Fact-checked by Grok 2 weeks ago

Steroidogenic factor 1

Steroidogenic factor 1 (SF-1), also known as subfamily 5 group A member 1 (NR5A1) or adrenal 4-binding protein (Ad4BP), is an that acts as a to regulate the development and function of steroidogenic tissues, including the and gonads. It plays a critical role in by controlling the expression of key enzymes such as those involved in conversion to . SF-1 binds to DNA as a via specific response elements, influencing endocrine and . Discovered independently in the early 1990s by Keith Parker and Ken-ichirou Morohashi, SF-1 was identified through its ability to bind promoter regions of steroidogenic genes like those encoding . Structurally, it features a with conserved P-box and A-box motifs for sequence-specific recognition, and a ligand-binding domain that, despite lacking a known , supports interactions with coactivators like p300 and GCN5 for transcriptional activation. Post-translational modifications, including , SUMOylation, and acetylation, modulate its activity and stability. SF-1 is expressed in a tissue-specific manner, prominently in the adrenal glands, gonads, , , and , with early onset during embryogenesis—around embryonic day 9 in mice and 32 days post-ovulation in humans. Functionally, it drives , survival, differentiation, and dynamics in endocrine tissues, while also contributing to and neuronal functions in the . In steroidogenesis, SF-1 coordinates the expression of genes across the biosynthetic pathway, ensuring proper hormone production in response to physiological demands. In development, SF-1 is indispensable for the formation of primary steroidogenic organs; studies in mice demonstrate complete of the adrenal glands and gonads, leading to embryonic due to and electrolyte imbalances. results in adrenal , while overexpression can induce ectopic tissue formation, highlighting its dosage-sensitive nature. Clinically, heterozygous NR5A1 mutations are a common cause, accounting for approximately 10-20% of 46,XY (DSD), and are also associated with and , with phenotypes ranging from mild to complete . Recent studies have identified over 200 NR5A1 variants, revealing a broader phenotypic spectrum including oligogenic effects in DSD (as of 2025). Overexpression or amplification of SF-1 is implicated in progression, correlating with poor prognosis in affected patients.

Genetics

Gene structure and location

The NR5A1 gene, which encodes steroidogenic factor 1 (SF-1), is located on the long arm of human at position 9q33.3, spanning approximately 26 kb from base pairs 124,481,236 to 124,507,399 (GRCh38.p14 assembly). The orthologous gene in mice, Nr5a1, maps to at position 2 B (24.42 cM). The gene consists of 7 exons, with exon 1 being non-coding and exons 2 through 7 containing the coding sequence that translates into the 461-amino-acid SF-1 protein. Exons 2–3 primarily encode the (DBD), exons 4–5 cover the hinge region, and exons 5–7 encode the ligand-binding domain (LBD), reflecting the modular architecture typical of genes. At least 15 alternative splice variants have been identified, some of which introduce frameshifts or deletions in the LBD, potentially impairing protein function; recent cohort analyses (e.g., in patients with ) have highlighted such variants, including intronic mutations that disrupt splicing and affect LBD integrity. The intron-exon boundaries of NR5A1 exhibit strong evolutionary conservation across vertebrate species, including mammals, birds, and fish, underscoring the ancient origin of this subfamily and the functional importance of its .

Nomenclature and discovery

Steroidogenic factor 1 (SF-1), encoded by the NR5A1 gene, was first identified in the early as a key regulating steroidogenic genes in adrenal and gonadal tissues. The factor was initially characterized as adrenal 4-binding protein (Ad4BP) through studies on the promoter of the bovine CYP11A1 gene (also known as P450scc), where it was found to bind a specific , termed the Ad4 site, essential for tissue-specific expression in steroidogenic cells. This discovery was reported by Morohashi et al. in 1992, who purified the 53 kDa protein from bovine adrenal nuclear extracts and demonstrated its role in activating multiple steroidogenic promoters. Concurrently, the laboratory cloned the homolog in and named it steroidogenic factor 1 (SF-1) due to its broad regulatory function across promoters, including those for CYP11A1, CYP11B1, and CYP17A1. The human gene was cloned in 1996 by et al. from an embryonic adrenal , revealing high sequence conservation with the versions. The official gene symbol NR5A1 ( subfamily 5, group A, member 1) was assigned by the in the mid-1990s, reflecting its classification within the nuclear receptor superfamily; common aliases include Ad4BP, FTZ-F1 ( factor 1 homolog), and (embryonic lethal protein). Key milestones in SF-1 research include the 1994 cloning and initial characterization as an orphan nuclear receptor without an identified ligand, followed by the generation of Nr5a1 knockout mice in 1994–1995 by independent groups, which revealed its indispensable role in adrenal and gonadal development, leading to agenesis of these organs in homozygotes. More recent advances, particularly from 2023 studies, have uncovered SF-1's interactions with phospholipids such as phosphatidylinositol-4,5-bisphosphate (PIP2), which modulate its structure and extend its functions to influence pre-mRNA splicing regulation in addition to transcriptional control. No pseudogenes of NR5A1 have been identified in the human genome, though it shares paralogy with NR5A2, which encodes the related liver receptor homolog-1 (LRH-1).

Protein structure

Domains and functional motifs

Steroidogenic factor 1 (SF-1), encoded by the NR5A1 gene, is a 461-amino-acid protein with an approximate molecular weight of 52 kDa. The protein exhibits the modular architecture typical of nuclear receptors, featuring an N-terminal A/B transactivation domain spanning residues 1-61, which contributes to transcriptional activation. This is followed by the DNA-binding domain (DBD), a conserved region encompassing residues 92-172 that includes two zinc finger motifs essential for DNA interaction. Within the DBD, the P-box facilitates recognition of the AGGTCA half-site sequence, while the A-box supports dimerization, although SF-1 predominantly functions as a monomer. The hinge region, approximately residues 200-250, links the DBD and ligand-binding domain (LBD) and harbors nuclear localization signals (NLS) that direct SF-1 to the , as well as phosphorylation sites that influence protein stability and activity. The C-terminal LBD (residues 219-461) adopts a canonical fold with 12 α-helices, including the helix 12 motif critical for recruiting coactivators via LXXLL motifs. As an lacking a known natural hormonal , SF-1's LBD instead accommodates phospholipids such as , with the lipid tails occupying a hydrophobic pocket to stabilize the active conformation. Specific motifs within SF-1, including LXXLL-binding sites for coactivators, modulate these regulatory associations. Structural insights into SF-1 have been derived from post-1980s X-ray crystallography studies, with key determinations of the LBD revealing monomeric binding to extended DNA sites and phospholipid occupancy. For instance, the 2005 crystal structure of the human SF-1 LBD (PDB ID: 1ZDT) at 2.1 Å resolution demonstrates how phospholipids like phosphatidylethanolamine embed in the ligand-binding pocket, promoting an agonist-like positioning of helix 12. These findings underscore SF-1's unique reliance on structural phospholipids for functional regulation rather than traditional ligands.

Evolutionary homology

Steroidogenic factor 1 (SF-1, encoded by ) belongs to the NR5A subfamily of nuclear receptors, with its closest homolog being (also known as liver receptor homolog-1 or LRH-1). These two receptors share approximately 70-80% sequence identity in their (DBD) and ligand-binding domain (LBD), reflecting their common origin from an ancestral duplicated early in evolution, while a third paralog, NR5A5, persists in teleost fish but was lost in mammals. SF-1 exhibits high evolutionary conservation, tracing its origins to the invertebrate ortholog fushi tarazu factor 1 (FTZ-F1, NR5A3) in , where it functions in embryonic segmentation and regulates signaling, the insect analog of steroid hormone pathways. This conservation extends to mammals, with the DBD showing over 90% identity across vertebrates, underscoring a preserved role in DNA recognition and . FTZ-F1 orthologs, such as those in , also hint at an ancestral function in , as NR5A family members modulate and steroid precursor handling in both flies and vertebrates. Among mammals, NR5A1 demonstrates remarkable sequence similarity, with the ortholog (Nr5a1) sharing 95% identity with the human protein, facilitating the use of models in functional studies. Comparative analyses reveal subtle divergences, particularly in LBD structural flexibility, which may influence ligand interactions and receptor activation across species, as highlighted in recent structural comparisons. Phylogenetically, the NR5A subfamily occupies a basal position relative to the steroid and thyroid hormone receptor clades within the nuclear receptor superfamily, emerging from an ancient duplication event in early metazoans. This positioning aligns with its conserved contributions to and development across amniotes, from reptiles to mammals, emphasizing an evolutionarily stable core function in endocrine .

Expression patterns

In adult steroidogenic tissues

In adult steroidogenic tissues, steroidogenic factor 1 (SF-1), encoded by the NR5A1 gene, exhibits high expression specifically in cells responsible for hormone production. Within the , SF-1 is prominently expressed in the steroidogenic cells of the and zona reticularis, where it supports and synthesis, respectively. In the gonads, expression is concentrated in cells of the ovarian follicles and Leydig cells of the testes, facilitating and biosynthesis. SF-1 maintains the steroidogenic capacity of these mature tissues by regulating the expression of key enzymes and transporters involved in production. In adults, SF-1 levels are generally stable, reflecting a constitutive role in tissue homeostasis, though they can be finely modulated by pituitary-derived trophic hormones such as (ACTH) in the and (LH) in the gonads. This modulation influences SF-1 transcriptional activity without substantially altering its baseline expression. RNA sequencing studies have quantified SF-1 mRNA abundance as 10- to 100-fold higher in adrenal and gonadal tissues compared to non-steroidogenic organs like the liver, , or , underscoring its specialized role in these sites. Notably, adult SF-1 expression patterns in these steroidogenic tissues show no significant differences between males and females.

During embryonic development

Steroidogenic factor 1 (SF-1, encoded by NR5A1) expression initiates during early embryogenesis in the coelomic of the urogenital ridge, marking the onset of steroidogenic tissue formation. In mice, Nr5a1 transcripts first appear at embryonic day 9.5 (E9.5), corresponding to approximately week 6 of , where SF-1 is detected in progenitor cells destined for adrenal and gonadal lineages. Expression peaks shortly thereafter in the fetal adrenal primordium around E10.5 in mice, with SF-1 localized to the nuclear compartment of nascent adrenocortical cells. In parallel, expression intensifies in the gonadal ridges by E11.5, exhibiting a bipotential pattern in undifferentiated gonads of both XX and XY embryos prior to sexual differentiation. In humans, SF-1 becomes detectable in the developing adrenals by gestational week 8, as evidenced by immunohistochemical and transcriptomic analyses of fetal tissues. This early expression is critical, as variants in NR5A1 manifest phenotypes in cohort studies of disorders of sex development, highlighting disruptions in adrenal and gonadal primordia formation. Recent 2024 analyses of patient cohorts underscore the temporal sensitivity of SF-1 during this window.00507-8/fulltext) Postnatally, SF-1 expression undergoes downregulation in non-gonadal sites, such as certain neural and ventral midline structures, while remaining sustained in the gonads through birth to support maturation. Spatial patterns during migration are confirmed by , revealing nuclear SF-1 in proliferating and migrating coelomic epithelial cells invading the urogenital ridge mesenchyme.

In non-steroidogenic tissues

Steroidogenic factor 1 (SF-1, also known as NR5A1) displays moderate expression in select non-steroidogenic tissues, notably the ventromedial (VMH) and pituitary gonadotropes, where it plays roles in endocrine regulation beyond primary production. Low levels of SF-1 expression have been detected in the liver, , and , while it is absent or undetectable in most other non-endocrine tissues such as muscle, , and intestine. Single-cell sequencing analyses confirm these patterns, highlighting its restricted but persistent ectopic distribution in adults. In the VMH, SF-1-positive neurons are integral to , responding directly to signaling to modulate feeding behavior and body weight. Studies from the mid-2000s demonstrated that depolarizes and increases firing rates in these neurons, and disruption of this pathway via SF-1 ablation leads to and impaired glucose regulation, underscoring its non-reproductive metabolic functions. Subsequent work in the 2010s reinforced this by linking VMH SF-1 activity to diet-induced and insulin sensitivity, with genetic models showing that SF-1 deletion exacerbates metabolic dysregulation under high-fat conditions. Recent 2024 investigations using single-cell RNA sequencing have identified SF-1 expression in reserves and neural progenitors outside classical steroidogenic contexts, where it influences alternative mRNA splicing to support and reserve maintenance. These findings, building on earlier observations of low , suggest broader regulatory roles for SF-1 in tissue resilience and progenitor dynamics without overlapping primary steroidogenic pathways.

Regulation

Transcriptional control

The NR5A1 promoter is activated during embryonic development by key transcription factors including WT1, which binds to four specific sites (WB1–WB4) within the proximal promoter region spanning nucleotides -589 to +85, utilizing its -KTS isoform to transactivate expression essential for indifferent formation at stages such as E10.5–E11.5 in mice. GATA4 cooperates with WT1 to enhance NR5A1 transcription in gonadal ridges, contributing to the initiation of steroidogenic cell differentiation. further supports this activation in the developing testis by integrating with upstream signals to sustain NR5A1 levels during sex determination. In contrast, the orphan COUP-TFII represses NR5A1 promoter activity in non-gonadal tissues, such as pituitary gonadotropes, preventing outside steroidogenic lineages. Several conserved enhancers within the 9q33 genomic region flanking the NR5A1 locus facilitate tissue-specific regulation. These elements, spanning over 30 kb around the gene's seven exons, include binding sites for developmental cues that drive expression in adrenal and gonadal primordia. A notable NR5A1-responsive enhancer, identified in 2024, integrates NR5A1 signaling to regulate SRY expression specifically in the nascent testis, reinforcing male gonadal commitment. In adult steroidogenic tissues, NR5A1 expression is induced tissue-specifically by hormones such as ACTH in the and LH in the s, acting through the cAMP-PKA signaling pathway to phosphorylate and stabilize promoter-bound factors. During embryogenesis, NR5A1 is induced in the bipotential by paracrine signals including FGF9 and , establishing a feed-forward that promotes specification. Epigenetic modifications tightly control NR5A1 accessibility, with enrichment of the active mark H3K27ac at promoter and enhancer regions in gonadal ridges facilitating open for binding. Conversely, DNA hypermethylation at CpG islands within the promoter silences NR5A1 in non-steroidogenic tissues, ensuring lineage-restricted expression. Promoter assays reveal cooperative binding dynamics among activators like WT1 and GATA4.

Post-translational modifications

Steroidogenic factor 1 (SF-1), also known as NR5A1, undergoes several post-translational modifications that dynamically regulate its stability, subcellular localization, and transcriptional potency in steroidogenic and endocrine cells. at serine 203 (S203) in the hinge region/ is primarily mediated by 7 (CDK7) within the TFIIH complex, with contributions from (MAPK/ERK) pathways. This modification stabilizes the ligand-binding domain through intramolecular interactions, enhances coactivator recruitment (e.g., GRIP1), and boosts transactivation of target promoters like CYP17, without directly altering DNA binding or subcellular localization. Although direct S203 by (PKA) is not observed, cAMP/PKA signaling indirectly promotes SF-1 activity via downstream and coactivator interactions. Additional sites in the A/B domain, targeted by CDK7, contribute to turnover , with inhibition of CDK7 reducing SF-1 and transcriptional output. SUMOylation occurs predominantly at lysine 194 (K194) in the hinge region and inhibits SF-1 by suppressing S203 , impairing target gene recognition, and sequestering the protein into transcriptionally inactive nuclear bodies. This modification is facilitated by SUMO ligases such as PIASx and PIASy, and its disruption in knockout models leads to ectopic signaling, endocrine organ malformations, and altered steroidogenesis. PIAS family members counteract excessive SUMOylation to fine-tune SF-1 activity in a context-dependent manner. Acetylation of SF-1, catalyzed by p300/CBP in response to signaling, occurs on multiple N-terminal lysines including K34, K38, and K72 within or near the motifs, promoting DNA binding affinity, nuclear clustering, and recruitment of coactivators like GCN5 to enhance of genes such as CYP11A1. This modification integrates hormonal signals to amplify SF-1 function in gonadal and adrenocortical cells. SF-1 stability is further modulated by ubiquitination via the , which targets the protein for proteasomal degradation and limits steroidogenic output, particularly under stress or nutrient deprivation conditions. The basal of SF-1 in adrenal and gonadal cells is extended by /PKA-mediated stabilization independent of S203 phosphorylation. analyses of SF-1 from adrenal cell lines (e.g., Y1 cells) have mapped these modifications, revealing dynamic phospho-sites in the A/B and hinge domains under forskolin stimulation, underscoring their role in signal-responsive turnover. Recent studies (2023-2025) highlight how binding (e.g., to PIP3 or ) in the LBD alters SF-1 conformation, influencing co-regulator affinity and potentially ubiquitination susceptibility, thereby linking to protein stability and broader physiological regulation.

Biological functions

Role in steroidogenesis

Steroidogenic factor 1 (SF-1, also known as NR5A1) serves as a master regulator of steroidogenesis, orchestrating the transcriptional activation of genes essential for the of hormones in adrenal and gonadal tissues. It initiates the process by binding to specific response elements in the promoters of the () and cytochrome P450 side-chain cleavage enzyme (CYP11A1), promoting transport to the mitochondrial inner membrane and its conversion to , the precursor of all hormones. This activation is critical for the rate-limiting first step in steroid production, with SF-1 collaborating with coactivators like CBP/p300 to enhance transcription. Beyond initiation, SF-1 coordinates the downstream multi-enzyme pathway by upregulating key steroidogenic enzymes, ensuring efficient progression from to bioactive hormones. In the adrenal glands, it drives expression of (3β-HSD), cytochrome P450 17α-hydroxylase/17,20-lyase (CYP17), and (CYP21), which are vital for and synthesis. In ovarian tissues, SF-1 similarly induces (CYP19), facilitating production from androgens. These regulatory actions maintain tissue-specific steroid profiles, with SF-1's monomeric DNA-binding enabling precise promoter occupancy. SF-1 also integrates feedback mechanisms to prevent dysregulated steroid output, including autoregulation of its own through intronic enhancers like the fetal adrenal enhancer (FadE), which sustains appropriate levels during steroidogenic demand. Interactions with corepressors such as DAX-1 (NR0B1) further modulate this by repressing SF-1 target genes, forming a loop that inhibits excess activity. In experimental lines, SF-1 overexpression leads to up to 20-fold of CYP11A1 transcription, correlating with enhanced production and underscoring its dosage-sensitive impact. Recent insights reveal SF-1's involvement in phospholipid-mediated fine-tuning of the acute steroid response, where phospholipids like (PI(4,5)P₂) and PI(3,4,5)P₃ bind its ligand-binding domain, stabilizing structure and altering co-regulator recruitment to rapidly adjust transcriptional output in response to hormonal signals. This mechanism, influenced by at serine 203, enables dynamic regulation beyond basal steroidogenesis. Beyond phosphoinositides, SF-1 also binds such as , which represses its transcriptional activity by impairing co-activator recruitment, and other phospholipids like and , further diversifying its modulation of steroidogenic in response to cellular lipid profiles (as of November 2025).

Role in sex determination and reproduction

Steroidogenic factor 1 (SF-1), encoded by the NR5A1 gene, is essential for the formation of the bipotential during early embryonic development, where it regulates the expression of key genes necessary for gonadal specification and initial cell differentiation. In the absence of SF-1, the urogenital fails to develop properly, leading to complete in both sexes. For testis differentiation, SF-1 cooperates with the sex-determining region Y (SRY) protein by synergistically activating the enhancer of , a critical that drives differentiation and subsequent testicular development. Additionally, SF-1 directly regulates SRY expression through a conserved NR5A1-responsive enhancer (E250) located approximately 5 kb upstream of the SRY gene, ensuring timely activation critical for testis determination; disruptions in this enhancer, such as specific base-pair substitutions, reduce SRY expression and are linked to 46,XY (DSD). Additionally, SF-1 interacts with dosage-sensitive sex reversal, adrenal hypoplasia congenita critical region on chromosome X gene 1 () to coordinately upregulate AMH () expression in s, promoting Müllerian duct regression and reinforcing male gonadal fate. In ovarian development and function, SF-1 plays a pivotal role in establishing the follicle reserve, which determines potential. A 2023 study demonstrated that conditional knockout of Nr5a1 in mice results in smaller ovaries with significantly fewer follicles due to increased death and disrupted - interactions, mediated by dysregulated expression and impaired KIT-KITL signaling. SF-1 also supports differentiation and steroidogenic capacity in growing follicles by regulating genes involved in hormone biosynthesis and survival. During , SF-1 is upregulated in periovulatory s, where it directly promotes PTGS2 (prostaglandin-endoperoxide synthase 2) expression in response to , facilitating production essential for cumulus expansion and follicle rupture. In the testis, SF-1 maintains function throughout adulthood, ensuring structural support for by sustaining expression of genes like and those involved in nutrient transport to s. It contributes to estrogen-mediated feedback in the hypothalamic-pituitary-gonadal axis by regulating (CYP19A1) in s, allowing local production that modulates sensitivity and maturation. In humans, SF-1 due to NR5A1 variants underlies in 10-20% of 46,XY (DSD) cases without , often presenting as partial or complete testicular regression and .

Role in neural and metabolic processes

Steroidogenic factor 1 (SF-1), encoded by the NR5A1 gene, is expressed in neurons of the (VMH), where it serves as a key regulator of energy balance and . These SF-1 neurons integrate peripheral signals such as and insulin to modulate feeding behavior, body weight, and systemic metabolism. Disruption of SF-1 in VMH neurons leads to increased susceptibility to diet-induced and impaired glucose regulation, highlighting their role in maintaining metabolic . Specifically, SF-1 neurons facilitate glucose sensing and interact with (POMC)-expressing neurons in the arcuate nucleus to suppress hepatic glucose production and promote insulin sensitivity. In the , SF-1 is indispensable for the maturation and function of gonadotrope cells, directly controlling the transcription of (LH) and (FSH). Conditional knockout of Nr5a1 in the pituitary results in severe , characterized by reduced expression and , underscoring SF-1's role in reproductive axis activation. This pituitary function links SF-1 to the timing of onset, as NR5A1 variants in humans are associated with altered pubertal progression through disrupted signaling and endocrine feedback. Beyond the hypothalamus and pituitary, SF-1 influences metabolic processes through central neural circuits that extend to peripheral tissues. , particularly in the VMH, regulate hepatic by modulating hormone action and stress pathways, such as JNK1, to prevent excessive accumulation and maintain expenditure. This central regulation overlaps functionally with liver receptor homolog-1 (LRH-1, encoded by NR5A2), a related highly expressed in hepatocytes that similarly governs and homeostasis, suggesting compensatory mechanisms in handling across NR5A family members. Recent analyses indicate that SF-1 also contributes to VMH neuronal during , enabling the nucleus's role in integrating metabolic cues for lifelong .

Target genes

In steroidogenic cells

In steroidogenic cells of the and gonads, steroidogenic factor 1 (SF-1, also known as NR5A1) acts as a master transcriptional regulator, directly activating key genes involved in mobilization and biosynthesis. Among its core targets are the (), which facilitates the rate-limiting transport of from the outer to the ; cytochrome side-chain cleavage enzyme (CYP11A1), which converts to ; and 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2), which isomerizes to progesterone and dehydroepiandrosterone to , enabling downstream steroid production. These genes contain SF-1 binding sites in their promoters, allowing SF-1 to coordinate the initial steps of steroidogenesis in response to hormonal stimuli such as (ACTH) in adrenals or in gonads. In adrenal steroidogenic cells, SF-1 further regulates zone-specific enzymes essential for synthesis, including (CYP21A2), which hydroxylates progesterone and 17-hydroxyprogesterone to their respective 11-deoxycorticosteroid intermediates, and 11β-hydroxylase (CYP11B1), which catalyzes the final step in production by converting 11-deoxycortisol to . These targets ensure efficient and output in the and fasciculata, respectively, with SF-1 binding directly to promoter elements to maintain basal and stimulated expression. In gonadal steroidogenic cells, such as Leydig cells in the testis and cells in the , SF-1 promotes the expression of 17α-hydroxylase/17,20-lyase (), which introduces a hydroxyl group at the 17-position of or progesterone and cleaves the to produce , and 17β-hydroxysteroid dehydrogenase (HSD17B family members, particularly HSD17B3 in testes), which reduces to testosterone or estrone to , thereby driving sex steroid biosynthesis. This regulation supports and reproductive function by fine-tuning and levels. SF-1 binds to genomic sites enriched for nuclear receptor motifs, facilitating broad control over steroidogenic and related pathways.

In gonadal support cells

In of the testis, steroidogenic factor 1 (SF-1, encoded by NR5A1) plays a critical role in maintaining testicular structure and function by directly regulating key genes essential for support cell identity and differentiation. Specifically, SF-1 cooperates with to bind the promoter of the (AMH) gene, driving its expression to promote Müllerian duct regression and Sertoli cell maturation during early gonadal development. This interaction ensures proper testis maintenance, as SF-1 and together activate AMH transcription in differentiating Sertoli cells. Additionally, SF-1 sustains expression in Sertoli cells, reinforcing their supportive role in and testicular cord formation. In ovarian granulosa cells, SF-1 supports follicle development by regulating genes involved in hormone responsiveness and follicular growth. SF-1 activates the promoter of the (FSHR) in a dose-dependent manner, enhancing sensitivity to FSH and thereby promoting antral follicle maturation. It also influences the expression of INHA, the gene encoding the inhibin α subunit, which modulates proliferation and inhibits FSH-stimulated production to fine-tune follicular dynamics. These actions underscore SF-1's importance in function beyond steroid production. SF-1 facilitates interactions between theca and granulosa cells in the by indirectly upregulating (CYP19A1) expression in granulosa cells through coordinated signaling pathways, such as those involving FSH and TGFβ1, which enhance from theca-derived androgens. Experimental knockdown of SF-1 in lines results in a substantial reduction in AMH expression, approximately a 10-fold decrease, highlighting its essential role in maintaining Sertoli-specific gene programs. More recently, a 2023 study using /Cas9-mediated activation of NR5A1 in human embryonic stem cell (hESC)-derived bipotential gonadal-like cells demonstrated that SF-1 overexpression steers cells toward an ovarian support cell fate, upregulating granulosa-like genes such as FOXL2 and WNT4 while suppressing male-biased markers.

In endocrine and neural cells

In pituitary gonadotropes, steroidogenic factor 1 (SF-1, also known as NR5A1) directly regulates the expression of key genes involved in gonadotropin synthesis and responsiveness to gonadotropin-releasing hormone (GnRH). SF-1 binds to specific response elements in the promoter of the luteinizing hormone β subunit (LHB) gene, where it is essential for basal promoter activity and GnRH-stimulated transcription, as demonstrated by mutagenesis studies showing near-complete loss of LHB promoter function upon disruption of the SF-1 binding site. Similarly, SF-1 interacts with the transcription factor NF-Y to synergistically activate the follicle-stimulating hormone β subunit (FSHB) promoter, enhancing FSHB expression critical for reproductive hormone regulation. For GnRH responsiveness, SF-1 binds to a gonadotrope-specific enhancer element in the gonadotropin-releasing hormone receptor (GNRHR) promoter, facilitating cell-specific expression and GnRH-mediated gonadotropin secretion; disruption of this site abolishes enhancer activity in gonadotrope cell lines. These regulatory actions underscore SF-1's role in coordinating pituitary gonadotrope function, with SF-1 knockout models exhibiting profound reductions in LHB, FSHB, and GNRHR expression, leading to infertility. In the ventromedial hypothalamus (VMH), SF-1 maintains its own expression through autoregulation via VMH-specific enhancers located in intron 6 of the NR5A1 gene, which drive tissue-restricted transcription essential for VMH neuronal and . This autoregulatory mechanism ensures sustained SF-1 levels in VMH neurons, which integrate metabolic and reproductive signals. SF-1 also governs melanocortin signaling in the VMH by promoting expression of the (MC4R) in SF-1-positive neurons, where MC4R mediates anorexigenic effects of α-melanocyte-stimulating hormone; conditional restoration of MC4R in SF-1 neurons rescues feeding and energy balance defects in MC4R knockout mice, highlighting SF-1's upstream control in this pathway. Beyond the , SF-1 influences neural gene expression in -producing neurons and broader processes. In neurons, SF-1 upregulates (TH), the rate-limiting enzyme in dopamine biosynthesis, by binding to conserved motifs in the TH promoter to support neuronal differentiation and catecholamine production during development. For , SF-1 directly activates (BDNF) expression in VMH neurons through SF-1-responsive elements in the BDNF promoter, promoting and neuronal survival; SF-1 leads to halved BDNF levels and impaired VMH circuit formation, as observed in heterozygous models. These targets link SF-1 to maturation and . SF-1 integrates with other transcription factors in pituitary development via a regulatory loop involving SOX3, where SOX3 binds the NR5A1 promoter to enhance SF-1 expression, while SF-1 reciprocally supports SOX3-dependent proliferation and gonadotrope ; disruptions in this loop, as seen in SOX3 mutants, result in pituitary and reduced SF-1-dependent . Recent enhancer studies have identified SF-1-responsive elements that parallel SRY-SF-1 synergies in gonadal sex determination, extending to neural contexts where SF-1 activates SRY-like targets to drive sex-dimorphic in developing neurons. A 2024 analysis of sex-biased neural enhancers revealed SF-1 binding motifs regulating dimorphic trajectories in induced pluripotent stem cell-derived neurons, contributing to sexually divergent and potentially underlying sex differences in neural disorders.

Experimental models

Knockout and loss-of-function studies

The initial targeted disruption of the NR5A1 gene encoding steroidogenic factor 1 (SF-1) in mice, generated in 1994, resulted in complete of the adrenal glands and gonads, accompanied by perinatal lethality due to . These homozygous null mice exhibited additional developmental defects, including of the ventromedial hypothalamic nucleus and ambiguous external genitalia with persistent Müllerian structures in individuals. A parallel study confirmed the absence of steroidogenic tissues and normal embryonic levels maintained by placental expression of steroidogenic enzymes, underscoring SF-1's essential role in rather than acute steroid production. To overcome the lethality of global knockouts and dissect SF-1 functions, transgenic rescue approaches were employed, where SF-1 expression driven by tissue-specific promoters restored adrenal development and steroidogenesis in null backgrounds. Specifically, SF-1 overexpression in steroidogenic lineages reactivated key target genes such as STAR (steroidogenic acute regulatory protein), enabling cholesterol transport and cortisol production that partially mitigated the agenesis phenotype. Conditional knockout models have further elucidated tissue-specific roles. Ventromedial hypothalamus (VMH)-specific SF-1 inactivation, achieved via SF-1-Cre recombination, produced viable mice with disrupted energy homeostasis, including increased adiposity, heightened susceptibility to high-fat diet-induced obesity, and impaired thermogenic responses to cold or β-adrenergic stimuli. Gonad-specific knockouts, bypassing embryonic lethality, revealed severe hypoplasia of testes and ovaries with failed gonadal descent; XY mutants displayed female-like external genitalia and Müllerian duct persistence, phenocopying aspects of disorders of sex development (DSD). In humans, NR5A1 haploinsufficiency from heterozygous loss-of-function variants manifests milder phenotypes than complete null states, often including isolated , , or mild in 46,XY individuals without full adrenal failure. Recent tissue-specific conditional knockouts in ovarian models (2023) using Amhr2-Cre demonstrated that SF-1 ablation in granulosa cells drastically reduces the primordial follicle reserve through defective follicle assembly and accelerated oocyte atresia, linking SF-1 to maintenance.

Gain-of-function and variant analyses

Overexpression of steroidogenic factor 1 (SF-1, encoded by NR5A1) in adrenocortical cell lines, such as the human H295R carcinoma line, promotes and alters production, reducing and aldosterone while maintaining dehydroepiandrosterone sulfate secretion. In transgenic mouse models with elevated SF-1 dosage, adrenocortical develops alongside ovarian , leading to disrupted follicular development and premature , contrasting with the observed in loss-of-function models. More than 218 NR5A1 variants have been documented in the Human Gene Mutation Database, with a substantial proportion consisting of missense mutations clustered in the (DBD) and ligand-binding domain (LBD). A 2024 international identified and characterized 35 novel NR5A1 variants in individuals with differences of sex development, revealing variable where some carriers exhibited severe gonadal phenotypes while others were unaffected. Functional assays of these variants, including transactivation on the CYP11A1 promoter in HEK293T cells, demonstrated reduced transcriptional activity for approximately 40% of DBD-located missense changes and variable impairment (often 20-60% reduction relative to wild-type) for LBD variants, with some showing disrupted nuclear translocation. Specific LBD missense variants, such as R255L, impair phospholipid ligand binding (e.g., to phosphoinositides like PIP2 and PIP3) within the hydrophobic core, compromising interdomain communication between the LBD and DBD and further attenuating activity by up to 80% in reporter assays. CRISPR/Cas9-mediated activation of endogenous NR5A1 in female human embryonic stem cells (hESCs), as reported in a 2023 study, promotes differentiation toward steroidogenic lineages by upregulating gonadal markers and enzymes like CYP11A1, facilitating the generation of bipotential gonadal-like cells biased toward steroid production. Dosage effects from NR5A1 duplications are rare in humans but mirror overexpression models, associating increased SF-1 levels with and metabolic disruptions in ovarian tissues.

Clinical significance

Adrenal and gonadal disorders

Mutations in the NR5A1 gene, which encodes steroidogenic factor 1 (SF-1), are a recognized cause of primary (PAI), accounting for approximately 10-15% of cases associated with 46,XY (DSD), though overall prevalence in isolated PAI is lower, around 1-2% in pediatric cohorts of unknown . Symptoms of SF-1-related PAI typically manifest in infancy or early childhood and include salt-wasting crises due to mineralocorticoid deficiency, from shortfall, and , often requiring urgent intervention to prevent life-threatening . Pathophysiologically, these mutations lead to impaired adrenal during embryonic development, resulting from SF-1 that disrupts the and survival of steroidogenic cells, as evidenced by complete adrenal in homozygous models. In the gonadal context, homozygous NR5A1 mutations produce a resembling lipoid (CAH), characterized by severe steroidogenic defects, , and profound adrenal failure due to near-total loss of SF-1 function, leading to accumulation of precursors and organ hypoplasia. Heterozygous mutations, more commonly reported, are associated with 46,XY or 46,XX , presenting with varying degrees of testicular or ovarian failure, ambiguous genitalia in 46,XY individuals, and primary amenorrhea in 46,XX cases, without consistent adrenal involvement. To date, over 300 cases of NR5A1-related disorders have been documented in international cohorts, highlighting the variable expressivity from complete to milder dysgenesis, mirroring embryonic SF-1 loss observed in loss-of-function animal models.00507-8/fulltext) Management of SF-1-related adrenal and gonadal disorders centers on lifelong , including glucocorticoids (e.g., ) and mineralocorticoids (e.g., ) for PAI, with dosing adjusted to mimic physiological rhythms and stress responses. For , supplemental sex hormones may be required post-puberty to support secondary sexual characteristics, alongside monitoring for associated risks like during illness. Currently, no or targeted molecular interventions are available, though ongoing research into SF-1 agonists holds potential for future therapeutic advances.

Disorders of sex development

Mutations in the NR5A1 gene, encoding steroidogenic factor 1 (SF-1), are a significant cause of 46,XY (DSD), accounting for approximately 10-20% of cases in specialized cohorts. These variants often lead to complete or partial , characterized by underdeveloped testes and resulting in phenotypes ranging from female-typical external genitalia to ambiguous genitalia with varying degrees of and . SF-1's critical role in gonadal development contributes to these disruptions by impairing testis differentiation and steroidogenesis. In 46,XX individuals, NR5A1 variants are rarer and typically associated with rather than overt DSD, though cases of testicular DSD or ovotesticular DSD have been reported, potentially leading to or due to aberrant gonadal function. These phenotypes arise from SF-1's influence on ovarian determination, where loss-of-function effects can disrupt normal female gonadal development. The clinical presentation of NR5A1 variants exhibits incomplete , with estimates suggesting 30-60% of carriers showing incomplete or mild DSD phenotypes, influenced by genetic modifiers such as variations in SOX9 dosage that affect SF-1-mediated enhancer activation. Recent studies highlight how SOX9 interactions modulate SF-1 function in sex determination pathways. Diagnosis of NR5A1-related DSD relies on next-generation sequencing (NGS) panels targeting DSD-associated genes, which enable identification of heterozygous loss-of-function variants in both 46,XY and 46,XX cases. Prenatal assessment may involve evaluating expression levels, which are often reduced in affected fetuses, aiding early detection through genetic screening. Management focuses on multidisciplinary care, including hormone replacement and surgical options for genital reconstruction, with fertility preservation strategies such as testicular sperm extraction recommended at for 46, individuals to mitigate progressive gonadal failure. Emerging therapeutic approaches, including /Cas9-mediated activation of NR5A1 in models, show promise for correcting gonadal defects as of 2025, potentially enabling steroidogenic cell fate restoration.

Reproductive and metabolic conditions

Steroidogenic factor 1 (SF-1, encoded by NR5A1) plays a critical role in reproductive function, with variants contributing to through disruptions in gonadal development and steroidogenesis. In males, heterozygous NR5A1 mutations are identified in approximately 4-12% of cases of idiopathic spermatogenic failure, often leading to impaired and due to reduced expression of genes essential for testicular and hormone production. In females, these variants are associated with (POI), affecting 2-8% of sporadic and familial POI cases, resulting in diminished ovarian follicle reserve and ovulation defects that manifest as premature amenorrhea and . Overall, NR5A1 variants appear in 2-5% of broader infertile cohorts, highlighting their underrecognized contribution to non-obstructive phenotypes. Recent studies have elucidated the impact of splicing variants on , with 2024 analyses identifying non-canonical splicing alterations in NR5A1 among men, leading to aberrant protein isoforms that compromise gonadal steroidogenic pathways and exacerbate spermatogenic arrest. For (PCOS), polymorphisms and overactivity in SF-1 are implicated in increased disease risk through androgen dysregulation, as excess NR5A1 expression promotes and ovarian dysfunction akin to PCOS features, though direct causal variants remain rare. Therapeutic approaches for SF-1-related include assisted reproductive technologies like fertilization (IVF), where success rates vary based on residual ovarian or testicular function, often necessitating oocyte or cryopreservation for preservation. Beyond reproduction, SF-1 influences metabolic , particularly through its expression in ventromedial (VMH) neurons, where loss-of-function disrupts energy balance. Conditional of Nr5a1 in VMH SF-1 neurons in mice recapitulates and glucose intolerance, mimicking features of and via impaired insulin and signaling. Human carriers of deleterious NR5A1 variants exhibit elevated risks for and adverse metabolic outcomes, including links to non-alcoholic (NAFLD) through dysregulation of targets like those involved in hepatic . Emerging metabolic therapies targeting the NR5A1 pathway, such as modulators, show promise in preclinical models for mitigating and , though clinical applications remain investigational with a focus on and interventions.

References

  1. [1]
    Steroidogenic factor-1 (SF-1, NR5A1) and human disease - PMC
    Steroidogenic factor-1 (SF-1, Ad4BP, encoded by NR5A1) is a key regulator of adrenal and reproductive development and function.
  2. [2]
    Steroidogenic Factor 1, a Goldilocks Transcription ... - PubMed Central
    Feb 10, 2023 · A nuclear receptor transcription factor that plays a crucial role in the regulation of adrenal and gonadal development, function and maintenance.
  3. [3]
    NR5A1 nuclear receptor subfamily 5 group A member 1 [ (human)]
    Aug 19, 2025 · The NR5A1 (previously called Ad4BP or SF-1) gene which is exclusively expressed in the adrenal cortex and steroidogenic tissue can form atypical ...
  4. [4]
    Nr5a1 nuclear receptor subfamily 5, group A, member 1 ... - NCBI
    Sep 24, 2025 · Enables RNA polymerase II cis-regulatory region sequence-specific DNA binding activity and chromatin binding activity.Missing: chromosomal | Show results with:chromosomal
  5. [5]
    184757 - NUCLEAR RECEPTOR SUBFAMILY 5, GROUP A ... - OMIM
    (1995) mapped the human NR5A1 gene to chromosome 9q33 by fluorescence in situ hybridization. ... (1996) determined that the NR5A1 gene contains 7 exons and spans ...
  6. [6]
    Phenotype and Molecular Characterizations of 30 Children From ...
    Oct 29, 2018 · The NR5A1 gene has one nontranslated exon (exon 1) and six other coding exons (exon 2–7). SF1 has two zinc finger DNA-binding domains (DBDs) ...Missing: details | Show results with:details
  7. [7]
    Can Non-Coding NR5A1 Gene Variants Explain Phenotypes of ...
    Feb 14, 2023 · Using biochemical experiments and DNA binding assays, they characterized 4 WT1 binding sites in the Nr5a1 promoter, which all share the GTGGG ...Subjects And Methods · In Vitro And In Silico... · Results
  8. [8]
    Gene: NR5A1 (ENSG00000136931) - Summary - Ensembl
    Chromosome 9: 124,481,231-124,507,444 reverse strand. ... This gene has 15 transcripts (splice variants), 275 orthologues, 1 paralogue and is associated with 10 ...
  9. [9]
    Clinical spectrum and molecular basis in 19 Chinese patients with ...
    Dec 2, 2024 · This study aimed to provide a comprehensive analysis of the clinical characteristics and molecular defects observed in 19 Chinese patients with NR5A1 variants.
  10. [10]
    Genomic Analysis of the Nuclear Receptor Family: New Insights Into ...
    The exon structure of the ligand-binding domain suggests that exon shuffling has played a role in the evolution of this family. An invariant splice junction in ...Figure 3 · Exon Structures Of Dbd And... · Figure 5
  11. [11]
    A common trans-acting factor, Ad4-binding protein, to the ... - PubMed
    Sep 5, 1992 · The competition experiments revealed that Ad4BP bound not only to (C/T)CAAGG(T/C)(C/T), which was originally identified as the Ad4 binding site, ...
  12. [12]
    Cloning and sequence analysis of the human gene encoding ...
    Cloning and sequence analysis of the human gene encoding steroidogenic factor 1. in Journal of Molecular Endocrinology. Authors: M Wong.
  13. [13]
  14. [14]
    Steroidogenic Factor-1 form and function: from phospholipids to ...
    Jan 1, 2025 · SF-1 was shown to bind and be regulated by phospholipids, one of the most interesting aspects of SF-1 regulation is the manner in which SF-1 ...Missing: splicing | Show results with:splicing
  15. [15]
    NR5A1 Gene - GeneCards | STF1 Protein | STF1 Antibody
    The protein encoded by this gene is a transcriptional activator involved in sex determination. The encoded protein binds DNA as a monomer.
  16. [16]
    NR5A1 - Steroidogenic factor 1 - Homo sapiens (Human) | UniProtKB
    Transcriptional activator. Essential for sexual differentiation and formation of the primary steroidogenic tissues (PubMed:27378692).
  17. [17]
  18. [18]
  19. [19]
    Sex-specific splicing of Z- and W-borne nr5a1 alleles suggests sex ...
    Three transcript isoforms of Z-nr5a1 were detected in gonads of adult ZZ males, two of which encode a functional protein. However, ZW females produced 16 ...
  20. [20]
    A functionally conserved member of the FTZ‐F1 nuclear receptor ...
    Oct 30, 2002 · Within the coding region the intron–exon structure of Smftz-f1 is conserved in relation to members of the NR5A2 family (including human FTF/LRH ...
  21. [21]
    Expression and Regulation of Fushi Tarazu factor-1 and ... - PubMed
    Teleost fushi tarazu factor-1 (FTZ-F1) is a potential regulator of steroidogenesis. The present study shows sex-specific regulation of Arctic char fushi ...Missing: ancestral | Show results with:ancestral
  22. [22]
  23. [23]
    [PDF] The wide clinical expression spectrum presented in two families with ...
    NR5A1 is highly conserved among different species, and the homology of mice is up to 95% of human NR5A1 gene [6]. In 46, XY individuals, SF-1 activates the.<|separator|>
  24. [24]
    Evolutionary Genomics of Nuclear Receptors: From Twenty-Five ...
    This phylogenetic reconstruction shows that receptors for similar ligands do not group in the tree, but are interspersed with receptors for totally different ...
  25. [25]
    Steroidogenic factor 1 messenger ribonucleic acid expression in ...
    Northern blot analyses of peripheral tissues revealed high hSF-1 mRNA expression in the adrenal cortex and the gonads, but no hSF-1 mRNA was detected in the ...
  26. [26]
    The role of steroidogenic factor 1 (SF-1) in steroidogenic cell ...
    Dec 2, 2022 · The nuclear receptor steroidogenic factor 1 (SF-1) is essential for mature mouse gonad steroidogenic gene expression, for Leydig and Sertoli cell function.
  27. [27]
    POD-1/Tcf21 overexpression reduces endogenous SF-1 and StAR ...
    ... adrenal cortex. Sf-1 mRNA levels were significantly higher in adrenal cells by 1.53±0.31 (P=0.045) and 2.48±0.32 (P=0.0014) fold in G and F/R cells ...
  28. [28]
    SF-1 a key player in the development and differentiation of ...
    While trying to uncover the molecular mechanisms controlling steroidogenic genes expression, two independent research teams identified an AGGTCA (Ad4) motif in ...
  29. [29]
    Impact of ACTH Signaling on Transcriptional Regulation of ... - PMC
    Mar 29, 2016 · Steroidogenic factor-1 (SF-1; NR5A1) is an orphan member of the nuclear receptor superfamily, which acts as a key regulator of adrenogonadal ...
  30. [30]
    Steroidogenic Acute Regulatory Protein Expression in the ... - Frontiers
    Regulation of StAR Expression. In endocrine tissues, trophic hormones like LH and ACTH control StAR expression and hence, steroid production. The identities ...
  31. [31]
    An integrated single-cell analysis of human adrenal cortex ...
    At a global level (bulk RNA-Seq), the developing adrenal gland showed a well-defined transcriptomic profile compared with control tissues. This transcriptome ...
  32. [32]
    The conditional deletion of steroidogenic factor 1 (Nr5a1) in Sox9 ...
    Feb 24, 2021 · ... urogenital ridge in both sexes also expresses Nr5a1 from E9.5. However, around the time of testicular morphogenesis at E12.5, Nr5a1 ...
  33. [33]
    Regulation of the adrenocortical stem cell niche - NIH
    SF-1 expression starts at E9.0 in the urogenital ridge, where it specifies precursors of the steroidogenic lineage and leads to the formation of the AGP. Mice ...
  34. [34]
    Steroidogenic Factor 1 (Nr5a1) is Required for Sertoli Cell Survival ...
    Mar 14, 2019 · The elevated level of Steroidogenic Factor 1 (Nr5a1, Sf-1) expression in the male gonadal development pathway, post sex determination, ...
  35. [35]
    Role of NR5A1 Gene Mutations in Disorders of Sex Development
    May 9, 2024 · NR5A1 interacts with the WT1-KTS complex already bound to the DNA-binding domain of AMH and synergistically up-regulates AMH expression [12].
  36. [36]
    Temporal and spatial expression of liver receptor homologue‐1 ...
    Jul 7, 2005 · During testis development, SF-1–expressing cells of coelomic epithelium rapidly proliferate and then migrate into the urogenital ridge where ...<|control11|><|separator|>
  37. [37]
    Role of NR5A1 Gene Mutations in Disorders of Sex Development
    May 9, 2024 · Several transcription factors, including NR5A1, SOX9, GATA4 and WT1, regulate AMH transcription. SOX9 and NR5A1 bind to a conserved site in the ...
  38. [38]
    Single-cell roadmap of human gonadal development - Nature
    Jul 6, 2022 · This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis.
  39. [39]
    Leptin Directly Activates SF1 Neurons in the VMH, and This Action ...
    In the present study, we show that leptin depolarizes and increases the firing rate of steroidogenic factor-1 (SF1)-positive neurons in the VMH.
  40. [40]
    Steroidogenic factor 1 directs programs regulating diet-induced ...
    Jun 2, 2011 · Recently, several studies have suggested that neurons expressing SF-1 in the VMH play critical roles in regulating energy balance and glucose ...
  41. [41]
    NR5A1 and cell population heterogeneity - Wiley Online Library
    Dec 9, 2024 · Abstract. Background: NR5A1 plays essential roles in the development of various tissues, including the ventromedial hypothalamus, ...Missing: ortholog | Show results with:ortholog<|separator|>
  42. [42]
    Steroidogenic factor 1 (SF-1; Nr5a1) regulates the formation ... - PNAS
    In this study, we identified the orphan nuclear receptor Steroidogenic factor 1 (SF-1) as a regulator of the formation of the ovarian follicle reserve, ...
  43. [43]
    Phosphorylation of Steroidogenic Factor 1 Is Mediated by Cyclin ...
    Sep 27, 2007 · We found that SF1 interacts with a CDK7-containing complex and that SF1 is phosphorylated in a TFIIH-dependent fashion on S203. Furthermore, we ...
  44. [44]
    Steroidogenic Factor 1: Its Roles in Differentiation, Development ...
    Our contributions to SF-1 biology (and connections to Parker) began in 1984 when we met at a symposium. White had recently cloned the cytochrome P450 (CYP)21 ...
  45. [45]
    SUMOylation Inhibits SF-1 Activity by Reducing CDK7-Mediated ...
    S203 phosphorylation serves to enhance coactivator binding and the transactivation potential of this receptor. Recent data indicate that SF-1 can be ...
  46. [46]
  47. [47]
    SF-1 (nuclear receptor 5A1) activity is activated by cyclic AMP via ...
    SF-1 transcriptional activity is regulated by p300 in response to the cAMP signaling pathway by way of increased acetylation, DNA binding, and recruitment to ...Missing: K27 | Show results with:K27
  48. [48]
    Steroidogenic factor-1: a key regulator of gonadotroph gene ...
    ... half-life of SF-1 protein in its target tissues (Aesoy et al. 2002). The control of SF-1 expression in the pituitary is poorly elucidated. Ovariectomised ...
  49. [49]
    Sex determination involves synergistic action of SRY and SF1 on a ...
    Aug 9, 2025 · The Y chromosome sex-determining region (SRY) gene triggers testicular differentiation from early bipotential gonads by up-regulating SRY-box ...
  50. [50]
    Nuclear receptors Sf1 and Dax1 function cooperatively to mediate ...
    May 15, 2005 · In the embryonic gonad, we found that Sf1 and Dax1 act coordinately to enhance the expression of the Sertoli-derived factors Dhh and Amh. Dhh is ...
  51. [51]
    The role of steroidogenic factor 1 (SF-1) in steroidogenic cell ...
    Dec 2, 2022 · The nuclear receptor steroidogenic factor 1 (SF-1) is essential for mature mouse gonad steroidogenic gene expression, for Leydig and Sertoli cell function.
  52. [52]
    Steroidogenic Factor 1 Regulation of the Hypothalamic-Pituitary ...
    The orphan nuclear receptor steroidogenic factor-1 (SF-1 or NR5A1) is an indispensable regulator of adrenal and gonadal formation.<|control11|><|separator|>
  53. [53]
    Estrogen and Spermatogenesis* | Endocrine Reviews
    FSH targets receptors within the Sertoli cell to regulate spermatogenesis by stimulating the production of numerous Sertoli cell factors. The roles of ...Iv. Ers, Aromatase, And... · A. Ers And Aromatase In The... · C. Ers And Aromatase In The...
  54. [54]
    Case Report: Severe Gonadal Dysgenesis Causing 46,XY Disorder ...
    Jul 4, 2022 · Mutations in the nuclear receptor subfamily 5 group A member 1 (NR5A1) are the underlying cause of 10–20% of 46,XY disorders of sex development ...
  55. [55]
    The Roles of SF-1 Neurons in Energy Homeostasis - PubMed Central
    Steroidogenic factor 1 regulates expression of the cannabinoid receptor 1 in the ventromedial hypothalamic nucleus. Mol. Endocrinol. 22, 1950–1961 10.1210 ...
  56. [56]
    New Insights of SF1 Neurons in Hypothalamic Regulation of Obesity ...
    Jun 8, 2021 · SF1 neurons in the ventromedial hypothalamus are a specific lead in the brain's ability to sense glucose levels and conduct insulin and leptin signaling.
  57. [57]
    Steroidogenic factor 1 (SF1) is essential for pituitary gonadotrope ...
    Jan 15, 2001 · The pituitary-specific SF1 knockout mice are a novel genetic model of hypogonadotropic hypogonadism that establishes essential role(s) of SF1 in ...Missing: modulation | Show results with:modulation
  58. [58]
    7 Broader impact and outcome of human NR5A1/SF1 variants
    This review highlights the expanding phenotypic spectrum associated with NR5A1/SF-1 variants, encompassing not only differences of sex development (DSD) and ...<|control11|><|separator|>
  59. [59]
    JNK1 in SF1 neurons regulates the central action of thyroid ...
    Our data demonstrated for the first time that JNK1 in SF1 neurons is necessary for the regulation of hepatic lipid metabolism, cold adaptation and central T3 ...
  60. [60]
    LRH-1 regulates hepatic lipid homeostasis and maintains ...
    Mar 8, 2018 · Small molecule agonists of the orphan nuclear receptors steroidogenic factor-1 (SF-1, NR5A1) and liver receptor homologue-1 (LRH-1, NR5A2).
  61. [61]
    Contributions of Steroidogenic Factor 1 to the Transcription ... - NIH
    SF-1 antibodies precipitated greater amounts of promoter DNA from the Star, Cyp11b1 and Cyp11a1 genes in parent Y1 cells than did a control IgG (Fig. 4A), ...
  62. [62]
    Steroidogenic differentiation of human amniotic membrane-derived ...
    Sep 17, 2024 · SF-1 considerably increased the expression of StAR, CYP11A1, and HSD3B2 in hAmMSCs. In contrast, WT1+KTS and WT1−KTS did not alter the mRNA ...
  63. [63]
    Transcriptional Regulation of Steroidogenic Genes - Sage Journals
    Aug 1, 2009 · This review will provide an overview of the hormonal and transcriptional regulation of the STARD1, CYP11A1 and specific steroidogenic HSD3B ...
  64. [64]
    Genetics and Pathophysiology of Classic Congenital Adrenal ...
    Jan 21, 2025 · The most important transcription factor for adrenal-specific expression of CYP21A2 is steroidogenic factor-1 (SF-1, Ad4BP, NR5A1). This protein ...
  65. [65]
    Expression Profiles of SF-1, DAX1, and CYP17 in the Human Fetal ...
    In this study, we investigate the regulation of the human CYP17 gene by two orphan nuclear receptors, steroidogenic factor 1 (SF-1) and DAX1.Missing: gonadal HSD17B
  66. [66]
    GSE186606 - Steroidogenic factor 1 (NR5A1) induces multiple ...
    Genomics Genome-wide SF-1 (NR5A1) binding sites in NCI-H295R cells. Project description:The nuclear receptor steroidogenic factor-1 (SF-1, NR5A1) is a key ...
  67. [67]
    Expression of Steroidogenic Factor 1 in the Testis Requires an E ...
    The promoters of these genes contain a common regulatory motif (AGGTCA) through which SF-1 binds and activates transcription. More recently, SF-1's role in ...Missing: ChIP- | Show results with:ChIP-
  68. [68]
    Evaluating the role of aldosterone synthesis on adrenal cell fate
    Aug 6, 2024 · This review aims to summarize the development of the adrenal glands, the key regulators of adrenal cortex homeostasis, and the dysregulation of this ...
  69. [69]
    Direct interaction of SRY-related protein SOX9 and steroidogenic ...
    In this study, we demonstrate that the canonical SOX-binding site within the human AMH proximal promoter can bind the transcription factor SOX9.
  70. [70]
    SOX9 and SF1 are involved in cyclic AMP-mediated upregulation of ...
    In the Sertoli cell line SMAT1 cAMP upregulates SOX9, SF1, and GATA4 expression and induces SOX9 and SF1 nuclear translocation mainly through PKA.
  71. [71]
  72. [72]
    Switching of NR5A proteins associated with the inhibin alpha ...
    The inhibin alpha-subunit gene is transcriptionally activated by FSH in ovarian granulosa cells during follicular growth. We have investigated the roles of the ...Missing: NR5A1 regulates
  73. [73]
    Calcineurin and CRTC2 mediate FSH and TGFβ1 upregulation of ...
    Aromatase is also regulated by the NR5A family members LRH1/NR5A2 and SF1/NR5A1, as both are expressed in granulosa cells and could bind to the two NR5A- ...Aromatase Activity Assay · Results · Fsh And Tgfβ1 Regulation Of...<|separator|>
  74. [74]
    Steroidogenic Factor 1 (Nr5a1) is Required for Sertoli Cell Survival ...
    Mar 14, 2019 · A strong reduction in AMH expression in SC-SF-1−/− Sertoli cells. To determine Nr5a1 role in the regulation of AMH in Sertoli cells, we ...Missing: knockdown | Show results with:knockdown
  75. [75]
    CRISPR/Cas9-mediated activation of NR5A1 steers female human ...
    NR5A1 plays an important role in the development and function of steroidogenic tissues, gonads and adrenal glands [17–19]. NR5A1 knock-out (KO) mice completely ...
  76. [76]
    Increased Steroidogenic Factor-1 Dosage Triggers Adrenocortical ...
    A frequent feature of childhood adrenocortical tumors is SF-1 amplification and overexpression. Here we show that an increased SF-1 dosage can by itself augment ...
  77. [77]
    Constitutive expression of Steroidogenic factor-1 (NR5A1) disrupts ...
    In the mouse, NR5A1 is expressed in somatic cells of undifferentiated gonads of both XX and XY embryos and loss of Nr5a1 results in gonadal agenesis (6, 9).
  78. [78]
    Characterization of 35 Novel NR5A1/SF-1 Variants Identified in ...
    Novel, likely disease-causing NR5A1/SF-1 variants from the SF1next cohort of individuals with DSD were characterized to elucidate their pathogenic effect.
  79. [79]
  80. [80]
    CRISPR/Cas9-mediated activation of NR5A1 steers female human ...
    Sep 20, 2023 · ... NR5A1 induction did not seem to affect the bipotential gonadal markers gata binding protein 4 (GATA4) and Wilms' tumour suppressor 1 (WT1) ...
  81. [81]
    Analysis of DAX1 (NR0B1) and Steroidogenic Factor-1 (NR5A1) in ...
    Our objective was to investigate the prevalence of DAX1 and SF1 mutations in children and adults with primary adrenal failure of unknown etiology.Missing: pathophysiology | Show results with:pathophysiology
  82. [82]
    Clinical and genetic characteristics of a large international cohort of ...
    Among 197 individuals with NR5A1/SF-1 variants, we confirmed diverse phenotypes. Over 70% of 46, XY individuals had a severe DSD phenotype, while 90% of 46, ...
  83. [83]
    A Novel NR5A1 Mutation in a Thai Boy with 46, XY DSD - PMC
    It has been discovered to be responsible for 10 to 20% of 46, XY DSD cases. Here, we described a 2-month-old infant who had ambiguous genitalia and 46, XY.
  84. [84]
    Clinical Spectrum, Surgical Management, and Outcomes of NR5A1 ...
    In the sex differentiation stage, the roles of NR5A1 are: regulation of AMH and AMHR2 expression by Sertoli cells, promoting the transcription of steroidogenic ...
  85. [85]
    Novel Variant in NR5A1 Presenting as 46,XY Difference of Sex ...
    Sep 15, 2023 · The NR5A1 gene located on chromosome 9q33.3 encodes steroidogenic factor-1 (SF-1), a key transcription factor involved in the regulation of ...
  86. [86]
    NR5A1 is a novel disease gene for 46,XX testicular and ... - Nature
    Aug 4, 2016 · A conserved NR5A1-responsive enhancer regulates SRY in testis-determination. Article Open access 30 March 2024. Expanding the phenotype of ...<|separator|>
  87. [87]
    recurrent p.Arg92Trp variant in steroidogenic factor-1 (NR5A1) can ...
    A recurrent p.Arg92Trp variant in steroidogenic factor-1 (NR5A1) can act as a molecular switch in human sex development.Results · Clinical Phenotypes · Nr5a1 Is Expressed In...
  88. [88]
    Mutant NR5A1/SF‐1 in patients with disorders of sex development ...
    Mutant NR5A1/SF‐1 in patients with disorders of sex development shows defective activation of the SOX9 TESCO enhancer · 13 Citations · 47 References.
  89. [89]
    Case Report: Severe Gonadal Dysgenesis Causing 46,XY Disorder ...
    Jul 5, 2022 · The STRING protein interaction shows that there is a strong interaction between NR5A1 and several other DSD proteins such as SOX9 (Figure 3C).
  90. [90]
    Diagnostic Application of Targeted Next-Generation Sequencing of ...
    Mar 15, 2017 · This study supports the notion that next-generation sequencing can be an efficient tool in the clinical diagnosis and variant discovery in DSD.
  91. [91]
    Invitae Disorders of Sex Development Panel | Test catalog
    Genetic testing for genes associated with disorders of sex development, characterized by atypical gonadal or anatomical sex development often presenting ...
  92. [92]
    Case Report: Longitudinal follow-up and testicular sperm extraction ...
    Jun 19, 2023 · We report a case with a new NR5A1 variant. The fertility preservation protocol proposed at the end of puberty did not allow any sperm retrieval for future ...
  93. [93]
    CRISPR/Cas9-mediated activation of NR5A1 steers female human ...
    Apr 16, 2025 · This corrects the article "CRISPR/Cas9-mediated activation of NR5A1 steers female human embryonic stem cell-derived bipotential gonadal-like cells towards a ...
  94. [94]
    Human Male Infertility Associated with Mutations in NR5A1 ...
    We identified seven men with severe spermatogenic failure who carried missense mutations in NR5A1. Functional studies indicated that these mutations impaired ...
  95. [95]
    Steroidogenic Factor-1 and Human Disease - PMC - PubMed Central
    Oct 8, 2012 · This factor was discovered in the early 1990s through the pioneering work of Keith L.
  96. [96]
    Decrypting the Hidden Genetic Architecture of Idiopathic Male Infertility
    Oct 30, 2025 · This study highlights the significance of non-canonical splicing variants in male infertility, a factor often overlooked during the analysis ...
  97. [97]
    DAX-1 (NR0B1) and steroidogenic factor-1 (SF-1, NR5A1) in human ...
    Overactivity of SF-1 has been proposed to be associated with other conditions, such as hyperandrogenic polycystic ovarian syndrome (PCOS) and endometriosis [70] ...
  98. [98]
    The Roles of SF-1 Neurons in Energy Homeostasis - Frontiers
    May 6, 2013 · This review focuses on the emerging homeostatic roles of the SF-1 neurons in the VMH discovered through the use of genetic models.Missing: gonadotropes tegmental VTA
  99. [99]
    Population-Based Study of Rare Coding Variants in NR5A1/SF-1
    , et al. SF1 and spleen development: new heterozygous mutation, literature review and consequences for NR5A1-mutated patient's management . Clin Genet . 2017.Missing: prevalence | Show results with:prevalence<|separator|>