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

Wnt family member 5A OKDB#: 1546
 Symbols: WNT5A Species: human
 Synonyms: hWNT5A  Locus: 3p14.3 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 Wnt genes belong to a family of protooncogenes with at least 13 known members that are expressed in species ranging from Drosophila to man. The name Wnt denotes the relationship of this family to the Drosophila segment polarity gene 'wingless' and to its vertebrate ortholog, Int1, a mouse protooncogene.

NCBI Summary: The WNT gene family consists of structurally related genes which encode secreted signaling proteins. These proteins have been implicated in oncogenesis and in several developmental processes, including regulation of cell fate and patterning during embryogenesis. This gene encodes a member of the WNT family that signals through both the canonical and non-canonical WNT pathways. This protein is a ligand for the seven transmembrane receptor frizzled-5 and the tyrosine kinase orphan receptor 2. This protein plays an essential role in regulating developmental pathways during embryogenesis. This protein may also play a role in oncogenesis. Mutations in this gene are the cause of autosomal dominant Robinow syndrome. Alternate splicing results in multiple transcript variants. [provided by RefSeq, Jan 2012]
General function Ligand, Growth factor
Comment
Cellular localization Secreted
Comment
Ovarian function Germ cell migration, Oocyte maturation
Comment Non-canonical WNT5a regulates Epithelial-to-Mesenchymal Transition in the mouse ovarian surface epithelium. Abedini A et al. (2020) The ovarian surface epithelium (OSE) is a monolayer that covers the ovarian surface and is involved in ovulation by rupturing and enabling release of a mature oocyte and by repairing the wound after ovulation. Epithelial-to-mesenchymal transition (EMT) is a mechanism that may promote wound healing after ovulation. While this process is poorly understood in the OSE, in other tissues wound repair is known to be under the control of the local microenvironment and different growth factors such as the WNT signaling pathway. Among WNT family members, WNT4 and WNT5a are expressed in the OSE and are critical for the ovulatory process. The objective of this study was to determine the potential roles of WNT4 and WNT5a in regulating the OSE layer. Using primary cultures of mouse OSE cells, we found WNT5a, but not WNT4, promotes EMT through a non-canonical Ca2+-dependent pathway, up-regulating the expression of Vimentin and CD44, enhancing cell migration, and inhibiting the CTNNB1 pathway and proliferation. We conclude that WNT5a is a stimulator of the EMT in OSE cells, and acts by suppressing canonical WNT signaling activity and inducing the non-canonical Ca2+ pathway.////////////////// Fam70A binds Wnt5a to regulate meiosis and quality of mouse oocytes. Zhang NN et al. (2020) Little is known about the roles of integral membrane proteins beyond channels, carriers or receptors in meiotic oocytes. The transmembrane protein Fam70A was previously identified as a likely "female fertility factor" in Fox3a-knockout mouse ovaries where almost all follicles underwent synchronous activation and the mice became infertile very early. However, whether Fam70A functions in oocyte meiosis remains unknown. Therefore, the present study aimed to address this question. Co-immunoprecipitation, immunogold labelling-electron microscopy, co-localization and yeast two-hybrid assays were used to verify the interaction. Antibody or small interfering RNA transfection was used to deplete the proteins. Immunofluorescence, immunohistochemistry and live tracker staining were used to examine the localization or characterize phenotypes. Western blot was used to examine the protein level. Fam70A was enriched in oocyte membranes important for normal meiosis. Fam70A depletion remarkably disrupted spindle assembly, chromosome congression and first polar body extrusion, which subsequently increased aneuploidy and abnormal fertilization. Moreover, Fam70A directly bound Wnt5a, the most abundant Wnt member within oocytes. Depletion of either Fam70A or Wnt5a remarkably increased adenomatous polyposis coli (APC), which stabilizes active β-catenin and microtubules. Consequently, depletion of either Fam70A or Wnt5a remarkably increased p-β-catenin (inactive form) and acetylated tubulin, while APC knockdown remarkably decreased these two. Furthermore, Fam70A depletion remarkably reduced Akt phosphorylation. Fam70A regulates meiosis and quality of mouse oocytes through both canonical and non-canonical Wnt5a signalling pathways.//////////////////
Expression regulated by
Comment
Ovarian localization Oocyte, Granulosa, Surface epithelium
Comment The WNT/-catenin signaling pathway and expression of survival promoting genes in luteinized granulosa cells: endometriosis as a paradigm for a dysregulated apoptosis pathway. Sanchez AM 2014 et al. OBJECTIVE To analyze the WNT/-catenin signaling pathway in luteinized granulosa cells from women with and without endometriosis in relation to cellular apoptosis. DESIGN Basic. SETTING University hospital. PATIENT(S) Patients with a laparoscopic diagnosis of endometriosis (n=30) and women undergoing intracytoplasmic sperm injection for male infertility (control group n=39). INTERVENTION(S) Isolation of luteinized granulosa cells. MAIN OUTCOME MEASURE(S) Gene expression analysis of components of the WNT/-catenin pathway, protein expression levels of -catenin, and cell cycle studies in luteinized granulosa cells. RESULT(S) Compared with luteinized granulosa cells from control women, cells derived from endometriosis patients had significantly higher transcript levels of the -catenin-independent molecules WNT4 and WNT5a and lower levels of the -catenin-dependent molecule WNT1. A decrease of total -catenin as well as of its dephosphorylated active form, together with an aberrant gene expression of the downstream targets survivin and BMP4, was detected in cells from affected women. Flow cytometry analysis confirmed an enhanced apoptosis of luteinized granulosa cells from patients with endometriosis. CONCLUSION(S) The concomitant dysregulation of specific members of the WNT pathway and of its pivot molecule -catenin in granulosa cells characterized by an increased apoptosis suggests that the WNT/-catenin signaling pathway might be involved in leading to granulosa cell atresia. ///////////////////////// Albert Ricken et al reported the Wnt Signaling in the Ovary and the identification and Compartmentalized Expression of wnt-2, wnt-2b, and Frizzled-4 mRNAs . Using RT-PCR with degenerate primers on RNA from ovaries of hormone-stimulated immature rats, the authors identified transcripts for wnt-2 and wnt-2b. RT-PCR and in situ hybridization (ISH) demonstrated that granulosa cells express wnt-2 mRNA.. RT-PCR analysis, using primers designed from this wnt-2b cDNA sequence, failed to detect transcripts in the ovarian follicular compartment (granulosa and oocyte). ISH revealed that the ovarian surface epithelium expresses wnt-2b mRNA. Using a similar degenerate RT-PCR approach, the authors detected expression of a putative wnt receptor, frizzled-4 (fzd-4), and a cytoplasmic component of the wnt signaling cascade, disheveled-2 (dsh-2), in the rat ovary. Further analyses using both RT-PCR and ISH indicated that granulosa cells express fzd-4 mRNA. RT-PCR, using degenerate and specific primers for wnts, on RNA from five ovarian cancer cell lines confirmed the expression of transcripts for wnt-2b. Two additional wnt transcripts (wnt-5a and wnt-11) were detected in the cancer cell lines and in the rat ovary. These results demonstrate that transcripts corresponding to components of the wnt signaling cascade are expressed in the immature rat ovary. The localization of these transcripts in specific ovarian compartments suggests that this signal transduction pathway may be involved in follicular development and ovarian function. Furthermore, because wnts have been implicated in the oncogenic transformation of epithelial cells, our results raise the possibility that aberrant wnt expression may be involved in ovarian tumorigenesis in humans.
Follicle stages
Comment Up-Regulated Expression of WNT5a Increases Inflammation and Oxidative Stress via PI3K/AKT/NF-κB Signaling in the Granulosa Cells of PCOS Patients. Zhao Y et al. (2015) Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder accompanied by chronic low-grade inflammation, but the molecular mechanism remains unclear. We investigated the action of WNT5a in the development of chronic inflammation in PCOS and the related molecular signaling pathways. This was a prospective study conducted at the Division of Reproduction Center, Peking University Third Hospital. A total of 35 PCOS patients and 87 control women who reported to the clinic for the in vitro procedure and the cause of marital infertility was male azoospermia were included. Mural granulosa cells (GCs) of 35 PCOS patients and 37 controls were collected during oocyte retrieval and gene expression was analyzed. The human KGN cells and mural GCs from 50 control subjects (six to eight samples were pooled together for each experiment) were cultured in vitro. The regulation of inflammation and oxidative stress was confirmed by quantitative PCR, flow-cytometric assay, and dual-luciferase reporter assay after inflammatory stimuli or WNT5a overexpression. Relevant signaling pathways were identified using specific inhibitors. Our data demonstrate significantly elevated WNT5a expression in the mural GCs of PCOS patients compared with the controls. Lipopolysaccharide stimulation increased WNT5a expression in KGN cells and mural GCs, and BAY-117082 and pyrrolidinedithiocarbamic acid [nuclear factor-κB (NF-κB) inhibitor] treatments suppressed WNT5a mRNA below the control level. WNT5a overexpression also enhanced the expression of inflammation-related genes and increased intracellular reactive oxygen species, whereas both BAY-117082 and LY-294002 (phosphatidylinositol 3-kinase inhibitor) significantly inhibited WNT5a-induced inflammation and oxidative stress. WNT5a acts as a proinflammatory factor in human ovarian GCs. The up-regulated expression of WNT5a in PCOS increases inflammation and oxidative stress predominantly via the phosphatidylinositol 3-kinase/AKT/NF-κB signaling pathway. The proinflammatory cytokines induced might further enhance WNT5a expression via NF-κB-dependent regulation, indicating a novel regulatory system for chronic inflammation in PCOS.//////////////////
Phenotypes PCO (polycystic ovarian syndrome)
Mutations 2 mutations

Species: mouse
Mutation name:
type: None
fertility: None
Comment: Loss of Wnt5a Disrupts Primordial Germ Cell Migration and Male Sexual Development in Mice. Chawengsaksophak K et al. Disruptions in the regulatory pathways controlling sex determination and differentiation can cause disorders of sex development (DSD), often compromising reproductive function. Although extensive efforts have been channeled into elucidating the regulatory mechanisms controlling the many aspects of sexual differentiation, the majority of DSD phenotypes are still unexplained at the molecular level. In this study, we have analyzed the potential involvement of Wnt5a in sexual development and show in mice that Wnt5a is male-specifically up-regulated within testicular interstitial cells at the onset of gonad differentiation. Homozygous deletion of Wnt5a affected sexual development in male mice, causing testicular hypoplasia and bilateral cryptorchidism despite the Leydig cells producing factors such as Hsd3b1 and Insl3. Additionally, Wnt5a-null embryos of both sexes showed a significant reduction in gonadal germ cell numbers, which was caused by aberrant primordial germ cell migration along the hindgut endoderm prior to gonadal colonization. Our results indicate multiple roles for Wnt5a during mammalian reproductive development, and help to clarify further the aetiology of Robinow syndrome (OMIM 268310), a disease previously linked to the WNT5A pathway.

Species: mouse
Mutation name:
type: null mutation
fertility: subfertile
Comment: WNT5a is required for normal ovarian follicle development and antagonizes gonadotropin responsiveness in granulosa cells by suppressing canonical WNT signaling. Abedini A et al. (2015) Whereas the roles of the canonical wingless-type MMTV (mouse mammary tumor virus) integration site family (WNT) signaling pathway in the regulation of ovarian follicle growth and steroidogenesis are now established, noncanonical WNT signaling in the ovary has been largely overlooked. Noncanonical WNTs, including WNT5a and WNT11, are expressed in granulosa cells (GCs) and are differentially regulated throughout follicle development, but their physiologic roles remain unknown. Using conditional gene targeting, we found that GC-specific inactivation of Wnt5a (but not Wnt11) results in the female subfertility associated with increased follicular atresia and decreased rates of ovulation. Microarray analyses have revealed that WNT5a acts to down-regulate the expression of FSH-responsive genes in vitro, and corresponding increases in the expression of these genes have been found in the GCs of conditional knockout mice. Unexpectedly, we found that WNT5a regulates its target genes not by signaling via the WNT/Ca(2+) or planar cell polarity pathways, but rather by inhibiting the canonical pathway, causing both β-catenin (CTNNB1) and cAMP responsive element binding (CREB) protein levels to decrease via a glycogen synthase kinase-3β-dependent mechanism. We further found that WNT5a prevents follicle-stimulating hormone and luteinizing protein from up-regulating the CTNNB1 and CREB proteins and their target genes, indicating that WNT5a functions as a physiologic inhibitor of gonadotropin signaling. Together, these findings identify WNT5a as a key regulator of follicle development and gonadotropin responsiveness.-Abedini, A., Zamberlam, G., Lapointe, E., Tourigny, C., Boyer, A., Paquet, M., Hayashi, K., Honda, H., Kikuchi, A., Price, C., Boerboom, D. WNT5a is required for normal ovarian follicle development and antagonizes gonadotropin responsiveness in granulosa cells by suppressing canonical WNT signaling.//////////////////

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: July 11, 2002, 3:02 p.m. by: hsueh   email:
home page:
last update: Oct. 19, 2020, 1:57 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