Fact-checked by Grok 2 weeks ago

NFAT

Nuclear Factor of Activated T-cells (NFAT) is a family of transcription factors that regulate in response to , primarily through by the phosphatase , enabling their translocation to the where they cooperate with other factors to control diverse cellular processes. Originally identified in activated T lymphocytes for their role in production, NFAT proteins are expressed across various cell types and are essential for immune responses, development, and adaptation to environmental cues. The NFAT family comprises five members: NFAT1 (also known as NFATc2), NFAT2 (NFATc1), NFAT3 (NFATc4), NFAT4 (NFATc3), and NFAT5 (TonEBP), with NFAT1–4 being calcium/calcineurin-dependent and sharing a conserved Rel domain for DNA binding to consensus sequences like (A/T)GGAAA, while NFAT5 operates independently and is constitutively nuclear. Activation occurs upon antigen receptor or other stimuli-induced calcium influx, which binds to activate ; this enzyme then dephosphorylates serine-rich regions in the NFAT regulatory , promoting nuclear import and interaction with partners such as AP-1 (Fos-Jun) to drive transcription of target genes. This process is reversible, as kinases like GSK-3 rephosphorylate NFAT, exporting it back to the , and is inhibited by immunosuppressive drugs such as cyclosporine A and FK506. In the , NFAT proteins are pivotal for T-cell development, activation, differentiation into effector subsets (e.g., Th1, Th2, Treg), and tolerance mechanisms, including the induction of anergy-associated genes like and ; they also influence expression in B cells, dendritic cells, cells, and natural killer T cells. Beyond immunity, NFAT regulates cardiac and differentiation, formation, neuronal development, osteogenesis, function, and responses to stress. Dysregulated NFAT activity contributes to pathologies, including autoimmune diseases, , and cancers such as lymphomas, breast, and pancreatic tumors, where it promotes proliferation, survival, migration, and angiogenesis. Additionally, NFAT modulates progression by controlling cyclins (e.g., , A2) and CDK inhibitors (e.g., p21, p15), as well as through targets like FasL (pro-apoptotic) and (anti-apoptotic).

Family and Structure

Members of the NFAT Family

The NFAT (Nuclear Factor of Activated T-cells) family comprises five principal members: NFAT1 (encoded by NFATC2), NFAT2 (encoded by NFATC1), NFAT3 (encoded by NFATC4), NFAT4 (encoded by NFATC3), and NFAT5 (also known as TONEBP, encoded by NFAT5). These proteins are transcription factors that regulate in response to cellular signals, primarily in immune and stress contexts. In humans, the genes are located as follows: NFATC1 on 18q23, NFATC2 on 20q13.2, NFATC3 on 16q22.1, NFATC4 on 14q12, and NFAT5 on 16q22.1. Their predicted molecular weights vary due to isoforms and post-translational modifications, but representative sizes include approximately 100 for NFAT1, 101 for NFAT2, 95 for NFAT3, 115 for NFAT4, and 166 for NFAT5.
MemberGeneChromosomal LocationApproximate Molecular Weight (kDa)
NFAT1NFATC220q13.2100
NFAT2NFATC118q23101
NFAT3NFATC414q1295
NFAT4NFATC316q22.1115
NFAT5NFAT516q22.1166
NFAT1 through NFAT4 are primarily inducible, with expression upregulated in immune cells such as T lymphocytes upon activation, whereas NFAT5 is constitutively expressed across various tissues and particularly responsive to changes, such as hyperosmotic in renal cells. Evolutionarily, all NFAT family members belong to the Rel superfamily and share a conserved (RHD) responsible for DNA binding and dimerization, though the RHD shows lower sequence conservation in NFAT compared to classical Rel/ proteins. They differ in their domains (TADs), which vary in length and composition, influencing transcriptional potency; notably, NFAT5 lacks the regulatory motifs for calcineurin-dependent that control the translocation of NFAT1-4. NFAT1 and NFAT2 were first identified in the late 1980s through studies of inducible nuclear complexes binding to promoters, such as IL-2, in activated T cells. NFAT5 was discovered in the late 1990s as a tonicity-responsive factor regulating osmotic stress genes in mammalian cells.

Protein Structure and Domains

NFAT proteins exhibit a modular characterized by a central Rel homology domain (RHD) and flanking regulatory and domains that govern their localization and activity. The RHD, spanning approximately 300 (e.g., residues 411–685 in NFATc2), is highly conserved across the family and consists of two subdomains: an N-terminal and a C-terminal domain involved in dimerization and interaction with inhibitory proteins. This domain enables sequence-specific binding to consensus DNA sites such as GGAAA and facilitates heterodimerization with other transcription factors. The (TAD) is primarily located at the and, to a lesser extent, the , serving to recruit co-activators like CBP/p300 for transcriptional enhancement. The N-terminal TAD contains intrinsically disordered regions that allow flexible interactions with partner proteins, contributing to the versatility of NFAT-mediated gene regulation. Unlike NFAT1–4, NFAT5 features distinct TAD organization adapted for osmotic stress responses. Regulation of NFAT localization is mediated by an N-terminal regulatory region rich in phosphorylation sites, including serine-rich motifs (SRR1 and SRR2) and SP-repeats (three serine-proline motifs: SP1, SP2, SP3). These motifs encompass up to 14 serine/proline-directed sites in NFAT1–4, targeted by kinases such as glycogen synthase kinase 3β (GSK3β) and (CK1), which maintain cytoplasmic retention in resting cells. Embedded within this region are a nuclear localization signal (NLS) and a (NES), whose exposure is modulated by status. NFAT5 notably lacks these serine motifs, rendering it insensitive to calcineurin-mediated . Structural insights into the RHD derive from crystal structures of NFAT-DNA complexes, revealing how the domain clamps onto DNA via β-strands and loops from the RHR-N subdomain, while the RHR-C supports dimer interfaces. For instance, the structure of the NFAT1 RHD bound to DNA (PDB: 1OWR) shows monomeric binding, but cooperative heterodimerization with NF-κB p50 on composite sites has been modeled based on Rel family homology, highlighting interprotein contacts in the RHR-C. These models underscore the RHD's role in both homotypic and heterotypic interactions essential for promoter specificity.

Activation and Signaling

Canonical Calcineurin-Dependent Pathway

The canonical calcineurin-dependent pathway represents the primary mechanism for activating nuclear factor of activated T cells (NFAT) proteins, particularly NFAT1 through NFAT4, in response to in immune cells. This pathway is initiated by antigen receptor stimulation on lymphocytes, such as the (TCR) or (BCR), which engages -presenting cells and activates receptor-associated protein tyrosine kinases. These kinases phosphorylate and activate phospholipase C-γ1 (PLC-γ1), leading to the hydrolysis of (PIP₂) into inositol 1,4,5-trisphosphate (IP₃) and diacylglycerol (DAG). IP₃ then binds to IP₃ receptors on the (ER), triggering the release of stored Ca²⁺ into the . This initial Ca²⁺ transient is amplified and sustained by store-operated Ca²⁺ entry (SOCE), where ER Ca²⁺ depletion activates stromal interaction molecule 1 (STIM1) on the ER membrane, which interacts with Orai1 channels on the plasma membrane to facilitate Ca²⁺ influx through Ca²⁺ release-activated Ca²⁺ (CRAC) channels. The elevated cytosolic Ca²⁺ binds to , forming a Ca²⁺- complex that allosterically activates (also known as 2B or PP2B), a serine/ composed of catalytic (CnA) and regulatory (CnB) subunits. Activated then dephosphorylates multiple serine residues within the N-terminal regulatory domain of NFAT1-4, specifically in the serine-rich region (SRR) and serine-proline (SP)-repeat motifs. This dephosphorylation induces a conformational change, exposing the nuclear localization signal (NLS) and promoting rapid nuclear import of NFAT via importin-β-mediated transport. Once in the nucleus, dephosphorylated NFAT drives transcription of target genes, but its activity is tightly regulated by opposing s that promote rephosphorylation and . Key among these are kinase 3 (GSK3), (CK1), and dual-specificity tyrosine phosphorylation-regulated kinase (DYRK), which sequentially phosphorylate NFAT upon declining Ca²⁺ levels, masking the NLS and exposing the (NES). This facilitates CRM1 (exportin 1)-dependent , recycling NFAT to the for rephosphorylation. Temporally, the pathway exhibits rapid dynamics: nuclear entry of NFAT occurs within minutes of Ca²⁺ elevation, enabling quick transcriptional responses, while and rephosphorylation follow Ca²⁺ cessation, often within tens of minutes, creating oscillatory cycles that fine-tune . This mechanism is predominantly active in lymphocytes, where sustained SOCE and signaling are essential for functions like production (e.g., IL-2). The net activation can be conceptually modeled as dependent on activity, which rises with Ca²⁺ influx, inversely proportional to the activity of Ca²⁺-inhibited opposing kinases: [\text{NFAT active}] \propto \frac{[\text{Ca}^{2+}] \cdot [\text{Calcineurin}]}{[\text{Ca}^{2+}-\text{inhibited kinases}]} This simplification highlights the balance between and activities in controlling NFAT localization.

Alternative Activation Pathways

In addition to the canonical -dependent , NFAT proteins can be activated through kinase-mediated events that promote nuclear translocation or enhance transcriptional activity. For instance, in cardiac myocytes, ERK/MAPK signaling phosphorylates NFAT2 (also known as NFATc1) at specific serine residues, facilitating its nuclear accumulation and contributing to hypertrophic independent of calcium flux. Similarly, in T lymphocytes, θ (PKCθ) initiates an alternative pathway by enhancing calcium mobilization and directly influencing NFAT nuclear import, thereby supporting production while partially circumventing full reliance during T cell receptor stimulation. NFAT5, the tonicity-responsive member of the family, is uniquely regulated by osmotic stress through (MAPK) pathways, particularly p38 and JNK, without requiring . Hypertonicity activates p38/JNK kinases, which phosphorylate NFAT5 to promote its nuclear translocation and induction of osmoprotective genes such as those encoding and sodium/myo-inositol . Recent studies have revealed that NFAT5 directly senses intracellular via its C-terminal prion-like domain (PLD), a ~450-amino-acid intrinsically disordered region that undergoes into condensates under elevated ionic conditions, recruiting coactivators like to drive the hypertonicity response. This mechanism ensures cellular adaptation in hypertonic environments, such as the , and highlights NFAT5's role in ionic homeostasis. Other non-canonical routes include (ROS)-induced activation, particularly for NFAT3 and NFAT4, where trigger their translocation to mitochondria and subsequent nuclear signaling in response to . In developmental contexts, Wnt signaling modulates NFAT via inhibition of glycogen synthase kinase 3β (GSK3β), which otherwise phosphorylates NFAT proteins leading to their proteasomal degradation; Wnt-mediated GSK3β suppression stabilizes and activates NFAT, influencing processes like vascular development. NFAT activation often integrates with other transcription factors through cross-talk, such as with or AP-1 to amplify inflammatory or hypertrophic responses in a context-dependent manner. For example, in T cells, NFAT and dynamically co-regulate downstream of TCR/ signaling, ensuring balanced immune activation. These interactions underscore the versatility of alternative pathways in fine-tuning NFAT function across tissues.

DNA Binding and Transcriptional Activity

NFAT proteins recognize specific DNA sequences through their Rel homology domain (RHD), which confers sequence-specific binding to a core consensus motif of (A/T)GGAAA found in the regulatory regions of target genes, including promoters of cytokines such as IL-2 and TNF-α. This motif often appears within composite elements, exemplified by the ARRE-2 site in the IL-2 promoter, where the NFAT core GGAAA is spaced by approximately 3-4 nucleotides from an adjacent AP-1 binding site (e.g., TTCC-like sequences in dyad-symmetric arrangements), enabling integrated regulation. The binding affinity of the isolated RHD to DNA is typically in the range of 10-100 nM, allowing for dynamic association that is enhanced in the context of multi-protein complexes. NFAT family members primarily bind DNA as monomers but can form homodimers in certain contexts, such as on κB-like sites (e.g., TGGAGTTCCC), and cooperate with Rel family proteins like c-Rel through binding to adjacent sites in enhancers, promoting cooperative occupancy and stability on DNA. Additionally, NFAT exhibits with other transcription factors, including the AP-1 heterodimer (Fos/) at composite NFAT:AP-1 elements and factors at synergistic sites, where direct protein-protein contacts via the RHD's N-terminal subdomain facilitate ternary complex formation and increased DNA affinity. These interactions are essential for the architectural organization of enhancers, as seen in gene loci where clustered composite elements allow NFAT to bridge multiple regulatory modules. In enhancer architecture, NFAT contributes to at loci by recruiting histone acetyltransferases such as p300, which acetylates to promote an open state conducive to transcription initiation. This recruitment enhances accessibility at composite elements, enabling sustained through modifications like H3K27ac.46193-9/fulltext) NFAT's transcriptional activity is highly context-dependent, functioning primarily in when partnered with coactivators like AP-1 or p300, but capable of repression in other settings through interactions with corepressors or deacetylases, thereby fine-tuning gene output based on cellular signals. The RHD, comprising DNA-binding and dimerization subdomains, underlies these versatile interactions without requiring upstream signaling details.

Roles in Immune Cells

T Lymphocytes

In T lymphocytes, nuclear factor of activated T cells (NFAT) proteins play a pivotal role in activation by transducing signals from the (TCR) complex, leading to the and nuclear translocation of NFAT via activation. This process is essential for the transcription of key cytokines such as interleukin-2 (IL-2) and interferon-gamma (IFN-γ), which drive T cell proliferation and effector functions. NFAT binds to consensus sites in the IL-2 promoter, facilitating its expression in response to antigenic stimulation, as initially demonstrated in studies of the human Jurkat T cell line. Similarly, NFAT regulates IFN-γ production by directly binding to its promoter and enhancing transcription in activated T cells, particularly in + and + subsets. Among the NFAT family, NFAT1 (NFATc2) and NFAT2 (NFATc1) are the predominant isoforms expressed in T lymphocytes, where they cooperatively mediate these responses; NFAT1 is constitutively present, while NFAT2 is upregulated upon activation. NFAT also influences T helper (Th) cell , balancing the development of Th1, Th2, Th17, and regulatory T (Treg) subsets through interactions with lineage-specific s. In Th1 differentiation, NFAT1 promotes IFN-γ expression, supporting the anti-viral and anti-bacterial effector program, while its absence biases toward Th2 responses with reduced IFN-γ and elevated IL-4. For Th17 cells, NFATc1 (NFAT2) is critical for IL-17 production, coupling TCR signal strength to the expression of IL-17A via direct binding to its promoter, thereby contributing to pro-inflammatory responses in . In contrast, NFAT interacts with the forkhead box P3 () in Tregs to repress IL-2 and promote suppressive genes like CTLA4 and CD25, enabling ; this cooperative binding is indispensable for Treg function and stability. Genetic studies underscore NFAT's necessity in T cell responses. Mice deficient in both NFAT1 and NFAT2 exhibit severely impaired IL-2 production and defective T cell proliferation upon TCR stimulation, highlighting their redundant yet essential roles in early activation events. Single NFAT1 knockout mice, however, display compensatory mechanisms with normal IL-2 levels but altered profiles, such as reduced IFN-γ. Recent investigations have linked dysregulated NFAT activity to T cell exhaustion in chimeric receptor ( therapies. Persistent NFAT signaling, particularly NFAT1, drives the expression of exhaustion markers like PD-1 and TOX in chronically stimulated CAR-T cells, limiting their anti-tumor efficacy against solid tumors; inhibiting NFAT has been shown to rejuvenate these cells and enhance persistence as of 2024 studies.

B Lymphocytes and Other Immune Cells

In B lymphocytes, NFAT family members play essential roles in antigen-driven responses, particularly through (BCR) signaling. NFAT2 (NFATc1) and NFAT4 (NFATc3) are activated downstream of BCR engagement via the /PLC-γ2 pathway, promoting proliferation and survival. Deficiency in NFAT2 impairs BCR-mediated by dysregulating genes such as CD22 and RCAN1, leading to reduced expansion. Similarly, combined NFAT1/NFAT4 deficiency alters numbers and enhances certain immunoglobulin isotypes, underscoring their contribution to proliferative responses. NFAT also regulates antibody class switching in B cells. NFATc1 controls isotype switching, particularly to IgG3, by modulating calcineurin-dependent transcriptional programs that support germline transcription and recombination. Although direct regulation of activation-induced cytidine deaminase (AID) expression by NFAT remains context-specific, NFATc1 deficiency results in defective class switching and reduced cytokine production essential for humoral immunity. Furthermore, NFATc1 influences plasma cell differentiation; its short isoform (NFATc1/αA) suppresses plasmablast formation by inhibiting Prdm1 (Blimp-1) expression via chromatin modifications, thereby fine-tuning terminal B cell maturation. In other immune cells such as and dendritic cells (DCs), NFAT isoforms respond to environmental stresses and modulate innate responses. NFAT5, activated independently of , drives osmotic stress responses in these cells by inducing osmoprotective genes like those for aquaporins and organic osmolytes, enabling adaptation to hypertonic conditions in inflamed tissues. NFAT1 (NFATc2) contributes to ; it promotes M2-like phenotypes in tumor-associated contexts and potentiates production, including IL-6 and IL-12, via P2Y6 receptor signaling to enhance innate antiviral and inflammatory responses. NFAT signaling exhibits crosstalk with in innate immunity, particularly in macrophages and DCs, where coordinated activation amplifies TLR-induced for cytokines and molecules. This interaction sustains inflammatory homeostasis but can drive if dysregulated. Knockout studies highlight NFAT's impact on ; NFATc1-deficient mice show reduced antibody responses due to impaired B cell proliferation, class switching, and T cell help. Recent findings link NFATc1 to bone destruction in rheumatoid arthritis (RA), where it drives osteoclast differentiation from monocyte/macrophage precursors, exacerbating joint erosion through RANKL-mediated pathways. Targeting this axis offers potential for mitigating RA-associated bone loss.

Tolerance and Dysfunction

In T cell tolerance, nuclear factor of activated T cells (NFAT) plays a pivotal role in inducing anergy, a state of functional unresponsiveness that prevents excessive immune activation. Anergy arises from chronic low-level T cell receptor (TCR) signaling without costimulation, leading to sustained NFAT nuclear translocation in the absence of sufficient AP-1 activity. This results in the transcriptional activation of anergy-associated genes, including Egr2 and Egr3, which repress interleukin-2 (IL-2) production and promote hyporesponsiveness. Specifically, NFAT dimers bind to the Il2 promoter to induce Egr2 and Egr3, forming repressive complexes that inhibit IL-2 expression and enforce the anergic phenotype. T cell exhaustion, a progressive dysfunction observed in chronic infections and cancer, is similarly regulated by dysregulated NFAT signaling. The PD-1 pathway inhibits activation, thereby limiting NFAT dephosphorylation and nuclear entry, which contributes to reduced effector functions and sustained expression of inhibitory receptors. Partnerless NFAT binding—lacking cooperation with AP-1—directly upregulates exhaustion markers such as PD-1 (Pdcd1), TIM-3 (), and LAG-3 (Lag3), with NFAT2 particularly promoting TIM-3 expression to exacerbate dysfunction. This mechanism underlies the transcriptional program of exhaustion, where NFAT induces TOX, a key driver of the exhausted state.00032-1) In regulatory T cells (Tregs), NFAT synergizes with to maintain by promoting immunosuppressive production. NFAT enhances transcriptional activity through direct interaction and recruitment to target promoters, facilitating the expression of IL-10 and TGF-β, which suppress effector T cell responses. This synergy is critical for Treg suppressive function, as NFAT deficiency impairs induction and reduces IL-10/TGF-β secretion in induced Tregs. Mouse models have elucidated NFAT's role in these processes, with transgenic expression of constitutively active NFAT1 recapitulating anergy by driving partnerless NFAT binding and upregulation of anergic genes like Dtx1. , NFAT1 supports tumor-induced anergy in + T cells, as its deficiency enhances antitumor responses by preventing hyporesponsiveness. Recent studies as of 2025 highlight NFAT's contribution to resistance; for instance, the SFRP2-NFAT/TOX axis in high-m1A gastric tumors promotes T cell exhaustion and poor responses to inhibitors, while dysregulated calcium-NFAT signaling correlates with resistance in solid tumors by sustaining exhaustion programs.

Functions in Non-Immune Systems

Neural Development and Function

NFAT transcription factors, particularly through the calcineurin-dependent pathway, play essential roles in neural development by regulating neuronal , migration, and survival. In neural precursor cells derived from the , NFAT activation promotes proliferation, migration, and into neurons and , as demonstrated by reduced neurosphere size and cell density upon inhibition with the NFAT-blocking VIVIT. Additionally, calcineurin-NFAT signaling mediates (BDNF) expression in developing neurons; BDNF activates NFATc4 in hippocampal progenitors, driving transcription of genes necessary for neuronal maturation and survival. In growth and guidance, dephosphorylation of NFAT by promotes outgrowth in response to guidance cues. Neurotrophins such as BDNF and netrins stimulate embryonic extension in Xenopus spinal neurons via calcineurin-mediated NFAT nuclear translocation, with NFAT-deficient models exhibiting severely impaired elongation.00390-8) This mechanism extends to post-injury regeneration, where NFAT activation facilitates axonal regrowth. In a non-canonical context, GSK3β phosphorylates NFAT to promote its nuclear export, thereby terminating signaling. NFAT also contributes to in mature neural circuits, particularly in the . NFATc1 and NFATc3 isoforms respond to calcium influx through L-type channels, translocating to the to regulate activity-dependent that underlies synaptic strengthening; repetitive calcium spikes in hippocampal neurons potently activate NFATc3, linking synaptic input to transcriptional changes essential for (LTP).30312-2) Disruptions in NFAT signaling, such as in models, lead to behavioral deficits; for example, NFATc4-deficient mice show reduced adult hippocampal and impaired survival of new neurons, resulting in deficits in and anxiety-like behaviors reminiscent of neurodevelopmental disorders.

Cardiovascular and Other Organ Systems

In the cardiovascular system, NFAT transcription factors play critical roles in and adaptation to stress. NFATc3 and NFATc4 are essential for embryonic formation, where they mediate calcineurin-dependent signaling in the myocardium and to repress (VEGF) expression, thereby initiating valve morphogenesis. Disruption of NFATc3 leads to defective development and embryonic lethality, highlighting its non-redundant in this process. In response to hypertrophic stimuli, NFATc4 (also known as NFAT3) translocates to the upon calcineurin activation, inducing expression of genes such as myocyte-enriched calcineurin-interacting protein 1 (MCIP1), which provides negative feedback to limit excessive remodeling, and (ANP), a marker of cardiac . This pathway ensures balanced growth but can contribute to pathological enlargement when dysregulated. Beyond the heart, NFATs influence physiology, particularly fiber type specification and repair. NFATc1 and NFATc2 respond to nerve activity and calcium signals via , promoting a switch toward slow-twitch oxidative fibers by upregulating slow heavy chain isoforms while repressing fast-twitch genes. In muscle regeneration, these isoforms facilitate satellite cell differentiation and fusion into myofibers, with NFATc2 specifically supporting myonuclear accretion and during overload or injury recovery. Loss of NFATc2 results in reduced muscle mass and impaired regenerative capacity, underscoring its role in maintaining fiber . In the , NFAT5 (also called TonEBP) is pivotal for adaptation to hypertonic environments, particularly in the inner medulla. Under osmotic stress, NFAT5 activates transcription of osmoprotective genes, including (AQP2), which enhances water reabsorption in collecting duct cells to restore cellular . Recent studies indicate that NFAT5 also senses ionic imbalances, such as high sodium or , coordinating broader renal responses to prevent injury in hypertonic regions. Segment-specific of NFAT5 disrupts this adaptation, leading to tubular damage and altered expression of stress-response genes beyond osmolyte accumulation. NFATs further contribute to functions in other non-immune tissues, such as and the . In osteoclasts, NFATc1 acts as a master regulator of and activation, induced by RANKL signaling to drive expression of genes essential for fusion, motility, and , thereby maintaining skeletal . Overexpression or constitutive activation of NFATc1 enhances resorptive activity, while its inhibition reduces bone loss in models of . In pancreatic β-cells, NFAT family members, particularly NFATc2 and NFATc4, link calcium influx from glucose stimulation to insulin secretion and gene transcription. Constitutively active NFAT boosts insulin release and supports β-cell proliferation, ensuring adaptive responses to metabolic demands.

Pathophysiological Roles

Inflammation and Autoimmunity

NFAT transcription factors significantly contribute to cytokine dysregulation in autoimmune diseases, particularly through their regulation of pro-inflammatory cytokine production in immune cells. In rheumatoid arthritis (RA), NFATc1 serves as a master regulator of osteoclast differentiation, where it is activated downstream of RANKL/RANK signaling involving NF-κB and MAPK pathways, leading to the expression of genes that promote bone resorption and joint destruction. A 2024 review highlights how NFATc1 amplification by co-stimulatory signals, such as those from SIRPβ1 and OSCAR receptors, exacerbates RA-associated bone erosion, with potential therapeutic targeting of this pathway offering promise for mitigating osteoclastogenesis. Similarly, in multiple sclerosis (MS), NFAT1 and NFAT2 directly bind to the IL-17 promoter, driving Th17 cell differentiation and IL-17 production, which promotes neuroinflammation and disease progression in experimental autoimmune encephalomyelitis (EAE) models of MS. Hyperactivation of NFAT1 in T cells further enhances IL-17 and IL-21 expression, underscoring its role in pathogenic Th17 responses. Beyond adaptive immunity, NFAT family members influence innate inflammatory responses in conditions like and (IBD). NFAT5, responsive to hyperosmotic stress, is required for and by cooperating with in myeloid cells to enhance pro-inflammatory upon LPS stimulation. In , osmotic imbalances trigger NFAT5 activation, promoting macrophage-derived cytokines such as TNF and IL-6, which amplify the cascade. In IBD, NFAT signaling intersects with pathways to regulate intestinal ; for instance, NFATc2/RAG2 double-deficient mice develop a spontaneous ulcerative colitis-like mediated by dysregulated innate immune cells, characterized by severe mucosal damage and infiltration, highlighting NFATc2's role in adaptive immunity to maintain gut by suppressing excessive innate responses. Mouse models of further illustrate NFAT's context-dependent functions. Conditional of NFAT5 in intestinal epithelial cells exacerbates dextran sulfate sodium (DSS)-induced and spontaneous by impairing renewal, reducing mucus (e.g., Muc2), and altering composition, resulting in heightened gut permeability and . In EAE models, while NFAT1 or NFAT2 deficiency attenuates severity by limiting pathogenic Th17 cells and IL-17/IL-21 , NFAT5 under high-salt conditions promotes Th17 pathogenicity, linking osmotic stress to worsened .

Cancer and Cardiovascular Diseases

NFAT transcription factors exhibit dual roles in cancer, promoting oncogenic processes such as and in many contexts while acting as suppressors in others. In particular, NFAT1 and NFAT2 drive tumor and by upregulating pro-metastatic factors like (VEGF) and matrix metalloproteinase 9 (MMP9). For instance, NFAT1 enhances cell invasion through integrin-mediated signaling and induction of (COX-2), which indirectly supports MMP9 activity in extracellular matrix degradation. Similarly, NFAT2 overexpression in correlates with increased via TGF-β/SMAD pathway activation, exacerbating tumor dissemination. NFAT5 contributes to tumor adaptation under hypoxic conditions, a hallmark of solid tumors, by rewiring metabolism toward . In pancreatic ductal , NFAT5 directly transcribes phosphoglycerate kinase 1 (PGK1), elevating glycolytic flux and enhancing cell survival and proliferation in low-oxygen environments, which correlates with poor patient prognosis and increased tumor aggressiveness. Conversely, certain NFAT isoforms function as tumor suppressors; NFATc3 (also known as NFAT4) downregulation promotes aggressive tumor development in and , where its deficiency impairs and worsens outcomes. NFAT also facilitates essential for tumor vascularization through cooperative interactions. The NFAT-AP-1 complex binds to the VEGF promoter, amplifying VEGF expression in endothelial cells and tumor-associated stroma, thereby driving . Genetic or pharmacological inhibition of NFAT signaling, such as via blockade, reduces VEGF-mediated vessel formation; for example, upregulates Down syndrome critical region 1 (DSCR1) to suppress NFAT activity, decreasing microvessel density and inhibiting metastasis in xenograft models. Recent studies highlight NFAT's involvement in CAR-T resistance, where NFAT upregulation induces T-cell exhaustion markers like PD-1 and LAG-3, limiting antitumor efficacy; targeted NFAT inhibition with antagonist peptides enhances CAR-T persistence and clearance in preclinical models. In cardiovascular diseases, NFAT signaling exacerbates and vascular pathologies. In , NFATc1 and NFATc3 activation in endothelial cells promotes by inducing proinflammatory cytokines like IL-33 and adhesion molecules such as , particularly under hyperglycemic conditions that trigger calcineurin-dependent NFAT nuclear translocation. This contributes to plaque formation and progression in diabetic , where NFAT inhibition restores endothelial barrier integrity and reduces inflammatory infiltration. NFAT drives pathological cardiac , a precursor to heart failure, through links to and mitochondrial impairment. Activated by calcium overload, NFAT1 upregulates NADPH oxidases (/NOX4) to generate (ROS), while NFAT3 induces SUMO-specific protease 1 (SENP1), disrupting mitochondrial and in cardiomyocytes. These 2023 findings underscore NFAT's role in amplifying ROS-mitochondria crosstalk, leading to maladaptive remodeling and systolic dysfunction. NFAT further contributes to cardiac fibrosis by promoting fibroblast activation and extracellular matrix deposition. In models of pressure overload-induced hypertrophy, NFATc4 integrates with TGF-β/Smad2 signaling to elevate collagen I/III expression, exacerbating interstitial fibrosis and ventricular stiffness. Recent 2024 advances reveal that NFATc3 activation downstream of TRPV4 channels sustains fibrotic remodeling in pathological hypertrophy, distinct from physiological growth, highlighting its potential as a therapeutic target to halt progression to heart failure.

Therapeutic Implications

NFAT as a Drug Target

Direct targeting of NFAT transcription factors has emerged as a promising strategy to modulate their activity in disease contexts, bypassing upstream regulators like calcineurin to avoid broad immunosuppressive effects. Small molecules that bind to the Rel-homology domain (RHD) of NFAT, responsible for DNA binding and nuclear localization, represent early preclinical advances. For instance, eutomer 2, an S-conformer of a racemic mixture identified through structural studies, binds the C-terminal RHD of NFAT1 with a dissociation constant (KD) of approximately 729 μM, occupying a hydrophobic pocket involving residues Phe603, Val628, and Leu638. This binding validates the ligandability of the RHD and suggests potential for developing higher-affinity inhibitors or PROTACs to degrade NFAT proteins in immune-modulating therapies. Additionally, Compound 10, a drug-like small molecule (N-(3-acetamidophenyl)-2-[5-(1H-benzimidazol-2-yl)pyridin-2-yl]sulfanylacetamide), selectively disrupts the NFAT:AP-1:DNA ternary complex at the antigen receptor response element-2 (ARRE-2) site by binding DNA in a sequence-specific manner, with an IC50 of about 2-5 μM in biochemical assays. This inhibition suppresses NFAT-dependent transcription without affecting individual NFAT or AP-1 DNA binding, offering a precise mechanism to block cooperative gene activation. Peptide-based approaches to disrupt NFAT-AP-1 interactions have also been explored, though primarily in proof-of-concept studies focusing on interface mutations rather than therapeutic peptides. Structural analyses reveal that limited substitutions at the NFAT1-Fos-Jun contact points can abolish and , inspiring designs for cell-permeable peptides that mimic these interfaces to prevent complex formation on promoters like IL-2. Such strategies aim to inhibit NFAT-driven production in T cells while preserving other NFAT functions. In parallel, decoys that sequester NFAT from endogenous DNA sites have shown efficacy in preclinical models. For example, (AAV)-mediated delivery of NFAT decoy neutralizes NFATc1, NFATc2, NFATc3, and NFATc4 in cardiomyocytes, reducing pathological , , and progression in mouse models of pressure overload, with significant improvements in and survival. These decoys bind NFAT with high affinity, preventing transcription of pro-hypertrophic genes like BNP and ANP. In cancer, direct NFAT targeting holds potential for anti-angiogenic effects by curbing (VEGF) expression and endothelial cell migration. Inhibition of NFATc1, for instance, suppresses in tumor microenvironments, as demonstrated in preclinical models where NFAT blockade reduces vessel formation and tumor progression without relying on upstream inhibition. Similarly, disrupting the NFAT:AP-1 complex with Compound 10 attenuates NFAT-mediated pro-angiogenic signaling in invasive cancer cells. For NFAT5, which responds to osmotic stress rather than , preclinical modulators are being developed to address hypertonicity-related pathologies. In , NFAT5 is upregulated in renal medullary cells under water restriction, contributing to impaired urinary concentration and osmotic imbalance, suggesting that selective NFAT5 inhibitors could mitigate dehydration-induced complications by downregulating osmoprotective genes like AQP2 and UT-A. Structural studies published in 2025 revealed NFAT5's direct sensing of ionic imbalances through its C-terminal prion-like domain in the region, enabling and activation under hypertonic stress; this mechanism supports development of small-molecule drugs targeting ionic sensors for renal injury or . Despite these advances, targeting NFAT presents significant challenges due to its ubiquitous expression across immune and non-immune tissues, raising risks of off-target effects such as impaired or metabolic dysregulation. The context-dependent nature of NFAT—promoting survival in some cells (e.g., cardiomyocytes) while driving in others (e.g., tumor cells)—complicates isoform-specific inhibition, necessitating nuanced approaches like tissue-targeted delivery via nanoparticles or AAV vectors to enhance specificity and minimize systemic toxicity. Ongoing efforts focus on optimizing these delivery systems to realize NFAT's therapeutic potential in precision medicine.

Calcineurin Inhibitors and Emerging Therapies

Calcineurin inhibitors represent a cornerstone of therapies targeting the NFAT pathway, primarily by blocking the upstream activation of transcription factors in immune cells. , discovered in the 1970s, binds to the intracellular protein , forming a complex that inhibits the activity of , thereby preventing the and nuclear translocation of NFAT proteins essential for T-cell activation and production. Similarly, , introduced in the 1980s, binds to FK506-binding protein 12 (FKBP12) to achieve the same inhibitory effect on . These drugs have revolutionized , serving as first-line treatments for preventing organ and managing autoimmune conditions such as , where they suppress pathogenic T-cell responses. Despite their clinical success, inhibitors are limited by dose-dependent toxicities that impact long-term use. , a primary concern, manifests as acute of renal and chronic interstitial , affecting up to 30-50% of transplant recipients within the first year and contributing to graft loss over time. , occurring in over 50% of patients, results from sodium retention, , and sympathetic activation, exacerbating cardiovascular risk. These side effects necessitate careful monitoring and dose adjustments, often leading to regimen switches in chronic . Emerging therapies aim to mitigate these limitations by developing more selective modulators of the calcineurin-NFAT axis. Peptides such as VIVIT, derived from the NFAT docking site, selectively disrupt the calcineurin-NFAT interaction without broadly inhibiting calcineurin phosphatase activity, preserving its roles in other cellular processes and potentially reducing nephrotoxicity. Preclinical studies in animal models of transplantation and autoimmunity have demonstrated potent immunosuppression with this approach, prompting investigations into cell-permeable variants for clinical translation. Additionally, gene-editing technologies like CRISPR/Cas9 are being explored to create NFAT-knockout T cells for adoptive cell therapies, enabling targeted immune modulation in conditions like graft-versus-host disease while avoiding systemic exposure. In vitro and ex vivo studies have achieved over 85% knockout efficiency in primary T cells, enhancing their therapeutic potential in personalized medicine. The calcineurin-NFAT pathway also plays a key role in pathological cardiac hypertrophy, and preclinical models suggest that selective inhibition could prevent myocyte growth, though clinical translation remains challenging due to off-target effects observed with existing inhibitors. Recent advances also focus on NFAT5, a calcineurin-independent NFAT family member responsive to ionic and osmotic stress, as a novel therapeutic target. The 2025 structural studies confirm NFAT5's direct sensing of ionic imbalances through its transactivation domain, suggesting opportunities for small-molecule drugs that modulate this pathway to counteract stress-induced pathologies like renal injury or . As of 2025, preclinical efforts are underway to develop NFAT5-targeted ionic sensors for drug screening, with potential applications in protecting against inhibitor toxicities or treating stress-related disorders independently. Clinical trials continue to evaluate NFAT pathway modulators, building on the established use of calcineurin inhibitors. In rheumatoid arthritis, cyclosporine remains a second-line option, with ongoing studies assessing its combination with biologics to optimize efficacy while minimizing toxicity; for instance, a phase III trial demonstrated reduced disease activity scores when paired with methotrexate. These trials underscore the pathway's therapeutic promise, though challenges in selectivity persist.

References

  1. [1]
    NFAT, immunity and cancer: a transcription factor comes of age
    Aug 20, 2010 · NFAT transcription factors are expressed by most immune cells and have critical roles in regulating immune responses. This Review describes ...
  2. [2]
    Nfat: ubiquitous regulator of cell differentiation and adaptation - PMC
    The nuclear factor of activated T cells (NFAT) proteins are a family of transcription factors whose activation is controlled by calcineurin, a Ca2+-dependent ...
  3. [3]
    Cell cycle and apoptosis regulation by NFAT transcription factors
    Apr 21, 2016 · The NFAT (nuclear factor of activated T cells) family of transcription factors consists of five members (NFAT1–NFAT5) encoded by different genes ...
  4. [4]
    Transcriptional regulation by calcium, calcineurin, and NFAT
    Diverse modes of DNA binding by NFAT. Structural relation to the Rel/NFκB family. All five proteins of the NFAT family, NFAT1-NFAT4 and NFAT5/TonEBP, may be ...Cellular Inputs Affecting... · Other Biological Systems In... · Conclusion
  5. [5]
    NFAT Gene Family in Inflammation and Cancer - PMC
    The N-terminal region contains regulatory domains, including the casein kinase 1 (CK1) docking site, the transactivation domain (TAD), and a calcineurin (Cn) ...
  6. [6]
    Review NFAT signaling dysregulation in cancer: Emerging roles in ...
    The amino-terminal regulatory domain of NFAT protein is composed of the NFAT homology region (NHR), the DNA-binding domain (also known as the REL-homology ...
  7. [7]
    Expression, Fermentation and Purification of a predicted Intrinsically ...
    The transactivating domain of NFAT5 is predicted to be intrinsically disordered under normal tonicity, and under high NaCl, the activity of this domain is ...
  8. [8]
    A Conserved Docking Motif for CK1 Binding Controls the Nuclear ...
    Here we demonstrate that NFAT1 regulation involves the concerted effort of distinct kinases to phosphorylate each of the two types of serine motifs. CK1 ...Results · Ck1 And Gsk3 Synergize To... · Ck1 Controls Nfat1 Nuclear...Missing: proline NFAT5
  9. [9]
    Concerted Dephosphorylation of the Transcription Factor NFAT1 ...
    The SRR-1 region was phosphorylated on only five of its seven conserved serines, a second serine-rich region (SRR-2245SRSSS249) contained only a single ...Article · Results · The Srr-1 Region Regulates...
  10. [10]
    1OWR: CRYSTAL STRUCTURE OF HUMAN NFAT1 ... - RCSB PDB
    Feb 10, 2004 · We have determined the crystal structure of the entire Rel homology region (RHR) of human NFAT1 (NFATc2) bound to DNA as a monomer.Missing: domain p50 heterodimer
  11. [11]
    Extracellular signal-regulated kinase 1/2-mediated phosphorylation ...
    In addition, the DNA binding domain shares >70% sequence homology among NFAT proteins and <20% sequence homology to the DNA binding domain of Rel/NF-κB factors, ...
  12. [12]
  13. [13]
  14. [14]
  15. [15]
  16. [16]
    Calcineurin–NFAT signaling regulates the cardiac hypertrophic ...
    An emerging paradigm suggests that calcineurin–NFAT and MAPK signaling pathways are inter-dependent and together orchestrate the cardiac hypertrophic response.Missing: NFAT2 | Show results with:NFAT2
  17. [17]
    Protein Kinase C θ Affects Ca2+ Mobilization and NFAT Activation in ...
    Our finding suggests that PKCθ plays a critical and nonredundant role in T cell receptor–induced NFAT activation.
  18. [18]
    Fyn and p38 signaling are both required for maximal hypertonic ...
    This is achieved by the activation of the transcription factor called OREBP (also known as TonEBP or NFAT5) that specifically binds to the osmotic response ...Missing: JNK | Show results with:JNK
  19. [19]
    Direct ionic stress sensing and mitigation by the transcription factor ...
    Feb 19, 2025 · NFAT5 activates the transcription of many osmoadaptive genes, including those that promote osmolyte accumulation and those that encode heat ...Missing: JNK | Show results with:JNK
  20. [20]
    Toll-Like Receptors Promote Mitochondrial Translocation of Nuclear ...
    Jul 29, 2015 · The other members of the NFAT family, including NFAT2, NFAT3 and NFAT4, also translocate into mitochondria in LPS-treated microglia. Confocal ...
  21. [21]
    GSK-3β/NFAT Signaling Is Involved in Testosterone-Induced ...
    Our results show that testosterone increases NFAT activity through calcineurin activation and GSK-3β inhibition. In addition, AR signaling modulates the ...
  22. [22]
    Interaction Between NFκB and NFAT Coordinates Cardiac ...
    Mar 8, 2012 · All Rel family members are conserved throughout evolution and share a Rel homology domain ... NFAT and NFκB contain a Rel homology domain ...
  23. [23]
    NFAT and NF-κB dynamically co-regulate TCR and CAR signaling ...
    Jul 25, 2023 · We show that two key pathways in T cell signaling, calcium/nuclear factor of activated T cells (NFAT) and protein kinase C (PKC)/nuclear factor κB (NF-κB), ...
  24. [24]
    Page not found | Nature
    Insufficient relevant content.
  25. [25]
    Characterization of DNA binding, transcriptional activation, and ...
    Nov 6, 2000 · NFATp is one member of a family of transcriptional activators whose nuclear accumulation and hence transcriptional activity is regulated in ...Missing: repression RHD-<|control11|><|separator|>
  26. [26]
    Nuclear Factor of Activated T Cells (NFAT) - NIH
    We show that transcription factors of the nuclear factor of activated T cells (NFAT) family bind p300/CBP and recruit histone acetyltransferase activity from T ...Missing: composite loci
  27. [27]
    B-cell Antigen Receptor Activates Transcription Factors NFAT ...
    Engagement of the B-cell antigen receptor (BCR) induces the activation of various transcription factors, including NFAT (nuclear factor of activated T-cells) ...
  28. [28]
    Role of NFAT in Chronic Lymphocytic Leukemia and Other B-Cell ...
    Apr 1, 2021 · Specifically, the defect in BCR-induced proliferation is determined by NFAT2-dependent expression of CD22, Rcan1, Tnfsf14, FasL and other key ...Missing: class | Show results with:class
  29. [29]
    NFATc1 affects mouse splenic B cell function by controlling the ...
    Mouse B cells lacking NFATc1 exhibit defective proliferation, survival, isotype class switching, cytokine production, and T cell help.
  30. [30]
    NFATc1 affects mouse splenic B cell function by controlling the ...
    Mouse B cells lacking NFATc1 exhibit defective proliferation, survival, isotype class switching, cytokine production, and T cell help.Nfatc1 Ablation Suppresses... · Nfatc1 Controls Numerous... · Nfatc1 Affects Ig Class...
  31. [31]
    Induction of Short NFATc1/αA Isoform Interferes with Peripheral B ...
    Jan 24, 2018 · In this study, we investigated the role of transcription factor NFATc1 (also designated as NFAT2) in plasma cell formation. NFATc1 is ...
  32. [32]
    Gene expression induced by Toll-like receptors in macrophages ...
    Feb 6, 2012 · We report that NFAT5, previously characterized as an osmostress responsive factor, regulates the expression of multiple TLR-induced genes in ...Results · Nfat5 Regulates Gene... · Tlr-Induced Nfat5 Expression...
  33. [33]
    NFATc2 promotes lactate and M2 macrophage polarization through ...
    Jun 1, 2024 · According to single-cell sequence data, NFATc2 was closely associated with infiltrating immune cells and was related to macrophage polarization.
  34. [34]
    Macrophage P2Y6 Receptor Signaling Selectively Activates ... - NIH
    NFATC2 mediates the P2Y6-potentiated IL-12 production and IFN-γ expression. Macrophage P2Y6 and NFATC2 control innate and type 2 immune responses. Introduction.Missing: M1 M2 polarization
  35. [35]
    Regulation and dysregulation of innate immunity by NFAT signaling ...
    Jun 18, 2012 · Therefore, it is conceivable that evolutionary conserved signaling pathways, such as the NF-κB pathway, globally dominate innate responses.
  36. [36]
    Recent advances of NFATc1 in rheumatoid arthritis-related bone ...
    Feb 3, 2024 · This review provides a comprehensive summary of recent advances in understanding the mechanism of NFATc1 in the context of RA-related bone destruction
  37. [37]
    Transcriptional complexes formed by NFAT dimers regulate the ...
    Mar 23, 2009 · This mutant showed almost no capacity to form dimers, but retained its ability to bind DNA and to form complexes with AP-1 (Fig. 1, F–H). Direct ...
  38. [38]
    Regulation of T‐cell tolerance by calcium/NFAT signaling
    Sep 10, 2009 · ... NFAT on other anergy-associated genes might be indirect. Expression of the early growth response gene 2 (Egr2) and Egr3 is dependent on NFAT ...
  39. [39]
    The transcription factor NFAT promotes exhaustion of activated ...
    The cells were then fixed and sorted to separate the exhausted PD-1+TIM3+ cell and control PD-1TIM3- cell populations (Figure S2A), and stained for endogenous ...
  40. [40]
    Dependence on nuclear factor of activated T-cells (NFAT ... - PNAS
    Several lines of evidence suggest nuclear factor of activated T-cells (NFAT) to control regulatory T cells: thymus-derived naturally occurring regulatory T ...Results · Nfat Is Essential For Itreg... · Nfat Is Dispensable For...<|separator|>
  41. [41]
    Control of regulatory T‐cell differentiation and function by T‐cell ...
    Feb 5, 2020 · Here I show the current understanding of how Foxp3 transcription factor complexes regulate the differentiation, maintenance and functional maturation of Treg.From Treg Classification To... · Foxp3 Seizes Runx... · Foxp3-Inducing Transcription...<|separator|>
  42. [42]
    ICOS signal facilitates Foxp3 transcription to favor suppressive ...
    Apr 3, 2018 · Mechanistic studies demonstrated that ICOS could induce the transcription activity of Foxp3, by facilitating the nuclear factor of activated T ...
  43. [43]
    Deltex1 Is a Target of the Transcription Factor NFAT that Promotes T ...
    Jul 17, 2009 · Here we show that Dtx1 was a transcription target of nuclear factor of activated T cells (NFAT) and participated in T cell anergy.
  44. [44]
    NFAT1 supports tumor-induced anergy of CD4+ T cells - PMC
    Clonal anergy in CD4+ T cells is established as a result of the activation of a program of gene expression that is dependent on the transcription factor NFAT.
  45. [45]
    The m1A-SFRP2-NFAT/TOX axis governs T cell exhaustion in ...
    Aug 11, 2025 · Our study reveals a novel mechanism of immunotherapy resistance mediated by SFRP2 overexpression in m1A high-risk gastric tumors. While SFRP2 ...
  46. [46]
    Differential regulation of calcium-NFAT signaling pathway by Akt ...
    Mar 26, 2025 · NFAT signaling serves a pivotal role in T-cell activation on TCR triggering, but it becomes involved in T-cell exhaustion when persistent ...
  47. [47]
    Brain-Derived Neurotrophic Factor Activation of NFAT (Nuclear ...
    Sep 3, 2003 · Here we delineate a novel pathway whereby BDNF signaling, through activation of the transcription factor NFATc4, triggers the induction of gene ...
  48. [48]
    Identification of a Novel Axon Regeneration Role for Noncanonical ...
    Aug 1, 2022 · ... NFAT ... Future studies will determine the effects of Wnt5a-induced PKC dephosphorylation on RGC protection, neurite growth and axonal regrowth.
  49. [49]
    NFAT signaling in neural development and axon growth
    Nov 17, 2007 · Elevated Ca2+ levels then activates NFAT transcription via calcineurin (Fig. 1). Moreover, neurotrophin signaling through NFAT appears to ...
  50. [50]
    Wnt signaling pathways in biology and disease - Nature
    Apr 4, 2025 · The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination ...
  51. [51]
    Nuclear factor of activated T cells (NFATc4) is required for BDNF ...
    May 14, 2012 · We show that NFATc4 calcineurin-dependent activity is required selectively for survival of adult-born neurons in response to BDNF signaling.
  52. [52]
    Targeted Disruption of NFATc3, but Not NFATc4, Reveals an ... - NIH
    Disruption of the NFATc1 gene resulted in embryonic lethality due to aberrant heart valve formation and cardiac insufficiency (6, 48). ... NFATc3 and NFATc4 in ...
  53. [53]
    A field of myocardial-endocardial NFAT signaling underlies heart ...
    Sep 3, 2004 · We show that the initiation of heart valve morphogenesis in mice requires calcineurin/NFAT to repress VEGF expression in the myocardium ...
  54. [54]
    miR-23a functions downstream of NFATc3 to regulate cardiac ... - NIH
    We report that miR-23a is a pro-hypertrophic miRNA, and its expression is regulated by the transcription factor, nuclear factor of activated T cells (NFATc3).
  55. [55]
    Opposing Roles of FoxP1 and Nfat3 in Transcriptional Control ... - NIH
    Sustained elevation of calcium signaling can induce cardiac hypertrophy through activation of Nfat family transcription factors. FoxP family transcription ...
  56. [56]
    Telmisartan suppresses cardiac hypertrophy by inhibiting ...
    NFAT has been observed to participate in pathological cardiac hypertrophy (39), and activated NFAT promotes ANP or BNP release and induces cell apoptosis (41,44) ...
  57. [57]
    NFAT is a nerve activity sensor in skeletal muscle and controls ... - NIH
    NFAT is a nerve activity sensor in skeletal muscle and controls activity-dependent myosin switching.
  58. [58]
    NFAT isoforms control activity-dependent muscle fiber type ... - PNAS
    NFATc1 knockout is embryonic lethal due to defects in cardiogenesis (15), whereas the knockouts of NFATc2 and c3 result in muscle atrophy and only minor, if any ...
  59. [59]
    The Functional Role of Calcineurin in Hypertrophy, Regeneration ...
    Genetic loss of calcineurin blocks mechanical overload-induced skeletal muscle fiber type switching but not hypertrophy. Journal of Biological Chemistry ...
  60. [60]
    Regulation of Muscle Cell Growth by Nfatc2 Pathway
    Here, we examine the role of NFATC2 in skeletal muscle by analyzing adult NFATC2−/− mice. These mice exhibit reduced muscle size due to a decrease in ...
  61. [61]
    NFAT5 in cellular adaptation to hypertonic stress – regulations and ...
    This review aims to summarize our current knowledge on the functional roles of NFAT5 in osmotic stress adaptation and the signaling pathways that regulate its ...
  62. [62]
    How do kinases contribute to tonicity-dependent regulation of the ...
    NFAT5 activates expression of water channels aquaporin-2 (AQP-2), which dictates the apical water permeability of the collecting ducts[4-6] and aquaporin-1 ...
  63. [63]
    Segment specific loss of NFAT5 function in the kidneys is sufficient ...
    Jan 31, 2025 · We confirmed that the loss of NFAT5 results in kidney injury that extends beyond hypertonic areas. Renal injury correlates with the expression level of genes ...
  64. [64]
    The nuclear factor of activated T cells 5 (NFAT5) contributes ... - Nature
    Nov 24, 2022 · We show that NFAT5 is a master regulator of gene expression in the kidney collecting duct and in vivo loss of NFAT function induces a kidney injury like ...
  65. [65]
    NFATc1: functions in osteoclasts - PubMed
    It is a master regulator of RANKL-induced osteoclast differentiation and plays a pivotal role in osteoclast fusion and osteoclast activation.
  66. [66]
    Regulation of NFATc1 in Osteoclast Differentiation - PMC - NIH
    [3] However, excessive bone resorption by osteoclasts compared to bone formation by osteoblasts, results in osteopenic disorders such as osteoporosis ...
  67. [67]
    NFAT Targets Signaling Molecules to Gene Promoters in Pancreatic ...
    These calcium transients induce insulin secretion and increase insulin gene expression in β-cells in response to metabolic fuels, which are amplified by gut ...
  68. [68]
    The Transcription Factor Nfatc2 Regulates β-Cell Proliferation and ...
    Dec 9, 2016 · In mouse islets, ca-Nfatc1 and ca-Nfatc2 significantly enhanced insulin secretion in response to high glucose (16.7 mM), or a depolarizing ...
  69. [69]
    Calcineurin/NFAT Signalling Regulates Pancreatic Beta-Cell Growth ...
    Sep 21, 2006 · Conditional NFAT activation also induces the expression of genes critical for beta-cell endocrine function, including all six genes mutated in ...
  70. [70]
    NFAT Proteins: Emerging Roles in Cancer Progression - PMC - NIH
    Note that NFAT1-4 differ in the size of the carboxyl-terminal domain, and alternative splice variants of NFAT1 (isoforms A, B, C), NFAT2 (isoforms A, B, C) and ...
  71. [71]
    NFAT2 Induces Tumor Cell Proliferation and Metastasis by Acting as ...
    Mar 4, 2025 · NFAT2 acts as a transcriptional co-activator of the TGF-β1/SMAD signaling pathway, promoting liver cancer progression and metastasis.
  72. [72]
    Transcription factor NFAT5 contributes to the glycolytic phenotype ...
    Dec 11, 2019 · These findings indicated that NFAT5 has a controversial role in cancer, and the exact role of NFAT5 in pancreatic cancer has not been reported.
  73. [73]
    Expression, Prognosis and Gene Regulation Network of NFAT ... - NIH
    Apr 9, 2021 · NFAT2/3/4/5 act as tumor suppressors in several types of cancers. NFAT2 and NFAT3 induce apoptosis in HCC [6] and glioma cells [22], ...
  74. [74]
    Vascular Endothelial Growth Factor Activates Nuclear Factor of ...
    VEGF activates NFAT and AP-1-dependent transcription and increases c-fos and c-jun mRNA levels. NFAT has been shown to cooperate with AP-1 transcription ...
  75. [75]
    Arsenic Trioxide Restrains Lung Cancer Growth and Metastasis by ...
    Conclusion: As2O3 can suppress lung cancer growth and metastasis through anti-angiogenesis effects by blocking the calcineurin-NFAT pathway by upregulating ...<|separator|>
  76. [76]
    NFAT and NFκB Inhibitors to Enhance CAR T-Cell Therapy
    In preclinical studies, NFAT inhibition improved T-cell persistence, enhanced anti-leukemia activity, and significantly extended survival. NFκB inhibition ...
  77. [77]
    Role of NFAT in the Progression of Diabetic Atherosclerosis - PMC
    Mar 11, 2021 · The role of Ca2+/ NFAT in dysfunction and inflammation of human coronary endothelial cells induced by sera from patients with Kawasaki disease.
  78. [78]
    Inhibition of NFAT Signaling Restores Microvascular Endothelial ...
    Dec 5, 2020 · More recently, we demonstrated a role for NFAT in diabetes-driven atherosclerotic plaque formation via regulation of inflammatory mediators and ...
  79. [79]
  80. [80]
    Dohongsamul-tang inhibits cardiac remodeling and fibrosis through ...
    Dohongsamul-tang inhibits cardiac remodeling and fibrosis through calcineurin/NFAT and TGF-β/Smad2 signaling in cardiac hypertrophy. J Ethnopharmacol. 2024 ...
  81. [81]
    TRPV4 Channels Promote Pathological, but Not Physiological ...
    TRPV4 channels contribute to pathological remodeling by promoting fibrosis and inducing TRPC6 upregulation through the activation of Ca 2+ -dependent CN/NFATc3 ...
  82. [82]
    Ligandability assessment of the C‐terminal Rel‐homology domain ...
    Feb 23, 2024 · NFAT1 is a multidomain protein comprising the NFAT homology region (NHR), the Rel-homology domain (RHD) domain, and the C-terminal domain ( ...Missing: transactivation | Show results with:transactivation<|separator|>
  83. [83]
    Targeting the NFAT:AP-1 transcriptional complex on DNA ... - PNAS
    Apr 24, 2019 · We performed a high-throughput screen and identified Compound 10, a small drug-like molecule that inhibits the NFAT:AP-1:DNA interaction on the ARRE-2 element.
  84. [84]
    Gene expression elicited by NFAT in the presence or absence of ...
    In this study we have addressed the question of whether NFAT–AP‐1 cooperation is mandatory for the expression of NFAT target genes. Using mutant NFAT proteins ...
  85. [85]
    AAV-mediated expression of NFAT decoy oligonucleotides protects ...
    Jun 4, 2021 · Here, we aimed at neutralizing four members of the NFAT family of transcription factors as a therapeutic strategy for myocardial hypertrophy ...
  86. [86]
    Nuclear Factor of Activated T Cells (NFAT) Proteins as Targeted ...
    Dec 16, 2024 · NFAT signaling is implicated in developing multiple diseases, such as cancer progression, that are associated with angiogenesis.
  87. [87]
    Sickle cell disease up‐regulates vasopressin, aquaporin 2, urea ...
    Apr 29, 2019 · However, under water restriction, SCD mice struggle to concentrate urine despite activating NFAT5. SCD‐induced urinary concentration defect ...
  88. [88]
    Inhibitors of the calcineurin/NFAT pathway - PubMed
    As substrates of calcineurin, transcription factors of the NFAT family play an essential role in lymphocyte activation, and it follows that their function is ...
  89. [89]
    Tacrolimus/Cyclosporine Pathway, Pharmacodynamics - ClinPGx
    Action on calcineurin and NFAT. Upon entering T cells, both cyclosporine and tacrolimus bind with high affinity to proteins known as immunophilins. Cyclosporine ...
  90. [90]
    Novel inhibitors of the calcineurin/NFATc hub - alternatives to CsA ...
    Oct 27, 2009 · Different steps in this pathway are targeted by certain compounds to finally suppress NFATc-dependent gene expression. A selection of the most ...
  91. [91]
    Calcineurin Inhibitors - StatPearls - NCBI Bookshelf - NIH
    Nov 12, 2023 · Adverse Effects · Hypertension is another significant adverse effect of these drugs, stemming from renal vasoconstriction and sodium retention.Missing: NFAT5 | Show results with:NFAT5
  92. [92]
    The Many Faces of Calcineurin Inhibitor Toxicity – What the FK? - PMC
    By 10 years, virtually all allografts will have evidence of CNI nephrotoxicity. CNI have been strongly associated with hypertension, dyslipidemia, and new onset ...Missing: NFAT5 | Show results with:NFAT5
  93. [93]
    Selective inhibition of NFAT activation by a peptide spanning the ...
    Peptides spanning the region inhibit the ability of calcineurin to bind to and dephosphorylate NFAT proteins, without affecting the phosphatase activity of ...
  94. [94]
    A novel peptide exerts potent immunosuppression by blocking the ...
    A novel peptide exerts potent immunosuppression by blocking the two-site interaction of NFAT with calcineurin.
  95. [95]
    Rapid and Efficient Gene Editing for Direct Transplantation of Naive ...
    Jul 20, 2021 · Taken together, we achieved above 85 % knockout efficiency of the transcription factor NFAT by applying gRNA-only nucleofection, which we ...
  96. [96]
    Direct ionic stress sensing and mitigation by the transcription factor ...
    This is a preprint. It has not yet been peer reviewed by a journal. The National Library of Medicine is running a pilot to include preprints that result ...Missing: 2025 | Show results with:2025
  97. [97]
    NFAT5: a stress-related transcription factor with multiple functions in ...
    May 22, 2025 · Thus, NFAT5 not only controls osmolyte regulation in response to hyperosmotic stress but also modulates genes involved in water transport and ...
  98. [98]
    Increased regulatory activity of the calcineurin/NFAT pathway in ...
    Calcineurin enzymatic activity was increased by 80% in human dilated cardiomyopathy compared with non-failing human hearts.Missing: modulators rheumatoid arthritis