Fact-checked by Grok 2 weeks ago

Immune response

The immune response is the coordinated physiological reaction mounted by the body's to detect, neutralize, and eliminate harmful foreign substances, such as pathogens (including , viruses, fungi, and parasites), toxins, and abnormal cells like cancer cells, while distinguishing self from non-self to prevent damage to healthy tissues. This multifaceted defense mechanism relies on a of cells, tissues, organs, and molecules that work together to maintain and protect against and . The immune response operates through two primary arms: innate immunity, which provides rapid, nonspecific protection as the first line of defense, and adaptive immunity, which offers targeted, antigen-specific responses with long-term memory for faster and more effective future encounters with the same threat. Innate immunity includes physical barriers like and mucous membranes, chemical defenses such as proteins in and , and cellular components including (e.g., neutrophils and macrophages) that engulf invaders, as well as natural killer cells that target virus-infected or cancerous cells. In contrast, adaptive immunity involves lymphocytes—primarily B cells and T cells—that recognize specific antigens via receptors, leading to processes like production for (mediated by B cells) and direct cell killing or helper functions for (mediated by T cells). Key organs and tissues, including the (where immune cells originate), (for T cell maturation), lymph nodes, , and mucosal-associated lymphoid tissues, facilitate the coordination and amplification of these responses through mechanisms like , signaling, and . Upon activation, the immune response triggers to recruit immune cells, to destroy pathogens, complement activation to lyse microbes, and in adaptive phases, clonal expansion of specific lymphocytes and generation of memory cells that confer lifelong immunity. Dysregulation of the immune response can lead to immunodeficiencies (impaired defense), allergies (overreaction to harmless substances), or autoimmune diseases (attack on self-tissues), highlighting its critical balance for health.

Overview

Definition and Importance

The immune response refers to the coordinated physiological reaction of the to foreign substances, termed antigens, or to damaged self-cells, encompassing processes of recognition, activation, and effector-mediated elimination to safeguard the host. This multifaceted defense integrates innate mechanisms, which provide rapid, nonspecific protection, and adaptive mechanisms, which offer antigen-specific targeting and immunological memory. The immune response is essential for preventing microbial infections, including those caused by , viruses, fungi, and parasites, thereby preserving integrity and overall . Dysregulation of this response can lead to immunodeficiencies, chronic inflammation, or autoimmune disorders, underscoring its in and disease resistance. Evolutionarily, these mechanisms are highly conserved among vertebrates, reflecting their indispensable function in survival against diverse environmental threats. In scope, the immune response builds upon but is distinct from passive non-immune barriers, such as or mucosal linings, by actively mobilizing cellular and soluble factors to detect and neutralize invaders rather than merely providing a physical shield.

Key Components and Organs

The immune system's cellular components primarily consist of leukocytes, a diverse group of derived from hematopoietic stem cells in the . Key leukocyte subtypes include neutrophils, which constitute the majority of circulating and serve as rapid responders; macrophages, which are tissue-resident ; dendritic cells, specialized for capture; and lymphocytes, encompassing T cells and B cells that are central to adaptive immunity. These cells differentiate from common myeloid and lymphoid progenitors, enabling a coordinated network for immune surveillance. Soluble molecules form another critical layer of the immune response, facilitating communication and direct pathogen neutralization. Antibodies, also known as immunoglobulins, are Y-shaped proteins produced by B cells that bind specifically to antigens on or infected cells. Cytokines are a broad class of small signaling proteins, including and interferons, secreted by various immune cells to regulate , , and recruitment. The comprises over 30 plasma proteins that amplify immune reactions through cascades leading to pathogen and opsonization. Anatomical sites are essential for immune cell development, maturation, and encounter with antigens. Primary lymphoid organs include the , where B cells mature and all leukocytes originate, and the , a site for T cell education and selection. Secondary lymphoid organs, such as lymph nodes, the , and (MALT), act as hubs for immune cell interactions; lymph nodes filter lymph and facilitate T and B cell responses, while the spleen monitors blood for pathogens, and MALT protects mucosal surfaces like the gut and . Immune cells and molecules are distributed via circulatory systems to ensure systemic coverage. Blood transports leukocytes and soluble factors like antibodies and cytokines throughout the body, while the collects interstitial fluids, antigens, and cells from tissues, directing them to lymph nodes. fluids in tissues provide a local milieu for initial immune detection, enabling rapid trafficking of cells like dendritic cells to lymphoid organs.

Innate Immune Response

First-Line Defenses

The first-line defenses of the consist of non-specific physical, chemical, and mechanical barriers that prevent pathogens from entering the body, acting as the initial shield before any cellular responses are triggered. These barriers are present at all times and function without prior exposure to antigens, providing immediate protection at potential entry sites such as , , gastrointestinal system, and genitourinary tract. By blocking or inhibiting microbial and proliferation, they significantly reduce the risk of from , viruses, and other invaders. Physical barriers form the outermost layer of defense, primarily through the intact and mucous membranes that line internal cavities exposed to the external . The 's and keratinized outer layer create a tough, impermeable shield that resists penetration by most pathogens, while — the shedding of dead skin cells—further removes attached microbes. Mucous membranes, found in the respiratory, digestive, and urogenital tracts, are coated with a viscous layer that traps particles and microorganisms, preventing direct contact with underlying tissues. In the , cilia—microscopic hair-like projections on epithelial cells—beat rhythmically to propel and trapped pathogens upward toward the throat for expulsion. Chemical barriers complement physical ones by producing substances that directly damage or inhibit pathogens. , such as produced by epithelial cells, disrupt microbial cell membranes and exhibit broad-spectrum activity against , fungi, and enveloped viruses at mucosal surfaces. , an enzyme abundant in tears, saliva, and nasal secretions, hydrolyzes in bacterial cell walls, effectively lysing and contributing to the sterility of ocular and oral environments. Low environments provide additional antimicrobial action; the stomach's maintains a of 1.5–3.5, denaturing proteins and killing ingested pathogens, while the vaginal mucosa's acidic of 3.8–4.5, maintained by from lactobacilli, inhibits the growth of opportunistic microbes like and bacterial vaginosis-associated species. Mechanical barriers enhance clearance through dynamic processes that physically remove pathogens from entry points. , the wave-like muscular contractions in the gastrointestinal and urinary tracts, propels contents along, dislodging and expelling adhered microbes before they can colonize tissues. flow in the respiratory and gastrointestinal tracts continuously sweeps trapped pathogens away, often aided by coughing or . Flushing mechanisms, such as the flow of through the urinary tract and across the eyes, wash away microbes attempting to ascend or adhere to these surfaces, maintaining sterility in otherwise vulnerable areas. These barriers are exemplified in their roles at key portals: in the , the combination of , cilia, and blocks inhaled and viruses like from reaching the lungs, while in the , low , peristalsis, and neutralize and expel ingested pathogens such as . If breached, these defenses signal the activation of subsequent innate cellular mechanisms.

Cellular and Molecular Mechanisms

The innate immune system's cellular and molecular mechanisms rely on receptors (PRRs) to detect invading pathogens through conserved molecular signatures known as pathogen-associated molecular patterns (PAMPs). Among these, Toll-like receptors (TLRs) form a major family of PRRs, with TLR4 recognizing (LPS) from and TLR5 detecting from motile bacteria.00122-5) Upon PAMP binding, TLRs trigger intracellular signaling cascades, such as the MyD88-dependent pathway, leading to the production of proinflammatory cytokines and activation of antimicrobial responses. Phagocytes, including macrophages and neutrophils, serve as key cellular effectors in the innate response by engulfing and destroying pathogens via .30065-6) Macrophages patrol tissues and initiate upon PRR engagement, forming a that fuses with lysosomes to degrade the engulfed material. Neutrophils, rapidly recruited to infection sites, exhibit enhanced phagocytic capacity and release (NETs) to ensnare and kill extracellular pathogens. Natural killer (NK) cells contribute cytotoxicity against virus-infected cells and tumor cells by recognizing the absence of class I molecules, releasing perforin and granzymes to induce target cell . The acts as a soluble molecular effector, activated through three pathways to amplify innate defenses. The classical pathway initiates via binding to antigens, the pathway spontaneously activates on microbial surfaces, and the recognizes carbohydrate patterns on pathogens, all converging on formation.81503-2) promotes opsonization by C3b tagging pathogens for , direct via the membrane attack complex, and through anaphylatoxins like C5a that recruit immune cells. Pathogen elimination involves diverse killing mechanisms orchestrated by innate cells, particularly neutrophils. Within phagosomes, neutrophils generate (ROS) via the complex, creating a toxic environment that damages microbial proteins and DNA. (NO), produced by inducible , further contributes to nitrosative stress and pathogen inhibition. Additionally, neutrophils deploy antimicrobial granules containing enzymes like and cationic peptides such as , which disrupt microbial membranes and amplify ROS effects. These mechanisms collectively drive pathogen clearance and link to downstream inflammatory responses.

Inflammatory Response

The inflammatory response is a fundamental component of the , representing a localized, rapid reaction to tissue injury, , or foreign invaders that aims to eliminate the threat and initiate repair processes. This response is orchestrated by resident immune cells and vascular , bridging immediate physical barriers to more targeted defenses. It is triggered by receptors (PRRs) detecting microbial or damage signals, setting off a cascade of events to contain and resolve the insult. The process unfolds in distinct stages, beginning with vascular changes that enhance delivery of immune effectors to the affected site. , mediated by local release of factors, increases blood flow, causing (redness) and warmth, while elevated allows plasma proteins and fluid to leak into tissues, leading to (swelling). These alterations are rapidly induced within minutes to hours following injury. Subsequently, cellular recruitment occurs through , where —small signaling proteins—guide neutrophils and monocytes from the bloodstream to the site via gradients, enabling and clearance. Key mediators drive these stages, primarily released by mast cells, macrophages, and endothelial cells. Mast cells degranulate to liberate , which promotes immediate and permeability, while prostaglandins, synthesized via pathways, amplify these effects and contribute to . Macrophages and other innate cells produce pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-α), which further recruit leukocytes, induce fever, and activate endothelial adhesion molecules to facilitate cell . The cardinal signs of inflammation—redness, heat, swelling, and —reflect these underlying changes and were first described by the Roman physician . arises from nerve stimulation by prostaglandins and , alerting the host to protect the injured area. Acute inflammation typically resolves within days once the trigger is neutralized, serving a protective role; in contrast, inflammation persists for weeks or longer due to unresolved stimuli or dysregulated signaling, leading to and tissue remodeling rather than repair. Resolution of inflammation is an active, programmed process essential for preventing excessive tissue damage and restoring . cytokines like IL-10, secreted by regulatory macrophages and T cells, suppress pro-inflammatory mediator production, inhibit leukocyte , and promote apoptotic clearance of spent neutrophils, thereby limiting injury. This phase ensures that the response does not escalate into , highlighting inflammation's dual nature as both defensive and potentially destructive.

Adaptive Immune Response

Antigen Recognition and Processing

Antigens in the adaptive immune response are broadly classified into exogenous and endogenous types. Exogenous antigens originate from extracellular pathogens, such as , and are typically internalized by antigen-presenting cells (APCs) through or . Endogenous antigens, in contrast, arise from intracellular sources like proteins synthesized within infected cells or abnormal self-proteins in cancer cells. These distinctions determine the specific pathways that prepare antigenic peptides for presentation on (MHC) molecules, enabling T cell recognition. The pathway processes endogenous antigens for presentation to + cytotoxic T cells. Cytosolic proteins are ubiquitinated and degraded by the into peptides, which are then transported into the (ER) via the transporter associated with (TAP). In the ER, peptides bind to nascent molecules with assistance from chaperones such as and tapasin, stabilizing the complex for transport to the cell surface. This pathway surveys intracellular threats, alerting + T cells to eliminate infected or malignant cells. In parallel, the pathway handles exogenous antigens for + helper T cells. Antigens are taken up into endosomal vesicles, where acidic conditions and proteases like cathepsin S degrade them into peptides. molecules, synthesized in the and protected by the invariant chain () to prevent premature peptide binding, traffic to late endosomal compartments called MHC class II compartments (MIICs). There, is proteolytically removed, leaving the CLIP fragment, which is exchanged for antigenic peptides facilitated by ; the resulting peptide- complexes are displayed on the surface. This process amplifies immune coordination by activating + T cells. Professional APCs, including dendritic cells, macrophages, and , are central to , as they efficiently process and display peptides on both and II molecules. Dendritic cells excel in capturing s via innate mechanisms and migrating to lymph nodes for T cell priming, while macrophages handle phagocytosed s and present antigens specific to their B cell receptors. Full T cell activation requires co-stimulatory signals alongside ; notably, the interaction between B7 molecules (/) on APCs and on T cells provides this essential second signal, preventing anergy and promoting proliferation.30149-2) Innate immune responses facilitate delivery to these APCs by initial capture.

Lymphocyte Activation and Differentiation

Lymphocyte activation begins when naive and encounter their specific antigens, typically presented by antigen-presenting cells in secondary lymphoid organs such as lymph nodes. This process triggers through the (TCR) or (BCR), leading to clonal expansion and into specialized subsets that amplify and direct the adaptive immune response. T cell activation primarily occurs in lymph nodes, where naive + helper T cells interact with dendritic cells presenting peptide-MHC class II complexes. Upon activation, these cells proliferate via clonal expansion, driven by interleukin-2 (IL-2), which binds to the high-affinity to promote progression and survival. Differentiation of naive + T cells into effector subsets—such as Th1 (promoted by IL-12 for intracellular ), Th2 (driven by IL-4 for extracellular parasites), Th17 (induced by IL-6 and TGF-β for fungal and bacterial responses), and regulatory T cells (Tregs, fostered by TGF-β for immune suppression)—is orchestrated by the milieu and transcription factors like T-bet, GATA3, RORγt, and , respectively. + cytotoxic T cells similarly undergo activation and clonal expansion in lymph nodes, differentiating into effectors that target infected cells, with a subset persisting as memory cells for long-term immunity. Ultimately, activated T cells differentiate into short-lived effector cells that execute immediate responses or long-lived central and effector memory cells that patrol tissues and rapidly reactivate upon re-exposure. B cell activation contrasts by occurring through two main pathways: T-dependent and T-independent. In T-dependent activation, B cells in lymphoid follicles recognize antigens via BCR, internalize and present them to CD4+ helper T cells in a process called linked recognition, leading to T cell-dependent proliferation and differentiation. This pathway drives formation in lymph nodes or , where B cells undergo and affinity maturation, selecting high-affinity clones through interactions with and T follicular helper cells. T-independent activation, often triggered by repetitive microbial structures like , bypasses T cell help and results in rapid, low-affinity responses without germinal centers. Like T cells, activated B cells clonally expand, with IL-2 and other signals supporting growth, before differentiating into effector plasma cells or memory B cells that provide durable .

Effector Functions

Effector functions represent the terminal phase of the adaptive immune response, where differentiated lymphocytes execute targeted actions to eliminate pathogens and infected cells. Following lymphocyte activation and differentiation, effector T and B cells deploy specialized mechanisms to resolve infections, with cytotoxic CD8+ T cells directly lysing target cells and CD4+ helper T cells amplifying broader immune coordination. Cytotoxic CD8+ T cells, also known as killer T cells, induce apoptosis in infected or malignant cells through the perforin-granzyme pathway. Upon recognizing antigen-MHC class I complexes on target cells, these effectors release perforin, which forms pores in the target cell membrane, allowing granzymes to enter the cytoplasm and activate caspases that trigger programmed cell death. This granule exocytosis mechanism is the primary mode of cytotoxicity for CD8+ T cells, ensuring precise elimination of threats without widespread tissue damage. CD4+ helper T cells, in contrast, primarily coordinate immune responses via cytokine secretion rather than direct killing. Differentiated into subsets such as Th1, Th2, Th17, or Tfh cells, they produce signature cytokines like (for macrophage activation and antiviral effects), (for class switching and humoral responses), (for recruitment), and (for enhancing CD8+ T cell and functions). These cytokines orchestrate , production, and cytotoxic responses, amplifying the overall efficacy of adaptive immunity against viruses and other pathogens. B cell effectors, primarily long-lived plasma cells, secrete antibodies of various isotypes that neutralize and facilitate their clearance. IgM serves as the initial response isotype, effective for and complement due to its pentameric structure; IgG, the most abundant in , promotes opsonization, neutralization, and (ADCC) through strong binding; IgA protects mucosal surfaces by preventing adhesion and promoting bacterial aggregation in the gut; IgE mediates anti-parasitic defenses and allergic responses by triggering ; while IgD's secreted form has a less defined role but may contribute to early immune regulation. These isotype-specific functions are programmed during differentiation, enabling tailored . Antibody-dependent cellular cytotoxicity (ADCC) bridges humoral and cellular arms, where cells recognize antibody-coated targets via FcγRIIIa () receptors. Binding triggers NK cell , releasing perforin and granzymes to lyse the opsonized cell, enhancing clearance of virus-infected or tumor cells without requiring . This mechanism is crucial for therapeutic antibodies, as seen in monoclonal treatments that exploit ADCC for antitumor effects.00188-0) Antibodies further promote clearance through opsonization, where their regions bind phagocytes like macrophages and neutrophils via receptors, markedly enhancing efficiency. Additionally, IgM and IgG recruit the via the classical pathway, generating C3b for further opsonization and the membrane attack complex for direct , thereby integrating adaptive specificity with innate effector amplification. These processes ensure rapid removal of immune complexes and infected debris, resolving infections and preventing chronic inflammation.00086-4)

Integration and Types

Interaction Between Innate and Adaptive

The innate immune system plays a pivotal role in priming the adaptive immune response, primarily through antigen-presenting cells such as dendritic cells (DCs), which link the recognition of pathogen-associated molecular patterns (PAMPs) to effective antigen presentation. Upon encountering PAMPs via pattern recognition receptors like Toll-like receptors (TLRs), DCs internalize and process antigens, presenting them as peptide-major histocompatibility complex (MHC) complexes to naïve T cells in lymphoid organs, thereby initiating adaptive immunity. Additionally, damage-associated molecular patterns (DAMPs) released from damaged cells enhance DC maturation and upregulate co-stimulatory molecules such as CD80 and CD86, providing essential signals for T cell activation and preventing tolerance. This priming ensures that adaptive responses are tailored to the specific threat detected by the innate system. Cytokines serve as critical bridges between the two arms of immunity, with innate cells producing factors that activate and direct adaptive effectors. Type I interferons, secreted by innate immune cells like plasmacytoid DCs in response to viral PAMPs, not only activate natural killer () cells for early but also prime + T cells for enhanced and effector function during subsequent adaptive phases. Similarly, interleukin-12 (IL-12) produced by conventional DCs promotes the differentiation of T helper 1 (Th1) cells, fostering against intracellular , while coordinating with cells to amplify interferon-gamma production. These signals create a milieu that shapes adaptive responses for optimal clearance. Feedback loops further integrate the systems, allowing adaptive outputs to enhance innate functions. Antibodies generated by B cells during the adaptive phase bind pathogens via their regions and engage Fc receptors on innate effectors like macrophages and neutrophils, promoting opsonization and to accelerate clearance. This antibody-dependent enhancement also triggers (ADCC) by cells, linking humoral adaptive responses back to innate killing mechanisms. Evolutionarily, the innate system functions as a rapid , providing immediate defense while instructing the slower, specific, and memory-capable adaptive arm, a paradigm first articulated by Charles Janeway Jr. in 1989. This integration has been conserved across vertebrates, balancing speed and precision to optimize host survival against diverse threats, as evidenced by the coordinated roles of DCs and cytokines in both ancient and modern immune contexts.

Humoral vs. Cell-Mediated Responses

The employs two primary branches—humoral and cell-mediated responses—to combat distinct types of , with the humoral response focusing on extracellular threats and the cell-mediated response targeting intracellular ones. The humoral immune response is antibody-mediated and primarily defends against extracellular , such as and toxins, by neutralizing them in bodily fluids to prevent or spread. It involves that differentiate into plasma cells upon recognition, secreting immunoglobulins (antibodies) of various classes, including IgM (the initial response for complement activation), IgG (for long-term neutralization and opsonization), IgA (mucosal protection), IgE (parasite and allergy responses), and IgD ( regulation). These antibodies bind to , marking them for destruction by or complement-mediated , effectively halting dissemination without directly invading host cells. In contrast, the cell-mediated immune response is orchestrated by T lymphocytes and targets intracellular threats, including viruses, intracellular bacteria, and cancer cells, by directly eliminating infected or abnormal host cells. Cytotoxic recognize antigens presented on molecules and induce in infected cells through perforin and granzyme release, preventing replication. Helper , particularly Th1 subsets, activate macrophages to enhance their killing capacity against engulfed intracellular pathogens, such as by promoting phagolysosome formation. The balance between humoral and cell-mediated responses is regulated by cytokine profiles from + T helper cells, which direct into Th1 or Th2 subsets. Th1 cells produce interferon-gamma (IFN-γ) to promote by enhancing activation and function, while Th2 cells secrete interleukin-4 (IL-4) to drive humoral responses by supporting proliferation, , and class switching.
AspectHumoral ResponseCell-Mediated Response
Primary MediatorsAntibodies (immunoglobulins)T lymphocytes (CD4+ and CD8+)
Key Cells InvolvedB cells and plasma cellsCD8+ cytotoxic T cells, CD4+ helper T cells, s
Main TargetsExtracellular pathogens (e.g., , toxins)Intracellular pathogens (e.g., viruses, intracellular ), cancer cells
Cytokine DriversIL-4 (Th2 profile)IFN-γ (Th1 profile)
Effector MechanismNeutralization, opsonization, complement activationDirect , macrophage activation
A classic example of is the response to , where the stimulates production of neutralizing IgG antibodies that bind and inactivate the tetanus toxin, providing long-term protection without cellular invasion. In , predominates, with CD4+ T cells producing IFN-γ to activate macrophages, enabling control of the intracellular bacterium within granulomas.

Regulation and Outcomes

Immune Tolerance and Memory

Central tolerance establishes self-tolerance during lymphocyte development by eliminating self-reactive cells in primary lymphoid organs. In the thymus, negative selection targets autoreactive T cells: thymocytes with high-affinity recognition of self-antigens presented on MHC molecules by medullary thymic epithelial cells (mTECs) or dendritic cells undergo apoptosis, preventing their maturation into the periphery. This process is facilitated by the autoimmune regulator (AIRE), which promotes ectopic expression of tissue-specific self-antigens (TSAs) in mTECs to broaden the repertoire of antigens surveyed. For B cells, central tolerance occurs primarily in the bone marrow, where immature B cell progenitors encountering self-antigens with high avidity are subjected to negative selection mechanisms, including clonal deletion or receptor editing to alter the B cell receptor (BCR) specificity.00611-9) Approximately half of newly generated B cells are autoreactive and retained in the bone marrow for these tolerance checkpoints.00611-9) Peripheral tolerance complements central mechanisms by inactivating or suppressing self-reactive lymphocytes that escape into secondary lymphoid tissues and circulation. A key process is T cell anergy, a reversible state of unresponsiveness induced in CD4+ T cells by suboptimal antigen presentation without co-stimulatory signals, involving transcriptional silencing, epigenetic modifications, and downregulation of pathways like mTORC1/Ras/MAPK. Regulatory T cells (Tregs), characterized by Foxp3 expression, play a central role in peripheral tolerance by suppressing autoreactive responses through cell-contact-dependent and cytokine-mediated mechanisms. Tregs express CTLA-4, which competes with CD28 on effector T cells for CD80/CD86 ligands on antigen-presenting cells, thereby inhibiting co-stimulation and promoting an immunosuppressive environment via trogocytosis and reduced effector T cell activation. Similarly, PD-1 on Tregs interacts with PD-L1 to dampen immune responses, enhancing Treg stability and function in peripheral tissues. Tregs also induce anergy in CD4+Foxp3- T cells via inhibitory receptors like CTLA-4 and PD-1, and secrete cytokines such as IL-10 and TGF-β to further modulate immunity. Immunological memory provides long-term protection by enabling faster and more robust secondary immune responses upon re-exposure, forming the basis of efficacy. Long-lived memory B cells and T cells persist after primary or , residing in lymphoid tissues, , or peripheral sites to mount amplified responses. Memory B cells rapidly differentiate into plasma cells producing high-affinity antibodies, while memory CD8+ T cells proliferate to clear infected cells more efficiently than naive counterparts. Vaccines exploit this by priming memory formation through exposure, leading to reduced disease severity; for instance, conjugate vaccines against induce memory via T cell help, unlike plain forms. Memory formation involves specialized processes that enhance response quality and longevity. In B cells, affinity maturation occurs in germinal centers (GCs) of secondary lymphoid organs, where activated B cells proliferate in the dark zone, accumulate somatic hypermutations in BCR genes, and compete for in the light zone to receive survival signals from . High-affinity clones are selected, leading to class-switched, high-avidity antibodies; a subset differentiates into long-lived plasma cells that home to niches for lifelong antibody secretion, independent of . For T cells, memory development includes epigenetic modifications that establish stable landscapes, distinguishing tissue-resident memory T (T_RM) cells from circulating ones through accessible regulatory elements for genes like Klf2 and S1pr1. These changes, involving transcription factors such as BACH2 and AP-1, support self-renewal and rapid effector function without persistent stimulation.

Dysregulation and Disorders

Dysregulation of the immune response occurs when regulatory mechanisms fail, resulting in exaggerated, misguided, or deficient reactions that contribute to various disorders. These failures can manifest as reactions, where the immune system overreacts to harmless antigens; , in which self-tolerance breaks down leading to attacks on the body's own tissues; or , characterized by inadequate defenses against pathogens. Such dysregulation often stems from a combination of genetic predispositions, environmental factors, and disruptions in immune , ultimately causing significant morbidity. Hypersensitivity reactions are classified into four types based on the Gell and Coombs , each involving distinct immune mechanisms. Type I, or immediate , is IgE-mediated and involves and degranulation upon binding, releasing and other mediators within minutes; examples include , allergic , and . Type II, antibody-dependent cytotoxicity, features IgG or IgM antibodies targeting cell-surface antigens, activating complement or natural killer cells, as seen in and Goodpasture syndrome. Type III involves immune complex deposition in tissues, triggering complement activation and inflammation 4–10 hours post-exposure, exemplified by systemic lupus erythematosus (SLE) and . Type IV, delayed-type , is T cell-mediated, with release causing tissue damage 48–72 hours after exposure, such as in and skin tests. Autoimmunity arises from the loss of , where self-reactive lymphocytes escape suppression and initiate chronic inflammation against host tissues. Diseases like (RA) and (MS) exemplify this, with RA involving destruction driven by autoantibodies and T cells, while MS features demyelination in the due to autoreactive T cells. Genetic factors, particularly (HLA) alleles, confer susceptibility; for instance, HLA-DRB104:01 increases RA risk through enhanced presentation of arthritogenic peptides, and HLA-DRB115:01 is strongly associated with MS, promoting myelin-specific T cell activation. Environmental triggers, such as infections via molecular mimicry or smoking, interact with these genetic risks to precipitate disease onset. Immunodeficiencies impair the immune system's ability to combat infections, divided into primary (genetic) and secondary (acquired) forms. Primary immunodeficiencies, like (SCID), result from mutations affecting development, leading to profound T- and B-cell deficiencies and susceptibility to opportunistic infections from infancy. Secondary immunodeficiencies, such as , arise from external factors depleting + T cells, causing progressive vulnerability to viral, bacterial, and fungal pathogens; untreated HIV can reduce CD4 counts below 200 cells/μL, defining AIDS. Both types compromise innate and adaptive responses, increasing mortality from otherwise controllable infections. Recent research highlights the gut microbiome's role in immune dysregulation, with —characterized by reduced microbial diversity—linked to heightened and . In conditions like and , altered composition promotes Th17 cell expansion and barrier dysfunction, facilitating release and loss of ; meta-analyses indicate a 0.11 decrease in alpha-diversity across autoimmune diseases. Interventions like fecal transplantation show promise in restoring balance and mitigating disease progression by modulating regulatory T cells and reducing .

References

  1. [1]
    Physiology, Immune Response - StatPearls - NCBI Bookshelf - NIH
    The immune response is the body's ability to stay safe by protecting against harmful agents. The response involves lines of defense against most microbes.
  2. [2]
    Definition of immune response - NCI Dictionary of Cancer Terms
    The way the body defends itself against substances it sees as harmful or foreign. In an immune response, the immune system recognizes the antigens.
  3. [3]
    Immune System | NIH - Clinical Info .HIV.gov
    A complex network of specialized cells, tissues, and organs that recognize and defend the body from foreign substances, primarily disease-causing ...<|control11|><|separator|>
  4. [4]
    Immunity In Depth | Linus Pauling Institute | Oregon State University
    Overview of the Immune System. The immune system protects the body against infection and disease. It is a complex and integrated system of cells, tissues, and ...
  5. [5]
    Parts of the Immune System | Children's Hospital of Philadelphia
    The immune system provides two levels of defense: innate and adaptive immunity. This discussion will begin with a brief description of the organs and tissues ...
  6. [6]
    Immune System and Disorders | Autoimmune Disease - MedlinePlus
    Dec 26, 2023 · When your immune system recognizes an antigen, it attacks it. This is called an immune response. Part of this response is to make antibodies.
  7. [7]
    An introduction to immunology and immunopathology - PMC - NIH
    Sep 12, 2018 · This article provides a practical overview of innate and adaptive immunity, and describes how these host defense mechanisms are involved in both heath and ...The Immune System: Innate... · Innate Immunity · Adaptive Immunity
  8. [8]
    Overview of the Immune Response - PMC - PubMed Central
    Memory responses, defined as rapid induction of high levels of high affinity antibody after secondary antigen challenge, are characterized by production of IgG, ...
  9. [9]
    The components of the immune system - Immunobiology - NCBI - NIH
    The myeloid progenitor is the precursor of the granulocytes, macrophages, dendritic cells, and mast cells of the immune system. Macrophages are one of the ...1-1. The white blood cells of... · 1-3. The peripheral lymphoid...
  10. [10]
    In brief: The innate and adaptive immune systems - NCBI - NIH
    Aug 14, 2023 · Antibodies are made by B cells. Germs and substances that can trigger the production of antibodies are called "antigens." An antibody only ...Bookshelf · The Adaptive Immune System... · T Cells<|control11|><|separator|>
  11. [11]
    The complement system and innate immunity - Immunobiology - NCBI
    The complement system is one of the major mechanisms by which pathogen recognition is converted into an effective host defense against initial infection.
  12. [12]
    In brief: What are the organs of the immune system? - NCBI - NIH
    Aug 14, 2023 · The organs of the immune system include skin and mucous membranes, blood and the organs of the lymphatic system.
  13. [13]
    A physical wiring diagram for the human immune system - Nature
    Aug 3, 2022 · The human immune system is composed of a distributed network of cells circulating throughout the body, which must dynamically form physical ...
  14. [14]
    The role of pattern-recognition receptors in innate immunity - Nature
    Apr 20, 2010 · Here we describe the recent advances that have been made by research into the role of TLR biology in host defense and disease.
  15. [15]
    Pattern recognition receptors in health and diseases - Nature
    Aug 4, 2021 · Toll-like receptors (TLRs) are one of the earliest PRRs discovered in the innate immune system, which plays an important role in inflammatory ...
  16. [16]
    Macrophages in immunoregulation and therapeutics - Nature
    May 22, 2023 · Macrophages are crucial in innate immunity by regulating several homeostatic and evolutionary host defense immune responses.
  17. [17]
    Neutrophil diversity and function in health and disease - Nature
    Dec 6, 2024 · Neutrophil phagocytosis​​ Neutrophils are pivotal innate immune cells, particularly during the early stages of infection, owing to their ...
  18. [18]
    Functions of natural killer cells | Nature Immunology
    Apr 18, 2008 · Natural killer (NK) cells are effector lymphocytes of the innate immune system that control several types of tumors and microbial infections.
  19. [19]
    Complement and its role in innate and adaptive immune responses
    Dec 15, 2009 · The complement system plays a crucial role in the innate defense against common pathogens. Activation of complement leads to robust and ...
  20. [20]
    Neutrophils to the ROScue: Mechanisms of NADPH Oxidase ...
    Aug 25, 2017 · Reactive oxygen species (ROS) generated by NADPH oxidase play an important role in antimicrobial host defense and inflammation.
  21. [21]
    Roles of neutrophil granule proteins in orchestrating inflammation ...
    Here, we review the mechanism of neutrophil degranulation and the emerging roles of granule proteins in orchestrating neutrophil‐mediated immunity. We also ...
  22. [22]
    Innate immunity and inflammation - PMC - PubMed Central - NIH
    Aug 22, 2016 · It is now recognized that inflammation induced by microbial infection and tissue damage is an essential mechanism of innate immune response.
  23. [23]
    An introduction to immunology and immunopathology
    Sep 12, 2018 · An important function of innate immunity is the rapid recruitment of immune cells to sites of infection and inflammation through the production ...
  24. [24]
    Pathways of Antigen Processing - Annual Reviews
    Mar 21, 2013 · In this review, we highlight the aspects of MHC-I and MHC-II biosynthesis and assembly that have evolved to intersect these pathways and sample ...
  25. [25]
    Major Histocompatibility Complex (MHC) Class I and ... - Frontiers
    Here, we review the recent literature that describe MHC class I and II dynamics from a theoretical and experimental point of view.
  26. [26]
    Towards a systems understanding of MHC class I and MHC class II ...
    Nov 11, 2011 · This article provides a broad, timely review of our current understanding of MHC class I and class II antigen presentation and presents a ...
  27. [27]
    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 ...
  28. [28]
    Metabolic guidance and stress in tumors modulate antigen ... - Nature
    Oct 16, 2022 · DCs, macrophages, and B cells are usually referred to as professional antigen-presenting cells (APCs) and are critical “matchmakers” which ...
  29. [29]
    T cell responses: naïve to memory and everything in between - PMC
    This review will summarize the T cell response broken down into three major stages: activation, differentiation, and memory formation.
  30. [30]
    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.
  31. [31]
    Interleukin 2: from immunostimulation to immunoregulation and back ...
    In line with its in vitro function, IL-2 was also assumed to have a crucial role in vivo during antigen-driven clonal expansion of T cells. As IL-2 is mainly ...
  32. [32]
    Regulation of Human Helper T Cell Subset Differentiation by ... - NIH
    We propose that the differentiation of human Th subsets is largely regulated by the three cytokines, IL-12, IL-23, and TGF-β.
  33. [33]
    Effector and memory T-cell differentiation: implications for vaccine ...
    Apr 1, 2002 · The memory T-cell compartment consists of both CD4+ and CD8+ T-cells that can rapidly acquire effector functions to kill infected cells and/or ...
  34. [34]
    Lineage relationship of effector and memory T cells - PubMed Central
    In this model, rather than differentiating to memory cells, effector cells represent a terminally differentiated state that can only give way to more effectors ...
  35. [35]
    Germinal center-dependent and -independent immune responses of ...
    Jul 7, 2023 · In lymphoid organs, B-cell responses involve both germinal center (GC)-dependent and GC-independent pathways for Bmem cell and PC production.
  36. [36]
    GERMINAL CENTER B CELL DYNAMICS - PMC - NIH
    Germinal centers (GCs) are the site of antibody diversification and affinity maturation, and as such are vitally important for humoral immunity.
  37. [37]
    The Continuing Story of T-cell Independent Antibodies - PMC - NIH
    Participation of activated T cells is essential for GC responses and affinity maturation, and they can optimize class switching, but whether the resulting ...
  38. [38]
    Similarities and differences in CD4+ and CD8+ effector and memory ...
    Sep 1, 2003 · This review compares and contrasts how naive CD4 + and CD8 + T cells make the transition to effector and/or memory cells and discusses the implications of ...
  39. [39]
    A quarter century of granzymes | Cell Death & Differentiation - Nature
    Nov 4, 2011 · The primary cytolytic molecules, delivered to target cells upon contact with cytotoxic cells, are perforin and Grs. Currently, the mechanism ...
  40. [40]
    Mechanisms of granule-dependent killing | Cell Death & Differentiation
    Nov 2, 2007 · Recent work has suggested that Treg cells can kill immune effector cells either through a perforin/granzyme-dependent pathway or through ...Missing: CD8 | Show results with:CD8<|separator|>
  41. [41]
    Expanding roles for CD4+ T cells in immunity to viruses - Nature
    Jan 20, 2012 · CD4+ T cells are orchestrators, regulators and direct effectors of antiviral immunity. Neutralizing antibodies provide protection against ...
  42. [42]
  43. [43]
    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 ...
  44. [44]
    Natural killer cells in antitumour adoptive cell immunotherapy - Nature
    Jul 25, 2022 · Natural killer (NK) cells comprise a unique population of innate lymphoid cells endowed with intrinsic abilities to identify and eliminate virally infected ...
  45. [45]
  46. [46]
  47. [47]
  48. [48]
  49. [49]
  50. [50]
  51. [51]
    The Humoral Immune Response - Immunobiology - NCBI Bookshelf
    The humoral immune response uses antibodies from B cells to destroy extracellular microorganisms, preventing spread of intracellular infections. Antibodies ...
  52. [52]
    T Cell-Mediated Immunity - Immunobiology - NCBI Bookshelf - NIH
    Cell-mediated immune responses involve the destruction of infected cells by cytotoxic T cells, or the destruction of intracellular pathogens by macrophages ( ...
  53. [53]
    The distribution and functions of immunoglobulin isotypes - NCBI - NIH
    The first antibodies to be produced in a humoral immune response are always IgM, because IgM can be expressed without isotype switching (see Figs 4.20 and 9.8).
  54. [54]
    Humoral immune responses to infection: Common mechanisms and ...
    Feb 23, 2018 · Humoral immune responses are crucial for protection against invading pathogens and are the underlying mechanism of protection for most successful vaccines.
  55. [55]
    T cell-mediated cytotoxicity - Immunobiology - NCBI Bookshelf - NIH
    Armed effector cytotoxic CD8 T cells are essential in host defense against pathogens that live in the cytosol, the commonest of which are viruses.8-21. Cytotoxic T cells can... · 8-22. Cytotoxic effector...
  56. [56]
    IFNγ-producing CD4+ T lymphocytes: the double-edged swords in ...
    Jun 15, 2017 · A key role in CD4+ T cell-mediated macrophage activation can be attributed to IFNγ, which helps in Mtb clearance by inducing iNOS expression ...
  57. [57]
    Th1 and Th2 responses: what are they? - PMC - NIH
    Th1-type cytokines tend to produce the proinflammatory responses responsible for killing intracellular parasites and for perpetuating autoimmune responses.
  58. [58]
    Durability of Vaccine-Induced Immunity Against Tetanus and ... - NIH
    Mar 21, 2016 · Humoral immunity to tetanus as a function of age and time after vaccination. Tetanus-specific serum antibody responses were measured in adult ...Missing: examples | Show results with:examples
  59. [59]
    Cell mediated immune responses in Tuberculosis - PMC
    That CD4 T cells were the primary mediators of anti-tuberculous immunity was shown as a result of transfer models and then later by the use of gene-deficient ...
  60. [60]
    Cellular and molecular mechanisms breaking immune tolerance in ...
    Apr 1, 2021 · Central T-cell tolerance is achieved through the mechanism of negative selection, whereby thymocytes recognizing self-antigens displayed on MHC ...
  61. [61]
    Self and microbiota-derived epitopes induce CD4 + T cell anergy ...
    Nov 2, 2020 · Alternatively, anergy can be also induced in potentially autoreactive CD4+Foxp3− T cells upon binding of the inhibitory receptors PD-1 or CTLA4 ...
  62. [62]
    Regulatory T cells in homeostasis and disease - Nature
    Oct 14, 2025 · CTLA-4 and PD-1. Tregs express immune checkpoints such as CTLA-4 and PD-1, suppressing immunity via ligand interactions with effector T cells.Missing: anergy | Show results with:anergy
  63. [63]
  64. [64]
    A guide to vaccinology: from basic principles to new developments
    Dec 22, 2020 · Long-lived plasma cells that can continue to produce antibodies for decades travel to reside in bone marrow niches. CD8+ memory T cells can ...
  65. [65]
    CFP1 promotes germinal center affinity maturation and restrains ...
    Aug 27, 2025 · Cfp1 deficiency in activated B cells greatly impairs GC formation with diminished proliferation, somatic hypermutation and affinity maturation.
  66. [66]
  67. [67]
    Immunodeficiency - StatPearls - NCBI Bookshelf - NIH
    These immunodeficiencies can be either primary, such as Bruton disease, or secondary, as the one caused by HIV infection.[1][2]
  68. [68]
    Overview of Allergic and Atopic Disorders - Immunology
    Classification of Hypersensitivity Reactions. Hypersensitivity reactions are divided into 4 types (I through IV) by the Gell and Coombs classification.
  69. [69]
    HLA Association with Autoimmune Diseases - NCBI - NIH
    The HLA, as per almost all the ADs, has been shown to exert the strongest genetic association and effect on SLE to date. The top association was found at HLA- ...
  70. [70]
    Autoimmune disease: genetic susceptibility, environmental triggers ...
    This review integrates recent insights across genetic factors, environmental triggers, and immune dysregulation to build a comprehensive understanding of ...
  71. [71]
    The role of gut microbiota in autoimmune disease progression and ...
    May 15, 2025 · Over time, this dysfunction can trigger a systemic immune response, driving pathogenic immune cells and autoimmune disease. We, therefore ...