Stanford Home
Ovarian Kaleidoscope Database (OKdb)

Home

History

Transgenic Mouse Models

INFORGRAPHICS

Search
Submit
Update
Chroms
Browse
Admin

Hsueh lab

HPMR

Visits
since 01/2001:
176557

inhibin subunit alpha OKDB#: 145
 Symbols: INHA Species: human
 Synonyms:  Locus: 2q35 in Homo sapiens


For retrieval of Nucleotide and Amino Acid sequences please go to: OMIM Entrez Gene
Mammalian Reproductive Genetics   Endometrium Database Resource   Orthologous Genes   UCSC Genome Browser   GEO Profiles new!   Amazonia (transcriptome data) new!

R-L INTERACTIONS   MGI

DNA Microarrays
SHOW DATA ...
link to BioGPS
General Comment Inhibin exists in 2 forms, each of which shares the same alpha subunit and, when covalently linked to 1 of 2 distinct subunits called beta-a and beta-b, inhibits pituitary FSH secretion. On the other hand, the dimers of 2 beta subunits, termed activin, are potent stimulators of FSH secretion and release in vitro.

NCBI Summary: This gene encodes a member of the TGF-beta (transforming growth factor-beta) superfamily of proteins. The encoded preproprotein is proteolytically processed to generate multiple peptide products, including the alpha subunit of the inhibin A and B protein complexes. These complexes negatively regulate follicle stimulating hormone secretion from the pituitary gland. Inhibins have also been implicated in regulating numerous cellular processes including cell proliferation, apoptosis, immune response and hormone secretion. Mutations in this gene may be associated with male infertility and premature ovarian failure in female human patients. [provided by RefSeq, Aug 2016]
General function Ligand, Hormone, Cytokine, Cell death/survival, Apoptosis, Cell cycle regulation, Tumor suppressor
Comment Mason et al. (1986) isolated cDNAs for the 3 inhibin subunits. These beta subunits share extensive sequence homology with transforming growth factor-beta. Wiater E,et al reported that inhibin is an antagonist of bone morphogenetic protein signaling. Inhibins are endogenous antagonists of activin signaling, long recognized as important regulators of gonadal function and pituitary FSH release. Inhibin, in concert with its co-receptor, betaglycan, can compete with activin for binding to type II activin receptors and, thus, prevent activin signaling. Because bone morphogenetic proteins (BMPs) also utilize type II activin receptors, we hypothesized that BMP signaling might also be sensitive to inhibin blockade. Here we show that inhibin blocks cellular responses to diverse BMP family members in a variety of BMP-responsive cell types. Inhibin abrogates BMP-induced Smad signaling and transcription responses. Inhibin competes with BMPs for type II activin receptors, and this competition is facilitated by betaglycan. Betaglycan also enables inhibin to bind to and compete with BMPs for binding to the BMP-specific type II receptor BMPRII, which does not bind inhibin in the absence of betaglycan. Betaglycan can confer inhibin responsiveness on cells that are otherwise insensitive to inhibin. These findings demonstrate that inhibin, acting through betaglycan, can function as an antagonist of BMP responses, suggesting a broader role for inhibin and betaglycan in restricting and refining a wide spectrum of transforming growth factor beta superfamily signals.
Cellular localization Secreted
Comment
Ovarian function Follicle development, Preantral follicle growth, Antral follicle growth, Steroid metabolism
Comment Activin effects on follicular growth in in vitro preantral follicle culture. Tanaka Y et al. (2019) As the follicular environment transits from being activin dominant to inhibin dominant during folliculogenesis, it is assumed that activin plays an important role in the early stage of follicular growth. We examined the effects of activin on morphological, biochemical and molecular changes in isolated preantral follicles. Preantral follicles were mechanically isolated from 14-day old female C57BL/6 mice. Each follicle was cultured and observed for 14 days usingan in vitro follicle culture system containing FSH, FSH + activin A and FSH + inhibin in the culture medium. We subsequently examined FSH receptor (FSH-R) mRNA expression in isolated follicle cultures with or without activin on days 0 and 2. Activin was observed to significantly stimulate follicle enlargement on days 2, 4, 6 and 8, accelerate morphological changes and increase estradiollevels in culture medium on days 4, 12 and 14. In contrast, inhibin did not alter follicular growth. Additionally, activin stimulated the expression of FSH-R mRNA in isolated granulosa cells. It was demonstrated that activin stimulated the growth of preantral follicles, mainly during the early stage of folliculogenesis, by inducing FSH-R expression, in an isolated follicle culture system. J. Med. Invest. 66 : 165-171, February, 2019.////////////////// RNAi-mediated knockdown of inhibin α subunit increased apoptosis in granulosa cells and decreased fertility in mice. Kadariya I et al. (2015) Inhibin α (INHα), a member of TGFβ superfamily, is an important modulator of reproductive function that plays a vital role in follicular changes, cell differentiation, oocyte development, and ultimately in mammalian reproduction. However, the role of inhibin α in female fertility and ovarian function remains largely unknown. To define its role in reproduction, transgenic mice of RNAi-INHα that knock down the INHα expression by shRNAi were used. Inhibin α subunit gene was knocked down successfully at both transcriptional and translational levels by RNAi PiggyBac transposon (Pbi) mediated recombinant pshRNA vectors and purified DNA fragments were microinjected into mouse zygotes. Results showed that transgenic female mice were sub-fertile and exhibited 35.28% reduction in litter size in F1 generation relative to wild type. The decreased litter size associated with the reduction in the number of oocytes ovulated after puberty. Serum INHα level was significantly decreased in both 3 and 6 weeks; whereas, FSH was significantly increased in 3 weeks but not in 6 weeks. Furthermore, suppression of INHα expression significantly promoted apoptosis by up-regulating Caspase-3, bcl2, INHβB and GDF9 and down regulated Kitl and TGFβRIII genes both at transcriptional and translational levels. Moreover, it also dramatically reduced the progression of G1 phase of cell cycle and the number of cells in S phase as determined by flow cytometer. These results indicate that suppression of INHα expression in RNAi-transgenic mice leads to disruption of normal ovarian regulatory mechanism and causes reproductive deficiencies by promoting cellular apoptosis, arresting cellular progression and altering hormonal signaling.////////////////// Inhibins are one of the most important protein hormone secreted by the ovary and responsible for both endocrine feedback regulation of the pituitary FSH release and paracrine regulation of local ovarian functions.The literature on inibibin and ovarian function has been summarized by Mather et al. (1997) , Woodruff et al. (1995) and Hillier et al. (1993). Hsueh et al. (1987) tested the intragonadal paracrine actions of heterodimers and homodimers of inhibin subunits. In cultured ovarian theca-interstitial cells and theca explants, the alpha beta heterodimer of inhibin enhances androgen biosynthesis stimulated by LH, whereas the activin beta beta homodimer suppresses androgen production. These data indicate that the inhibin-related gene products synthesized by granulosa cells may form heterodimers or homodimers to serve as intragonadal paracrine signals in the modulation of LH-stimulated androgen biosynthesis. Hillier et al. (1993) reported that inhibin exerts potent and selective stimulation of human thecal cell androgen synthesis in vitro. Forage et al. (1987) demonstrate that neutralization of inhibin can be effected by immunization with bovine inhibin alpha subunit and that such immunization results in increased ovulation rates as predicted from the biological role of inhibin as a suppressor of FSH.
Expression regulated by FSH, LH, Steroids, Growth Factors/ cytokines, Eicosanoids
Comment Recombinant BMP4 and BMP7 increase activin A production by up-regulating inhibin βA subunit and furin expression in human granulosa-lutein cells. Chang HM et al. (2015) Context: Granulosa cell-derived activins play important roles in the regulation of ovarian functions. To date, there is limited information pertaining to the intracellular regulation, assembly and secretion of endogenous activin A in human granulosa cells. Objective: The aim of this study was to examine the effects of BMP4 and BMP7 on furin expression and activin A production as well as the underlying mechanisms of action in human granulosa cells. Design: An established immortalized human granulosa cell line (SVOG) and primary granulosa-lutein cells were used as study models. Expression of inhibin subunits and furin as well as activin A accumulation were examined after exposure to recombinant human BMP4 or BMP7. A BMP type I receptor inhibitor (dorsomorphin), a furin inhibitor (Dec-RVKR-CMK), and small interfering RNAs targeting SMAD4 and furin were used to verify the specificity of the effects and investigate potential mechanisms. Setting: The study was conducted in an academic center. Main Outcome Measures: Specific mRNA and protein levels were examined using real time-quantitative PCR and Western blot. Activin A levels were measured using enzyme immunoassay. Results: Treatment with BMP4 and BMP7 significantly increased furin mRNA and protein, inhibin βA mRNA and activin A accumulation. Pre-treatment with dorsomorphin or SMAD4 knockdown reversed the stimulatory effects of BMP4 and BMP7 on furin and inhibin βA expression. In addition, furin knockdown or pre-treatment with a furin inhibitor attenuated the BMP4- and BMP7-induced accumulation of activin A. Conclusion: Recombinant BMP4 and BMP7 increase the production of bioactive mature activin A by up-regulating both the production and proteolytic processing of inhibin βA subunit in human granulosa cells. The enhancement of inhibin βA subunit processing is attributable to a SMAD-dependent up-regulation of its proprotein convertase, furin. These findings provide a potential mechanism by which theca cells can regulate neighboring granulosa cells in the ovary.////////////////// DNA Methylation and Histone Modifications Are Associated with Repression of the Inhibin a Promoter in the Rat Corpus Luteum. Meldi KM et al. The transition from follicle to corpus luteum after ovulation is associated with profound morphological and functional changes and is accompanied by corresponding changes in gene expression. The gene encoding the a subunit of the dimeric reproductive hormone inhibin is maximally expressed in the granulosa cells of the preovulatory follicle, is rapidly repressed by the ovulatory LH surge, and is expressed at only very low levels in the corpus luteum. Although previous studies have identified transient repressors of inhibin a gene transcription, little is known about how this repression is maintained in the corpus luteum. This study examines the role of epigenetic changes, including DNA methylation and histone modification, in silencing of inhibin a gene expression. Bisulfite sequencing reveals that methylation of the inhibin a proximal promoter is low in preovulatory and ovulatory follicles but is elevated in the corpus luteum. Increased methylation during luteinization is observed within the cAMP response element in the promoter, and EMSA demonstrate that methylation of this site inhibits cAMP response element binding protein binding in vitro. Chromatin immunoprecipitation reveals that repressive histone marks H3K9 and H3K27 trimethylation are increased on the inhibin a promoter in primary luteal cells, whereas the activation mark H3K4 trimethylation is decreased. The changes in histone modification precede the alterations in DNA methylation, suggesting that they facilitate the recruitment of DNA methyltransferases. We show that the DNA methyltransferase DNMT3a is present in the ovary and in luteal cells when the inhibin a promoter becomes methylated and observe recruitment of DNMT3a to the inhibin promoter during luteinization. The hormonal regulation of inhibin production by cultured granulosa cells from immature hypophysectomized, estrogen-treated rats was examined using a specific RIA by Bicsak et al. (1986) Granulosa cell inhibin secretion is stimulated by FSH and LH but not by PRL, presumably via a cAMP-mediated pathway. Bergada I, et al 2000 reported the role of inhibins in childhood and puberty. The determination of serum dimeric inhibins from birth through adulthood reflects a distinct pattern of both inhibins in males and females. Concomitantly with the gonadotrophin surge, an important production of inhibin B is observed during the first months of life. In girls, high serum levels of inhibin A are observed during the first two months of life; thereafter, they are undetectable until puberty. An active secretion of inhibin B persists in both males and females in the period of maximal LHRH pulse generator restraint; however, the possible gonadotrophin dependence of this production remains controversial. At puberty, a progressive rise in serum inhibin B occurs concomitantly with the increased production of sex steroids in both males and females. A similar secretion pattern of inhibin A is observed in girls. This increment is mainly exerted by gonadotrophins and modulated by multiple paracrine/autocrine mechanisms within the ovary and the testis that regulate the dimerization of the inhibin subunits throughout pubertal maturation. The differences observed in males and females between circulating dimeric inhibins in relation to gonadotrophins and sex steroid concentrations from birth through puberty has opened a new perspective for research in human reproduction.
Ovarian localization Granulosa, Luteal cells
Comment Meunier et al (1989) studied immature female rats treated with PMSG and human CG to induce ovulation for inhibin subunit mRNA localization. Administration of PMSG led to a sharp increase in the expression of all three subunits in large preovulatory follicles whereas injection with human CG caused a decrease in the levels of the respective mRNAs. In contrast to mature females, shortly before ovulation, levels of inhibin alpha-subunit mRNA were low in small antral follicles. In addition, at that time, inhibin beta A- and beta B-subunits mRNAs were present in several large follicles (greater than 500 microns). More than 2 days after ovulation, inhibin beta A- and beta B-subunit mRNAs could not be detected in small antral size follicles of hormonally induced females. On the other hand, hybridization signals for the inhibin alpha-subunit were observed in some small antral and preantral size follicles, while signals were very low or undetectable in a large number of atretic follicles. Smith et al. (1991) reported that the human corpus luteum is a significant source of immunoreactive inhibin during the normal human menstrual cycle. The specific localization within the granulosalutein cells of the corpus luteum suggests that inhibin alpha-subunit production may originate from a discrete cell population within the human corpus luteum. Lappohn et al. (1989) summarized data on the use of inhibin as a marker for granulosa-cell tumors. Danforth et al. (1998) reported that luteal phase inhibin-a and follicular phase inhibin-b were correlated inversely with age in perimenopausal women. In addition, luteal phase inhibin-a and follicular phase inhibin-b levels were correlated inversely with follicular phase FSH levels. These direct measures of ovarian function may be more sensitive indicators of "ovarian reserve" than indirect indicators such as pituitary FSH secretion. Changes in mouse granulosa cell gene expression during early luteinization. McRae RS et al. Changes in gene expression during granulosa cell luteinization have been measured using serial analysis of gene expression (SAGE). Immature normal mice were treated with pregnant mare serum gonadotropin (PMSG) or PMSG followed, 48 h later, by human chorionic gonadotropin (hCG). Granulosa cells were collected from preovulatory follicles after PMSG injection or PMSG/hCG injection and SAGE libraries generated from the isolated mRNA. The combined libraries contained 105,224 tags representing 40,248 unique transcripts. Overall, 715 transcripts showed a significant difference in abundance between the two libraries of which 216 were significantly down-regulated by hCG and 499 were significantly up-regulated. Among transcripts differentially regulated, there were clear and expected changes in genes involved in steroidogenesis as well as clusters of genes involved in modeling of the extracellular matrix, regulation of the cytoskeleton and intra and intercellular signaling. The SAGE libraries described here provide a base for functional investigation of the regulation of granulosa cell luteinization.
Follicle stages Secondary, Antral, Preovulatory, Corpus luteum
Comment Hsu et al. (1995) reported that different 5'-flanking regions of the inhibin-alpha gene target transgenes to the gonad and adrenal in an age-dependent manner in transgenic mice. Kananen et al. (1995) reported gonadal tumorigenesis in transgenic mice bearing the mouse inhibin alpha-subunit promoter/simian virus T-antigen fusion gene and the establishment of gonadotropin-responsive granulosa cell lines.
Phenotypes POF (premature ovarian failure)
Mutations 6 mutations

Species: mouse
Mutation name: None
type: null mutation
fertility: infertile - ovarian defect
Comment: Matzuk et al. (1992) generated inhibin-deficient mice, that developed sex cord stromal tumors at an early age with nearly 100% penetrance, demonstrating that inhibin functions in vivo as a tumor suppressor in the gonads of mice. These mice showed a severe wasting syndrome, which mimics the human cancer cachexia syndrome.

Species: human
Mutation name: None
type: naturally occurring
fertility: subfertile
Comment: INHA promoter polymorphisms are associated with premature ovarian failure Harris SE, et al . Inhibin is an important glycoprotein that is involved in folliculogenesis. INHA, the gene encoding the inhibin alpha subunit, was recently proposed as a candidate for premature ovarian failure (POF), a syndrome that leads to the cessation of ovarian function under the age of 40 years. 70 POF patients and 70 controls were screened for the previously identified INHA - 16C>T transition mutation. The T allele was found in 31/70 (44.3%) of controls, but only 18/70 (25.7%) of POF patients. This result indicates that the T allele is significantly underrepresented in the POF patient population (Fisher's exact test, two-tail: P = 0.033). Sequence analysis of the INHA promoter in 50 POF patients and 50 controls identified a highly polymorphic imperfect TG repeat at approximately -300 bp, that consisted of four common haplotypes (A, B, C and D). The -16T allele is linked to the shortest repeat haplotype (haplotype C). Despite the association between haplotype C and POF, no significant difference was found between the promoter activity of a luciferase reporter construct containing haplotype C, and most of the other haplotypes tested. Interestingly, haplotype B failed to show any promoter activity. We conclude that the inheritance of specific INHA promoter hap-lotypes predispose to the development of premature ovarian failure. Controversial role of inhibin {alpha}-subunit gene in the aetiology of premature ovarian failure. Sundblad V et al. BACKGROUND: Premature ovarian failure (POF) is characterized by hypergonadotropic amenorrhoea before the age of 40. Inhibin alpha-subunit (INH3) gene is proposed as a candidate gene due to its role in negative feedback control of FSH. METHODS: Polymorphism -16C>T of INHalpha gene was studied in 61 POF patients and 82 controls above 40 years old (C > 40). Substitution 769G>A was studied in 59 POF patients, 76 C > 40 and 73 controls below 40 years old (C < 40). RESULTS: No significant difference in risk of POF development for -16T allele was found when comparing idiopathic POF (I-POF) with C > 40 (Odds ratio = 1.46; 95% confidence interval = 0.63-3.19). Implication of -16C>T polymorphism in serum inhibin levels was analysed in 46 controls, and no significant differences (P > 0.05) were found between CC and CT + TT genotype groups when comparing either mid-follicular phase Pro-alphaC and inhibin B values or mid-luteal phase Proalpha3C and inhibin A values. Heterozygosity for substitution 769G>A was found in 1 of 59 POF woman, 2 of 76 C > 40 and 6 of 73 C < 40. Presence of this substitution in a relevant number of control subjects is herein described for the first time. CONCLUSION: Our results indicate that -16C>T and 769G>A variants in INH3 gene may not be associated to POF disease.

Species: mouse
Mutation name: None
type: null mutation
fertility: subfertile
Comment: Loss of inhibin alpha uncouples oocyte-granulosa dynamics and disrupts postnatal folliculogenesis. Myers M et al. Targeted disruption of the inhibin alpha gene (Inha(-/-)) in mice results in an ovarian phenotype of granulosa cell tumors that renders the animals infertile. Little is known about the reproductive defects prior to tumor development. Here, we report novel data on early follicle dynamics in Inha(-/-)mice, which demonstrate that inhibin alpha has important consequences upon follicle development. Morphological changes in both germ and somatic cells were evident in postnatal day 12 ovaries, with Inha(-/-)mice exhibiting numerous multilayered follicles that were far more advanced than those observed in age-matched controls. These changes were accompanied by alterations in follicle dynamics such that Inha(-/-)ovaries had fewer follicles in the resting pool and more committed in the growth phase. Absence of inhibin alpha resulted in advanced follicular maturation as marked by premature loss of anti-M黮lerian hormone (AMH) in secondary follicles. Additionally, gene expression analysis revealed changes in factors known to be vital for oocyte and follicle development. Together, these data provide key evidence to suggest that regulation of the inhibin/activin system is essential for early folliculogenesis in the prepubertal mouse ovary. GDF9 Modulates the Reproductive and Tumor Phenotype of Female Inha Null Mice. Myers M et al. Intraovarian factors play important roles in coordinating germ cell and somatic cell growth in the ovary. Prior to the onset of gonadotropin stimulation and reproductive cyclicity, follicle development is dependent upon locally produced growth factors, such as the transforming growth factor beta family members inhibin, activin, and GDF9. In the absence of inhibin in prepubertal mice (Inha-/-), there are marked alterations in preantral follicle growth, but no evidence of ovarian tumors characteristic of adult Inha null mice. To ascertain the contribution of GDF9 to the Inha null phenotype, we analyzed folliculogenesis in postnatal Inha Gdf9 double knockout mice. Deletion of Gdf9 from Inha-/- rescues the initial growth defects found at early follicle stages in Inha-/- ovaries, but surprisingly enhances the onset of pre-tumor lesions. The normalization of growth dynamics between granulosa cells and oocytes of Inha Gdf9 double knockout mice is also accompanied by a reduction in levels of the activin/inhibin beta B subunit, Inhbb, which is upregulated in Inha-/- ovaries. However, at later ages, Inha Gdf9 double knockout ovaries are similar to Inha-/- ovaries, and show upregulation of the activin/inhibin subunits and downregulation of the growth factor, kit ligand (Kitl), thus resulting in local environment that is growth-promoting for granulosa cells, but growth-inhibitory for oocytes. These data suggest a sequential mechanism of action initiated by GDF9 in the Inha knockout mouse that promotes defective folliculogenesis. These studies thus provide a novel role for GDF9 in causing reproductive defects and suppressing tumor initiation in the Inha-/- mouse model.

Species: human
Mutation name: None
type: naturally occurring
fertility: unknown
Comment: Germline Mutations of Inhibins in Early-Onset Ovarian Epithelial Tumors. Tournier I 2013 et al. To identify novel genetic bases of early-onset epithelial ovarian tumors, we used the trio exome sequencing strategy in a patient without familial history of cancer who presented metastatic serous ovarian adenocarcinomas at 21 years of age. We identified a single de novo mutation (c.1157A>G/p.Asn386Ser) within the INHBA gene encoding the A-subunit of inhibins/activins, which play a key role in ovarian development. In vitro, this mutation alters the ratio of secreted activins and inhibins. In a second patient with early-onset serous borderline papillary cystadenoma, we identified an unreported germline mutation (c.179G>T/p.Arg60Leu) of the INHA gene encoding the a-subunit, the partner of the A-subunit. This mutation also alters the secreted activin/inhibin ratio, by disrupting both inhibin A and inhibin B biosynthesis. In a cohort of 62 cases, we detected an additional unreported germline mutation of the INHBA gene (c.839G>A/p.Gly280Glu). Our results strongly suggest that inhibin mutations contribute to the genetic determinism of epithelial ovarian tumors. This article is protected by copyright. All rights reserved. /////////////////////////

Species: porcine
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Association of inhibin-α gene polymorphisms with follicular cysts in large white sows. Li W et al. (2015) Ovarian follicular cysts are anovulatory follicular structures that have been identified in sows and are known to cause infertility. The pathogenesis of follicular cysts remains poorly understood. Hormones play key roles in the formation and persistence of cysts. The hormone inhibin is a member of the TGF-β superfamily and is named for its negative regulation of FSH, another hormone that controls follicular recruitment and growth. In the present study, 48 sows with follicular cysts and 60 normal sows with no cysts were screened for mutations in the inhibin-α gene to examine the association of inhibin-α gene polymorphisms with the presence of follicular cysts. The results show that the c.-42G>A and c.3222G>A polymorphisms are significantly associated with follicular cysts and that sows with c.-42GG and c.3222GG genotypes have lower risk of developing cysts. Our findings may provide novel biological biomarkers and promising gene therapy candidates for follicular cyst formation in sows, which would greatly benefit pig breeding programs.//////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Functional analysis of the human inhibin alpha subunit variant A257T and its potential role in premature ovarian failure. Chand AL et al. (2008) A nucleotide substitution in the inhibin alpha subunit (INHA 769G>A, A257T) has been associated with premature ovarian failure (POF). We hypothesize this mutation causes a reduction in inhibin bioactivity, removing its suppression on the pituitary FSH secretion. The aim of this study is to establish if A257T inhibin has reduced bioactivity. Mouse LbetaT2 pituitary gonadotrope, human granulosa (COV434) and human embryonic kidney (HEK293) cells were co-transfected with an activin-responsive reporter and increasing amounts of wild-type or variant A257T inhibin alpha subunit, and the degree of inhibin antagonism of activin signalling determined. A 5-fold inhibition was observed with wild-type inhibin alpha subunit overexpression (P < 0.001) (confirmed in HEK293 cells), while the A257T inhibin showed no inhibitory activity. In human ovarian COV434 transfected cells, while wild-type and A257T inhibin A had similar bioactivities, there was a significant reduction in the bioactivity of A257T inhibin B compared with wild-type inhibin B (P < 0.005). In all the three cell systems, overexpression of wild-type and A257T alpha subunit resulted in a 2- to 6-fold increase in secretion of dimeric inhibin indicating the reduced inhibin response was not due to a failure of dimerization. This study supports the hypothesis that the INHA 769G>A variant may increase susceptibility to POF with impaired inhibin B bioactivity and provides insight into the complex aetiology of POF.//////////////////

Genomic Region show genomic region
Phenotypes and GWAS show phenotypes and GWAS
Links
OMIM (Online Mendelian Inheritance in Man: an excellent source of general gene description and genetic information.)
OMIM \ Animal Model
KEGG Pathways
Recent Publications
None
Search for Antibody


created: Sept. 25, 1999, midnight by: Hsueh   email:
home page:
last update: March 19, 2020, 1:19 p.m. by: hsueh    email:



Use the back button of your browser to return to the Gene List.

Click here to return to gene search form