Fact-checked by Grok 2 weeks ago

FOXP3

FOXP3 is a that encodes the forkhead box P3 (Foxp3) protein, a member of the forkhead/winged-helix family of transcription factors crucial for the development, maintenance, and suppressive function of regulatory T cells (Tregs). These + Tregs play a pivotal role in establishing and preserving , thereby preventing and excessive inflammatory responses while allowing effective immunity against pathogens. The FOXP3 gene is located on the at cytogenetic position Xp11.23 in humans. The Foxp3 protein features a conserved forkhead at its , which enables it to bind specific DNA sequences and regulate the transcription of target genes involved in T cell homeostasis and immune suppression. As a master regulator, Foxp3 acts both as a transcriptional activator and , interacting with other factors like NF-AT to inhibit pro-inflammatory production, such as interleukin-2 (IL-2), and to modulate metabolic pathways in Tregs, including and . Mutations in FOXP3 lead to immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome, a rare but severe X-linked recessive disorder characterized by early-onset affecting multiple endocrine glands, the gut, and other tissues, often resulting in fatal outcomes without intervention. In mice, Foxp3 mutations cause the Scurfy , mirroring IPEX with systemic autoimmunity and lymphoproliferation. Beyond its canonical role in Tregs, Foxp3 expression has been detected in other cell types, including some tumor cells, where it may function as a tumor suppressor by inhibiting and promoting in carcinomas such as those of the breast, , and . Research as of 2025 highlights post-translational modifications of Foxp3, such as and ubiquitination, as key regulators of its stability and activity, alongside influences from non-coding RNAs like miR-125b and a novel non-coding FOXP3 transcript isoform, offering potential avenues for therapeutic modulation in autoimmune diseases and , including CRISPR-mediated FOXP3 knockout to enhance function.

History and Discovery

Gene Identification

The FOXP3 was first identified in 2001 through genetic studies of patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, a severe autoimmune disorder characterized by early-onset , eczema, and gastrointestinal issues. Researchers in the Bennett laboratory, including Christopher L. Bennett and colleagues, discovered that mutations in FOXP3 were responsible for IPEX by performing linkage analysis and sequencing in affected families, revealing hemizygous mutations in male patients that disrupted the 's function. This finding established FOXP3 as a critical X-linked involved in immune . In parallel, that same year, Mary E. Brunkow, Fred Ramsdell, and their team at Darwin Molecular Corporation identified the mouse ortholog of FOXP3 as the causative gene for the scurfy mutation, an X-linked recessive condition in mice leading to fatal lymphoproliferation and multi-organ , using scurfy mice obtained from the Oak Ridge National Laboratory. Through positional cloning and functional assays, they demonstrated that loss-of-function mutations in the Foxp3 gene resulted in the scurfy phenotype, mirroring human IPEX and highlighting evolutionary conservation. These concurrent discoveries in humans and mice provided the initial characterization of FOXP3 as a forkhead box essential for preventing . The human FOXP3 gene was mapped to the at locus Xp11.23 and fully sequenced shortly after its identification, revealing an 11-exon structure spanning approximately 14 kb with a coding sequence encoding a 431-amino-acid protein. Annotation efforts, including those documented in the (OMIM) database, confirmed its role as a member of the FOXP family and detailed the spectrum of mutations linked to IPEX. The groundbreaking contributions of these early studies were recognized in 2025 when the in Physiology or Medicine was awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi for their discoveries concerning FOXP3's role in function and the prevention of . This accolade underscored the gene's pivotal importance in maintaining immune homeostasis through regulatory T cells. The link between FOXP3 and regulatory T cells (Tregs) was established through pivotal studies in the early 2000s, revealing FOXP3 as the master transcription factor governing Treg development and function. Building on Shimon Sakaguchi's earlier work in the 1990s identifying CD4+CD25+ Tregs as suppressors of autoimmunity, researchers in 2003 demonstrated that Foxp3 expression is highly specific to CD4+CD25+ Tregs in mice, marking these cells as a distinct lineage responsible for maintaining immune tolerance and preventing autoimmunity. This finding built on prior observations of CD25+ Tregs but pinpointed Foxp3 as their defining genetic marker, with its absence leading to uncontrolled T cell proliferation and systemic inflammation. Further experiments confirmed FOXP3's causal role in Treg suppressive activity via studies. Retroviral transduction of Foxp3 into naïve + T cells in models induced a phenotypic and functional conversion to Treg-like suppressors, capable of inhibiting effector T cell responses both and . These transformed cells expressed typical Treg markers and suppressed immune responses similarly to natural Tregs, establishing FOXP3 as sufficient to reprogram conventional T cells into regulatory phenotypes. Knockout studies provided definitive evidence of FOXP3's necessity for Treg differentiation and function. In scurfy mice, a natural disrupting the Foxp3 results in the complete absence of functional CD4+CD25+ Tregs, leading to fatal multi-organ shortly after birth. Complementary targeted Foxp3 in mice recapitulated this , confirming that FOXP3 is indispensable for Treg generation in the and their suppressive capacity in the periphery. In humans, mutations in the X-linked FOXP3 cause IPEX syndrome, characterized by Treg deficiency and severe immune dysregulation, mirroring the murine findings. FOXP3 exhibits strong evolutionary conservation across vertebrate species, including mammals and birds, correlating with the presence of FOXP3-dependent Tregs as a key mechanism for immune homeostasis. Sequence analysis shows high identity in the FOXP3 protein among primates and other mammals, with orthologs identified in avian species such as chickens, underscoring its role in Treg-mediated tolerance across vertebrates.

Molecular Biology

Gene Structure

The FOXP3 is located on the short arm of the X chromosome at cytogenetic band Xp11.23, spanning approximately 14.3 kb from positions 49,250,438 to 49,264,710 (GRCh38.p14 assembly). It consists of 12 s, with the first being entirely non-coding and the remaining 11 encoding the protein, while portions of exon 2 and the 3' end of exon 12 are also untranslated. Exon-intron boundaries are highly conserved between and orthologs. The FOXP3 promoter, situated upstream of the transcription start site (-511 to +176 bp), features core elements including a GC box at -138 bp, a at -44 bp, and a at -218 bp, which facilitate binding of transcription factors such as AP-1, NFAT, Sp1, and Sp3 to initiate basal transcription. Tissue-specific expression, particularly in regulatory T cells (Tregs), is tightly controlled by three intronic conserved non-coding sequences (CNS) acting as enhancers: CNS1 (~2 kb downstream of the transcription start site in 1) serves as a TGF-β-responsive element that recruits SMAD3 and NFAT to promote induced Treg ; CNS2 (~4 kb downstream, also in 1) stabilizes long-term FOXP3 expression in mature Tregs by binding factors like RUNX1, Ets-1, and FOXP3 itself; and CNS3 (~7 kb downstream in 2) functions as a element responsive to TCR stimulation via c-Rel binding, enabling initial opening for FOXP3 induction. These CNS elements collectively ensure Treg-restricted expression by integrating signals from cytokines and T cell receptors. Alternative splicing of FOXP3 transcripts generates multiple isoforms, primarily differing in the inclusion of exons 2 and 7, without causing frameshifts. The full-length isoform (FOXP3fl, NM_014009.4) includes all exons and constitutes 20-30% of FOXP3 in CD4+ CD25+ Tregs, featuring a nuclear export signal in exon 2 that supports IL-10 production via STAT3 interaction and robust suppressive function. The predominant isoform FOXP3Δ2 (lacking exon 2, ~70% of total) remains nuclear-localized, retains core suppressive activity but exhibits reduced stability and impaired STAT3-mediated functions compared to the full-length form. Less common variants include FOXP3Δ7 (lacking exon 7, which encodes a repressive domain) and the double-mutant FOXP3Δ2Δ7 (~1-3% of total), both of which diminish Treg suppressive capacity; the latter acts as a dominant-negative inhibitor, promoting pro-inflammatory Th17 differentiation instead. These isoforms modulate Treg plasticity and function, with full-length FOXP3 being essential for stable immunosuppression. A novel non-coding FOXP3 transcript isoform has been identified, potentially involved in regulatory mechanisms for Treg biology and associated with disease contexts. Epigenetic modifications further govern FOXP3 expression, notably through at the Treg-specific demethylated region (TSDR), a CpG-rich island within CNS2. In mature Tregs, demethylation of the TSDR during thymic differentiation stabilizes FOXP3 transcription by facilitating binding of transcription factors like FOXP3 and RUNX1, preventing silencing in peripheral tissues and ensuring heritable Treg identity. Hypermethylation of this region in conventional T cells or induced Tregs correlates with unstable or absent FOXP3 expression, while demethylating agents like 5-azacytidine can ectopically activate it. This mechanism underscores the epigenetic barrier to non-Treg FOXP3 expression.

Protein Structure

FOXP3 is a 431-amino acid protein with a calculated molecular weight of approximately 47 kDa in humans. The protein features several key structural domains that contribute to its transcriptional regulatory function. The N-terminal region contains a proline-rich domain (amino acids 1–97), which acts as a repression domain involved in recruiting co-repressors to silence target gene expression. Adjacent to this is a central region encompassing a C2H2-type zinc finger domain (amino acids 105–134) and a leucine zipper (Zip) domain (amino acids 193–229), which facilitate protein-protein interactions, including homodimerization and heterocomplex formation with other transcription factors. The C-terminal forkhead (FKH) domain (amino acids 335–428) is a winged-helix DNA-binding motif conserved across the FOX family of transcription factors. Crystallographic studies have elucidated the structure of the FOXP3 FKH domain, revealing its ability to bind DNA as a domain-swapped dimer. In this conformation, two FOXP3 monomers exchange their recognition helices to contact adjacent DNA sites, enabling cooperative binding to consensus sequences such as 5'-GTAAACAA-3'. The leucine zipper domain further supports dimerization by forming a coiled-coil structure that stabilizes these interactions, promoting enhanced affinity for target DNA elements. Recent structural analyses as of 2025 have shown that FOXP3 forms ultrastable and versatile multimeric ensembles on nucleosomal DNA, capable of bridging 2–4 DNA duplexes, with nucleosomes facilitating assembly through local DNA bending to enhance chromatin interactions and precise gene regulation in Tregs. This architectural arrangement allows FOXP3, primarily expressed in regulatory T cells, to exert precise control over gene transcription.

Physiological Role

Development of Regulatory T Cells

The development of thymic regulatory T cells (tTregs), also known as natural Tregs, occurs in the thymus where FOXP3 expression is induced in CD4+ single-positive thymocytes. This process requires coordinated signals from the T cell receptor (TCR) and interleukin-2 (IL-2) pathways. Strong TCR engagement by self-antigens with intermediate affinity selects thymocytes for the Treg lineage, while IL-2 signaling through its receptor activates signal transducer and activator of transcription 5 (STAT5), which directly binds to conserved non-coding sequences in the Foxp3 promoter and enhancer regions to drive FOXP3 transcription. Without IL-2/STAT5 signaling, Foxp3 expression fails, leading to a severe reduction in tTreg numbers and onset of autoimmunity. In contrast, peripheral regulatory T cells (pTregs) arise extrathymically from naive + T cells in peripheral lymphoid tissues or mucosal sites under tolerogenic conditions. FOXP3 expression in these cells is primarily induced by transforming growth factor-β (TGF-β), which activates SMAD proteins that bind to Foxp3 regulatory elements, promoting its transcription in TCR-stimulated naive T cells. The metabolite further enhances this process by upregulating TGF-β signaling and inhibiting pro-inflammatory production, thereby favoring pTreg over effector T cell fates in environments like the gut mucosa. pTregs are crucial for maintaining tolerance to environmental antigens and commensal microbes. FOXP3 plays a central role in maintaining Treg stability by enforcing an epigenetic landscape that resists toward effector phenotypes. High FOXP3 levels promote at the Foxp3 locus, ensuring sustained expression and preventing loss of suppressive identity even under inflammatory pressures. This stability is evident , where mature Tregs largely self-renew without significant conversion to pro-inflammatory cells, thereby preserving immune . The dosage of FOXP3 quantitatively influences Treg development and function, with higher expression correlating to increased Treg numbers and enhanced suppressive potency. In models with reduced Foxp3 levels, such as those expressing lower amounts due to genetic attenuation, Treg populations are smaller, and their ability to suppress effector T cell proliferation is diminished, leading to partial loss of tolerance. This dosage sensitivity underscores FOXP3's role as a rheostat for Treg fitness, where even moderate reductions impair lineage commitment and regulatory capacity. The critical physiological role of FOXP3 in the development and function of regulatory T cells was recognized by the 2025 in Physiology or Medicine, awarded on October 6, 2025, for discoveries concerning .

Immune Suppression Mechanisms

FOXP3, the master transcription factor in regulatory T cells (Tregs), directly the expression of pro-inflammatory genes such as IL-2 and IFN-γ by functioning as a transcriptional that recruits histone deacetylases (HDACs), including HDAC7, to target gene promoters, thereby promoting chromatin condensation and inhibiting transcription initiation. This mechanism is essential for dampening effector T cell responses, as demonstrated in studies where FOXP3 overexpression in conventional T cells led to reduced IL-2 and IFN-γ production through HDAC-dependent deacetylation of at these loci. In FOXP3-deficient models, such as scurfy mice, unchecked expression of these cytokines correlates with severe , underscoring the repressive role of FOXP3 in maintaining . Beyond direct gene repression, FOXP3 interacts with nuclear factor of activated T cells (NFAT) and NF-κB to inhibit effector T cell activation and cytokine production. FOXP3 physically associates with NFAT and NF-κB in the nucleus, blocking their transactivation domains without disrupting DNA binding, which prevents the induction of pro-inflammatory programs in activated T cells. This cooperative complex converts T cell activation signals into a suppressive program, as evidenced by structure-guided mutations in FOXP3 that disrupt NFAT binding and abolish Treg suppressive function in vitro and in vivo models of autoimmunity. Complementary studies show that FOXP3-NFAT interactions specifically repress IL-2 promoter activity, limiting clonal expansion of effector T cells. Treg-mediated suppression also occurs through contact-dependent mechanisms involving CTLA-4, which is upregulated in FOXP3-expressing cells and engages CD80/CD86 on antigen-presenting cells to trans-endocytose these ligands, depriving effector T cells of co-stimulatory signals. This CTLA-4-mediated inhibition reduces dendritic cell maturation and T cell priming, with seminal experiments in CTLA-4-deficient Tregs demonstrating restored effector responses and exacerbated inflammation. Concurrently, FOXP3 promotes the secretion of immunosuppressive cytokines like IL-10 and TGF-β by Tregs, which inhibit effector T cell proliferation and cytokine release in a paracrine manner; for instance, Treg-derived IL-10 limits IFN-γ production by Th1 cells, while surface-bound TGF-β enforces tolerance in mucosal tissues. At the metabolic level, FOXP3 enhances Treg fitness and suppressive capacity by downregulating c-Myc, a key driver of , thereby shifting metabolism toward and sustaining long-term Treg survival in inflammatory environments. This c-Myc repression prevents exhaustion and maintains FOXP3 stability, as shown in Treg-specific c-Myc knockout models where enhanced suppressive function correlates with reduced glycolytic flux. Additionally, FOXP3-expressing Tregs generate extracellular through coordinated expression of CD39 (ectonucleoside triphosphate diphosphohydrolase-1) and CD73 (ecto-5'-nucleotidase), which hydrolyze ATP to ; this activates A2A receptors on effector T cells, suppressing their and while promoting Treg dominance. In vivo, CD39/CD73-deficient Tregs exhibit impaired suppression in models of transplant , highlighting adenosine's role in contact-independent inhibition.

Pathological Implications

Autoimmune Disorders

Mutations in the FOXP3 gene cause immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, a rare but severe autoimmune disorder primarily affecting males due to its X-linked inheritance. This condition arises from loss-of-function mutations that impair the development and suppressive function of regulatory T cells (Tregs), leading to uncontrolled autoimmune attacks on multiple organs, including the pancreas, thyroid, intestines, and skin, often manifesting as fatal multi-organ autoimmunity in infancy. For instance, the R397W missense mutation in the forkhead (FKH) domain disrupts FOXP3's DNA-binding ability, resulting in defective Treg physiology and widespread inflammation. The discovery of FOXP3's role in IPEX syndrome was recognized by the 2025 Nobel Prize in Physiology or Medicine. The scurfy mouse model, characterized by a spontaneous Foxp3 , recapitulates key features of IPEX syndrome and has been instrumental in elucidating FOXP3's role in . These mice exhibit rapid lymphoproliferation driven by unrestrained CD4+ T cell activation, culminating in severe multi-organ infiltration and early death within 3-4 weeks of age, highlighting the essential suppressive function of Foxp3+ Tregs in maintaining immune . Studies of scurfy mice have demonstrated that the absence of functional Foxp3 leads to hyperactivation of effector T cells and overproduction of pro-inflammatory cytokines, underscoring the genetic parallels to human FOXP3 deficiencies. In more common autoimmune diseases, polymorphisms in the FOXP3 gene have been associated with reduced Treg numbers or impaired suppressive capacity, contributing to disease susceptibility. Similarly, FOXP3 polymorphisms like -3279C/A and -924A/G are linked to higher susceptibility to systemic lupus erythematosus (SLE), where they diminish Treg-mediated control of autoreactive B cells and immune complexes. In rheumatoid arthritis (RA), variants such as rs3761548 influence Treg frequency and correlate with disease severity, promoting synovial inflammation through inadequate suppression of Th17 responses. Therapeutic strategies targeting FOXP3+ Tregs offer promise for mitigating autoimmune disorders by enhancing their immunosuppressive activity. Low-dose interleukin-2 (IL-2) selectively expands and activates FOXP3+ Tregs without overly stimulating effector T cells, leading to improved clinical outcomes in trials for conditions like SLE, , and as a model for . Clinical studies have shown that this approach increases circulating Treg levels, restores immune balance, and reduces disease activity, with ongoing research exploring its efficacy in broader autoimmune contexts.

Role in Cancer

FOXP3 plays a in cancer through its expression in regulatory T cells (Tregs) and, paradoxically, in non-Treg tumor cells. In the , high densities of FOXP3+ tumor-infiltrating Tregs are frequently associated with poor clinical outcomes, as they suppress anti-tumor immune responses. For instance, in , elevated FOXP3+ Treg infiltration correlates with reduced overall survival (odds ratio 2.15, 95% 1.34–3.44) and disease-free survival (odds ratio 2.13, 95% 1.18–3.84). Similarly, in ovarian carcinoma, particularly in non-advanced stages, increased FOXP3+ Treg density predicts unfavorable and higher risk of recurrence, though outcomes can vary by disease stage. The immunosuppressive mechanisms of FOXP3+ Tregs in cancer involve inhibition of effector T cell activation and proliferation. FOXP3 drives Treg expression of checkpoint molecules such as PD-1 and CTLA-4, which dampen anti-tumor immunity by limiting and promoting (IDO) secretion. Additionally, Tregs secrete inhibitory cytokines like IL-10 and TGF-β, which deplete IL-2 availability and induce in effector T cells, thereby fostering tumor immune escape. These actions collectively reduce the of endogenous anti-tumor responses and contribute to resistance against immunotherapies. Beyond Tregs, FOXP3 expression in non-immune cancer cells exhibits oncogenic potential, particularly in promoting epithelial-mesenchymal transition (EMT), metastasis, and chemoresistance. In breast and prostate cancers, aberrant FOXP3 upregulation in tumor cells enhances migratory and invasive capabilities by activating pathways like Wnt/β-catenin, leading to downregulation of E-cadherin and upregulation of N-cadherin, vimentin, and matrix metalloproteinases. This FOXP3-driven EMT facilitates distant metastasis and confers resistance to chemotherapeutic agents, correlating with poorer patient survival. Recent advances highlight therapeutic targeting of the FOXP3 pathway to bolster . In 2025 studies, disruption of the HuR-FOXP3 axis—where the HuR stabilizes FOXP3 mRNA in Tregs—has shown promise; HuR inhibitors like KH-3 destabilize FOXP3 transcripts, impair Treg suppressive function, and enhance anti-tumor immunity without broad autoimmune risks. Such targeted approaches aim to reprogram tumor-infiltrating Tregs, improving responses to PD-1/CTLA-4 blockade in solid tumors.

Other Diseases

FOXP3 polymorphisms have been associated with the progression of Chagas disease, particularly influencing the transition to indeterminate clinical forms. A 2025 study identified that the heterozygous CT genotype of the rs3761548 polymorphism in the FOXP3 gene is twice as prevalent in women with the indeterminate form of chronic Chagas disease compared to those with cardiac involvement, suggesting a protective role against severe progression by modulating regulatory T cell (Treg) function. In infection, FOXP3-expressing Tregs contribute to viral latency through suppressive mechanisms that inhibit HIV-specific immune responses. FOXP3 directly suppresses HIV-1 long-terminal repeat (LTR) transcription in + T cells, promoting the establishment and maintenance of latent reservoirs, which complicates antiretroviral therapy eradication efforts. Dysregulation of FOXP3 expression impairs Treg function in allergic conditions such as , where reduced FOXP3+ Tregs in fluid correlate with heightened Th2-driven and airway hyperresponsiveness. Similarly, in (IBD), diminished FOXP3 levels in colonic Tregs exacerbate mucosal , leading to barrier dysfunction. Therapeutic strategies targeting peripheral Treg (pTreg) induction, such as through TGF-β and IL-2 signaling, show promise in restoring FOXP3 stability and suppressing aberrant immune responses in both and IBD models. Recent investigations have uncovered pathways modulating FOXP3 stability during , including the cGAS-STING axis. In 2025 research on models, UV-induced activation of cGAS-STING signaling triggered cytosolic DNA sensing, leading to type I production that enhanced FOXP3 expression in Tregs via acetylation and STING-MAPK-CREB pathways, thereby supporting immunosuppressive Treg function in skin immunity. This pathway highlights potential therapeutic targets for modulating -associated Treg activity in chronic conditions. FOXP3+ Tregs in adipose tissue play a critical role in obesity-related insulin resistance by regulating local inflammation. In visceral adipose tissue of obese individuals, expanded FOXP3+ Treg populations initially suppress pro-inflammatory macrophage activation to maintain metabolic homeostasis, but chronic obesity leads to Treg dysfunction, exacerbating adipose inflammation and impairing insulin signaling in adipocytes. Studies demonstrate that enhancing adipose-resident Treg function can alleviate insulin resistance by reducing cytokine-driven inflammation and promoting beiging of white adipose tissue.

References

  1. [1]
    50943 - Gene ResultFOXP3 forkhead box P3 [ (human)] - NCBI
    Aug 19, 2025 · According to this review, FoxP3 is an important tumor suppressor gene in carcinomas and has putative cancer suppressor gene function in ...
  2. [2]
    Advances in Foxp3+ regulatory T cells (Foxp3+ Treg) and key ...
    Jun 11, 2024 · This review primarily focuses on the properties and capabilities of Foxp3 and Foxp3+Treg. It emphasizes the advancement of research on the ...
  3. [3]
    FoxP3 in Treg cell biology: a molecular and structural perspective
    FoxP3 plays a dual role as both a transcriptional activator and repressor of its target genes in Treg cells 21. FoxP3 heterodimerize with FoxP1, which enforces ...
  4. [4]
    FoxP3: A Life beyond Regulatory T Cells - PMC - NIH
    Oct 2, 2008 · The FoxP3/JM2 gene was first cloned in 2000 because its mutation caused fatal autoimmune disease with early lethality in the Scurfy mice [1] and ...
  5. [5]
    The dual role of the X‐linked FoxP3 gene in human cancers - PMC
    The FoxP3 gene has important functions as a tumor suppressor gene in human carcinomas and recent data suggests it plays a role in melanoma as well.
  6. [6]
    The immune dysregulation, polyendocrinopathy, enteropathy, X ...
    The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001 Jan;27(1):20-1. doi: ...
  7. [7]
    Disruption of a new forkhead/winged-helix protein, scurfin ... - PubMed
    Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001 Jan;27(1):68- ...
  8. [8]
    Entry - *300292 - FORKHEAD BOX P3; FOXP3 - OMIM - (OMIM.ORG)
    FOXP3, a member of the fork-winged helix family of transcription factors, plays an important role in the development and function of naturally occurring CD4 ...Missing: annotation | Show results with:annotation
  9. [9]
    Press release: The Nobel Prize in Physiology or Medicine 2025
    Oct 6, 2025 · Mary Brunkow, Fred Ramsdell and Shimon Sakaguchi are awarded the Nobel Prize in Physiology or Medicine 2025 for their fundamental discoveries ...
  10. [10]
    Control of Regulatory T Cell Development by the Transcription ...
    Here we show that Foxp3, which encodes a transcription factor that is genetically defective in an autoimmune and inflammatory syndrome in humans and mice,
  11. [11]
    Disruption of a new forkhead/winged-helix protein, scurfin, results in ...
    Nevertheless, the discovery that mutations in human FOXP3 lead to a syndrome very similar to scurfy (see Wildin et al. ... Brunkow, Eric W. Jeffery ...
  12. [12]
    [PDF] Evolutionary analysis of the genomic region of FOXP3 in vertebrates
    The FOXP3 protein is highly conserved across mammalian species with highest identity and coverage among the various primate species.Missing: lacking Tregs
  13. [13]
    Phylogenomic Evidence for the Existence of Avian FoxP3 | PLOS One
    FoxP3 is believed to perform a regulatory role in all the vertebrate species in which it has been detected. The prevailing view is that FoxP3 is absent in birds ...
  14. [14]
    The molecular mechanisms of Foxp3 gene regulation - PMC
    The Foxp3 gene is found on the X chromosome. Critical for the initiation and regulation of Foxp3 transcription is the Foxp3 conserved promoter sequence, located ...
  15. [15]
    Transcriptional regulation of FOXP3 requires integrated activation of ...
    May 9, 2017 · In addition to the promoter region three conserved noncoding sequences (CNSs) in the FOXP3 locus, viz. CNS1, CNS2 and CNS3, also regulate and ...
  16. [16]
    Role of conserved non-coding DNA elements in the Foxp3 gene in ...
    Feb 11, 2010 · We proposed that conserved non-coding DNA sequence (CNS) elements at the Foxp3 locus encode information defining the size, composition and stability of the T ...
  17. [17]
    Alternative Splicing of FOXP3—Virtue and Vice - Frontiers
    This review summarizes current knowledge about alternative splicing of FOXP3 and the specific functions of FOXP3 isoforms in health and disease.
  18. [18]
  19. [19]
    FOXP3 - Forkhead box protein P3 - Homo sapiens (Human) - UniProt
    Gene Ontology (GO) annotations organized by slimming set. Aspect, Term ... Chromosome X. Organism-specific databases. AGR · HGNC:6106 · HGNC · HGNC:6106 FOXP3.
  20. [20]
    FoxP3 in T reg cell biology: a molecular and structural perspective
    Aug 6, 2019 · Structurally, FoxP3 contains three major domains: a proline-rich N-terminal domain (1–97 aa) responsible for transcriptional activation and ...
  21. [21]
    Molecular and biological role of the FOXP3 N-terminal domain in ...
    These mutations are dominantly located at three domains of FOXP3, the forkhead domain responsible for DNA binding, the zinc finger and leucine zipper region ...
  22. [22]
    FOXP3 and Tip60 Structural Interactions Relevant to IPEX ... - Frontiers
    Feb 2, 2021 · In humans, FOXP3 encodes a 431 amino acid (aa) containing protein with a molecular weight of 47.25 KDa. It consists of four distinct domains ...
  23. [23]
    DNA binding by FOXP3 domain-swapped dimer suggests ...
    In this study, we determined the crystal structure of the FOXP3 forkhead domain bound to DNA. ... FOXP3 consensus binding sequence (AAACA). ChIP-qPCR ...
  24. [24]
    ChIP-on-chip analysis identifies IL-22 as direct target gene of ...
    Dec 17, 2012 · ... FOXP3 binding to DNA [8]. The human FOXP3 gene consists of 11 exons encoding for various splice variants. Besides the full-length 431 aa ...<|control11|><|separator|>
  25. [25]
    Stability of the Regulatory T Cell Lineage in Vivo - Science
    Sep 24, 2010 · Our results suggest that self-renewal of mature T reg cells serves as a major mechanism of maintenance of the T reg cell lineage in adult mice.
  26. [26]
    Foxp3 interacts with nuclear factor of activated T cells and NF-κB to ...
    Apr 5, 2005 · Furthermore, complementation of Foxp3 in scurfy-derived T cells lowers the NFAT and NF-κB transcriptional activity to the physiological level.
  27. [27]
  28. [28]
  29. [29]
    Immunometabolism of regulatory T cells | Nature Immunology
    May 19, 2016 · Furthermore, the glycolytic enzyme enolase-1 can repress the FOXP3 splice variant containing exon 2 (E2) in human Treg cells, and its engagement ...Abstract · Main · T Cell Bioenergetics And...
  30. [30]
    Analyses of a Mutant Foxp3 Allele Reveal BATF as a ... - PubMed
    Aug 15, 2017 · The I363V and R397W mutations were loss-of-function mutations, causing multi-organ inflammation by globally compromising Treg cell physiology.<|separator|>
  31. [31]
    FOXP3 polymorphisms in type 1 diabetes and coeliac disease
    Our results indicated an association between FOXP3 and T1D (global p=0.004) and a possible interaction between FOXP3 and the HLA-DR3-DQ2 susceptibility ...
  32. [32]
    FOXP3 gene polymorphisms increase the risk of systemic lupus ...
    Jun 7, 2024 · This paper investigated whether the three SNPs in the FOXP3 gene (-3279 C/A, -924 A/G and -6054 del/ATT) are associated with systemic lupus erythematosus (SLE) ...
  33. [33]
    Genetic polymorphisms of Foxp3 in patients with rheumatoid arthritis
    Current findings indicated that the Foxp3 genetic polymorphism and the Foxp3 protein level may be associated with susceptibility to RA in the Polish ...
  34. [34]
    Low-Dose IL-2 Therapy in Transplantation, Autoimmunity ... - PubMed
    Dec 1, 2019 · Administration of low-dose IL-2 is shown to be a promising approach to prevent allograft rejection and to treat autoimmune and inflammatory conditions in ...
  35. [35]
    Low-dose interleukin-2 fosters a dose-dependent regulatory T cell ...
    We showed that low-dose interleukin-2 (ld-IL-2) safely expands/activates Tregs in patients with AID, such HCV-induced vasculitis and Type 1 Diabetes (T1D).Missing: clinical | Show results with:clinical
  36. [36]
    Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in ...
    Oct 14, 2015 · In high grade or advanced ovarian cancer, high numbers of FoxP3+ Tregs infiltrating were found to be associated with improved survival ...
  37. [37]
    Prognostic role of FOXP3+ regulatory T cells infiltrating human ...
    ... high density of FOXP3+ tumor-infiltrating cells is associated with unfavorable clinical prognosis ... FoxP3 is associated with poor prognosis in ovarian cancer.
  38. [38]
    Analysis of FoxP3+ T-Regulatory Cells and CD8+T-Cells in Ovarian ...
    Treg cells are able to modulate the anti- tumor response of CD8+ effector T-cells and are associated with poor prognosis in ovarian carcinoma [20]–[24].
  39. [39]
    Regulatory T cells in solid tumor immunotherapy: effect, mechanism ...
    Apr 11, 2025 · Hypoxia in solid tumors upregulates HIF-1α and FoxP3, promoting Treg differentiation and TGF-β-mediated suppression. Clinical Interventions ...
  40. [40]
    Review The expression of FOXP3 and its role in human cancers
    In some types of human cancers, the expression of FOXP3 is upregulated, and it can promote the development of cancers, leading to a poor prognosis.Missing: chemoresistance | Show results with:chemoresistance
  41. [41]
    Forkhead Box Transcription Factors: Double-Edged Swords in Cancer
    In various cancers, including ovarian, breast, and prostate cancer, FOX factors are implicated in cancer initiation, progression, and chemoresistance (6–9). In ...Cancer Metabolism · Pioneering Factors · Future Directions<|separator|>
  42. [42]
    FOXP3 promotes tumor growth and metastasis by activating Wnt/β ...
    Jul 17, 2017 · FOXP3 overexpression significantly induced cell proliferation, migration, and invasion, whereas its inhibition impaired its oncogenic function.
  43. [43]
    HuR ablation destabilizes Foxp3 mRNA and impairs regulatory T ...
    Sep 25, 2025 · Conclusion: These findings together suggest that ablation of HuR in Tregs disrupts Foxp3 expression and Treg function, likely through ...
  44. [44]
    FOXP3 gene polymorphisms are associated with indeterminate ...
    Jun 19, 2025 · This study investigated FOXP3 gene polymorphism (rs3761548) in patients with Chronic Chagas disease and the association between FOXP3 ...Missing: progression | Show results with:progression
  45. [45]
    Regulatory T cells: the ultimate HIV reservoir? - PMC
    In vitro studies provide evidence that FOXP3 expression can inhibit HIV-1 long-terminal repeat transcription, which could favour the establishment of latently ...
  46. [46]
    Regulatory T cells and their roles in immune dysregulation and allergy
    These data suggest that FOXP3 supports the maintenance of an immunosuppressive milieu. For a long time, FOXP3 was considered to be a master regulator for the ...
  47. [47]
    Regulatory T-cell therapy in Crohn's disease: challenges and ... - Gut
    21 The significance of this lies in the epigenetic differences in the Foxp3 locus rendering pTregs less stable and more likely to demonstrate plasticity toward ...
  48. [48]
    Photoaging: UV radiation-induced cGAS-STING signaling promotes ...
    Jun 20, 2025 · ... FOXP3 transcription factor, a master regulator of the immunosuppressive activity of Tregs. ... cGAS/STING inflammation during aging. Nat Commun 15 ...
  49. [49]
    Fat Treg cells: a liaison between the immune and metabolic systems
    Obesity is accompanied by chronic, low-grade inflammation of adipose tissue, which promotes insulin resistance and type-2 diabetes.
  50. [50]
    Tregs facilitate obesity and insulin resistance via a Blimp-1/IL-10 axis
    We show here that CD4+Foxp3+ regulatory T cells (Tregs) are a substantial source of IL-10 and that Treg-derived IL-10 can suppress adipocyte beiging.
  51. [51]
    The Science Behind the Nobel Prize
    Article detailing the affiliations of Brunkow and Ramsdell at Darwin Molecular Corporation during the 2001 FOXP3 discovery.
  52. [52]
    FOXP3 and scurfy: how it all began
    PubMed article confirming the origin of the scurfy mutation at Oak Ridge National Laboratory.