets and depletes key components, leading to profound and increased susceptibility to opportunistic infections and cancers. Conversely, overactivity can contribute to , where self-tissues are mistakenly attacked, as in or . Overall, its balance is essential for maintaining immune and preventing chronic diseases.

Cells Involved

Lymphocytes: T and B cells

Lymphocytes are the primary cellular effectors of the adaptive immune system, originating from hematopoietic stem cells in the . All lymphocytes begin as common lymphoid progenitors in the , where they differentiate into either T or B cell lineages. T lymphocytes, or T cells, migrate from the to the as early thymic progenitors, where they undergo maturation through stages involving gene rearrangement and selection processes to ensure self-tolerance and specificity. In contrast, B lymphocytes, or B cells, complete their maturation entirely within the , progressing through pro-B, pre-B, and immature B cell stages before becoming mature naive s. T cells are central to , coordinating responses against intracellular pathogens and abnormal cells by directly killing targets or activating other immune components through release. B cells, on the other hand, drive by producing antibodies that neutralize extracellular pathogens, mark them for destruction, or enhance . Both cell types achieve their specificity through diverse receptors generated via during development, enabling recognition of a vast array of antigens. The T cell receptor (TCR), a heterodimeric protein typically composed of α and β chains, is expressed on the surface of T cells and recognizes peptide antigens presented by major histocompatibility complex (MHC) molecules on cell surfaces. Similarly, the B cell receptor (BCR), an membrane-bound immunoglobulin (usually IgM or IgD), allows B cells to bind directly to soluble or surface-bound antigens without MHC restriction. These receptors, along with co-receptors like CD4 on helper T cells and CD8 on cytotoxic T cells, or CD19 and Igα/Igβ on B cells, facilitate signal transduction upon antigen engagement. Naive lymphocytes constantly recirculate between the blood, lymphoid tissues, and peripheral organs to surveil for antigens, a process essential for mounting rapid adaptive responses. T and B cells enter secondary lymphoid organs, such as lymph nodes and , via high endothelial venules, guided by adhesion molecules like and chemokines including CCL19, CCL21, and CXCL13. Upon exiting these sites, they return to circulation through lymphatic vessels, ensuring broad patrolling of the body while minimizing energy expenditure. This recirculation is tightly regulated, with naive cells expressing receptors like CCR7 and to home to specific lymphoid compartments.

Antigen-presenting cells

Antigen-presenting cells (APCs) serve as critical intermediaries that bridge the innate and adaptive arms of the by capturing, processing, and displaying antigens to lymphocytes, thereby initiating targeted adaptive responses. Professional APCs, which include dendritic cells, macrophages, and B cells, are specialized for this function due to their high expression of () class II molecules and ability to provide necessary costimulatory signals for effective T cell activation. In contrast, non-professional APCs, such as most other nucleated cells, primarily express and play a limited role in priming naive T cells, focusing instead on surveillance for endogenous threats. Dendritic cells (DCs) are the most potent professional APCs, excelling in the capture and of antigens from diverse sources, including pathogens and damaged tissues, to prime naive T cells in lymphoid organs. Macrophages, tissue-resident , act as professional APCs by engulfing cellular debris and microbes, contributing to both innate clearance and adaptive priming, particularly in inflammatory contexts. B cells function as professional APCs primarily for antigens recognized by their receptors, enabling them to internalize and present specific pathogens to helper T cells, which in turn supports . Antigen uptake by professional APCs occurs through specialized mechanisms tailored to the nature of the . allows macrophages and immature DCs to internalize large particulate antigens, such as or apoptotic cells, via pattern recognition receptors like Toll-like receptors. , including receptor-mediated and fluid-phase pathways, enables B cells and DCs to capture soluble antigens or immune complexes, facilitating targeted for subsequent into peptides suitable for MHC loading. Following uptake, professional APCs perform an initial overview of antigen degradation in endosomal compartments, preserving peptides for surface while integrating environmental cues to modulate the . Full activation of naive T cells by APC-presented antigens requires not only antigen recognition but also costimulatory signals to prevent anergy and promote and . The B7 family molecules and on professional APCs bind to on T cells, delivering a key signal that stabilizes the and amplifies T cell responses. This interaction, upregulated upon APC maturation, ensures that only relevant s in the context of danger signals trigger adaptive immunity. To prime lymphocytes effectively, professional APCs, particularly DCs, migrate from peripheral tissues to draining lymph nodes where naive T cells reside. This is orchestrated by the CCR7 on maturing DCs, which responds to ligands CCL19 and CCL21 in lymphatic vessels, enabling transport and in structured lymphoid environments. Macrophages and B cells also traffic to lymph nodes or via lymphatic or blood routes, positioning antigens for interaction with circulating lymphocytes.

Antigen Processing and Presentation

Exogenous antigens

Exogenous antigens are extracellular molecules, such as bacterial toxins, viral proteins released outside infected cells, or components from extracellular pathogens like certain parasites, that enter antigen-presenting cells (APCs) from the external environment. These antigens are distinct from intracellular threats and are primarily handled through the endosomal-lysosomal pathway to generate immune responses against infections occurring outside host cells. The processing of exogenous antigens begins with their uptake by APCs, such as dendritic cells, macrophages, or B cells, via mechanisms like , , or . Once internalized, the antigens are delivered to early endosomes, where the acidic environment promotes fusion with lysosomes containing proteolytic enzymes, including cathepsins. This degradation breaks down the antigens into fragments, typically 13-25 long, which are then transported to specialized compartments (MIICs). In these compartments, the invariant chain (Ii) dissociates from nascent molecules, allowing the fragments to bind to the MHC II groove, facilitated by , which edits the complex for optimal stability. The resulting peptide-MHC II complexes are transported to the cell surface for presentation. Presentation of exogenous antigens via activates + helper T cells, which recognize the complexes through their T cell receptors, leading to T cell proliferation and release. This activation is essential for initiating , as the helper T cells provide signals to B cells for production and class switching against extracellular pathogens. For example, protein subunit vaccines, such as those containing purified viral surface proteins like the hepatitis B surface antigen, are processed exogenously by APCs to generate MHC class II-restricted responses that drive protective antibody-mediated immunity.

Endogenous antigens

Endogenous antigens are intracellular proteins generated within host cells, such as proteins produced during or aberrant proteins arising from tumors, which are processed and presented to alert the to internal threats. These antigens are primarily handled through the cytosolic pathway, distinguishing them from extracellular threats managed via endosomal routes. The processing of endogenous antigens begins in the , where proteins—often including defective ribosomal products (DRiPs) that constitute a major source of peptides—are ubiquitinated and degraded by the 26S into short peptides, typically 8-10 long. These peptides are then transported across the (ER) membrane by the transporter associated with (TAP), a heterodimeric ATP-binding cassette protein composed of TAP1 and TAP2 subunits, which selectively binds and translocates peptides with appropriate hydrophobic anchors. In the ER lumen, the peptides are loaded onto newly synthesized (MHC-I) molecules within the peptide-loading complex (PLC), which includes chaperones like tapasin, , and ERp57 to edit and stabilize high-affinity peptide-MHC-I complexes, ensuring stable presentation. The assembled peptide-MHC-I complexes are transported through the Golgi apparatus to the cell surface, where they are displayed on nearly all nucleated cells, enabling surveillance by circulating CD8+ T cells for signs of or . Unlike MHC class II presentation, which is largely restricted to professional antigen-presenting cells (APCs), MHC-I expression is ubiquitous, allowing direct detection of compromised cells without intermediary processing. A specialized mechanism, , enables professional APCs such as dendritic cells to process and present exogenous —including those that originated as endogenous in neighboring cells (e.g., via uptake of infected cell debris or apoptotic bodies)—on to prime naive + T cells in lymph nodes. This process involves proteasomal degradation in the of the APC after antigen uptake, followed by TAP-dependent loading, and is crucial for initiating adaptive responses against viruses or tumors that do not directly infect APCs.

T Cell Mediated Immunity

CD8+ cytotoxic T cells

CD8+ cytotoxic T cells, also known as cytotoxic T lymphocytes (CTLs), are a subset of T lymphocytes that play a central role in by directly eliminating infected, cancerous, or abnormal cells. These cells express the coreceptor, which stabilizes their interaction with class I (MHC I) molecules on target cells presenting endogenous antigens, such as viral peptides or tumor-associated antigens. Upon activation, CD8+ T cells differentiate into effector cells capable of inducing target cell through specialized cytotoxic mechanisms. Activation of naive + T cells occurs primarily in secondary lymphoid organs, where they encounter antigen-presenting cells, such as dendritic cells, displaying peptides on MHC I via the (TCR). This TCR-MHC I interaction provides signal 1, while costimulatory signals from on the T cell binding to / on the deliver signal 2, preventing anergy and promoting full activation. Cytokines like IL-12 further drive differentiation by activating transcription factors such as T-bet and Eomesodermin, which upregulate effector genes. In the context of endogenous , this process ensures precise recognition of intracellular threats. Following , naive + T cells undergo rapid clonal expansion and differentiate into short-lived effector cells, with a subset surviving to form long-lived cells. This is influenced by the strength and duration of antigenic stimulation, as well as cytokines like IL-2, IL-7, and IL-15; brief exposure (2-24 hours) commits cells to effector fates, while prolonged signals favor formation. Effector + T cells acquire cytotoxic capabilities, including expression of perforin and granzymes, while cells, such as central and effector subsets, persist in lymphoid tissues or peripheral sites, enabling faster recall responses upon re-exposure. The transition involves downregulation of inhibitory receptors and upregulation of survival molecules like those in the TNF receptor family. The primary effector functions of CD8+ T cells involve inducing in target cells through two main pathways. In the perforin-granzyme pathway, activated CTLs release perforin to form pores in the target , allowing granzymes to enter and activate , leading to DNA fragmentation and cell death. Alternatively, CD8+ T cells express (FasL), which binds Fas receptors on target cells, triggering the extrinsic pathway via death-inducing signaling complex formation and activation. These mechanisms are calcium-dependent for and ensure without widespread tissue damage. CD8+ T cells are essential for viral clearance by recognizing and lysing virus-infected cells expressing viral peptides on MHC I, as demonstrated in infections like and where their depletion leads to persistent . In tumor surveillance, they patrol tissues to detect and eliminate neoplastic cells displaying mutated or overexpressed antigens, with high CD8+ infiltration in "" tumors correlating with better and response to . Additionally, CD8+ T cells contribute to by mounting alloreactive responses against mismatched MHC I on donor tissues, driving acute graft destruction through effector infiltration and cytokine production like IFN-γ and TNF-α.

CD4+ helper T cells

CD4+ helper T cells, also known as CD4+ T cells, are activated when their (TCR) recognizes peptide antigens presented by class II (MHC II) molecules on antigen-presenting cells, such as dendritic cells, in the context of costimulatory signals like CD28 binding to CD80/CD86. This activation occurs primarily in response to exogenous antigens processed by professional antigen-presenting cells. Upon activation, naive CD4+ T cells proliferate and differentiate into distinct subsets based on the milieu, transcription factors, and environmental signals, enabling them to coordinate tailored adaptive immune responses. The major subsets include Th1, Th2, Th17, and regulatory T (Treg) cells, each characterized by unique cytokine profiles and functions. Th1 cells differentiate under the influence of IL-12 and IFN-γ, driven by the T-bet, and secrete IFN-γ and IL-2 to promote macrophage activation and against intracellular pathogens. Th2 cells arise in the presence of IL-4, regulated by GATA3, producing IL-4, IL-5, and IL-13 to support and defense against extracellular parasites, though they also contribute to allergic responses. Th17 cells develop via TGF-β, IL-6, IL-21, and IL-23, with RORγt as the master regulator, secreting IL-17A/F and to recruit neutrophils and combat extracellular and fungi, but implicated in when dysregulated. Treg cells, induced by TGF-β and IL-2 under control, produce IL-10 and TGF-β to suppress excessive immune activity and maintain tolerance to self-antigens. Through secretion, + T cells orchestrate broader immune responses: Th1-derived IFN-γ activates macrophages for enhanced and promotes + T cell , while Th2 s like IL-4 drive class switching to IgE and recruitment. Th17 IL-17 induces proinflammatory to amplify innate responses, and Treg s dampen inflammation to prevent tissue damage. These helper functions are critical for activating s toward production and licensing + cytotoxic T cells for effective viral clearance. CD4+ T cells play pivotal roles in vaccine-induced immunity by providing help for long-lived antibody responses and formation, as seen in vaccines against viruses like and where robust CD4+ responses correlate with protection. In chronic infections, such as , sustained antigen exposure leads to CD4+ T cell exhaustion, marked by reduced production, upregulated inhibitory receptors like PD-1, and impaired , contributing to progression. Similarly, in cancer, exhausted CD4+ T cells fail to sustain antitumor immunity, allowing tumor evasion, though their partial retention of effector functions underscores potential therapeutic targets like checkpoint blockade.

Gamma delta T cells

Gamma delta (γδ) T cells represent a distinct subset of T lymphocytes characterized by their heterodimeric (TCR) composed of γ and δ chains, in contrast to the α and β chains found in conventional αβ T cells. This γδ TCR enables recognition of antigens in an MHC-independent manner, allowing γδ T cells to respond directly to stress signals or pathogen-associated molecules without requiring by (MHC) molecules on antigen-presenting cells. Unlike αβ T cells, which primarily mediate adaptive immunity through MHC-restricted recognition, γδ T cells bridge innate and adaptive responses by acting as rapid sentinels in peripheral tissues. A hallmark of γδ T cells is their ability to recognize non-peptide antigens, such as phosphoantigens produced by bacteria like or synthesized by host cells under stress. These small phosphorylated metabolites, including , are detected via the γδ TCR in conjunction with butyrophilin family molecules like BTN3A1 and BTN2A1, particularly in the Vγ9Vδ2 subset predominant in human peripheral blood. This recognition mechanism facilitates swift activation without the need for classical , enabling γδ T cells to mount early defenses against intracellular pathogens and tumors. γδ T cells play critical roles in early immune responses to infections, where they rapidly produce pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and interleukin-17 (IL-17) to recruit neutrophils and promote clearance. In , subsets like murine Vγ6Vδ1 cells in the and lungs secrete to support epithelial repair and tissue regeneration, while decidual γδ T cells in produce growth factors such as binding protein 2 (IGFBP2) and C (VEGFC) to aid development. For mucosal immunity, γδ T cells maintain barrier integrity at sites like the gut and by regulating composition—such as suppressing pathobionts like Aggregatibacter via IL-17—and defending against viruses, , and fungi through IFN-γ-mediated antiviral and antibacterial effects. These functions are supported by the tissue-resident nature of γδ T cells, which constitute 10-30% of T cells in and up to 40% in the , guided by receptors like and CCR6 in the or BTNL1-BTNL6 and α4β7 in the gut. Upon , resident γδ T cells exhibit innate-like rapid production, releasing IFN-γ and IL-17 within hours to orchestrate immediate inflammatory responses before adaptive immunity fully engages. This positioning and responsiveness position γδ T cells as key effectors in frontline mucosal and epithelial defense.

B Cell Mediated Immunity

B cell activation

B cells are activated upon recognition of antigens by their (BCR), a membrane-bound immunoglobulin that binds native, soluble, or membrane-associated antigens with high specificity. This binding triggers BCR clustering and initiates intracellular signaling cascades involving kinases such as Syk and family members, leading to the internalization of the antigen-BCR complex via clathrin-dependent . The internalized antigen is then processed into peptides within endosomal compartments and loaded onto major histocompatibility complex class (MHC II) molecules for presentation on the B cell surface. In the T-dependent activation pathway, antigen-activated B cells migrate to the T cell zones of secondary lymphoid organs, where they present processed peptides to CD4+ helper T cells via MHC II. Cognate recognition by activated (Tfh) delivers essential co-stimulatory signals to the B cell, primarily through the interaction of CD40 ligand (CD40L) on the T cell with CD40 on the B cell, which promotes B cell and . Additionally, cytokines secreted by Tfh cells, such as IL-4, IL-21, and IFN-γ, further modulate B cell responses by enhancing and directing . This interaction facilitates the formation of germinal centers, where B cells undergo clonal expansion, , and affinity maturation. T-independent activation occurs without T cell involvement and is typically triggered by multivalent antigens, such as bacterial , that extensively BCRs due to their repetitive epitopes. These antigens often engage additional innate receptors, including Toll-like receptors (TLRs) on B cells, providing co-stimulatory signals that amplify BCR-mediated activation and promote rapid differentiation into short-lived plasma cells. Marginal zone and B1 B cells are particularly responsive to such stimuli, enabling quick IgM production against blood-borne pathogens. Upon activation through either pathway, B cells differentiate into antibody-secreting plasma cells or memory B cells, with the former requiring transcription factors like Blimp-1 and XBP-1 to suppress proliferation and upregulate secretory machinery. In T-dependent responses, this differentiation is more robust and leads to long-lived plasma cells that reside in the , while T-independent activation primarily yields short-lived effectors.

Antibody production and classes

Antibodies, also known as immunoglobulins, are Y-shaped glycoproteins produced by plasma cells derived from activated B cells, consisting of two identical heavy chains and two identical light chains linked by bonds. The Y-shaped structure divides into the antigen-binding fragment () regions at the tips of the arms, which contain variable domains responsible for specific recognition, and the crystallizable fragment (Fc) region at the base, formed by constant domains that mediate interactions with immune cells and complement proteins. The variable domains in the Fab regions exhibit hypervariability in three complementarity-determining regions (CDRs) that form the antigen-binding site, while the constant domains determine the antibody's effector functions and isotype. Antibodies exist in five main isotypes in humans—Igm, IgG, IgA, IgE, and IgD—each defined by distinct constant regions of the heavy chain that confer unique structural and functional properties. IgM is the first isotype produced during an initial , existing primarily as a pentamer with high for multivalent antigens, while IgG predominates in secondary responses as a with strong penetration. IgA functions mainly in mucosal immunity as a dimer, IgE mediates allergic responses and anti-parasitic defense, and IgD's role remains less defined but involves regulation. Class switching, or isotype switching, allows to change from producing IgM (and IgD) to IgG, IgA, or IgE without altering the variable region, enabling tailored immune responses; this process is mediated by activation-induced cytidine deaminase (AID), which initiates DNA double-strand breaks in switch regions upstream of constant region genes, followed by repair. Cytokines from T helper cells, such as IL-4 for IgE switching or IFN-γ for IgG subclasses, direct the choice of isotype during reactions. The effector functions of antibodies primarily occur via the region and include neutralization, where antibodies bind pathogens to block their attachment to host cells; opsonization, which coats antigens to enhance by macrophages and neutrophils via Fc receptors; complement activation, initiating the classical pathway to form membrane attack complexes that lyse pathogens; and (ADCC), recruiting natural killer cells to destroy antibody-coated targets through Fcγ receptors. These functions vary by isotype: IgG subclasses excel in opsonization and ADCC, IgM is potent for complement activation due to its pentameric form, and IgA promotes mucosal opsonization but weakly activates complement. Neutralization and opsonization can occur independently of effector cells, providing rapid defense, while ADCC and complement activation amplify cellular and innate responses. Affinity maturation refines antibody specificity and strength during an through (SHM) and in germinal centers. SHM, also driven by , introduces point mutations at a high rate (about 10^{-3} per per generation) into the variable region genes of immunoglobulin loci, generating diversity in the CDRs. B cells with mutations yielding higher-affinity antibodies receive survival signals from presented on and T follicular helper cells, leading to selection and proliferation, while lower-affinity clones undergo ; this iterative process can increase affinity by 10- to 100-fold over days to weeks. Affinity maturation thus ensures robust, pathogen-specific .
IsotypeStructureKey Functions
IgMPentamer (or hexamer)Initial response; strong complement activation;
IgG (subclasses: IgG1-4)Opsonization; ADCC; neutralization; long-term immunity
IgADimer (secretory form)Mucosal immunity; opsonization in secretions
IgE; anti-parasitic; degranulation
IgDB cell surface receptor; unclear soluble role

Immunological Memory

Active immunity

Active immunity refers to the adaptive immune system's endogenous production of immunological following direct exposure to antigens, typically through natural , which confers long-term protection against subsequent encounters with the same . This process begins with the primary , where naive T and B lymphocytes recognize and respond to the for the first time, leading to a relatively slow phase that peaks after 7–10 days. During this phase, antigen-specific effector T cells and B cells proliferate, differentiate into short-lived effectors that eliminate the , and generate long-lived memory cells, establishing the foundation for future defenses. Upon re-exposure to the same , the secondary immune response, also known as the anamnestic response, is triggered by these memory cells, resulting in a faster onset (within 1–3 days) and greater magnitude compared to the primary response, with higher antibody titers and enhanced effector functions. This accelerated and amplified reaction effectively controls or prevents , highlighting the adaptive immune system's ability to "remember" prior threats. Memory T and B cells play pivotal roles here, rapidly expanding to produce antibodies and coordinate cellular immunity without the need for extensive naive cell recruitment. Memory cell formation occurs during the resolution of the primary response, where a subset of activated T and B cells survives and differentiates into distinct memory populations rather than undergoing . For T cells, central T cells (T_CM) primarily reside in secondary lymphoid organs, exhibit high proliferative potential, and maintain long-term surveillance, while effector T cells (T_EM) patrol peripheral tissues, providing immediate effector functions like production upon re-encounter. Similarly, memory B cells (MBCs) include central memory types in lymphoid tissues that undergo rapid class-switched antibody production and effector memory variants in mucosal or peripheral sites for swift local responses. These subsets ensure compartmentalized, efficient recall immunity tailored to the site's needs. The duration of active immunity varies by but can persist for decades or a lifetime, though it may wane over time due to , necessitating periodic boosting through re-exposure to sustain . For instance, natural infection induces lifelong immunity, as evidenced by adults with a history of during childhood not acquiring after re-exposure 65 years later. This enduring underscores the potency of endogenous formation in preventing severe recurrence.

Passive immunity

Passive immunity refers to the transfer of pre-formed antibodies from an external source to provide immediate but temporary protection against pathogens, without stimulating the recipient's own or generating . This form of immunity relies on the recipient's of exogenous antibodies, primarily (IgG) and (IgA), which neutralize pathogens or mark them for destruction by other immune components. Unlike adaptive responses that involve clonal expansion, passive immunity offers rapid onset but wanes as the transferred antibodies degrade. Natural passive immunity occurs primarily through maternal antibody transfer to the or newborn. During , IgG antibodies cross the placental barrier via the neonatal , providing the with protection against infections and during the early months of life. After birth, delivers IgA antibodies concentrated in and mature milk, which coat the infant's mucosal surfaces in the gastrointestinal and respiratory tracts to prevent adhesion and invasion. Artificial passive immunity involves the deliberate administration of antibodies, such as derived from immunized animals or s, or purified monoclonal antibodies, to confer immediate protection. For example, , often a combination of human or equine antibodies, is administered post-exposure alongside to neutralize the virus before it reaches the . Monoclonal antibodies, engineered for specificity, are increasingly used in targeted therapies, offering precise neutralization without the risks of polyclonal sera like . The duration of passive immunity is limited by the half-life of the transferred antibodies, typically lasting weeks to months. IgG antibodies have an average serum half-life of about 21 days, leading to gradual decline in protection until levels fall below effective thresholds, usually within 3 to 4 months. This short-lived nature makes suitable for bridging gaps in susceptibility rather than long-term defense. is particularly valuable for immediate protection in immunodeficient individuals, such as those with primary immunodeficiencies or undergoing immunosuppressive therapies, who cannot mount adequate responses to vaccines or infections. Antibody products like immune globulins provide a critical buffer against opportunistic pathogens during high-risk periods, such as post-transplant recovery or .

Immunization strategies

Immunization strategies represent artificial approaches to eliciting active immunity, leveraging the adaptive immune system's ability to generate long-term protection against pathogens through targeted exposure. These methods primarily involve , which introduce antigens in controlled forms to stimulate T cell and B cell responses without causing disease, thereby priming immunological memory for future encounters. By mimicking natural while minimizing risks, immunization has eradicated or controlled numerous infectious diseases globally. The foundation of modern traces back to Edward Jenner's 1796 development of the , the first successful vaccine in history, which used material from lesions to protect against the related variola virus; this breakthrough demonstrated the principle of cross-protective immunity and led to smallpox's global eradication in 1980. In the 20th and 21st centuries, vaccine innovation accelerated, with notable examples including the by in 1955 and the rapid deployment of mRNA-based COVID-19 vaccines in 2020, which encoded the to induce robust neutralizing production and were authorized for emergency use after demonstrating over 90% in phase 3 trials. These milestones highlight how strategies have evolved from empirical observations to sophisticated biotechnological interventions. Various vaccine types employ distinct mechanisms to present antigens and activate adaptive immunity. Live attenuated vaccines, such as those for , , and (MMR), use weakened s that replicate mildly in the host to provoke a strong, broad mimicking natural , conferring lifelong immunity in most recipients but contraindicated in immunocompromised individuals. Inactivated vaccines, like the inactivated polio vaccine (IPV), employ killed s or toxins to safely deliver antigens, stimulating primarily through production, though they may require boosters for sustained protection. Subunit vaccines, exemplified by the , isolate specific proteins (e.g., surface antigens) to target immune recognition without viral replication, offering precise and safe induction of B cell responses. mRNA vaccines, as in the Pfizer-BioNTech and COVID-19 formulations, deliver synthetic mRNA encoding antigens that cells translate into proteins, triggering both T cell and responses; their transient nature enhances safety and enables rapid adaptation to new variants. Viral vector vaccines, such as the COVID-19 vaccine using a modified adenovirus, ferry genes into host cells to produce antigens, eliciting cellular and while avoiding pre-existing immunity to common vectors. Achieving population-level protection often requires , where coverage reduces transmission to safeguard unvaccinated individuals; thresholds vary by , typically 80-95% for highly transmissible diseases like , as calculated from the (R0). Booster schedules reinforce waning immunity, such as annual vaccinations to counter antigenic drift or multiple doses in HPV programs to ensure durable protection against oncogenic strains, with ongoing 2025 initiatives expanding access in low-resource settings through WHO's Agenda 2030. Despite these advances, challenges persist, including driven by , which contributed to resurgence in several countries by 2023, and the need to update formulations against evolving variants, as seen in biennial COVID-19 booster recommendations targeting subvariants like JN.1 through 2025. Ongoing flu and HPV immunization programs face logistical hurdles in equitable distribution, yet demonstrate sustained impact, with HPV vaccination reducing incidence by up to 90% in vaccinated cohorts since introduction in 2006.

Generating Diversity

Genetic mechanisms

The adaptive immune system's capacity to recognize a vast array of antigens relies on germline-encoded genetic mechanisms that generate diversity in T cell receptors (TCRs) and B cell receptors (BCRs), also known as immunoglobulins, prior to any antigen encounter. These mechanisms primarily involve the rearrangement of gene segments within multigene families, ensuring a repertoire of receptors capable of binding diverse pathogens. Unlike the fixed, evolutionarily encoded receptors of the innate immune system, such as Toll-like receptors (TLRs), which recognize conserved pathogen-associated molecular patterns through limited germline variations, the adaptive system's genetic strategies enable combinatorial and junctional diversity on a massive scale. Central to this process is V(D)J recombination, a site-specific DNA rearrangement that assembles variable (V), diversity (D), and joining (J) gene segments to form functional receptor genes in developing lymphocytes. This recombination is mediated by the recombination-activating gene (RAG) proteins, RAG1 and RAG2, which form a complex that recognizes recombination signal sequences (RSSs) flanking the V, D, and J segments and introduces double-strand breaks at these sites. The broken DNA ends are then processed and ligated by the non-homologous end joining (NHEJ) pathway, resulting in the coding joint that encodes the variable region of the receptor and a signal joint that is discarded. For BCRs, V(D)J recombination occurs in the heavy chain locus (using V, D, and J segments) and light chain loci (using V and J segments only), while for TCRs, it involves similar segment usage, with D segments in the beta, delta, and gamma chains. The diversity generated by V(D)J recombination stems from two key features: combinatorial joining and junctional diversity. Combinatorial diversity arises from the random selection of one V, one or two D (where applicable), and one J segment from large multigene families; in humans, there are hundreds of such segments across the loci, including approximately 40-50 functional V segments, 25 D segments, and 6 J segments in the locus, with comparable scales in TCR loci. Junctional diversity further amplifies this by introducing variability at the segment junctions through the addition or removal of nucleotides by enzymes like (TdT), which adds non-templated N-nucleotides, and the imprecise processing of DNA ends. Together, these mechanisms can theoretically generate over 10^12 unique receptor specificities in humans, far exceeding the diversity possible from innate pattern recognition genes, which are encoded by fewer than 100 TLR family members recognizing broad microbial motifs. To maintain monospecificity, ensuring each expresses a single receptor specificity, regulates the recombination process such that only one of each receptor locus is productively rearranged per . This is achieved through a feedback mechanism where a functional protein product from the first successful rearrangement signals the inhibition of further recombination on the homologous , primarily via signaling pathways that enforce asynchronous replication timing and epigenetic silencing of the unrearranged locus. applies to both heavy and light chains in B cells and chains in T cells, preventing dual specificities that could lead to autoreactivity or inefficient responses. In contrast to the innate system's polyclonal activation via genes, this exclusion underpins the principle of adaptive immunity.

Somatic processes

Somatic hypermutation (SHM) is an antigen-driven process that introduces point mutations into the variable regions of immunoglobulin genes in activated B cells, primarily targeting the complementarity-determining regions (CDRs) to enhance antibody diversity and affinity. This process is initiated by the enzyme activation-induced cytidine deaminase (AID), which deaminates cytosine residues to uracil in single-stranded DNA during transcription, leading to a mutation rate of approximately 10^{-3} to 10^{-4} mutations per base pair per cell division—orders of magnitude higher than the spontaneous genomic mutation rate.00078-7) The resulting mismatches are processed by error-prone DNA repair pathways, such as base excision repair and mismatch repair, which incorporate mutations at both C/G and A/T pairs, thereby refining antibody specificity. Affinity maturation occurs concurrently with SHM in germinal centers of secondary lymphoid organs, where B cells proliferate and compete for antigen presented on follicular dendritic cells and T follicular helper cells. High-affinity B cell clones are positively selected through interactions that promote survival signals, while low-affinity clones undergo apoptosis, resulting in a progressive increase in average antibody affinity by 10- to 100-fold over the course of an immune response. This Darwinian selection process ensures that the humoral response evolves toward higher-affinity antibodies capable of more effective pathogen neutralization. In mammals, this refinement is almost exclusively driven by SHM, distinguishing it from the initial V(D)J recombination that establishes baseline diversity. In some non-mammalian , such as chickens, somatic diversification also involves gene conversion, where segments from upstream pseudogenes are copied into the functional region , introducing blocks of to expand diversity.90311-0) plays a central role here as well, facilitating the homologous recombination events that replace portions of the rearranged V with sequences from a library of pseudogenes, achieving diversification rates comparable to SHM in mammals. This mechanism predominates in avian B cells within the and contributes to broad coverage without relying heavily on junctional diversity. These somatic processes collectively broaden the adaptive immune response to accommodate evolving pathogens, such as viral variants, by generating antibodies with enhanced and potency through iterative and selection. For instance, SHM enables the adaptation of SARS-CoV-2-specific antibodies to emerging variants by introducing bystander mutations that improve binding to conserved epitopes.00485-5) This dynamic refinement is crucial for long-term protective immunity against pathogens that undergo rapid antigenic drift.

Regulation

Immune tolerance

Immune tolerance encompasses the adaptive immune system's mechanisms to discriminate self from non-self, preventing autoreactive responses that could lead to autoimmunity. These processes ensure that lymphocytes reactive to self-antigens are either eliminated or rendered inactive, maintaining immune homeostasis. Central tolerance operates during lymphocyte maturation in primary lymphoid organs, while peripheral tolerance provides backup suppression in mature cells that escape central checkpoints. Central tolerance for T cells occurs primarily in the through negative selection, where developing thymocytes with T cell receptors (TCRs) exhibiting high affinity for self-peptide-major histocompatibility complex (MHC) ligands undergo . This deletion targets double-positive thymocytes in the for ubiquitous antigens and single-positive cells in the medulla for tissue-specific antigens, presented by medullary thymic epithelial cells (mTECs) and dendritic cells. The process relies on proapoptotic molecules like Bim and Nur77 to induce the intrinsic pathway, eliminating up to 95% of thymocytes. For B cells, central tolerance takes place in the at the immature IgM+ stage, where B cell receptors (BCRs) binding self-antigens trigger checkpoints: receptor editing, involving secondary light-chain gene rearrangement to alter specificity (occurring in 20-35% of autoreactive cells), or via Bim-mediated pathways if editing fails. This reduces autoreactive B cells from 50-75% in early to about 10-40% in the mature repertoire. A key facilitator of central T cell tolerance is the autoimmune regulator (AIRE) gene, expressed in mTECs, which transcriptionally activates of thousands of tissue-restricted self-, such as insulin and . AIRE binds to unmethylated H3K4 marks, promoting stochastic TSA to just 1-2% of mTECs per , enabling effective negative selection of organ-specific autoreactive T cells. Defects in AIRE, as seen in Aire-knockout mice, impair this and lead to multi-organ , underscoring its role. Peripheral tolerance mechanisms act on autoreactive lymphocytes that evade central deletion, including anergy, where T cells encountering self-antigens without costimulatory signals (e.g., ligation) become hyporesponsive, upregulating inhibitory molecules like CTLA-4 and PD-1 while inhibiting and production. Deletion eliminates these cells via Fas- or Bim-dependent in secondary lymphoid organs, often induced by tolerogenic dendritic cells or stromal cells. Regulatory T cells (Tregs), primarily + + cells derived from the or periphery, suppress autoreactivity through direct contact, secretion (e.g., IL-10, TGF-β, IL-35), and metabolic disruption of effector T cells, maintaining balance in tissues. The discovery of these + Tregs and their role in peripheral tolerance by Shimon Sakaguchi, Mary Brunkow, and Fred Ramsdell was awarded the 2025 in or . Breakdowns in immune tolerance contribute to autoimmune diseases; for example, in , central failures such as reduced thymic insulin expression due to INS gene polymorphisms allow escape of β-cell-specific autoreactive T cells, while peripheral defects like impaired Treg function from low IL-2 signaling and reduced inhibitory receptor expression (e.g., CTLA-4, PD-1) enable their activation and pancreatic infiltration. These combined lapses result in progressive β-cell destruction, highlighting tolerance as a multi-layered safeguard.

Immune network hypothesis

The immune network hypothesis, also known as the idiotypic , was proposed by Niels Kaj Jerne in 1974 as a framework for understanding the self-regulation of the adaptive immune system. In this model, the variable regions of antibodies and T-cell receptors—termed idiotypes—act not only as sites for foreign antigens but also as antigens themselves, recognized by complementary anti-idiotypic immune components. This mutual creates an interconnected where an initial antibody response (Ab1) to an antigen elicits anti-idiotypic antibodies (Ab2) that bind to Ab1's idiotype, potentially suppressing or modulating the response; Ab2 can then stimulate further anti-anti-idiotypic antibodies (Ab3), forming a dynamic cascade that maintains without constant external antigenic stimulation. The theory has implications for stimulating adaptive immunity, particularly through anti-idiotype vaccines, where Ab2 mimics the three-dimensional structure of a pathogen's to elicit a targeted akin to the original . For instance, anti-idiotypic antibodies have been used experimentally to induce protective immunity against tumors and viruses by engaging the to amplify specific B- and T-cell clones. In the context of , network suppression occurs when anti-idiotypic interactions dampen autoreactive responses, preventing excessive activation and contributing to the balance between immunity and self-tolerance. Post-2000 refinements have integrated the idiotypic network with advances in regulatory T cells (Tregs) and cytokines, proposing that anti-idiotypic Tregs suppress effector responses via network interactions, while cytokines like IL-10 and TGF-β modulate network connectivity to fine-tune . This updated view posits the network as a layered system where Tregs bearing anti-idiotypic specificities enforce , bridging classical network dynamics with contemporary . Despite its foundational influence, the has drawn criticisms for assuming an overly connected given the vast of the immune , which may limit the feasibility of widespread idiotypic interactions, and for lacking predictive power in complex scenarios, rendering it largely of historical interest today. However, evidence from autoimmune models supports its relevance; for example, in experimental autoimmune encephalomyelitis, anti-idiotypic antibodies targeting pathogenic clones reduce disease severity by network-mediated suppression, and similar interactions modulate progression.

Acquired immunity during pregnancy

During pregnancy, the adaptive immune system undergoes specific modifications to accommodate the semi-allogeneic , balancing protection against pathogens with prevention of maternal rejection. These adaptations include shifts in T cell subsets and antibody dynamics that promote fetal while maintaining maternal immune competence. A key mechanism for fetal involves the expansion of regulatory T cells (Tregs), which suppress pro-inflammatory responses at the maternal-fetal interface. Tregs increase in number and function early in , peaking in the first and second trimesters, to inhibit effector T cell activity and promote an anti-inflammatory environment. Concurrently, there is a shift toward Th2 dominance, characterized by elevated levels of interleukin-4, -10, and -13, which favor and dampen Th1-mediated that could harm the fetus. This Th2 bias, alongside Treg activity, ensures immune and supports placental development. Maternal antibody transfer provides to the , primarily through the transplacental passage of (IgG). This process is mediated by the neonatal (FcRn) expressed on cells in the , which binds IgG in maternal circulation and facilitates its transport to the fetal compartment in a pH-dependent manner. IgG transfer increases exponentially from the second onward, peaking near term, and equips the neonate with protective antibodies against infections until its own matures. Disruptions in these adaptive mechanisms can lead to pathological conditions such as , where failure of Treg-mediated and excessive Th1/Th17 responses contribute to placental and vascular dysfunction. In , reduced Treg numbers and impaired balance result in endothelial damage and systemic immune dysregulation, often preceding clinical symptoms like . Gestational , manifesting as exacerbated autoimmune responses or flares during , similarly arises from breakdowns in adaptive , increasing risks for conditions like remission failure or with immune components. Postpartum, the adaptive immune system undergoes restoration, reverting from Th2 dominance to a balanced Th1/Th2 profile within weeks to months, with Treg levels declining to pre-pregnancy baselines. This recovery supports maternal defense against infections but can unmask latent . Breastfeeding aids this process by sustaining elevated and oxytocin levels, which modulate immune cell activity and enhance production in , indirectly supporting maternal immune while providing the with secretory IgA and other adaptive factors.

Evolution and Variations

Evolutionary origins

The adaptive immune system first emerged in jawed vertebrates, or gnathostomes, approximately 500 million years ago during the period, marking a pivotal innovation in vertebrate . This development coincided with the divergence from jawless vertebrates, such as lampreys and , and was facilitated by the acquisition of recombination-activating genes ( and RAG2), which originated from an ancient transposon. Concurrently, the (MHC) evolved to enable , allowing T cells to recognize foreign peptides in a self-restricted manner. These genetic elements provided the foundation for specific, memory-based immune responses absent in more primitive chordates. Central to this system's phylogeny were key innovations like V(D)J recombination, a somatic process that assembles variable (V), diversity (D), and joining (J) gene segments to generate diverse antigen receptors. This mechanism underpins the immunoglobulin (Ig) and T cell receptor (TCR) superfamily, whose members share structural domains and function in B and T lymphocyte recognition across all jawed vertebrates. The Ig/TCR superfamily diversified early, with evidence of both αβ and γδ TCR variants in basal gnathostomes, reflecting an ancient split that enhanced pathogen surveillance. The evolution of the adaptive immune system was driven by co-evolution with pathogens, exerting selective pressures that favored increased receptor diversity and affinity maturation. In early vertebrates, exposure to diverse microbial threats likely accelerated the refinement of RAG-mediated recombination and MHC polymorphism, as seen in the high variability of MHC loci in primitive fish species. This contributed to the system's robustness, enabling vertebrates to counter evolving infectious agents more effectively than innate defenses alone. Fossil and genetic evidence traces this system's progression from early gnathostomes to mammals. Phylogenetic analyses place its origin near the Cambrian-Ordovician boundary, inferred from the ~500-million-year-old divergence of chondrichthyans (), which possess functional Ig and TCR genes organized in clusters. In bony fish, such as , genomic sequencing reveals conserved V(D)J machinery and lymphoid organs, bridging to tetrapods. By the period (~400 million years ago), amphibians and subsequent amniotes (reptiles, birds, mammals) show elaborated structures like lymph nodes, underscoring a continuous refinement without major discontinuities.

Adaptive-like systems in other species

In jawless vertebrates such as lampreys and , an adaptive immune system analogous to that in jawed vertebrates operates through variable lymphocyte receptors (VLRs), which are somatically diversified to recognize antigens. Unlike the recombination-based immunoglobulin and systems in jawed vertebrates, VLRs achieve diversity via a gene conversion-like process involving the assembly of variable segments from flanking modules onto incomplete germ-line VLR s in s. This mechanism generates a vast repertoire of VLRs expressed on the surface of distinct lineages, enabling antigen-specific responses and immunological memory without the involvement of RAG-mediated recombination. In like , adaptive-like immune responses lack lymphocytes but incorporate sequence-specific mechanisms such as (RNAi) for antiviral defense. In , RNAi is triggered by viral double-stranded RNA, which is processed into small interfering RNAs (siRNAs) by Dicer-2; these siRNAs then guide Argonaute-2 to cleave complementary viral RNA, providing a targeted, heritable suppression of specific pathogens. Circulating hemocytes amplify and disseminate these siRNAs systemically, conferring a form of immunological memory that enhances resistance to reinfection. The phenoloxidase system complements this by activating melanization cascades in response to pathogens, though it operates more broadly through receptors rather than sequence specificity. Bacteria employ CRISPR-Cas systems as a form of adaptive antiviral immunity, acquiring short DNA sequences (spacers) from invading phages or plasmids and integrating them into CRISPR arrays to create heritable memory. Upon reinfection, CRISPR transcripts form RNA guides that direct Cas nucleases to cleave matching foreign DNA with high specificity, preventing viral replication while sparing self-genomes through protospacer-adjacent motif recognition. These adaptive-like systems in non-vertebrates illustrate of antigen-specific recognition and memory, distinct from the lymphocyte-based architecture in vertebrates, and highlight the modular origins of adaptive immunity across kingdoms. By comparing VLRs, RNAi, and CRISPR-Cas to vertebrate mechanisms, researchers gain insights into the selective pressures driving immune innovation, underscoring the uniqueness of RAG-dependent recombination in enabling scalable, dual B- and T-cell responses in vertebrates.

References

  1. [1]
    The Adaptive Immune System - Molecular Biology of the Cell - NCBI
    Our adaptive immune system saves us from certain death by infection. An infant born with a severely defective adaptive immune system will soon die.Missing: authoritative sources
  2. [2]
    In brief: The innate and adaptive immune systems - NCBI - NIH
    Aug 14, 2023 · The adaptive immune system specifically targets the type of germ that is causing the infection. But to do that, it first needs to recognize the ...Missing: authoritative | Show results with:authoritative
  3. [3]
    An introduction to immunology and immunopathology
    Sep 12, 2018 · The hallmark of adaptive immunity is the capacity for memory which enables the host to mount a more rapid and efficient immune response upon ...The Immune System: Innate... · Innate Immunity · Adaptive ImmunityMissing: authoritative | Show results with:authoritative
  4. [4]
    20.5: Adaptive Immune System - Biology LibreTexts
    Jul 7, 2023 · The adaptive immune system (also called specific immunity) is a subsystem of the overall immune system. It is composed of highly specialized cells and ...
  5. [5]
    Adaptive Immune System - an overview | ScienceDirect Topics
    The adaptive immune system is typified by the following four characteristics: specificity, diversity, memory, and ability to distinguish between self and ...
  6. [6]
    Ontogeny and Phylogeny of Adaptive Immunity - ScienceDirect
    The chapter discusses the ontogeny and phylogeny of adaptive immunity, including the acquisition of passive and active immunity by the developing fetus.
  7. [7]
    18.1 Overview of Specific Adaptive Immunity - Microbiology | OpenStax
    Nov 1, 2016 · Adaptive immunity is defined by two important characteristics: specificity and memory. Specificity refers to the adaptive immune system's ...<|control11|><|separator|>
  8. [8]
    50 million years of chordate evolution: Seeking the origins of ... - PNAS
    The immune mechanisms shared by all vertebrates are collectively termed “adaptive immunity,” “combinatorial immunity,” or the “anticipatory response” (1) for ...
  9. [9]
    Evolution of Alternative Adaptive Immune Systems in Vertebrates
    Apr 26, 2018 · Finstad J, Good RA. 2. 1964. The evolution of the immune response: III. Immunologic responses in the lamprey. J. Exp. Med. 120:1151–68 ...
  10. [10]
  11. [11]
    Does the Immune System Naturally Protect Against Cancer? - Frontiers
    Collectively, these studies demonstrate that the adaptive immune system is able to detect cancer by specifically recognizing the mutated proteins of the ...
  12. [12]
    A guide to vaccinology: from basic principles to new developments
    Dec 22, 2020 · This Review provides an introductory overview of vaccines, immunization and related issues and thereby aims to inform a broad scientific audience
  13. [13]
    Principles of innate and adaptive immunity - Immunobiology - NCBI
    Moreover, because there is a delay of 4–7 days before the initial adaptive immune response takes effect, the innate immune response has a critical role in ...1-5. Most infectious agents... · 1-7. Lymphocytes activated by...
  14. [14]
    HIV infection, aging, and immune function: implications for cancer ...
    This article documents cancer risk among HIV-infected persons, reviews immune system effects of HIV infection in relation to cancer risk, discusses implications ...
  15. [15]
    Principles and therapeutic applications of adaptive immunity
    Apr 25, 2024 · Adaptive immunity is essential for host protection from infectious and malignant diseases but also contributes to autoimmune and inflammatory ...
  16. [16]
    Lymphocytes and the Cellular Basis of Adaptive Immunity - NCBI - NIH
    T cells develop in the thymus, and B cells, in mammals, develop in the bone marrow in adults or the liver in fetuses. Despite their different origins, both T ...
  17. [17]
    Introduction to T and B lymphocytes - Autoimmunity - NCBI Bookshelf
    The process of development and maturation of the T Cells in mammals begins with the haematopoietic stem cells (HSC) in the fetal liver and later in the bone ...
  18. [18]
    Antigen presentation by dendritic cells and their instruction of CD4+ ...
    Dendritic cells are powerful antigen-presenting cells that are essential for the priming of T cell responses.
  19. [19]
    Role of B cells as antigen presenting cells - PMC - PubMed Central
    This review will discuss the pathways and mechanisms by which B cells present antigens, and how B cells differ from other professional APCs.
  20. [20]
    Review of Dendritic Cells, Their Role in Clinical Immunology, and ...
    The DCs then migrate to the lymph nodes and present the antigens to the T-cells, and eventually the DCs mature, losing their antigen-uptake capacity, and ...
  21. [21]
    Dendritic cell migration in inflammation and immunity - Nature
    Jul 23, 2021 · Dendritic cells (DCs) play crucial roles not only in initiating protective immunity against invading pathogens but also in inducing immune ...Dc Migration Routes And... · Signaling Transduction Of Dc... · Molecular Regulation Of Dc...<|control11|><|separator|>
  22. [22]
    Pathways of Antigen Processing - PMC - PubMed Central - NIH
    Exogenous proteins, however, are primarily presented by MHC-II molecules. Antigens are internalized by several pathways, including phagocytosis, ...
  23. [23]
    The ins and outs of MHC class II-mediated antigen processing and ...
    In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide–MHC class II ...
  24. [24]
    Exogenous Antigens Bind MHC Class II first, and Are Processed by ...
    Aug 5, 2015 · Pathogen-derived antigens bind to MHC class II as full-length proteins or large fragments, while DM facilitates the selection of the best fitting epitopes.
  25. [25]
    Immune complex–mediated antigen presentation induces tumor ...
    Processing of extracellular antigen via the exogenous pathway culminates in the presentation of antigenic peptides on MHC class II for the activation of CD4 ...
  26. [26]
    Enhanced Delivery of Exogenous Peptides into the Class I Antigen ...
    However, for vaccines composed of soluble protein antigens, immunization usually results in antigen uptake into an exogenous processing pathway that leads ...<|control11|><|separator|>
  27. [27]
  28. [28]
  29. [29]
  30. [30]
  31. [31]
  32. [32]
    CD8 T-cell subsets: heterogeneity, functions, and therapeutic potential
    Nov 1, 2023 · As typical cytotoxic CD8+ T cells, Tc1 cells produce perforin, granzyme B, IFN-γ, and TNF-α, which enable them to eliminate tumor and infected ...
  33. [33]
    Cytotoxic CD8 + T cells in cancer and cancer immunotherapy - Nature
    Sep 15, 2020 · Cytotoxic T cells expressing cell-surface CD8 are the most powerful effectors in the anticancer immune response and form the backbone of current successful ...
  34. [34]
    Effector and memory T-cell differentiation: implications for vaccine ...
    Apr 1, 2002 · This review will focus on the formation of effector and memory T cells, and in particular, we will examine some of the recent models that have been proposed.
  35. [35]
  36. [36]
    CD4+T Cells: Differentiation and Functions - PMC - PubMed Central
    This paper will focus on the cytokine-signaling and the network of transcription factors responsible for the differentiation of naive CD4 + T cells.
  37. [37]
    Immunologic self-tolerance maintained by activated T cells ...
    Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995 Aug 1;155(3):1151-64.
  38. [38]
    Expanding roles for CD4+ T cells in immunity to viruses - PMC
    A key role of CD4+ T cells is to ensure optimal responses by other lymphocytes. CD4+ T cells are necessary as helpers to promote B cell antibody production and ...
  39. [39]
    CD4 T-Cell Exhaustion: Does It Exist and What Are Its Roles in ... - NIH
    In chronic infections and in cancer, persistent antigen stimulation under suboptimal conditions can lead to the induction of T-cell exhaustion.
  40. [40]
    γδ T cells: origin and fate, subsets, diseases and immunotherapy
    Nov 22, 2023 · In this comprehensive review, we explore the origin and fate of γδ T cells, their subsets, their relevance to various diseases including infections, autoimmune ...
  41. [41]
    Function of γδ T cells in tumor immunology & cancer therapy
    Mar 12, 2021 · T cells of the γδ lineage are unconventional T cells with functions not restricted to MHC-mediated antigen presentation.
  42. [42]
    Double-edged sword: γδ T cells in mucosal homeostasis and disease
    Sep 11, 2023 · γδ T cells efficiently defend the mucosa from various pathogens, such as viruses, bacteria, and fungi. In addition, γδ T cells are necessary for the ...
  43. [43]
    Early BCR Events and Antigen Capture, Processing, and Loading ...
    Mar 10, 2014 · Here, we review the sequential steps required for BCR recognition of antigen, internalization, processing, and loading onto MHC II, with ...Antigen Capture By B Cells · Bcr Signaling And Antigen... · Antigen Processing And...
  44. [44]
    B-cell activation by armed helper T cells - Immunobiology - NCBI - NIH
    T-cell dependent antibody responses require the activation of B cells by helper T cells that respond to the same antigen; this is called linked recognition.
  45. [45]
  46. [46]
    The Continuing Story of T-cell Independent Antibodies - PMC - NIH
    The purpose of this article is to review the role of extrafollicular and T-cell independent antibody responses in humoral immunity.
  47. [47]
  48. [48]
    The structure of a typical antibody molecule - Immunobiology - NCBI
    Antibodies are Y-shaped with three equal parts, made of two heavy and two light chains. The arms have V regions for antigen binding, and the stem has C regions.
  49. [49]
    Antibody Structure and Function: The Basis for Engineering ...
    Human immunoglobulins are Y-shaped proteins composed of two identical light chains (LCs) and two identical heavy chains (HCs). In natural systems, the pairing ...
  50. [50]
    Antibody Structure - an overview | ScienceDirect Topics
    Antibodies are Y-shaped proteins with two light and two heavy chains, linked by disulfide bonds. The antigen-binding site is at the tips of the Y.
  51. [51]
    Immunoglobulin class‐switch recombination: Mechanism, regulation ...
    According to the different regions of the Ig H chain, Ig is classified into five isotypes, namely IgM, IgG, IgA, IgE, and IgD. IgM typically exists in ...
  52. [52]
    Functional switching | Nature Immunology
    Dec 2, 2016 · Each antibody isotype leads to distinct immunological functions: immunoglobulin M (IgM) and IgD are a first-line defense mechanism; IgA is ...
  53. [53]
    Molecular Mechanism of Class Switch Recombination: Linkage with ...
    Apr 1, 2002 · Annual Review of Immunology Volume 20, 2002 Molecular Mechanism of Class Switch Recombination: Linkage with Somatic Hypermutation
  54. [54]
    Functions of Antibodies - PMC - PubMed Central - NIH
    Antibodies neutralize infectivity, mediate phagocytosis, ADCC, and complement-mediated lysis. They can also neutralize independently of effector cells.
  55. [55]
    Fc-dependent antibody effector functions in SARS-CoV-2 infection
    Dec 19, 2022 · Antibody-dependent complement deposition​​ Complement is a highly conserved component of the immune system that functions by opsonizing pathogens ...
  56. [56]
    Insights into the Structural Basis of Antibody Affinity Maturation from ...
    Jan 31, 2018 · Affinity maturation is the process whereby the immune system generates antibodies of higher affinities during a response to antigen.<|control11|><|separator|>
  57. [57]
    The Use of Somatic Hypermutation for the Affinity Maturation of ... - NIH
    Here we describe the use of SHM coupled with mammalian cell surface display for the maturation of antibodies in vitro and the complementarity of these methods.
  58. [58]
    B cell memory: from generation to reactivation - Nature
    Mar 7, 2024 · Here, we reviewed literature on the development of MBCs and their reactivation, interaction with other cell types, strategies against invading pathogens, and ...
  59. [59]
    Understanding the generation and function of memory T cell subsets
    Memory T cells can be broadly divided into central memory and effector memory subsets, which are endowed with different capacities to home to lymphoid or ...
  60. [60]
    Effect of subclinical infection on maintaining immunity against ...
    Introduction. Natural measles infection induces lifelong immunity as shown during an epidemic in the Faroe Islands when only inhabitants who had had measles in ...
  61. [61]
    Origin and evolution of the adaptive immune system - NIH
    The adaptive immune system (AIS) in mammals, which is centred on lymphocytes bearing antigen receptors that are generated by somatic recombination, arose ...
  62. [62]
    The RAG proteins in V(D)J recombination: more than just a nuclease
    The mechanism requires precise cutting of the DNA at segment boundaries followed by rejoining of particular pairs of the resulting termini. The imprecision of ...
  63. [63]
    Molecular Mechanism of V(D)J Recombination from Synaptic RAG1 ...
    RAG recognizes specific recombination signal sequences (RSSs) flanking the 3′ end of the V, D, and J segments, which are composed of a conserved heptamer, a ...
  64. [64]
    Role of recombination activating genes in the generation of antigen ...
    The first part of the review discusses the basic mechanism of V(D)J recombination, and the last part focuses on how the RAG complex functions as a sequence- ...
  65. [65]
    V(D)J recombination, somatic hypermutation and class switch ...
    The recombination of the V(D)J gene is the key mechanism to produce antibody diversity. The recombinational process, including randomly choosing a pair of V, D, ...
  66. [66]
    The generation of diversity in immunoglobulins - NCBI - NIH
    The human V gene segments can be grouped into families in which each member shares at least 80% DNA sequence identity with all others in the family. Both the ...Missing: multigene | Show results with:multigene
  67. [67]
    The Complete Nucleotide Sequence of the Human Immunoglobulin ...
    The region contains 123 VH segments classifiable into seven different families, of which 79 are pseudogenes. Of the 44 VH segments with an open reading frame, ...
  68. [68]
    Allelic exclusion of immunoglobulin genes: models and mechanisms
    (A) Ig allelic exclusion guarantees the monospecificity of B cells. Ig allelically excluded B cells produce only one functional HC and one functional LC, giving ...
  69. [69]
    Antigen Receptor Allelic Exclusion: An Update and Reappraisal - PMC
    Antigen receptor allelic exclusion is defined as the surface expression of Ig or TCR chains from a single allelic copy of corresponding genetic loci.
  70. [70]
    T-Cell Tolerance: Central and Peripheral - PMC
    Clonal deletion and clonal diversion (Treg differentiation) are the major processes in the thymus that eliminate or control self-reactive T cells. Although ...Central Tolerance · Clonal Deletion · Molecular Mechanisms Of...
  71. [71]
    Central B-Cell Tolerance: Where Selection Begins - PMC
    Central B-cell tolerance, in fact, refers to the process that negatively selects newly generated immature B cells that react with a self-antigen in the bone ...
  72. [72]
    Mechanisms of central tolerance for B cells - PMC
    Central tolerance refers to the regulatory mechanisms that occur at the early stages of B cell development in the bone marrow, when B cells carry a surface ...
  73. [73]
    AIRE in the Thymus and Beyond - PMC - PubMed Central
    Aire promotes self-tolerance in part by inducing the transcription of a wide array of tissue-specific antigens (TSAs), particularly in the thymus.
  74. [74]
    Immunological mechanisms of tolerance: Central, peripheral and the ...
    Dec 11, 2023 · Peripheral tolerance mechanisms, such as Treg regulation and the exhaustion, anergy, or deletion of self-reactive T cells, are mainly relied ...
  75. [75]
    Breakdown in Peripheral Tolerance in Type 1 Diabetes in Mice and ...
    T1D is an example of one such autoimmune disease wherein the breakdown in tolerance leads to the initiation and progressive destruction of the insulin- ...Missing: failure review
  76. [76]
    Towards a network theory of the immune system - PubMed
    Towards a network theory of the immune system. Ann Immunol (Paris). 1974 Jan;125C(1-2):373-89. Author. N K Jerne. PMID: 4142565. No abstract available ...Missing: PDF | Show results with:PDF
  77. [77]
    On the Genesis of the Idiotypic Network Theory
    Dec 4, 2012 · The idiotypic network theory (INT) was conceived by the Danish immunologist Niels Kaj Jerne in 1973/1974. It proposes an overall view of the ...Missing: original | Show results with:original
  78. [78]
    The promise of the anti-idiotype concept - PMC - PubMed Central
    A central tenet of the network theory is that the interaction between Id+ and anti-Id immunoglobulin has regulatory consequences in addition to generating cross ...Missing: modern | Show results with:modern
  79. [79]
    The Promise of Anti-idiotype Revisited - Frontiers
    Apr 11, 2019 · The promise of idiotype-based therapeutics has been disappointing forcing a new look at the concept and its potential to generate an effective ...
  80. [80]
    Anti-idiotypic regulatory T cell theory for immune tolerance - Frontiers
    This review proposes a new hypothesis for immune tolerance mechanism by integrating the pre-existing “idiotypic network theory” and “Treg cell theory” into an ...Missing: modern | Show results with:modern
  81. [81]
    Regulatory T Cell-Targeted Immunomodulatory Therapy for Long ...
    “Jerne's idiotypic network theory” (Jerne, 1974) proposes that antigen-binding sites (idiotypes) of autologous immunoglobulins are sufficiently immunogenic to ...
  82. [82]
    Idiotypic Network - an overview | ScienceDirect Topics
    The idiotypic network (IN) refers to a system where cells can recognize each other and antigens, allowing for communication and reconfiguration in response ...Missing: modern | Show results with:modern
  83. [83]
    Evidence in favor of a role of idiotypic network in autoimmune ...
    Dec 10, 2007 · In the autoimmune reaction, the autoreactive clone, being anti-idiotypic, responded earlier than the clone reacting to the injected antigen.Missing: criticisms | Show results with:criticisms
  84. [84]
    Innate and Adaptive Immune Systems in Physiological and ... - NIH
    This review will present our current understanding of how innate and adaptive immune responses contribute to pregnancy, generalize the key roles of each system ...
  85. [85]
    Role of Regulatory T Cells in Regulating Fetal-Maternal Immune ...
    The immune-suppressive property of Tregs equips this T lymphocyte subpopulation with a pivotal role in the establishment and maintenance of maternal tolerance ...
  86. [86]
    The Complement System, T Cell Response, and Cytokine Shift in ...
    Dec 7, 2021 · Successful pregnancy requires an immunological shift with T helper CD4+ bias based on disbalance Th1/Th17 versus Th2/T regulatory (Tregs) ...
  87. [87]
    T Helper (Th) Cell Profiles in Pregnancy and Recurrent ... - Frontiers
    Th2 cells, then, induce local Th2 dominance by releasing Th2-type cytokines, which could promote maternal-fetal tolerance. In a rodent study, when Th2 ...
  88. [88]
    Factors Affecting the FcRn-Mediated Transplacental Transfer of ...
    Placental transfer of IgG occurs in an exponential fashion as pregnancy progresses, with minimal transfer in the first trimester (6). In the second trimester, ...
  89. [89]
    FcRn, but not FcγRs, drives maternal-fetal transplacental ... - PNAS
    FcRn has been previously identified as the major driver of IgG transplacental transport. Here we examine whether other receptors, such as FcγRs, also contribute ...
  90. [90]
    Disruption in the Regulation of Immune Responses in the Placental ...
    ... disease, highlighting a relationship between preeclampsia and cardiovascular disease (10). ... failure of immune regulation (75, 162). This is expected ...
  91. [91]
    Four Pathways Involving Innate Immunity in the ... - Frontiers
    Activation of adaptive immunity by innate immune ... Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype.
  92. [92]
    Editorial: Adaptive Immunity in Pregnancy - Frontiers
    Oct 3, 2021 · Various subsets of T cells are essential for pregnancy tolerance and interact in networks with innate immune cells to counteract inflammation ...
  93. [93]
    Inflammatory and anti-inflammatory markers in plasma: from late ...
    Feb 12, 2019 · After delivery, the immune system returns to a non-pregnant state. A shift towards Th1 dominance, and a fall in Th2 and Treg cells, followed by ...
  94. [94]
    Immune Changes and Dysphoric Moods across the Postpartum - NIH
    Pregnancy is characterized by profound changes in maternal physiology, which include alterations in both the innate and adaptive immune systems. There is ...
  95. [95]
    Breastfeeding Contributes to Physiological Immune Programming in ...
    This review will focus on the importance of breastfeeding and its immunological biocomponents that allow physiological immune programming in the newborn.Missing: postpartum restoration
  96. [96]
  97. [97]
  98. [98]
  99. [99]
  100. [100]
  101. [101]
    Adaptive immunity by convergent evolution - Nature
    Apr 25, 2018 · Cooper and colleagues discovered a new type of adaptive immunity in jawless vertebrates. This revolutionary discovery demonstrated three ...Missing: like non-
  102. [102]
    Discovery of an unconventional lamprey lymphocyte lineage ...
    Sep 3, 2024 · Lymphocyte receptors independently evolved in both jawed and jawless vertebrates with similar adaptive immune responses.
  103. [103]
    Mechanism of adaptive immunity found in the fruitfly - Nature
    Apr 26, 2017 · Antiviral immunity in D. melanogaster is mediated by RNA interference (RNAi), a process whereby small interfering RNAs (siRNAs) guide an ...
  104. [104]
    Hemolymph protease-5 links the melanization and Toll immune ...
    Sep 8, 2020 · Proteolytic activation of phenoloxidase (PO) and the cytokine Spätzle during immune responses of insects is mediated by a network of hemolymph ...
  105. [105]
    Molecular memory of prior infections activates the CRISPR/Cas ...
    Jul 10, 2012 · Our results explain how adaptive CRISPR/Cas immunity becomes specifically directed towards foreign DNA, allowing bacteria to efficiently counter individual ...