Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 392
Filter
Add more filters

Publication year range
1.
Physiol Rev ; 99(2): 1249-1279, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30810078

ABSTRACT

Nuclear receptors are intracellular proteins that act as transcription factors. Proteins with classic nuclear receptor domain structure lacking identified signaling ligands are designated orphan nuclear receptors. Two of these, steroidogenic factor-1 (NR5A1, also known as SF-1) and liver receptor homolog-1 (NR5A2, also known as LRH-1), bind to the same DNA sequences, with different and nonoverlapping effects on targets. Endogenous regulation of both is achieved predominantly by cofactor interactions. SF-1 is expressed primarily in steroidogenic tissues, LRH-1 in tissues of endodermal origin and the gonads. Both receptors modulate cholesterol homeostasis, steroidogenesis, tissue-specific cell proliferation, and stem cell pluripotency. LRH-1 is essential for development beyond gastrulation and SF-1 for genesis of the adrenal, sexual differentiation, and Leydig cell function. Ovary-specific depletion of SF-1 disrupts follicle development, while LRH-1 depletion prevents ovulation, cumulus expansion, and luteinization. Uterine depletion of LRH-1 compromises decidualization and pregnancy. In humans, SF-1 is present in endometriotic tissue, where it regulates estrogen synthesis. SF-1 is underexpressed in ovarian cancer cells and overexpressed in Leydig cell tumors. In breast cancer cells, proliferation, migration and invasion, and chemotherapy resistance are regulated by LRH-1. In conclusion, the NR5A orphan nuclear receptors are nonredundant factors that are crucial regulators of a panoply of biological processes, across multiple reproductive tissues.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Reproduction , Steroidogenic Factor 1/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endometriosis/metabolism , Endometriosis/pathology , Female , Gene Expression Regulation , Humans , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/pathology , Ligands , Male , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Pregnancy , Protein Conformation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Steroidogenic Factor 1/chemistry , Steroidogenic Factor 1/genetics , Structure-Activity Relationship , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
2.
Proc Natl Acad Sci U S A ; 120(32): e2220849120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37494420

ABSTRACT

The ovarian follicle reserve, formed pre- or perinatally, comprises all oocytes for lifetime reproduction. Depletion of this reserve results in infertility. Steroidogenic factor 1 (SF-1; Nr5a1) and liver receptor homolog 1 (LRH-1; Nr5a2) are two orphan nuclear receptors that regulate adult endocrine function, but their role in follicle formation is unknown. We developed models of conditional depletion of SF-1 or LRH-1 from prenatal ovaries. Depletion of SF-1, but not LRH-1, resulted in dramatically smaller ovaries and fewer primordial follicles. This was mediated by increased oocyte death, resulting from increased ovarian inflammation and increased Notch signaling. Major dysregulated genes were Iroquois homeobox 3 and 5 and their downstream targets involved in the establishment of the ovarian laminin matrix and oocyte-granulosa cell gap junctions. Disruptions of these pathways resulted in follicles with impaired basement membrane formation and compromised oocyte-granulosa communication networks, believed to render them more prone to atresia. This study identifies SF-1 as a key regulator of the formation of the ovarian reserve.


Subject(s)
Ovarian Reserve , Pregnancy , Female , Humans , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ovarian Reserve/genetics , Ovarian Follicle/metabolism , Ovary/metabolism , Granulosa Cells/metabolism
3.
BMC Genomics ; 25(1): 464, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741085

ABSTRACT

Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.


Subject(s)
Cell Lineage , Chromatin , Gonads , SOX9 Transcription Factor , Single-Cell Analysis , Animals , Chromatin/metabolism , Chromatin/genetics , Mice , Cell Lineage/genetics , Female , Male , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Gonads/metabolism , Gonads/cytology , Gonads/embryology , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Testis/metabolism , Testis/cytology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Ovary/metabolism , Ovary/cytology
4.
Acta Neuropathol ; 147(1): 16, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38228887

ABSTRACT

Pituitary neuroendocrine tumors (PitNETs) are classified according to cell lineage, which requires immunohistochemistry for adenohypophyseal hormones and the transcription factors (TFs) PIT1, SF1, and TPIT. According to the current WHO 2022 classification, PitNETs with co-expression of multiple TFs are termed "plurihormonal". Previously, PIT1/SF1 co-expression was prevailingly reported in PitNETs, which otherwise correspond to the somatotroph lineage. However, little is known about such tumors and the WHO classification has not recognized their significance. We compiled an in-house case series of 100 tumors, previously diagnosed as somatotroph PitNETs. Following TF staining, histopathological features associated with PIT1/SF1 co-expression were assessed. Integration of in-house and publicly available sample data allowed for a meta-analysis of SF1-associated clinicopathological and molecular features across a total of 270 somatotroph PitNETs. The majority (74%, 52/70) of our densely granulated somatotroph PitNETs (DGST) unequivocally co-expressed PIT1 and SF1 (DGST-PIT1/SF1). None (0%, 0/30) of our sparsely granulated somatotroph PitNETs (SGST) stained positive for SF1 (SGST-PIT1). Among DGST, PIT1/SF1 co-expression was significantly associated with scarce FSH/LH expression and fewer fibrous bodies compared to DGST-PIT1. Integrated molecular analyses including publicly available samples confirmed that DGST-PIT1/SF1, DGST-PIT1 and SGST-PIT1 represent distinct tumor subtypes. Clinicopathological meta-analyses indicated that DGST-PIT1 respond more favorably towards treatment with somatostatin analogs compared to DGST-PIT1/SF1, while both these subtypes show an overall less aggressive clinical course than SGST-PIT1. In this study, we spotlight that DGST with co-expression of PIT1 and SF1 represent a common, yet underrecognized, distinct PitNET subtype. Our study questions the rationale of generally classifying such tumors as "plurihormonal", and calls for a refinement of the WHO classification. We propose the term "somatogonadotroph PitNET".


Subject(s)
Adenoma , Neuroendocrine Tumors , Pituitary Neoplasms , Humans , Cell Lineage , Neuroendocrine Tumors/genetics , Pituitary Neoplasms/genetics , Transcription Factors , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism
5.
Gen Comp Endocrinol ; 353: 114512, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38582176

ABSTRACT

Eels are gonochoristic species whose gonadal differentiation initiates at the yellow eel stage and is influenced by environmental factors. We revealed some sex-related genes were sex dimorphically expressed in gonads during gonadal sex differentiation of Japanese eel (Anguilla japonica); however, the expression of sex-related genes in the brain-pituitary during gonadal sex differentiation in eels is still unclear. This study aimed to investigate the sex-related gene expressions in the brain-pituitary and tried to clarify their roles in the brain and gonads during gonadal sex differentiation. Based on our previous histological study, the control eels developed as males, and estradiol-17ß (E2) was used for feminization. Our results showed that during testicular differentiation, the brain cyp19a1 transcripts and aromatase proteins were increased significantly; moreover, the cyp19a1, sf-1, foxl2s, and esrs (except gperb) transcripts in the midbrain/pituitary also were increased significantly. Forebrain gnrh1 transcripts increased slightly during gonadal differentiation of both sexes, but the gnrhr1b and gnrhr2 transcripts in the midbrain/pituitary were stable during gonadal differentiation. The expression levels of gths and gh in the midbrain/pituitary were significantly increased during testicular differentiation and were much higher in males than in E2-feminized females. These results implied that endogenous estrogens might play essential roles in the brain/pituitary during testicular differentiation, sf-1, foxl2s, and esrs may have roles in cyp19a1 regulation in the midbrain/pituitary of Japanese eels. For the GnRH-GTH axis, gths, especially fshb, may be regulated by esrs and involved in regulating testicular differentiation and development in Japanese eels.


Subject(s)
Aromatase , Brain , Pituitary Gland , Sex Differentiation , Animals , Sex Differentiation/genetics , Sex Differentiation/physiology , Male , Aromatase/genetics , Aromatase/metabolism , Female , Brain/metabolism , Pituitary Gland/metabolism , Anguilla/genetics , Anguilla/metabolism , Anguilla/growth & development , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism , Gonads/metabolism , Gonads/growth & development
6.
Anim Biotechnol ; 35(1): 2351975, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38742598

ABSTRACT

The development of ovarian follicles in poultry is a key factor affecting the performance of egg production. Ovarian follicle development is regulated via the Wnt/ß-catenin signaling pathway, and ß-catenin, encoded by CTNNB1, is a core component of this pathway. In this study, using ovary GCs from laying hens, we investigated the regulatory role of CTNNB1 in steroid synthesis. We found that CTNNB1 significantly regulates the expression of StAR and CYP11A1 (key genes related to progesterone synthesis) and the secretion of progesterone (P4). Furthermore, simultaneous overexpression of CTNNB1 and SF1 resulted in significantly higher levels of CYP11A1 and secretion of P4 than in cells overexpressing CTNNB1 or SF1 alone. We also found that in GCs overexpressing SF1, levels of CYP11A1 and secreted P4 were significantly greater than in controls. Silencing of CYP11A1 resulted in the inhibition of P4 secretion while overexpression of SF1 in CYP11A1-silenced cells restored P4 secretion to normal levels. Together, these results indicate that synergistic cooperation between the ß-catenin and SF1 regulates progesterone synthesis in laying hen ovarian hierarchical granulosa cells to promote CYP11A1 expression.


Subject(s)
Chickens , Cholesterol Side-Chain Cleavage Enzyme , Granulosa Cells , Progesterone , beta Catenin , Animals , Female , Progesterone/biosynthesis , Progesterone/metabolism , beta Catenin/metabolism , beta Catenin/genetics , Granulosa Cells/metabolism , Chickens/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Gene Expression Regulation/physiology
7.
Cytogenet Genome Res ; 163(5-6): 307-316, 2023.
Article in English | MEDLINE | ID: mdl-38246151

ABSTRACT

INTRODUCTION: Testis differentiation is initiated by the SRY gene on the Y chromosome in mammalian species. However, the Amami spiny rat, Tokudaia osimensis, lacks both the Y chromosome and the Sry gene and acquired a unique Sox9 regulatory mechanism via a male-specific duplication upstream of Sox9, without Sry. In general mammalian species, the SRY protein binds to a testis-specific enhancer to promote SOX9 gene expression. Several enhancers located upstream of Sox9/SOX9 have been reported in mice and humans. In particular, the binding of SRY to the highly conserved enhancer Enh13 is thought to be a common mechanism underlying testis differentiation and sex determination in mammals. METHODS: Sequences of T. osimensis homologues of three Sox9 enhancers that were previously reported in mice, Enh8, Enh14, and Enh13, were determined. We performed in vitro assays to confirm enhancer activity involved in Sox9 regulation in T. osimensis. RESULTS: T. osimensis Enh13 showed enhancer activity when co-transfected with NR5A1 and SOX9. Mouse Enh13 was activated by NR5A1 and SRY; however, T. osimensis Enh13 did not respond to SRY, even though the binding sites of SRY and NR5A1 were conserved. To identify the key sequence that is present in mouse but absent from T. osimensis, we performed reporter gene assays using vectors in which partial sequences of T. osimensis Enh13 were replaced with mouse sequences. For T. osimensis Enh13 in which the second half (approximately 430 bp) was replaced with the corresponding mouse sequence, activity in response to NR5A1 and SRY was recovered. Further, reporter assays revealed that multiple regions in the second half of the mouse Enh13 sequence are required for the response to NR5A1 and SRY. The latter 49 bp was particularly important and contained four binding sites for three transcription factors, POU2F1, HOXA3, and GATA1. CONCLUSION: We showed that there are unknown sequences responsible for the interaction between NR5A1 and SRY and mEnh13 based on comparative analyses of Sry-dependent and Sry-independent species. Our comparative analyses revealed new molecular mechanisms underlying mammalian sex determination.


Subject(s)
Enhancer Elements, Genetic , SOX9 Transcription Factor , Sex-Determining Region Y Protein , Animals , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Mice , Male , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Rats , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism , Base Sequence
8.
Reproduction ; 165(1): 1-17, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36194434

ABSTRACT

In brief: The nuclear receptor steroidogenic factor 1 (SF-1) is essential for mature mouse gonad steroidogenic gene expression, for Leydig and Sertoli cell function, and depletion of SF-1 in steroidogenic cells of the testis compromises steroidogenesis, spermatogenesis and male fertility. Abstract: Steroidogenic factor 1 (SF-1 or NR5A1) plays an essential role in the development of fetal gonads and regulates genes involved in steroid biosynthesis. Since SF-1 is expressed in multiple cell types in mouse gonads, we developed three novel conditional knockout (cKO) mouse models employing Cre-recombinase and floxed alleles of SF-1 (Nr5a1f/f) to identify its role in testes and ovaries of mature mice: Cytochrome P450 17α-hydroxylase (Cyp17Cre/+;Nr5a1f/f, Leydig and theca cell-specific), aromatase (Cyp19Cre/+;Nr5a1f/f, Sertoli and granulosa cell-specific), as well as a combination of both (Cyp17+Cyp19-Cre;Nr5a1f/f). Compared to control animals, Cyp19-Cre;Nr5a1f/f cKO males showed normal fertility and testicular function. The Cyp17Cre/+;Nr5a1f/f cKO males had smaller testis, with drastically reduced Leydig cell volumes and impaired steroidogenesis, though their reproductive performance remained comparable to controls. Some 50% of Cyp17Cre/++Cyp19Cre/+;Nr5a1f/f double-cKO (dKO) males were infertile, while the remaining 50% showed significantly reduced fertility. These dKO males also had smaller testis with degenerative seminiferous tubules, abnormal Leydig cell morphology and lower levels of intra-testicular testosterone. Abnormal Sertoli cell localization was noted in dKO testes, with increased Sox9, p27 and inhibin subunit ßb and decreased androgen receptor expression. Female mice from all genotypes showed normal reproductive capacity, though steroidogenic gene expression levels were significantly decreased in both Cyp17Cre/+;Nr5a1f/f cKO and dKO females. These results show the essential role of SF-1 in mature mouse gonad steroidogenic gene expression, for Leydig and Sertoli cell function, and that depletion SF-1 in all steroidogenic cells of the testis compromises steroidogenesis, spermatogenesis and male fertility.


Subject(s)
Ovary , Steroidogenic Factor 1 , Testis , Animals , Female , Male , Mice , Aromatase/metabolism , Leydig Cells/metabolism , Mice, Knockout , Ovary/metabolism , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism , Testosterone
9.
Cell Biol Toxicol ; 39(5): 2051-2067, 2023 10.
Article in English | MEDLINE | ID: mdl-35246761

ABSTRACT

Dexamethasone is widely used to treat pregnancy disorders related to premature delivery. However, lots of researches have confirmed that prenatal dexamethasone exposure (PDE) could increase the risk of offspring multiple diseases. This study was designed to elucidate the epigenetic mechanism of adrenal developmental programming and explore its early warning marker in peripheral blood mononuclear cells (PBMC). We found the adrenal morphological and functional changes of PDE male offspring rats before and after birth, which were mainly performed as the decreased serum corticosterone concentration, steroidogenic acute regulatory (StAR) protein expression, and histone 3 lysine 27 acetylation (H3K27ac) level of steroidogenic factor 1 (SF1) promoter region and its expression. Simultaneously, the expressions of glucocorticoid receptor (GR) and histone acetylation enzyme 5 (HDAC5) in the PDE male fetal rats were increased. In vitro, dexamethasone reduced the expression of SF1, StAR, and cortisol production and still increased the expression of GR and HDAC5, the binding between GR and SF1 promoter region, and protein interaction between GR and HDAC5. GR siRNA or HDAC5 siRNA was able to reverse the above roles of dexamethasone. Furthermore, in vivo, we confirmed that H3K27ac levels of SF1 promoter region and its expression in PBMC of the PDE group were decreased before and after birth, showing a positive correlation with the same indexes in adrenal. Meanwhile, in clinical trials, we confirmed that prenatal dexamethasone application decreased H3K27ac of SF1 promoter region and its expression in neonatal PBMC. In conclusion, PDE-caused adrenal insufficiency of male offspring rats was related to adrenal GR activated by dexamethasone in uterus. The activated GR, on the one hand, increased its direct binding to SF1 promoter region to inhibit its expression, on the other hand, upregulated and recruited HDAC5 to decrease H3K27ac level of SF1 promoter region, and strengthened the inhibition of SF1 and subsequent StAR expression.


Subject(s)
Adrenal Insufficiency , Prenatal Exposure Delayed Effects , Pregnancy , Female , Humans , Rats , Male , Animals , Leukocytes, Mononuclear , Rats, Wistar , Acetylation , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Histones/metabolism , Corticosterone , Dexamethasone/pharmacology , Biomarkers/metabolism , RNA, Small Interfering/metabolism
10.
Proc Natl Acad Sci U S A ; 117(32): 19566-19577, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719118

ABSTRACT

The ventromedial hypothalamus (VMH) plays chief roles regulating energy and glucose homeostasis and is sexually dimorphic. We discovered that expression of metabotropic glutamate receptor subtype 5 (mGluR5) in the VMH is regulated by caloric status in normal mice and reduced in brain-derived neurotrophic factor (BDNF) mutants, which are severely obese and have diminished glucose balance control. These findings led us to investigate whether mGluR5 might act downstream of BDNF to critically regulate VMH neuronal activity and metabolic function. We found that mGluR5 depletion in VMH SF1 neurons did not affect energy balance regulation. However, it significantly impaired insulin sensitivity, glycemic control, lipid metabolism, and sympathetic output in females but not in males. These sex-specific deficits are linked to reductions in intrinsic excitability and firing rate of SF1 neurons. Abnormal excitatory and inhibitory synapse assembly and elevated expression of the GABAergic synthetic enzyme GAD67 also cooperate to decrease and potentiate the synaptic excitatory and inhibitory tone onto mutant SF1 neurons, respectively. Notably, these alterations arise from disrupted functional interactions of mGluR5 with estrogen receptors that switch the normally positive effects of estrogen on SF1 neuronal activity and glucose balance control to paradoxical and detrimental. The collective data inform an essential central mechanism regulating metabolic function in females and underlying the protective effects of estrogen against metabolic disease.


Subject(s)
Blood Glucose/metabolism , Estrogens/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Ventromedial Hypothalamic Nucleus/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Energy Metabolism , Female , Glutamate Decarboxylase/metabolism , Homeostasis , Lipid Metabolism , Male , Mice , Mice, Mutant Strains , Nerve Net , Neural Inhibition , Neurons/metabolism , Neurons/physiology , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Estrogen/metabolism , Sex Factors , Signal Transduction , Steroidogenic Factor 1/metabolism , Sympathetic Nervous System/metabolism , Synaptic Transmission , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
11.
Differentiation ; 128: 83-100, 2022.
Article in English | MEDLINE | ID: mdl-36114074

ABSTRACT

Nuclear receptor subfamily 5 group A member 1 (NR5A1) encodes steroidogenic factor 1 (SF1), a key regulatory factor that determines gonadal development and coordinates endocrine functions. Here, we have established a stem cell-based model of human gonadal development and applied it to evaluate the effects of NR5A1 during the transition from bipotential gonad to testicular cells. We combined directed differentiation of human induced pluripotent stem cells (46,XY) with activation of endogenous NR5A1 expression by conditionally-inducible CRISPR activation. The resulting male gonadal-like cells expressed several Sertoli cell transcripts, secreted anti-Müllerian hormone and responded to follicle-stimulating hormone by producing sex steroid intermediates. These characteristics were not induced without NR5A1 activation. A total of 2691 differentially expressed genetic elements, including both coding and non-coding RNAs, were detected immediately following activation of NR5A1 expression. Of those, we identified novel gonad-related putative NR5A1 targets, such as SCARA5, which we validated also by immunocytochemistry. In addition, NR5A1 activation was associated with dynamic expression of multiple gonad- and infertility-related differentially expressed genes. In conclusion, by combining targeted differentiation and endogenous activation of NR5A1 we have for the first time, been able to examine in detail the effects of NR5A1 in early human gonadal cells. The model and results obtained provide a useful resource for future investigations exploring the causative reasons for gonadal dysgenesis and infertility in humans.


Subject(s)
Induced Pluripotent Stem Cells , Infertility , Humans , Male , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Mutation , Induced Pluripotent Stem Cells/metabolism , Gonads/metabolism , Scavenger Receptors, Class A/genetics
12.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36835002

ABSTRACT

Steroidogenic factor-1 (SF-1, also termed Ad4BP; NR5A1 in the official nomenclature) is a nuclear receptor transcription factor that plays a crucial role in the regulation of adrenal and gonadal development, function and maintenance. In addition to its classical role in regulating the expression of P450 steroid hydroxylases and other steroidogenic genes, involvement in other key processes such as cell survival/proliferation and cytoskeleton dynamics have also been highlighted for SF-1. SF-1 has a restricted pattern of expression, being expressed along the hypothalamic-pituitary axis and in steroidogenic organs since the time of their establishment. Reduced SF-1 expression affects proper gonadal and adrenal organogenesis and function. On the other hand, SF-1 overexpression is found in adrenocortical carcinoma and represents a prognostic marker for patients' survival. This review is focused on the current knowledge about SF-1 and the crucial importance of its dosage for adrenal gland development and function, from its involvement in adrenal cortex formation to tumorigenesis. Overall, data converge towards SF-1 being a key player in the complex network of transcriptional regulation within the adrenal gland in a dosage-dependent manner.


Subject(s)
Adrenocortical Carcinoma , Steroidogenic Factor 1 , Humans , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/metabolism , Fushi Tarazu Transcription Factors , Homeodomain Proteins , Steroidogenic Factor 1/metabolism , Transcription Factors/metabolism
13.
Reproduction ; 164(2): 31-40, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35666805

ABSTRACT

In brief: The insulin-like 3 (INSL3) hormone produced by Leydig cells is essential for proper male sex differentiation, but the regulation of Insl3 expression remains poorly understood. This study describes a new physical and functional cooperation between the nuclear receptors SF1 and COUP-TFII in Insl3 expression. Abstract: INSL3, a hormone abundantly produced by Leydig cells, is essential for testis descent during fetal life and bone metabolism in adults. The mechanisms regulating Insl3 expression in Leydig cells have been studied in several species but remain poorly understood. To date, only a handful of transcription factors are known to activate the Insl3 promoter and include the nuclear receptors AR, NUR77, COUP-TFII, and SF1, as well as the Krüppel-like factor KLF6. Some of these transcription factors are known to transcriptionally cooperate on the Insl3 promoter, but the mechanisms at play remain unknown. Here, we report that COUP-TFII and SF1 functionally cooperate on the Insl3 promoter from various species but not on the Inha, Akr1c14, Cyp17a1, Hsd3b1, Star, Gsta3, and Amhr2 promoters that are known to be regulated by COUP-TFII and/or SF1. The Insl3 promoter contains species-conserved binding sites for COUP-TFII (-91 bp) and SF1 (-134 bp). Mutation of either the COUP-TFII or the SF1 sequence had no impact on the COUP-TFII/SF1 cooperation, but the mutation of both binding sites abolished the cooperation. In agreement with this, we found that COUP-TFII and SF1 physically interact in Leydig cells. Finally, we report that the transcriptional cooperation is not limited to COUP-TFII and SF1 as it also occurred between all NR2F and NR5A family members. Our data provide new mechanistic insights into the cooperation between the orphan nuclear receptors COUP-TFII and SF1 in the regulation of Insl3 gene expression in Leydig cells.


Subject(s)
COUP Transcription Factor II , Insulin , Leydig Cells , Proteins , Steroidogenic Factor 1 , Adult , Binding Sites , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Leydig Cells/metabolism , Male , Promoter Regions, Genetic , Proteins/genetics , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism
14.
FASEB J ; 35(6): e21660, 2021 06.
Article in English | MEDLINE | ID: mdl-34010469

ABSTRACT

In the mammalian testis, two distinct populations of Sertoli cells (SCs), the immature SCs (ISCs) and adult SCs (ASCs), play significant roles in regulating the development and function of Leydig cells. However, the effect of different SC types on the function of Leydig cells is poorly understood. Here, our study showed that miR-145-5p expression was significantly different in SCs at different stages, with the highest expression observed in ISCs. Exosomes mediate the transfer of miR-145-5p from ISCs to Leydig cells. Overexpression of miR-145-5p in Leydig cells significantly downregulated steroidogenic gene expression and inhibited testosterone synthesis. Additionally, miR-145-5p functioned by directly targeted steroidogenic factor-1 (Sf-1) and downregulated the expression of SF-1, which further downregulated the expression of steroidogenic genes, induced accumulation of lipid droplets, and eventually suppressed testosterone production. These findings demonstrate that SC-derived miR-145-5p plays a significant role in regulating the functions of Leydig cells and may therefore serve as a diagnostic biomarker for male hypogonadism developmental abnormalities during puberty.


Subject(s)
Exosomes/metabolism , Leydig Cells/metabolism , MicroRNAs/genetics , Sertoli Cells/metabolism , Steroidogenic Factor 1/antagonists & inhibitors , Steroids/biosynthesis , Testis/metabolism , Animals , Exosomes/genetics , Leydig Cells/pathology , Male , Mice , Mice, Inbred C57BL , Sertoli Cells/pathology , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/pathology
15.
Environ Toxicol ; 37(10): 2419-2433, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35762508

ABSTRACT

Phthalates may interfere with the biosynthesis of steroid hormones in the adrenal cortex. Bis (2-butoxyethyl) phthalate (BBOP) is a phthalate containing oxygen atoms in the alcohol moiety. In this study, 35-day-old male Sprague-Dawley rats were daily gavaged with BBOP (0, 10, 100, 250, and 500 mg/kg body weight) for 21 days. BBOP did not affect the weight of body and adrenal glands. BBOP significantly reduced serum corticosterone levels at 250 and 500 mg/kg, and lowered aldosterone level at 500 mg/kg without affecting adrenocorticotropic hormone. BBOP did not alter the thickness of the adrenal cortex. BBOP significantly down-regulated the expression of steroidogenesis-related genes (Scarb1, Star, Cyp11a1, Cyp21, Cyp11b1, Cyp11b2, Nr5a1, Nr4a1, and Nr4a2) and proteins, and antioxidant enzymes (Sod1, Sod2, Gpx1, and Cat) and their proteins, while up-regulating the expression of Mc2r and Agtr1a at various doses. BBOP reduced the phosphorylation of AKT1, AKT2, and ERK1/2, as well as the levels of SIRT1 and PGC1α without affecting the phosphorylation of AMPK. BBOP significantly induced the production of reactive oxygen species and apoptosis rate in H295R cells at 100 µM and higher after 24 h of treatment. In conclusion, male rats exposed to BBOP in puberty have significant reduction of steroid biosynthesis with a potential mechanism that is involved in the decrease in the phosphorylation of AKT1, AKT2, ERK1/2, as well as SIRT1 and PGC1α and increase in ROS.


Subject(s)
Sexual Maturation , Sirtuin 1 , Animals , Corticosterone/metabolism , Male , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phthalic Acids , Rats , Rats, Sprague-Dawley , Sirtuin 1/metabolism , Steroidogenic Factor 1/metabolism , Steroids
16.
Development ; 145(23)2018 12 05.
Article in English | MEDLINE | ID: mdl-30518625

ABSTRACT

Previous studies have established that fetal Leydig cells (FLCs) and adult Leydig cells (ALCs) show distinct functional characteristics. However, the lineage relationship between FLCs and ALCs has not been clarified yet. Here, we reveal that a subset of FLCs dedifferentiate at fetal stages to give rise to ALCs at the pubertal stage. Moreover, the dedifferentiated cells contribute to the peritubular myoid cell and vascular pericyte populations in the neonatal testis, and these non-steroidogenic cells serve as potential ALC stem cells. We generated FLC lineage-specific Nr5a1 (Ad4BP/SF-1) gene-disrupted mice and mice lacking the fetal Leydig enhancer (FLE) of the Nr5a1 gene. Phenotypes of these mice support the conclusion that most of the ALCs arise from dedifferentiated FLCs, and that the FLE of the Nr5a1 gene is essential for both initial FLC differentiation and pubertal ALC redifferentiation.


Subject(s)
Adult Stem Cells/cytology , Cell Dedifferentiation , Fetus/cytology , Leydig Cells/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Lineage , Enhancer Elements, Genetic/genetics , Fibrosis , Integrases/metabolism , Leydig Cells/metabolism , Male , Mice , Models, Biological , Phenotype , Sequence Deletion/genetics , Steroidogenic Factor 1/metabolism , Testis/cytology , Testis/transplantation
17.
Neuropathol Appl Neurobiol ; 47(3): 415-427, 2021 04.
Article in English | MEDLINE | ID: mdl-33128255

ABSTRACT

AIMS: The gonadotroph tumour (GT) is the most frequently resected pituitary neuroendocrine tumour. Although many symptomatic GT are successfully resected, some recur. We sought to identify histological biomarkers that may predict recurrence and explore biological mechanisms that explain this difference in behaviour. METHODS: SF-1 immunohistochemistry of 51 GT, a subset belonging to a longitudinal prospective cohort study (n = 25), was reviewed. Four groups were defined: Group 1-recently diagnosed GT (n = 20), Group 2-non-recurrent GT with long-term follow up (n = 11), Group 3-initial resections of GT that recur (n = 7) and Group 4-recurrent GT (n = 13). The percentage of SF-1 immunolabelling in the lowest staining fields (SF-1 labelling index (SLI)) was assessed and RNA sequencing was performed on 5 GT with SLI <80% and 5 GT with SLI >80%. RESULTS: Diffuse, strong SF-1 immunolabelling was the most frequent pattern in Groups 1/2, whereas patchy SF-1 staining predominated in Groups 3/4. There was a lower median SLI in Groups 3/4 than 1/2. Overall, GT with SLI <80% recurred earlier than GT with SLI >80%. Differential expression analysis identified 89 statistically significant differentially expressed genes (FDR <0.05) including over-expression of pituitary stem cell genes (SOX2, GFRA3) and various oncogenes (e.g. BCL2, ERRB4) in patchy SF-1 GT. Gene set enrichment analysis identified significant enrichment of genes involved in the PI3K-AKT pathway. CONCLUSIONS: We speculate that patchy SF-1 labelling in GT reflects intratumoural heterogeneity and are less differentiated tumours than diffusely staining GT. SF-1 immunolabelling patterns may have prognostic significance in GT, but confirmatory studies are needed for further validation.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasm Recurrence, Local/pathology , Neuroendocrine Tumors/pathology , Pituitary Neoplasms/pathology , Steroidogenic Factor 1/metabolism , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
18.
Endocr J ; 68(11): 1283-1291, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34176817

ABSTRACT

The functional role of the transcription factors NR5A1 and NR5A2 and their interaction with Clock gene and bone morphogenetic proteins (BMPs) were investigated in human granulosa KGN cells. Treatment with BMP-15 and GDF-9 suppressed forskolin (FSK)-induced steroidogenesis as shown by the mRNA expression levels of StAR and P450scc but not the mRNA expression level of P450arom. Of interest, treatment with BMP-15 and GDF-9 also suppressed FSK-induced NR5A2 mRNA expression. Treatment with BMP-15 suppressed NR5A2 mRNA and protein expression but increased Clock mRNA and protein expression levels by granulosa cells. The mRNA expression levels of NR5A1, but not those of NR5A2, were positively correlated with the levels of Clock mRNA, while the mRNA levels of Id-1, the target gene of BMP signaling, were positively correlated with those of NR5A1 but not with those of NR5A2. It was also demonstrated that the mRNA expression levels of NR5A1 were positively correlated with those of P450arom and 3ßHSD, whereas the mRNA expression level of NR5A2 was correlated with those of StAR and P450scc. Furthermore, inhibition of Clock gene expression by siRNA attenuated the expression of NR5A1, and the mRNA levels of Clock gene were significantly correlated with those of NR5A1. Collectively, the results suggested a novel mechanism by which Clock gene expression induced by BMP-15 is functionally linked to the expression of NR5A1, whereas NR5A2 expression is suppressed by BMP-15 in granulosa cells. The interaction between Clock NR5A1/NR5A2 and BMP-15 is likely to be involved in the fine-tuning of steroidogenesis by ovarian granulosa cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , CLOCK Proteins/metabolism , Granulosa Cells/metabolism , Ovary/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1/metabolism , Bone Morphogenetic Protein 15/pharmacology , Bone Morphogenetic Proteins/genetics , CLOCK Proteins/genetics , Female , Gene Expression/drug effects , Granulosa Cells/drug effects , Growth Differentiation Factor 9/pharmacology , Humans , Ovary/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/drug effects , Steroidogenic Factor 1/genetics
19.
Gynecol Endocrinol ; 37(6): 546-553, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33150797

ABSTRACT

In this study, the polycystic ovary syndrome (PCOS) mice model was randomly divided into 6 groups: blank control group, Clomiphene group, PCOS group, and Yulin mixture high-/medium-/low-dose group. Rats were killed after 5 weeks of administration. The expression levels of serum E2,T,Insulin and LH were detected by ELISA. The localizations and quantities of Steroid-generating factor-1 (SF-1) and Cytochrome protein P450 a1 (Cyp19a1) were detected by immunohistochemistry and western blot. The quantities of miR-320 were detected by RT-PCR. The results showed that the mechanism of Yulin mixture inhibiting the growth of polycystic ovary on mouse PCOS model may be through the decreasing of serum T and LH levels and then reducing local estrogen content to make the polycystic ovary atrophy. Yulin mixture can decrease the level of miR-320 and increase the expression of SF-1 and Cyp19a1 in ovary, thereby regulating the ovarian granulosa cell proliferation and apoptosis.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Gene Expression Regulation/drug effects , Polycystic Ovary Syndrome/drug therapy , Animals , Aromatase/genetics , Aromatase/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Estradiol/blood , Female , Insulin/blood , Luteinizing Hormone/blood , Medicine, Chinese Traditional , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovary/drug effects , Ovary/metabolism , Phytotherapy , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Preliminary Data , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testosterone/blood
20.
Int J Mol Sci ; 22(12)2021 Jun 08.
Article in English | MEDLINE | ID: mdl-34201257

ABSTRACT

Despite the substantial role played by the hypothalamus in the regulation of energy balance and glucose homeostasis, the exact mechanisms and neuronal circuits underlying this regulation remain poorly understood. In the last 15 years, investigations using transgenic models, optogenetic, and chemogenetic approaches have revealed that 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 in energy expenditure and glucose homeostasis, with minor feeding control. Deletion of hormonal receptors, nutritional sensors, or synaptic receptors in SF1 neurons triggers metabolic alterations mostly appreciated under high-fat feeding, indicating that SF1 neurons are particularly important for metabolic adaptation in the early stages of obesity. Although these studies have provided exciting insight into the implications of hypothalamic SF1 neurons on whole-body energy homeostasis, new questions have arisen from these results. Particularly, the existence of neuronal sub-populations of SF1 neurons and the intricate neurocircuitry linking these neurons with other nuclei and with the periphery. In this review, we address the most relevant studies carried out in SF1 neurons to date, to provide a global view of the central role played by these neurons in the pathogenesis of obesity and diabetes.


Subject(s)
Diabetes Mellitus/pathology , Hypothalamus/pathology , Neurons/pathology , Obesity/pathology , Steroidogenic Factor 1/metabolism , Animals , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Humans , Hypothalamus/metabolism , Neurons/metabolism , Obesity/etiology , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL