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

SMAD family member 4 OKDB#: 642
 Symbols: SMAD4 Species: human
 Synonyms: JIP, DPC4, MADH4, MYHRS  Locus: 18q21.2 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 The Mad (for 'mothers against decapentaplegic') gene in Drosophila and the related Sma genes in C. elegans are implicated in signal transduction by members of the IGF-beta family in these organisms and also in vertebrates. Derynck et al. (1996) proposed a revised nomenclature for the Mad-related products in vertebrates. Their proposed root symbol was SMAD, a merger of Sma and Mad, which serves to differentiate these proteins from unrelated gene products previously called Mad.

NCBI Summary: This gene encodes a member of the Smad family of signal transduction proteins. Smad proteins are phosphorylated and activated by transmembrane serine-threonine receptor kinases in response to transforming growth factor (TGF)-beta signaling. The product of this gene forms homomeric complexes and heteromeric complexes with other activated Smad proteins, which then accumulate in the nucleus and regulate the transcription of target genes. This protein binds to DNA and recognizes an 8-bp palindromic sequence (GTCTAGAC) called the Smad-binding element (SBE). The protein acts as a tumor suppressor and inhibits epithelial cell proliferation. It may also have an inhibitory effect on tumors by reducing angiogenesis and increasng blood vessel hyperpermeability. The encoded protein is a crucial component of the bone morphogenetic protein signaling pathway. The Smad proteins are subject to complex regulation by post-translational modifications. Mutations or deletions in this gene have been shown to result in pancreatic cancer, juvenile polyposis syndrome, and hereditary hemorrhagic telangiectasia syndrome. [provided by RefSeq, Aug 2017]
General function Intracellular signaling cascade, Nucleic acid binding, DNA binding, Transcription factor
Comment Relative expression of genes encoding SMAD signal transduction factors in human granulosa cells is correlated with oocyte quality. Kuo FT et al. PURPOSE: To determine the expression of SMAD transcripts in human granulosa cells. METHODS: Luteinized mural granulosa cells were harvested from forty women undergoing oocyte retrieval, and RNAs were isolated. SMAD expression levels were determined by polymerase chain reaction (PCR) and quantitative real-time PCR (q-RTPCR). RESULTS: SMAD1-7 and 9 are expressed in human granulosa cells, with SMAD2, 3 and 4 showing the highest expression levels. Peak estradiol (E2) levels correlated with the number of oocytes retrieved during IVF. Oocyte number showed no correlation with SMAD expression levels or ratios. Fertilization rates also did not correlate with the expression levels of individual SMADs, but did correlate with higher SMAD4:SMAD3 ratios (p?=?0.0062) and trended with SMAD4:SMAD2 (p?=?0.0698). CONCLUSIONS: SMAD transcripts are differently expressed in human granulosa cells, where they may mediate TGF-beta superfamily signaling during folliculogenesis and ovulation. Further, the relative expression ratios of SMAD2, 3 and 4 may differentially affect fertilization rate.
Cellular localization Cytoplasmic, Nuclear
Comment
Ovarian function Antral follicle growth, Follicle atresia, Early embryo development
Comment SMAD4 activates Wnt signaling pathway to inhibit granulosa cell apoptosis. Du X et al. (2020) The TGF-β and Wnt signaling pathways are interrelated in many cell types and tissues, and control cell functions in coordination. Here, we report that SMAD4, a downstream effector of the TGF-β signaling pathway, induces FZD4, a receptor of the Wnt signaling pathway, establishing a novel route of communication between these two pathways in granulosa cells (GCs). We found that SMAD4 is a strong inducer of FZD4, not only initiating FZD4 transcription but also activating FZD4-dependent Wnt signaling and GC apoptosis. Furthermore, we identified the direct and indirect mechanisms by which SMAD4 promotes expression of FZD4 in GCs. First, SMAD4 functions as a transcription factor to directly bind to the FZD4 promoter region to increase its transcriptional activity. Second, SMAD4 promotes production of SDNOR, a novel lncRNA that acts as a sponge for miR-29c, providing another mean to block miR-29c from degenerating FZD4 mRNA. Overall, our findings not only reveal a new channel of crosstalk between the TGF-β and Wnt signaling pathways, SMAD4-FZD4 axis, but also provide new insights into the regulatory network of GC apoptosis and follicular atresia. These RNA molecules, such as miR-29c and lnc-SDNOR, represent potential targets for treatment of reproductive diseases and improvement of female fertility.////////////////// SMAD4 feedback regulates the canonical TGF-β signaling pathway to control granulosa cell apoptosis. Du X et al. (2018) Canonical TGF-β signals are transduced from the cell surface to the cytoplasm, and then translocated into the nucleus, a process that involves ligands (TGF-β1), receptors (TGFBR2/1), receptor-activated SMADs (SMAD2/3), and the common SMAD (SMAD4). Here we provide evidence that SMAD4, a core component of the canonical TGF-β signaling pathway, regulates the canonical TGF-β signaling pathway in porcine granulosa cells (GCs) through a feedback mechanism. Genome-wide analysis and qRT-PCR revealed that SMAD4 affected miRNA biogenesis in GCs. Interestingly, TGFBR2, the type II receptor of the canonical TGF-β signaling pathway, was downregulated in SMAD4-silenced GCs and found to be a common target of SMAD4-inhibited miRNAs. miR-425, the most significantly elevated miRNA in SMAD4-silenced GCs, mediated the SMAD4 feedback regulation of the TGF-β signaling pathway. This was accomplished through a direct interaction between the transcription factor SMAD4 and the miR-425 promoter, and a direct interaction between miR-425 and the TGFBR2 3'-UTR. Furthermore, miR-425 enhanced GC apoptosis by targeting TGFBR2 and the canonical TGF-β signaling pathway, which was rescued by SMAD4 and TGF-β1. Overall, our findings demonstrate that a positive feedback mechanism exists within the canonical TGF-β signaling pathway. This study also provides new insights into mechanism underlying the canonical TGF-β signaling pathway, which regulates GC function and follicular development.////////////////// Oocyte-derived Smad4 is not required for development of the oocyte or the preimplantation embryo. Kaune H et al. (2014) The generation of a competent egg requires complex molecular interactions between the oocyte and the ovary, and transforming growth factor β (TGF-β) is a major signaling pathway. Smad4 is a central regulator of the TGF-β signaling pathway as it mediates gene expression triggered by activation of TGF-β receptors. Deletion of Smad4 in granulosa cells disrupts follicle development; however, the role of Smad4 in the oocyte has not been confirmed. Furthermore, the role of Smad4 in embryo development has not been confirmed because previous studies of Smad4(del/del) embryos were generated from heterozygous parents, and thus it is possible that maternal transcripts rescue development before embryonic day 6.5 (E6.5) when Smad4(del/del) embryos die. To determine the role of TGF-β signaling in oocyte and embryo development, mice with oocyte-specific deletion of Smad4 were studied. Fertility was evaluated in Mutant (Smad4(F/F):ZP3Cre) and Control (Smad4(F/F)) females mated continuously with control males during a 6-month period. Surprisingly, Mutant females were fertile with the same litter size (Mutants, 9.23 ± 0.4; Controls, 9.42 ± 0.4) and interlitter period as Controls. Ovulation rate induced using a superovulation regime did not differ between Controls and Mutants at both 6 weeks and 6 months. Embryo development was assessed at E6.5 using Control and Mutant females mated with heterozygous males. Development of Smad4(del/del) embryos at E6.5 was retarded consistent with previous studies of embryos generated from heterozygous parents indicating that there is no rescue of preimplantation development by maternal transcripts. The numbers of implanted embryos at 6.5 dpc also did not differ (Control: 9.1 ± 0.4; Mutant: 7.0 ± 0.9). However, only 26.3% of E6.5 embryos carried by Mutant females were Smad4(del/del) compared with the expected ratio of 50%. Since litter size was not decreased, this indicates that either the number of Smad4(del) sperm fertilizing the oocytes is reduced or implantation of Smad4(del/del) embryos is suboptimal. In summary, we have shown that Smad4 in the oocyte, and thus TGF-β signaling, is not required for oocyte or follicle development, ovulation, fertilization, preimplantation development, or implantation.////////////////// Evidence Supporting a Functional Requirement of SMAD4 for Bovine Preimplantation Embryonic Development: A Potential Link to Embryotropic Actions of Follistatin. Lee KB 2014 et al. Transforming growth factor beta (TGFbeta) superfamily signaling controls various aspects of female fertility. However, the functional roles of TGFbeta-superfamily cognate signal transduction pathway components (e.g. SMAD2/3, SMAD4, SMAD1/5/8) in early embryonic development are not completely understood. We have previously demonstrated pronounced embryotropic actions of the TGFbeta superfamily member-binding protein, follistatin, on oocyte competence in cattle. Given SMAD4 is a common 'SMAD' required for both SMAD2/3 and SMAD1/5/8 signaling pathways, the objectives of the present studies were to determine the temporal expression and functional role of SMAD4 in bovine early embryogenesis and whether embryotropic actions of follistatin are SMAD4-dependent. SMAD4 mRNA is increased in bovine oocytes during meiotic maturation, is maximal in 2-cell stage embryos, remains elevated through the 8-cell stage and is decreased and remains low through the blastocyst stage. Ablation of SMAD4 via siRNA microinjection of zygotes reduced proportions of embryos cleaving early and development to the 8- to 16-cell and blastocyst stages. Stimulatory effects of follistatin on early cleavage, but not on development to 8- to 16-cell and blastocyst stages were observed in SMAD4-depleted embryos. Therefore, results suggest SMAD4 is obligatory for early embryonic development in cattle and embryotropic actions of follistatin on development to 8- to 16-cell and blastocyst stages are SMAD4 dependent. ///////////////////////// Interference RNA (RNAi)-Based Silencing of Endogenous Smad4 in Porcine Granulosa Cells Resulted in Decreased FSH-mediated GCs Proliferation and Steroidogenesis. Wang W et al. FSH plays a critical role in granulosa cell (GC) proliferation and steroidogenesis through modulation by factors including bone morphogenetic proteins (BMPs) family, which belongs to Transforming growth factor ?(TGF-? superfamily. TGF-? are the key factors in maintaining cell growth and differentiation in ovaries. However, the interaction of FSH and TGF-?on the GCs' proliferation and steroidogenesis remains to be elucidated. Here, we have investigated the role of Smad4, a core molecule mediating the intracellular TGF-?Smad signal transduction pathway, in FSH-mediated proliferation and steroidogenesis of porcine GCs. In this study, Smad4 was knockdown by utilizing interference RNA (RNAi) in porcine GCs. Our results showed that Smad4-siRNA cause specific inhibition of Smad4 mRNA and protein expression after transfection. Knockdown Smad4 significantly inhibited FSH-induced porcine granulosa cell proliferation and E2 production and changed the expression of Cyclin D2, CDK2, CDK4, CYP19a1 and CYP11a1. Thus these observations establish an important role of Smad4 in the regulation of the response of porcine GCs to FSH.
Expression regulated by
Comment xyz
Ovarian localization Oocyte, Granulosa, Theca, Luteal cells
Comment Expression and localization of Smad2 and Smad4 proteins in the porcine ovary. Xing N 2014 et al. The objective of the present study was to investigate the temporal and spatial expression of Smad2 and Smad4 proteins, the downstream signaling molecules of the transforming growth factor beta (TGF-? superfamily, in the porcine ovary. Cellular localization of Smad2 and Smad4 proteins was examined using immunohistochemistry. The specificity of the antibodies was examined using Western blot assay. Western blot analyses demonstrated that 52kDa Smad2 and 60kDa Smad4 proteins were expressed in the porcine ovary. Immunohistochemistry revealed that Smad2 and Smad4 were widely expressed in the porcine ovary, mainly localized in the oocyte, granulosa and thecal cells at different stages of folliculogenesis. Within the primordial and primary follicles, Smad2 and Smad4 showed strong staining in oocytes and follicular cells. In the antral follicle, strong staining was observed in oocytes, granulosa and theca cells. These findings suggest that Smad2 and Smad4 may be a key regulator of follicular development and growth of oocytes in the porcine ovary. ///////////////////////// Jaatinen R, et al reported the activation of the bone morphogenetic protein signaling pathway induces inhibin beta(B)-subunit mRNA and secreted inhibin B levels in cultured human granulosa-luteal cells. They show that human granulosa luteal (GL) cells express mRNAs for distinct serine/threonine kinase receptors (BMP-RIA and BMIP-RII) and Smad signaling proteins (Smad1, Smad4, and Smad5) involved in the mediation of cellular effects of BMPs. Subsequently, they determined in hGL cell cultures the effects of distinct members of the BMP family previously found to be expressed in mammalian ovaries. Recombinant BMP-2 induces potently in a time- and concentration-dependent manner the expression of the inhibin beta(B)-subunit mRNAs in hGL cells without affecting the levels of alpha- or beta(A)-subunit mRNAs. BMP-6 has a similar, but weaker, effect than BMP-2, whereas BMP-3 and its close homolog, BMP-3b (also known as growth differentiation factor-10) had no effect on inhibin subunit mRNA expression. hCG treatment of hGL cells was previously shown to abolish the stimulatory effect of activin on beta(B)-subunit mRNA levels, and here hCG is also shown to suppress the effect of BMP-2. Furthermore, BMP-2 stimulates hGL cell secreted dimeric inhibin B levels in a concentration-dependent manner. Depending on the experiment, maximal increases in inhibin B levels of 6- to 28-fold above basal levels were detected during a 72-h culture period. Billiar RB, et al reported the localization and developmental expression of the activin signal transduction proteins smads 2, 3, and 4 in the baboon fetal ovary. Western blot analyses demonstrated that the 59 kDa Smad 2, 54 kDa Smad 3, and 64 kDa Smad 4 proteins were expressed in fetal ovaries of untreated baboons at both mid and late gestation and that the level of expression was not significantly altered in late gestation by in vivo treatment with CGS 20267 or CGS 20267 and estrogen. Immunocytochemistry localized Smads 2/3 and 4 to cytoplasm of oocytes and pregranulosa cells at midgestation and oocytes and granulosa cells of primordial follicles in late gestation. Smad 4 was also detected in granulosa cell nuclei in late gestation, and nuclear expression appeared to be decreased in fetal ovaries of baboons deprived of estrogen. The site of localization of Smads correlated with localization of the activin receptors IA and IIB, which we previously showed were abundantly expressed in oocytes and (pre)granulosa cells at both mid and late gestation and unaltered by estrogen deprivation.
Follicle stages Primordial, Primary, Secondary, Antral, Preovulatory, Corpus luteum
Comment Billiar RB, et al determined whether the primate fetal ovary expressed Smads 2/3 and 4 and whether expression of these activin-signaling proteins was altered in fetal ovaries of baboons in which estrogen production was suppressed. Western blot analyses demonstrated that the 59 kDa Smad 2, 54 kDa Smad 3, and 64 kDa Smad 4 proteins were expressed in fetal ovaries of untreated baboons at both mid and late gestation and that the level of expression was not significantly altered in late gestation by in vivo treatment with CGS 20267 or CGS 20267 and estrogen. Immunocytochemistry localized Smads 2/3 and 4 to cytoplasm of oocytes and pregranulosa cells at midgestation and oocytes and granulosa cells of primordial follicles in late gestation. Smad 4 was also detected in granulosa cell nuclei in late gestation and nuclear expression appeared to be decreased in fetal ovaries of baboons deprived of estrogen. The site of localization of Smads correlated with localization of the activin receptors IA and IIB which we previously showed were abundantly expressed in oocytes and (pre)granulosa cells at both mid and late gestation and unaltered by estrogen deprivation. In summary, the results of the current study are the first to show that the intracellular signaling molecules required to transduce an activin signal are expressed in the baboon fetal ovary and that expression was not altered by estrogen-deprivation in utero. These findings coupled with our previous observations showing that estrogen-deprivation reduced follicle numbers and up-regulated/induced expression of inhibin but not activin or the activin receptors, lend further support to the hypothesis that estrogen regulates fetal ovarian folliculogenesis by controlling the intraovarian activin:inhibin ratio.
Phenotypes
Mutations 7 mutations

Species: mouse
Mutation name: None
type: null mutation
fertility: embryonic lethal
Comment: Sirard et al. (1998) demonstrated that homozygous Smad4 mutant mice died before embryonic day 7.5. Mutant embryos have reduced size, fail to gastrulate or express a mesodermal marker, and show abnormal visceral endoderm development. Growth retardation of the Smad4-deficient embryos results from reduced cell proliferation rather than increased apoptosis. Aggregation of mutant Smad4 embryonic stem cells with wildtype tetraploid morulae rescued the gastrulation defect.

Species: mouse
Mutation name: None
type: null mutation
fertility: embryonic lethal
Comment: Differential requirements for Smad4 in TGF{beta}-dependent patterning of the early mouse embryo Chu GC, et al . Genetic and biochemical data have identified Smad4 as a key intracellular effector of the transforming growth factor beta (TGFbeta superfamily of secreted ligands. In mouse, Smad4-null embryos do not gastrulate, a phenotype consistent with loss of other TGFbeta-related signaling components. Chimeric analysis reveals a primary requirement for Smad4 in the extra-embryonic lineages; however, within the embryo proper, characterization of the specific roles of Smad4 during gastrulation and lineage specification remains limited. We have employed a Smad4 conditional allele to specifically inactivate the Smad4 gene in the early mouse epiblast. Loss of Smad4 in this tissue results in a profound failure to pattern derivatives of the anterior primitive streak, such as prechordal plate, node, notochord and definitive endoderm. In contrast to these focal defects, many well-characterized TGFbeta- and Bmp-regulated processes involved in mesoderm formation and patterning are surprisingly unaffected. Mutant embryos form abundant extra-embryonic mesoderm, including allantois, a rudimentary heart and middle primitive streak derivatives such as somites and lateral plate mesoderm. Thus, loss of Smad4 in the epiblast results not in global developmental abnormalities but instead in restricted patterning defects. These results suggest that Smad4 potentiates a subset of TGFbeta-related signals during early embryonic development, but is dispensable for others.

Species: mouse
Mutation name: None
type: null mutation
fertility: subfertile
Comment: Premature luteinization and cumulus cell defects in ovarian-specific Smad4 knockout mice. Pangas SA et al. SMAD4 is a central component of the TGFbeta superfamily signaling pathway. Within the ovary, TGFbeta-related proteins play crucial roles in controlling granulosa cell growth, differentiation, and steroidogenesis. To study the in vivo roles of SMAD4 during follicle development, we generated an ovarian conditional knockout of Smad4 using the cre/loxP recombination system. Smad4 ovarian-specific knockout mice are subfertile with decreasing fertility over time and multiple defects in folliculogenesis. Regulation of steroidogenesis is disrupted in the Smad4 conditional knockout, leading to increased levels of serum progesterone. In addition, severe cumulus cell defects are present both in vivo and when assayed in vitro. These findings demonstrate that disrupting signaling through SMAD4 in the ovarian granulosa cells leads to premature luteinization of granulosa cells and eventually premature ovarian failure, thereby demonstrating key in vivo roles of TGFbeta superfamily signaling in the timing of granulosa cell differentiation.

Species: mouse
Mutation name: None
type: null mutation
fertility: subfertile
Comment: Minimal Fertility Defects in Mice Deficient in Oocyte-Expressed Smad4. Li X et al. Bidirectional signaling between oocytes and granulosa cells is required for normal folliculogenesis. Oocyte-secreted members of the transforming growth factor beta (TGFB) family, growth and differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), are well known mediators of granulosa cell function, and deletion in granulosa cells of Smad4, the common SMAD mediating all canonical TGFB-related protein signals, results in infertility. Reciprocal signaling by granulosa cell-expressed TGFB family ligands, such as activin, to the oocyte during follicle development has been proposed but not tested in vivo using conditional knockout mice. Therefore, we generated two oocyte-specific conditional knockout models for the common SMAD, Smad4, using cre recombinase expression from either the zona pellucida 3 (Zp3) or Gdf9 promoter. Cre expression from the Gdf9 promoter occurs at a slightly earlier time point in follicle development than from Zp3. Deletion of Smad4 using Zp3cre had no effect on fertility, while deletion of Smad4 with Gdf9icre resulted in a slight, but significant, reduction in litter size. These mouse models suggest a novel, although minor, role for Smad4 in the oocyte restricted to the primordial follicle stage.

Species: mouse
Mutation name: None
type: null mutation
fertility: subfertile
Comment: Selective Smad4 Knockout in Ovarian Preovulatory Follicles Results in Multiple Defects in Ovulation. Yu C et al. The TGF-?signaling pathway is involved with multiple processes in the mammalian ovary, including primordial follicle formation, granulosa cell (GC) proliferation, follicle atresia, ovulation, and feedback regulation between the pituitary and ovary. The transcriptional factor SMAD4 is the central component of the canonical TGF-?signaling pathway. Smad4 knockout using Amhr2-Cre, which is expressed in GCs of immature developing follicles, causes premature luteinization. In this study, we specifically depleted Smad4 in GCs of preovulatory follicles using Cyp19-Cre mice. As different from results with Smad4(fl/fl);Amhr2-Cre mice, Smad4 depletion in preovulatory follicles did not cause premature luteinization or suppress GC proliferation; rather, it increased follicle atresia. In addition, Nppc and Npr2 expressions were reduced by Smad4 depletion; thus, their effect of maintaining oocyte meiotic arrest was weakened in Smad4 conditional knockout mice. Smad4(fl/fl);Cyp19-Cre female mice were subfertile and had irregular estrous cycles and ovulation defects. Smad4 knockout also blocked luteinizing hormone (LH)-induced cumulus expansion and follicle rupture, but not oocyte meiotic resumption. Our results also indicated that SMAD4 was required for LH-stimulated activation of ERK1/2 and the expressions of ovulation-related genes. The defects arising from SMAD4 depletion could not be rescued by intra-ovarian mediators of LH actions, such as EGF-like factors and prostaglandin E2. Furthermore, corpus lutea did not form in Smad4(fl/fl);Cyp19-Cre female mice, indicating that SMAD4 was crucial for GCs terminal differentiation. Thus, by characterizing the ovarian phenotypes of preovulatory follicle-specific Smad4 knockout mice, we identified the developmental stage-specific functions of the canonical TGF-?signaling pathway in ovulation and luteinization.

Species: mouse
Mutation name: None
type: null mutation
fertility: infertile - ovarian defect
Comment: TGF signaling promotes juvenile granulosa cell tumorigenesis by suppressing apoptosis. Mansouri-Attia N 2014 et al. Molecular changes that give rise to granulosa cell tumors of the ovary (GCT) are not well understood. Previously, we showed that deletion in granulosa cells of the bone morphogenetic protein (BMP) receptor-signaling transcription factors, Smad1 and Smad5, causes development of metastatic granulosa cell tumors that phenocopies the juvenile form of granulosa cell tumors (JGCT) in humans. The transforming growth factor (TGF)-SMAD2/3 pathway is active in JGCT but its role is unknown. We tested the in vivo contribution of TGF-SMAD signaling to JGCT development by genetically deleting the common Smad4 from Smad1/5 double knockout mice. Smad1/5/4 triple knockout mice were sterile, and had significantly increased survival and delayed tumor development compared to the Smad1/5 double knockout mice. The few tumors that did develop were smaller, showed no evidence of metastasis, and had increased apoptosis. In the human JGCT cell line (COV434), TGF1 increased viability by inhibiting apoptosis through a TGF type I receptor (TGFRI)-dependent repression of caspase activity and inhibition of PARP cleavage. These data support a tumor-promoting function of TGF in JGCT through its ability to repress apoptosis. /////////////////////////

Species: mouse
Mutation name:
type: null mutation
fertility: infertile - ovarian defect
Comment: Sexual Fate Change of XX Germ Cells Caused by the Deletion of SMAD4 and STRA8 Independent of Somatic Sex Reprogramming. Wu Q et al. (2016) The differential programming of sperm and eggs in gonads is a fundamental topic in reproductive biology. Although the sexual fate of germ cells is believed to be determined by signaling factors from sexually differentiated somatic cells in fetal gonads, the molecular mechanism that determines germ cell fate is poorly understood. Herein, we show that mothers against decapentaplegic homolog 4 (SMAD4) in germ cells is required for female-type differentiation. Germ cells in Smad4-deficient ovaries respond to retinoic acid signaling but fail to undergo meiotic prophase I, which coincides with the weaker expression of genes required for follicular formation, indicating that SMAD4 signaling is essential for oocyte differentiation and meiotic progression. Intriguingly, germline-specific deletion of Smad4 in Stra8-null female germ cells resulted in the up-regulation of genes required for male gonocyte differentiation, including Nanos2 and PLZF, suggesting the initiation of male-type differentiation in ovaries. Moreover, our transcriptome analyses of mutant ovaries revealed that the sex change phenotype is achieved without global gene expression changes in somatic cells. Our results demonstrate that SMAD4 and STRA8 are essential factors that regulate the female fate of germ cells.//////////////////

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
http://www.ncbi.nlm.nih.gov/UniGene/clust.cgi?ORG=Rn&CID=9774
Search for Antibody


created: Jan. 31, 2000, midnight by: uni   email:
home page:
last update: May 19, 2020, 12:50 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