Fact-checked by Grok 2 weeks ago

Immune privilege

Immune privilege refers to specialized anatomical compartments in the body where immune responses are suppressed or modulated to prevent destructive , thereby protecting irreplaceable tissues such as those in the eye and from immune-mediated damage. This phenomenon enables the prolonged survival of foreign grafts, such as allografts, in these sites compared to conventional locations like . First described by in 1948 through experiments demonstrating reduced rejection of homografts in the anterior chamber of the eye and the , immune privilege represents an evolutionary balancing immunity against the risk of or excessive in vital, low-regenerative organs. Classical sites of immune privilege include the anterior chamber of the eye, central nervous system (CNS), testis, and fetomaternal interface during pregnancy, with emerging evidence for additional locations like the liver and gastrointestinal mucosa. In the eye, for instance, the avascular vitreous humor and retina are shielded, while the testis maintains separation via the blood-testis barrier to support spermatogenesis without immune attack. The CNS benefits from the blood-brain barrier, which restricts immune cell infiltration, meningeal lymphatics that enable controlled immune surveillance, and active presentation of endogenous self-peptides on MHC class II molecules, which induce regulatory T cells to dampen autoreactive responses. At the fetomaternal interface, semi-allogeneic fetal tissues evade maternal rejection through localized immunosuppression. Mechanisms underlying immune privilege combine passive barriers and active immunoregulatory processes. Passive elements include anatomical seclusion, such as the absence of lymphatic drainage in the eye's anterior chamber and the tight junctions of endothelial barriers that limit and leukocyte access. Active suppression involves the local expression of immunosuppressive molecules like transforming growth factor-β (TGF-β), which dampens pro-inflammatory production, and (FasL), which triggers in Fas-expressing immune cells attempting infiltration. In the eye, anterior chamber-associated immune deviation (ACAID) induces systemic by generating regulatory T cells and altering function upon exposure. Similarly, (IDO) in privileged sites depletes to inhibit T-cell proliferation. These mechanisms collectively foster a tolerogenic environment, often extending systemically to prevent widespread immune activation. Contemporary understanding reframes immune privilege not as an absolute, site-restricted trait but as a tunable property, inducible in non-privileged areas like or allografts through similar regulatory pathways. This perspective highlights its role in broader immunoregulation, including tumor evasion and chronic infection tolerance, with therapeutic implications for enhancing graft survival in transplantation or disrupting to bolster anti-tumor immunity. Ongoing research into glymphatic clearance in the CNS, mucosal tolerance in the gut, self-peptide-mediated in the CNS (as of 2024), and molecular maintaining ocular (as of 2025) further elucidates how these processes integrate with peripheral immune surveillance.

Fundamentals

Definition

Immune privilege refers to the phenomenon in which specific anatomical sites or tissues within the body actively suppress or limit immune responses to prevent inflammatory damage to vulnerable, non-renewable structures. This localized immune regulation protects essential functions, such as and neural processing, by mitigating the destructive potential of unchecked during pathogen encounters or immune challenges. Key characteristics of immune privilege include its relative , meaning it is not an barrier to immunity but a modulated suppression that allows controlled responses while prioritizing . Unlike passive , it involves active regulatory processes, such as the induction of tolerogenic immune cells and expression of immunosuppressive molecules, which collectively create an environment favoring antigen-specific over aggressive rejection. Evolutionarily, this has emerged to safeguard critical organs and reproductive processes, balancing the need for immune defense against the risk of autoimmune or inflammatory harm. Immune privilege is distinct from systemic , which establishes broad unresponsiveness across the body to self-antigens, whereas privilege operates through site-specific mechanisms that confine suppression to privileged tissues like the eye and . A notable illustration is the enhanced survival of allografts transplanted into these sites, where foreign tissues persist without systemic , as seen in corneal transplants achieving over 90% success rates in low-risk cases.

Physiological Significance

Immune privilege serves as a critical that safeguards essential s from the destructive potential of unchecked immune responses, thereby preserving vital physiological functions. In delicate structures such as the eye and (CNS), it prevents that could lead to irreversible damage, for instance by averting vision loss due to inflammatory swelling in the eye or neural destruction in the . This protective mechanism ensures the longevity and functionality of non-regenerative s, minimizing the risk of collateral harm during immune activation against threats. From an evolutionary perspective, immune privilege enhances organismal survival by facilitating key reproductive and sensory processes that would otherwise be vulnerable to immune-mediated rejection. It supports successful fetal development at the by tolerating semi-allogeneic tissues, protects gametes in the testes from autoimmune assaults on antigens, and maintains sensory integrity in sites like the eye to sustain environmental interaction. These adaptations reflect a selective pressure favoring tissues that prioritize over aggressive immunity, thereby promoting reproductive and long-term viability. Beyond localized protection, immune privilege contributes to systemic by modulating immune activity to avoid widespread disruption in interconnected physiological networks. Dysregulation of this balance has been associated with disorders such as in the eye and in the CNS, where excessive compromises tissue integrity. Overall, it fosters a stable internal environment conducive to health maintenance. At its core, immune privilege embodies a nuanced interplay between pathogen defense and self-protection, allowing controlled immune surveillance while curtailing responses that could harm privileged sites. This equilibrium reduces the likelihood of autoimmune attacks on irreplaceable tissues, enabling the to neutralize threats without sacrificing essential functions.

Mechanisms

Physical and Anatomical Barriers

Physical and anatomical barriers form the foundational structural components of immune privilege, limiting the ingress of immune cells and antigens into sensitive tissues while permitting essential nutrient and . These barriers primarily consist of specialized endothelial linings reinforced by tight junctions, which create a selective permeability profile that favors small molecules over larger immune effectors such as leukocytes and immunoglobulins. In privileged sites, this compartmentalization prevents the initiation of inflammatory responses that could compromise tissue function, such as vision in the eye or in the testes. The blood-brain barrier (BBB) exemplifies these features in the (CNS), where brain capillary endothelial cells are interconnected by complex tight junctions involving proteins like and claudins, supported by endfeet and . This structure restricts paracellular diffusion and leukocyte trafficking, while selective transporters enable nutrient passage. Similarly, the blood-aqueous barrier in the eye, formed by tight junctions in the non-fenestrated endothelium of and ciliary body vessels, along with the blood-retinal barrier's and endothelial tight junctions, maintains intraocular homeostasis by limiting immune cell entry and . In the testes, the blood-testis barrier (BTB) arises from tight junctions between Sertoli cells, creating basal and adluminal compartments that sequester post-meiotic germ cells from systemic circulation and immune surveillance. The blood-placental barrier, comprising and layers with tight junctions, isolates the by preventing maternal leukocyte migration while allowing selective transport of maternal antibodies and nutrients. Beyond vascular barriers, anatomical isolation enhances privilege through acellular zones and compartmentalization. For instance, the vitreous humor in the eye serves as an avascular gel that physically separates retinal tissues from anterior structures, reducing immune cell mobility. In the CNS, the (CSF) and meningeal compartments provide additional layering, with the dura and limiting direct blood-tissue interfaces. Testicular compartmentalization via the BTB and tunica albuginea further isolates seminiferous tubules, while placental trophoblast layers create a multi-tiered separation between maternal and fetal . These features collectively minimize exposure and access, fostering . Functionally, these barriers exhibit selective permeability that prioritizes physiological needs over immune activation; for example, the and BTB allow glucose and influx via specific carriers but block most plasma proteins and lymphocytes, thereby suppressing to T cells. This restriction curtails adaptive immune responses, as evidenced by reduced expression on barrier endothelial cells compared to peripheral vessels. In the eye and , similar mechanisms limit proinflammatory diffusion while permitting anti-inflammatory factors. Complementing these, anatomical features like the vitreous and CSF reduce convective flow of immune components. Evidence for barrier integrity derives from classic dye leakage experiments, which demonstrate restricted penetration in privileged sites. Paul Ehrlich's 1885 intravenous dye injections revealed brain exclusion, later confirmed by tracer studies showing minimal BBB leakage under normal conditions, with dye confined to vasculature. In the eye, tracers injected intravenously fail to cross the blood-aqueous barrier, but leakage occurs in inflammatory models like endotoxin-induced , highlighting barrier disruption's role in privilege loss. Analogous BTB studies using radiolabeled tracers confirm compartmental isolation, with adluminal compartment exclusion of systemic markers. Placental barrier experiments with fluorescent dextrans similarly show size-dependent permeability, restricting larger immune complexes. These findings underscore the barriers' role in maintaining immune quiescence.

Immunoregulatory Molecules and Cells

Immune privilege is actively maintained through the expression of specific immunoregulatory molecules that suppress inflammatory responses and promote of potentially harmful immune cells. (FasL), a member of the family, is prominently expressed in privileged sites and induces Fas-mediated in invading lymphocytes, thereby limiting their accumulation and . Transforming growth factor-β (TGF-β) isoforms, particularly TGF-β2, are secreted by resident cells in these tissues and foster an immunosuppressive environment by inhibiting pro-inflammatory cytokine production and enhancing regulatory pathways. (IDO) is another key enzyme expressed in privileged sites, including the and testis, where it catalyzes depletion along the , starving T cells of essential and inhibiting their proliferation. Additionally, neuropeptides such as α-melanocyte-stimulating hormone (α-MSH) contribute to tolerance by binding to on immune cells, downregulating inflammatory mediators like and pro-inflammatory cytokines. Complement regulatory proteins, including (DAF, also known as CD55), play a crucial role in preventing complement-mediated damage to host tissues within privileged sites. These proteins accelerate the decay of complement convertases, thereby inhibiting the classical and alternative pathways and protecting local cells from bystander lysis during any residual immune activity. Immunosuppressive cytokines such as interleukin-10 (IL-10) are often elevated in privileged tissues compared to systemic circulation, with studies showing up to several-fold higher concentrations that correlate with reduced T-cell proliferation and enhanced tolerance. At the cellular level, regulatory T cells (Tregs), particularly +CD25+Foxp3+ subsets, are enriched in immune-privileged environments and actively suppress effector T-cell responses through direct cell contact and secretion of inhibitory cytokines like TGF-β and IL-10. In the eye, anterior chamber-associated immune deviation (ACAID) represents a specialized where antigen exposure in the anterior chamber leads to systemic tolerance, involving the generation of regulatory T cells and modification of function to promote responses. Resident macrophages in these sites often adopt an M2-like , characterized by high expression of IL-10 and arginase-1, which promotes repair and dampens Th1-driven rather than exacerbating it. These cellular mechanisms integrate with molecular signals to sustain a tolerogenic milieu. Key pathways underlying this regulation involve the inhibition of pro-inflammatory Th1 and Th17 responses, which are critical for but detrimental in privileged contexts. TGF-β and IL-10 signaling suppresses STAT4 and RORγt transcription factors, respectively, thereby reducing interferon-γ and IL-17 production. Conversely, these molecules promote Th2 and Th3 (TGF-β-producing) responses, which favor and oral tolerance-like mechanisms without aggressive inflammation. This balanced pathway modulation ensures that immune surveillance persists at a low level while preventing destructive .

Privileged Sites

Eye

The eye maintains immune privilege through specialized barriers that limit inflammatory responses and promote tolerance, ensuring the preservation of visual function. A key ocular-specific barrier is the anterior chamber-associated immune deviation (ACAID), an active process induced when antigens are introduced into the anterior chamber, leading to systemic tolerance via the generation of regulatory T cells and suppression of delayed-type hypersensitivity. ACAID involves antigen-presenting cells in the and that process antigens in the presence of immunosuppressive factors, resulting in a deviant that favors regulatory mechanisms over effector responses. Complementing this, the blood-ocular barrier comprises tight junctions in the (forming the blood-retinal barrier) and non-pigmented ciliary epithelium (forming the blood-aqueous barrier), which restrict the influx of immune cells and molecules while allowing selective transport to maintain intraocular . These barriers collectively minimize exposure to systemic immunity, with the blood-retinal barrier particularly safeguarding the neural from circulating leukocytes. Local regulators further enforce ocular privilege by actively suppressing immune activation. The and express high levels of (FasL), a that induces in Fas-expressing inflammatory cells, thereby eliminating potential threats without widespread . In the , constitutive FasL expression acts as a barrier to infiltrating lymphocytes, promoting immune quiescence in the posterior segment. Additionally, the aqueous humor contains transforming growth factor-β2 (TGF-β2), which suppresses T-cell proliferation and activation by inhibiting production and promoting regulatory T-cell differentiation, often in a latent form that is activated locally to fine-tune responses. This TGF-β2 dominance in the anterior chamber microenvironment shifts immune dynamics toward . Ocular immune responses exhibit patterns that prioritize over cellular-mediated ones, reflecting the privilege's emphasis on non-destructive handling. Antigens encountered in the eye elicit stronger production while dampening cytotoxic T-cell activity, as seen in ACAID where splenic B cells are primed for humoral responses without robust Th1 effector functions. to ocular antigens is maintained through peripheral mechanisms, including regulatory T cells that suppress autoreactive responses to retinal proteins, preventing in the avascular vitreous and . This selective response ensures that intraocular threats are neutralized without collateral damage to delicate structures like the or photoreceptors. Experimental evidence underscores these mechanisms, with allografts placed in the anterior chamber demonstrating prolonged survival compared to subcutaneous sites, attributed to ACAID induction and local FasL-mediated of effector cells. In models, skin allografts in the anterior chamber persist indefinitely even in presensitized hosts, highlighting the chamber's tolerogenic capacity. Conversely, models reveal privilege breakdown when barriers are compromised; for instance, in experimental autoimmune induced by interphotoreceptor retinoid-binding protein, disruption of the blood- barrier allows T-cell infiltration, leading to and loss of . These models show that neutralizing TGF-β2 or FasL exacerbates , confirming their roles in maintaining privilege.

Central Nervous System

The central nervous system (CNS), comprising the brain and spinal cord, exhibits immune privilege through a combination of physical barriers and local regulatory mechanisms that limit inflammatory responses to protect delicate neural tissue. The blood-brain barrier (BBB) serves as the primary anatomical safeguard, formed by endothelial cells with tight junctions, basement membranes, and ensheathing processes from astrocytes and pericytes, which collectively restrict the passage of circulating immune cells, pathogens, and pro-inflammatory molecules from the bloodstream into the CNS parenchyma. Astrocytes contribute by inducing endothelial tight junction formation via secreted factors like agrin, while pericytes regulate vascular stability and permeability, further enhancing the barrier's selectivity to maintain a controlled microenvironment essential for neuronal function. Additionally, cerebrospinal fluid (CSF) dynamics, including bulk flow and glymphatic clearance pathways, facilitate the removal of waste and antigens while directing limited immune surveillance through meningeal lymphatics, preventing unchecked peripheral immune infiltration. Within the CNS, local immunoregulatory elements further enforce tolerance. Microglial cells, the resident macrophages of the brain, function as tolerogenic sentinels by monitoring the and modulating responses to potential threats, often promoting phenotypes that suppress excessive T-cell activation and release. These cells express (IDO), an enzyme that catabolizes , thereby depleting this and inhibiting T-cell proliferation and differentiation, which contributes to local immune suppression. IDO activity is upregulated in microglia, , and neurons in response to inflammatory signals, creating a metabolic environment that favors regulatory over effector immune responses. Immune patterns in the CNS are characterized by subdued and biased regulatory signaling. Neurons and most parenchymal cells display limited (MHC) class I and II expression under homeostatic conditions, minimizing by cytotoxic T cells and restricting adaptive immune within the . This low MHC profile, combined with preferential of regulatory T cells upon exposure to CNS antigens, directs immune encounters toward rather than rejection, preserving synaptic and neural signaling. Empirical evidence underscores these mechanisms' efficacy and limitations. Allogeneic tissue grafts implanted into the brain parenchyma demonstrate prolonged survival compared to those in peripheral sites, attributable to the BBB's isolation and local tolerogenic factors, as first observed in classic transplantation studies. In experimental autoimmune (EAE), a model of CNS , immune privilege delays but does not fully prevent T-cell infiltration and demyelination, revealing that while barriers and regulators attenuate responses, robust peripheral priming can overwhelm CNS tolerance.

Testes

The testes represent a key site of immune privilege in the , safeguarding developing gametes from immune-mediated damage to preserve . This privilege arises from a combination of physical barriers and active immunoregulatory mechanisms that prevent the recognition and attack of autoantigenic cells, which emerge post-puberty and express novel antigens absent during maturation. The compartmentalization of within the seminiferous tubules ensures that haploid germ cells are isolated from circulating immune components, allowing for the production of genetically unique spermatozoa without triggering systemic . A primary structural feature of testicular immune privilege is the blood-testis barrier (BTB), formed by specialized tight junctions between adjacent Sertoli cells near the of the seminiferous . These junctions create a selective permeability barrier that restricts the passage of immune cells, antibodies, and potentially harmful molecules into the adluminal compartment where post-meiotic germ cells develop, thereby sequestering sperm antigens from immune surveillance. This compartmentalization divides the seminiferous into basal and adluminal regions, with Sertoli cells enveloping and nurturing germ cells in the protected adluminal space during . Complementing these barriers, Sertoli cells actively contribute to by expressing (FasL), which induces in Fas-bearing infiltrating T cells, and by secreting transforming growth factor-β (TGF-β), a that dampens pro-inflammatory responses and promotes activity. Testicular macrophages, comprising a significant portion of the leukocyte , further reinforce this environment through their immunosuppressive ; unlike typical inflammatory macrophages, they produce anti-inflammatory cytokines such as IL-10 and exhibit reduced expression of pro-inflammatory mediators, helping to maintain local tolerance. This privileged state manifests in specific immune patterns, including tolerance to sperm antigens that arise after , when the is already established and could otherwise mount autoimmune responses against these novel self-s. The BTB and local regulators collectively prevent the formation of antisperm antibodies by limiting exposure and suppressing activation, ensuring that the majority of healthy males do not develop against their own spermatozoa. Experimental evidence underscores the robustness of testicular immune privilege: allogeneic skin grafts placed on the testicular surface in exhibit prolonged survival, often indefinitely, compared to grafts in non-privileged sites, demonstrating the site's capacity to inhibit allograft rejection. Similarly, models in rats reveal that immune privilege persists despite sperm leakage into the interstitial space; regulatory T cells are recruited to control potential autoimmune responses, maintaining without widespread .

Placenta and Fetus

The and represent a critical site of immune privilege, enabling the survival of a semi-allogeneic graft—the , which inherits paternal antigens foreign to the mother—without eliciting maternal rejection responses throughout . This privilege is essential for successful , as the maternal must tolerate fetal antigens while protecting against pathogens. The hemochorial structure of the human facilitates this by allowing direct contact between maternal and fetal in the intervillous space, yet maintains immunological separation. Physical barriers at the maternal-fetal interface play a foundational role in immune privilege. The layer, comprising and , forms the outermost fetal component, directly interfacing with maternal blood while limiting and immune cell trafficking. The underlying , a transformed endometrial layer rich in immune cells, further supports this barrier by regulating gradients that restrict leukocyte infiltration. In humans, this hemochorial configuration ensures efficient nutrient exchange without compromising tolerance, as the lacks classical and II molecules, reducing visibility to maternal T cells. Local immunoregulatory molecules and cells actively suppress potential rejection. Extravillous trophoblasts express non-classical , which binds inhibitory receptors such as KIR2DL4, ILT-2, and ILT-4 on decidual natural killer (dNK) cells, inhibiting their and promoting or anergy in these cells. This HLA-G-mediated interaction creates a tolerogenic environment, suppressing T cell proliferation and enhancing activity through cytokines like IL-10 and TGF-β. Complementing this, progesterone, elevated during , dampens maternal immune responses by inhibiting pro-inflammatory Th1 cytokines (e.g., TNF-α, IFN-γ) and promoting Th2 cytokines (e.g., IL-4, IL-10), while inducing the progesterone-induced blocking factor (PIBF) to bias immunity toward . Immune cell dynamics in the further exemplify this privilege, with uterine NK cells (uNK cells) shifting from to supportive roles. Comprising up to 70% of decidual leukocytes, uNK cells (CD56^bright CD16^dim) secrete angiogenic factors like VEGF and PlGF, driving spiral artery remodeling and placental vascularization essential for fetal growth, rather than mounting attacks on trophoblasts. Their low toward fetal cells stems from interactions with and on trophoblasts, which engage inhibitory receptors like CD94/NKG2A, fostering fetal . This pattern ensures that maternal immunity supports without rejecting paternal antigens. Evidence of this privilege is evident in the sustained survival of the semi-allogeneic , where maternal prevents graft-versus-host-like rejection despite paternal MHC disparities, as demonstrated in human and murine models of . Disruptions in these mechanisms underlie conditions like , where failed trophoblast invasion and HLA-G downregulation lead to inadequate spiral artery remodeling, placental ischemia, and a shift to pro-inflammatory Th1/Th17 dominance, resulting in systemic and . Thus, immune privilege at the and not only sustains but also highlights the consequences of its breach.

Clinical Implications

Transplantation Medicine

In transplantation medicine, immune privilege is leveraged to enhance graft survival, particularly in sites like the eye and (CNS), where natural barriers and immunoregulatory mechanisms reduce rejection risks without requiring systemic . Corneal allografts exemplify this application, as the avascular and immune-quiescent nature of the anterior chamber allows over 90% survival rates for first-time, low-risk transplants in humans, even without HLA matching or potent drugs, relying primarily on topical corticosteroids. In contrast, non-privileged allografts without are universally rejected within 10-14 days due to robust T-cell responses, highlighting the stark advantage of ocular privilege. Similarly, CNS implants for , such as neural precursor cell grafts, benefit from partial immune privilege in regions like the , where 50-70% graft survival is observed at two months in models, compared to complete rejection in less protected striatal sites. Strategies to harness immune privilege include deliberate site selection for grafts to exploit inherent protections; for instance, subretinal placement of cells extends privilege to allogeneic transplants by limiting T-cell infiltration and promoting , as demonstrated in models of retinal degeneration. Adjunct therapies mimicking privilege, such as FasL overexpression on graft surfaces, induce local of infiltrating lymphocytes and boost regulatory T cells, enabling prolonged islet allograft survival (>6 months) in nonhuman primates under short-term rapamycin, without systemic . These approaches draw from natural mechanisms like FasL expression in privileged tissues, adapting them to non-privileged grafts to foster localized . Outcomes underscore the efficacy of these tactics: corneal transplants achieve 86-90% one-year survival in low-risk cases, far surpassing the <10% long-term success of unmatched skin grafts without intervention, with reduced rejection episodes in privileged ocular beds. For CNS applications in Parkinson's, hippocampal implants show 50-70% viability at months post-transplant in preclinical studies, minimizing the need for heavy and preserving function. Despite these benefits, challenges persist, as immune privilege is incomplete in inflamed or vascularized sites, where prior rejection or elevates failure rates to over 60% for corneal grafts, necessitating combined like agents alongside site-specific strategies. In CNS transplants, surgical trauma can disrupt the blood-brain barrier, triggering innate that erodes privilege and accelerates rejection, underscoring the need for adjunctive therapies to maintain graft integrity.

Autoimmune and Inflammatory Disorders

Breakdowns in immune privilege can lead to autoimmune and inflammatory disorders in privileged sites, where the loss of protective mechanisms allows aberrant immune responses against self-antigens, resulting in tissue damage. In the eye, failure of ocular immune privilege contributes to , a group of inflammatory conditions characterized by intraocular inflammation that can cause vision loss if untreated. Similarly, in the (CNS), breaches in the blood-brain barrier () disrupt CNS immune privilege, enabling pathogenic immune cell infiltration that underlies (MS), an autoimmune . In the testes, compromise of testicular immune privilege can precipitate , involving leukocyte infiltration and seminiferous damage, often as part of autoimmune responses. The pathophysiology of these disorders often involves the diminution of key immunoregulatory factors, such as Fas ligand (FasL) and transforming growth factor-beta (TGF-β), which normally induce apoptosis in effector immune cells or promote tolerance. Loss of FasL-mediated apoptosis allows activated T cells to survive and infiltrate privileged sites, exacerbating inflammation in conditions like uveitis and orchitis. Reduced TGF-β signaling impairs the suppression of pro-inflammatory cytokines, facilitating effector cell entry and amplifying autoimmune damage in the CNS, as seen in MS where BBB disruption permits encephalitogenic T cell migration. This inflammatory cascade further erodes local barriers, creating a vicious cycle of tissue injury and immune activation. Epidemiologically, trauma to privileged sites heightens the risk of autoimmune disorders; for instance, penetrating ocular injury can trigger , a bilateral granulomatous occurring in up to 0.5% of cases, due to exposure of sequestered uveal antigens that breach immune privilege. Therapeutic interventions aim to restore immune privilege by targeting these breakdowns. Biologic agents like anti-tumor necrosis factor (anti-TNF) therapies, such as , effectively suppress by inhibiting pro-inflammatory cytokines, thereby reinstating ocular tolerance and reducing relapse rates in non-infectious cases. For broader autoimmune conditions affecting privileged sites, (MSC) therapies show promise in modulating regulators like TGF-β and FasL; MSCs exert immunomodulatory effects by promoting regulatory T cells and suppressing effector responses, as demonstrated in experimental autoimmune models where they ameliorate inflammation. In MS, MSC infusions have improved neurological outcomes in clinical trials by enhancing BBB integrity and dampening CNS inflammation, while emerging applications in testicular autoimmunity focus on stem cell-mediated regeneration and immune resetting to preserve .

Research History

Early Discoveries

The concept of immune privilege emerged in the late through observations by ophthalmologists studying the eye's unique immunological environment. In 1873, Dutch ophthalmologist van Dooremaal reported that a graft implanted into the anterior chamber of a dog eye survived for an extended period, unlike typical graft rejection in other sites, suggesting a protective environment in the avascular ocular compartment. This finding, along with similar reports from ophthalmologists attributing privilege to the eye's lack of vascular and lymphatic drainage, laid the groundwork for recognizing certain tissues as immunologically sheltered. Early 20th-century surgical advances further highlighted ocular immune privilege. By the 1900s, corneal transplants were being attempted with notable success rates, far exceeding those of other tissue grafts, due to the avascular nature of the minimizing immune recognition. The first successful full-thickness human corneal allograft was performed by Eduard Zirm in 1905, restoring vision in a patient without systemic , demonstrating the clinical implications of this phenomenon. These outcomes contrasted sharply with the routine rejection of vascularized grafts elsewhere in the body, prompting inquiries into site-specific immune suppression. Pioneering experiments in the mid-20th century formalized the concept. In 1948, coined the term "immune privilege" while investigating skin allografts placed in the anterior chamber of rabbit eyes, where they exhibited prolonged survival compared to subcutaneous sites, which he attributed to the absence of lymphatic drainage. Medawar's contemporaneous work on fetal tolerance, including his 1953 formulation of the "Medawar paradox" explaining how the fetus evades maternal rejection, linked developmental immunology to privileged sites like the , earning him the 1960 in Physiology or Medicine for discovering acquired immunological tolerance. Further foundational work in the explored immune deviation in the eye. J. Wayne Streilein and colleagues demonstrated that antigens injected into the anterior chamber of mouse eyes induced systemic rather than rejection, a phenomenon termed anterior chamber-associated immune deviation (ACAID) in their seminal 1981 , though initial experiments began in the late 1970s. These studies built on Medawar's tolerance framework, showing how privileged sites actively modulate immune responses to prevent inflammation in delicate tissues.

Modern Developments

The molecular era of immune privilege research, beginning in the , marked a shift toward identifying key genetic and molecular regulators that actively suppress immune responses in privileged sites. A pivotal discovery was the cloning and characterization of (FasL), a member of the family, which induces in Fas-expressing immune cells, thereby preventing inflammatory damage. This mechanism was demonstrated in the eye and testis, where constitutive FasL expression on parenchymal cells confers local immune suppression without systemic effects. Concurrently, the identification of (HLA-G), a non-classical molecule, revealed its role in placental ; expressed on extravillous trophoblasts, HLA-G inhibits cytotoxicity and promotes expansion, ensuring fetal allograft survival. Advancing into the 2010s and 2020s, high-throughput technologies like single-cell sequencing have elucidated the heterogeneity and site-specific adaptations of regulatory T cells (Tregs) in privileged tissues. These studies highlight tissue-resident Tregs with unique transcriptional profiles, such as elevated expression of immunosuppressive genes like and Il10 in and mucosal sites, which maintain privilege by dampening effector T cell responses and supporting niches. For instance, in the , Tregs cluster around the bulge region to enforce immune quiescence during anagen phases. Recent findings have extended immune privilege concepts to pathological contexts, notably cancer, where tumor microenvironments mimic privileged sites to evade immunity. Cancer stem cells exploit mechanisms like FasL upregulation and expression to induce in infiltrating lymphocytes and foster an immunosuppressive niche, contributing to therapy resistance in solid tumors. In the , studies have reaffirmed and expanded privileged status to additional sites: hair follicles, where collapse of privilege via re-expression triggers , and articular in joints, where low MHC expression and chondrocyte-derived factors limit T cell infiltration, protecting against autoimmune arthritis despite proximity to synovial inflammation. Technological advances have enabled precise manipulation and visualization of these dynamics. CRISPR-Cas9 editing of privilege regulators, such as knocking out PD-1 in Tregs or enhancing in grafts, shows promise for therapeutic immune modulation, particularly in transplantation by creating hypoimmunogenic cells that integrate into privileged sites without rejection. In vivo imaging techniques, including two-photon microscopy, have captured real-time Treg patrols in the niche and corneal stroma, revealing dynamic suppression of autoreactive cells and barrier enforcement during . Addressing longstanding gaps, integration with systems immunology approaches has modeled privilege as a network of cellular interactions and signaling pathways, incorporating multi-omics to predict responses in tissues like the CNS. This framework supports emerging applications in , where leveraging CNS immune privilege facilitates engraftment; recent preclinical studies using induced pluripotent stem cell-derived neurons have demonstrated sustained repair in models by minimizing alloimmune rejection. As of 2025, ongoing clinical trials, including FDA-cleared iPSC-based therapies for , continue to explore immune modulation strategies to enhance engraftment and efficacy.

References

  1. [1]
    Ocular immune privilege - PMC - NIH
    Jan 18, 2010 · The eye has a special relationship with the immune system, known as immune privilege. The term was coined in the 1940s by Sir Peter Medawar, who ...
  2. [2]
  3. [3]
    The privileged immunity of immune privileged organs - Frontiers
    Such immune privilege is thought to be an evolutionary adaptation to protect tissues that are indispensable, yet have limited regeneration capacity, like the ...
  4. [4]
    Immune privilege: a recurrent theme in immunoregulation? - 2006
    Sep 14, 2006 · Our goal in this issue is to assess contemporary thinking on classical immune privilege and to determine if the concept of immune privilege ...
  5. [5]
    Revisiting the concept of CNS immune privilege - PMC
    CNS immune privilege is the idea that the brain is tolerogenic, unable to induce an efficient immune response, and is immunologically unique.Missing: definition | Show results with:definition
  6. [6]
    Article Immune privilege or privileged immunity? - ScienceDirect.com
    Immune privilege can be generated in non-privileged sites such as the skin and allografts, and is a property of the tissue itself.
  7. [7]
    Immune Privilege: Keeping an Eye on Natural Killer T Cells - PMC
    Immune privilege is thought to reflect an evolutionary adaptation to protect vital structures from damage by inflammatory responses directed against pathogens.
  8. [8]
    Immune privilege or privileged immunity? | Mucosal Immunology
    Jul 2, 2008 · Immune privilege can be generated in non-privileged sites such as the skin and allografts, and is a property of the tissue itself.
  9. [9]
    Immune Privilege of Corneal Allografts - PMC - NIH
    Normally, HLA typing and systemic immunosuppressive drugs are not utilized, yet 90% of corneal allografts survive. In rodents, corneal allografts representing ...
  10. [10]
    Structural, cellular and molecular aspects of immune privilege in the ...
    Jun 10, 2012 · Moreover, the BTB can be comprised of three components, namely anatomical, physiological, and immunological barriers (Mital et al., 2011).
  11. [11]
    Ocular immune privilege - PMC - NIH
    The mechanisms of ocular immune privilege include unique anatomical features of a blood barrier and a lack of direct lymphatic drainage.
  12. [12]
    Immunology at the Maternal-Fetal Interface - StatPearls - NCBI - NIH
    Feb 22, 2025 · Apoptosis also contributes to immune privilege. ... Antiviral cytokines secreted by the placenta also play a crucial role in immune defense.Continuing Education Activity · Introduction · Function · Clinical Significance
  13. [13]
    CNS immune privilege: hiding in plain sight - PMC - PubMed Central
    The existence of BBB was first intimated by Paul Ehrlich in 1885 by his observation that systemically injected dye failed to penetrate the brain as readily as ...Missing: leakage | Show results with:leakage
  14. [14]
    Applications of the Role of α-MSH in Ocular Immune Privilege - PMC
    This chapter will cover what is known about how α-MSH is part of the mechanisms of ocular immune privilege, about the expression of melanocortin receptors.
  15. [15]
    Chronic Low Level Complement Activation within the Eye Is ... - IOVS
    We investigated the role of the complement system and complement regulatory proteins in an immune privileged organ, the eye. In the past, several in vitro ...
  16. [16]
    Interleukin‐10: A Multi‐Faceted Agent of Pregnancy - Thaxton - 2010
    May 10, 2010 · Levels of IL-10 from first and second trimester placental tissues were significantly higher than levels found in third trimester tissues, ...
  17. [17]
    Cellular and Molecular Mechanisms of Anterior Chamber ...
    The ACAID mechanism is an immunomodulatory phenomenon induced following an injection of an antigen into the anterior chamber of an eye. The antigen (depicted ...
  18. [18]
    Anterior chamber associated immune deviation (ACAID) - PubMed
    This review gives a historical perspective of the immune privilege research and provides up-to-date information of molecules, cells, and concepts newly ...
  19. [19]
    Blood–Retinal Barrier, Immune Privilege, and Autoimmunity - PMC
    The blood–ocular barrier and the DIE are mechanisms that depend on each other's proper function. They are central to preserving the immune privilege of the eye.
  20. [20]
    Ocular immune privilege | Eye - Nature
    Jan 9, 2009 · The first experimental description of ocular immune privilege was made by Medawar in the 1940s. He described the prolonged survival of skin ...
  21. [21]
    Ocular immune privilege: a review - ScienceDirect.com
    Abstract. The definition of the term 'immune privilege' has evolved over the last century. Current usage refers to a state within a particular organ or tissue ...
  22. [22]
    The Role of Retinal Pigment Epithelial Cells in Regulation of ...
    The RPE have an important role in ocular immune privilege to regulate the behavior of immune cells within the retina. Reviewed is the current understanding of ...
  23. [23]
    Ocular Immune Privilege and Transplantation - Frontiers
    The phrase immune privilege is a transplantation term defined by Peter Medawar and colleagues in the 1940s ( · There are resident immune cells with the potential ...
  24. [24]
    Decreased active TGF-β2 levels in the aqueous humour during ...
    Nov 2, 2007 · In this study, we tested the hypothesis whether the activated form of TGF-β2 is decreased in eyes with immune reactions following PK. Methods.
  25. [25]
    Ocular Immune Privilege and Transplantation - PMC
    Feb 8, 2016 · This review presents the current understanding of how the mechanism of ocular immune privilege promotes tolerogenic activity by APC, and T cells.
  26. [26]
    Ocular Immune Privilege and Uveitis - PMC - PubMed Central - NIH
    In 1948 Medawar described the prolonged survival of skin allografts within the anterior chamber of the rabbit eye. He observed that the grafts survived as along ...
  27. [27]
    Immunologic privilege in the central nervous system and the blood ...
    Oct 17, 2012 · We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors.
  28. [28]
    Blood-Brain Barrier and Its Role in Immune Privilege in the Central ...
    In this review, we discuss those characteristics of the BBB that play an important role in maintaining immune privilege in the CNS, as well as factors that ...Missing: prevention | Show results with:prevention
  29. [29]
    IDO (indolamine 2,3-dioxygenase) expression and function in the CNS
    The mechanism maintaining immune privilege are poorly understood and are apparently site-specific. In the placenta, inhibition of IDO leads to spontaneous ...
  30. [30]
    CNS immune privilege: hiding in plain sight - PubMed
    Tissues that are rapidly rejected by the immune system when grafted in sites, such as the skin, show prolonged survival when grafted into the CNS.
  31. [31]
    Revisiting the Mechanisms of CNS Immune Privilege - ScienceDirect
    We examine the notion of immune privilege of the CNS in light of both earlier findings and recent studies revealing a functional meningeal lymphatic system.Opinion · Introduction · Immune Privilege Of The Cns<|control11|><|separator|>
  32. [32]
  33. [33]
  34. [34]
  35. [35]
  36. [36]
  37. [37]
  38. [38]
  39. [39]
  40. [40]
  41. [41]
  42. [42]
    Comparative aspects of trophoblast development and placentation
    Based on the number of tissues separating maternal from fetal blood, placentas are classified as epitheliochorial, endotheliochorial or hemochorial.
  43. [43]
    Immune responses at the maternal-fetal interface - Science
    The placenta has a number of innate immune mechanisms to protect the fetus from congenital infections of all types, including the expression of pattern ...
  44. [44]
    Multi-Layered Mechanisms of Immunological Tolerance at the ...
    Jul 15, 2024 · Placental barrier restricts the transfer of fetal antigen to maternal circulation. Maternal immune cells including dNKs, antigen-presenting ...
  45. [45]
    HLA-G: An Important Mediator of Maternal-Fetal Immune-Tolerance
    The placenta is a more specific immunologically privileged site that prevents the fetus that contains paternal heredity from maternal immunological attack. It ...
  46. [46]
    Progesterone: A Unique Hormone with Immunomodulatory Roles in ...
    Jan 25, 2022 · The authors hypothesized that progesterone helps protect the conceptus from maternal immunologic rejection by suppressing maternal immune ...
  47. [47]
    Natural killer cell-triggered vascular transformation: maternal care ...
    Aug 16, 2010 · The uterus of early pregnancy is a non-lymphoid organ enriched in NK cells that are specifically recruited to placental attachment sites.Missing: vascularization | Show results with:vascularization
  48. [48]
    Disruption in the Regulation of Immune Responses in the Placental ...
    These include insufficient control of inflammation, failure of tolerance toward paternal antigens at the fetal–maternal interface, and subsequent over- or ...
  49. [49]
    Immune recognition and rejection of allogeneic skin grafts - PMC
    Many attempts to induce tolerance to allogeneic skin transplants (indefinite survival in the absence of chronic immunosuppression) have been unsuccessful, ...
  50. [50]
    Brain Region-Dependent Rejection of Neural Precursor Cell ...
    Apr 29, 2018 · We show here that the hippocampus is a partially immune privileged site which protects NPC grafts, as compared to the complete immune-rejection ...
  51. [51]
    Subretinal space and vitreous cavity as immunologically ... - PubMed
    Allogeneic newborn neural retinal grafts implanted in the subretinal space and vitreous cavity experience immune privilege and induce deviant immune ...
  52. [52]
    FasL microgels induce immune acceptance of islet allografts in ...
    May 13, 2022 · Islet transplantation to treat insulin-dependent diabetes is greatly limited by the need for maintenance immunosuppression.
  53. [53]
    High-risk corneal allografts: A therapeutic challenge - PubMed Central
    The corneal graft survival rate is 86% at 1-year for penetrating keratoplasty (PK), despite the fact that corneal grafts are rarely tissue matched for ...
  54. [54]
    The immune response of stem cells in subretinal transplantation
    Sep 14, 2015 · We then summarize methods that can suppress the immune response of the host and improve graft survival. Immune privilege of the subretinal space.
  55. [55]
    Immune Privilege: The Microbiome and Uveitis - PMC
    Jan 25, 2021 · Immune privilege (IP) was conceived as a protective response to immune challenge by tissues with limited capacity for renewal such as the eye ...
  56. [56]
    Testicular defense systems: immune privilege and innate ... - NIH
    Jun 23, 2014 · Testicular immune privilege protects immunogenic germ cells from systemic immune attack, and local innate immunity is important in preventing testicular ...
  57. [57]
    Fas Ligand-Induced Apoptosis as a Mechanism of Immune Privilege
    Inflammatory cells entering the anterior chamber of the eye in response to viral infection underwent apoptosis that was dependent on Fas (CD95)-Fas ligand (FasL) ...
  58. [58]
    Sympathetic Ophthalmia - StatPearls - NCBI Bookshelf - NIH
    The exact pathogenesis of sympathetic ophthalmia is unclear. The immune-privileged status of the eye occurs after a penetrating injury and exposure of ...Introduction · Pathophysiology · Treatment Planning · Staging
  59. [59]
    Ocular immunosuppressive microenvironment and novel drug ...
    This review discusses mechanisms of ocular immune privilege, followed by an overview of uveitis treatments and ongoing clinical trials.
  60. [60]
    Mesenchymal stromal cells for the treatment of ocular autoimmune ...
    Mesenchymal stem cells: immune evasive, not immune privileged. Nat ... stem/stromal cell-induced immune tolerance in mice with experimental autoimmune uveitis.
  61. [61]
    Recent advances in mesenchymal stem cell therapy for multiple ...
    Mesenchymal stem cell (MSC) therapy offers a promising alternative, leveraging its immunomodulatory, neuroprotective, and regenerative capabilities.4.4. 1 Exosomes · 5 Clinical Trials · Table 2
  62. [62]
    Ocular Immune Privilege and Ocular Melanoma: Parallel ... - Frontiers
    The roots of immune privilege reach back two centuries to an observation made by the Dutch ophthalmologist van Dooremaal (1873). In an attempt to identify the ...
  63. [63]
    Ocular Immune Privilege and Ocular Melanoma - PubMed Central
    Jun 13, 2012 · The roots of immune privilege reach back two centuries to an observation made by the Dutch ophthalmologist van Dooremaal (1873). In an attempt ...
  64. [64]
    Centennial review of corneal transplantation - PubMed
    Abstract One hundred years ago, on 7 December 1905, Dr Eduard Zirm performed the world's first successful human corneal transplant.Missing: early 1900s
  65. [65]
    A brief history of corneal transplantation: From ancient to modern - NIH
    Indeed, the first successful human corneal transplant was not performed by Eduard Zirm until 1905.Missing: 1900s | Show results with:1900s
  66. [66]
    Ocular immune privilege - PubMed - NIH
    It has been over 60 years since the phrase immune privilege was used by Sir Peter Medawar to describe the lack of an immune response against allografts ...Missing: 1948 paper
  67. [67]
    Peter Medawar – Nobel Lecture - NobelPrize.org
    Nobel Lecture, December 12, 1960. Immunological Tolerance. “Immunological tolerance” may be described as a state of indifference or non-reactivity towards a ...
  68. [68]
    Medawar's legacy to cellular immunology and clinical transplantation
    Medawar's earlier research had focused on the rejection of skin grafts by burns ... His own earlier work using outbred rabbits, and George Snell and Peter ...