Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Biol Reprod ; 107(1): 269-274, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35244683

ABSTRACT

Wilms' tumor 1 (Wt1) encodes a zinc finger nuclear transcription factor which is mutated in 15-20% of Wilms' tumor, a pediatric kidney tumor. Wt1 has been found to be involved in the development of many organs. In gonads, Wt1 is expressed in genital ridge somatic cells before sex determination, and its expression is maintained in Sertoli cells and granulosa cells after sex determination. It has been demonstrated that Wt1 is required for the survival of the genital ridge cells. Homozygous mutation of Wt1 causes gonad agenesis. Recent studies find that Wt1 plays important roles in lineage specification and maintenance of gonad somatic cells. In this review, we will summarize the recent research works about Wt1 in gonadal somatic cell differentiation.


Subject(s)
Cell Differentiation , Gonads , WT1 Proteins , Animals , Female , Genes, Wilms Tumor , Gonads/growth & development , Humans , Male , Mice , WT1 Proteins/genetics , WT1 Proteins/physiology
2.
Int J Mol Sci ; 22(14)2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34299295

ABSTRACT

Nearly three decades ago, the Wilms' tumor suppressor Wt1 was identified as a crucial regulator of heart development. Wt1 is a zinc finger transcription factor with multiple biological functions, implicated in the development of several organ systems, among them cardiovascular structures. This review summarizes the results from many research groups which allowed to establish a relevant function for Wt1 in cardiac development and disease. During development, Wt1 is involved in fundamental processes as the formation of the epicardium, epicardial epithelial-mesenchymal transition, coronary vessel development, valve formation, organization of the cardiac autonomous nervous system, and formation of the cardiac ventricles. Wt1 is further implicated in cardiac disease and repair in adult life. We summarize here the current knowledge about expression and function of Wt1 in heart development and disease and point out controversies to further stimulate additional research in the areas of cardiac development and pathophysiology. As re-activation of developmental programs is considered as paradigm for regeneration in response to injury, understanding of these processes and the molecules involved therein is essential for the development of therapeutic strategies, which we discuss on the example of WT1.


Subject(s)
Heart/growth & development , Myocardium/metabolism , WT1 Proteins/metabolism , Animals , Epithelial-Mesenchymal Transition/physiology , Gene Expression/genetics , Heart/embryology , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Pericardium/embryology , Pericardium/pathology , Transcription Factors/metabolism , WT1 Proteins/genetics , WT1 Proteins/physiology
3.
IUBMB Life ; 73(6): 866-882, 2021 06.
Article in English | MEDLINE | ID: mdl-33759343

ABSTRACT

Gene expression profiling has been broadly performed in the field of cancer research. This study aims to explore the key gene regulatory network and focuses on the functions of microRNA (miR)-216a in pancreatic cancer (PC). PC datasets GSE15471, GSE16515, and GSE32676 were used to screen the differentially expressed genes (DEGs) in PC. A miRNA microarray analysis and gene oncology analysis suggested miR-216a as an important differentially expressed miRNA in PC. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis suggested that miR-216a and the DEGs are largely enriched on the phosphatidyl inositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway. miR-216a targeted Wilms Tumor 1 (WT1), while WT1 promoted transcription activity of keratin 7 (KRT7). Upregulation of miR-216a reduced proliferation and invasiveness of PC cells, while further upregulation of WT1 blocked the functions of miR-216a. Silencing of KRT7 diminished the oncogenic role of WT1. The in vitro results were reproduced in vivo. High expression of miR-216a while poor expression of WT1 indicated better prognosis of PC patients. The miR-216a/WT1/KRT7 axis influenced the activity of the PI3K/AKT pathway. To conclude, this study evidenced that miR-216a suppressed WT1 expression and blocked KRT7 transcription, which inactivated the PI3K/AKT signaling and reduced PC progression.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , Keratin-7/biosynthesis , MicroRNAs/genetics , Neoplasm Proteins/biosynthesis , Pancreatic Neoplasms/genetics , RNA, Neoplasm/genetics , Transcriptome , WT1 Proteins/biosynthesis , Adult , Aged , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Ontology , Genes, Wilms Tumor , Heterografts , Humans , Keratin-7/genetics , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Pancreas/chemistry , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Protein Interaction Mapping , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , WT1 Proteins/genetics , WT1 Proteins/physiology
4.
Domest Anim Endocrinol ; 74: 106521, 2021 01.
Article in English | MEDLINE | ID: mdl-32739762

ABSTRACT

Wilms' tumor gene WT1 encodes a nuclear transcriptional factor, which has been shown to regulate granulosa cell steroidogenesis in bovine; however, it is not known whether the functions of theca cells are regulated by WT1. Here, we determined the effects of this gene on theca cell proliferation, apoptosis, and steroidogenesis in vitro. In cultured bovine theca cells, the downregulation of WT1 increased the secretion of progesterone but had no effect on proliferation and apoptosis. WT1 includes the variants WT1(+KTS) and WT1(-KTS), which differ by 3 amino acids KTS (lysine, threonine, and serine). WT1(±KTS) upregulation increased the messenger RNA (mRNA) expression of STAR and CYP17A1 and decreased the progesterone secretion and CYP11A1 mRNA expression. In contrast to WT1(+KTS), WT1(-KTS) upregulation also decreased the mRNA expression of 3ß-HSD. In both variants, WT1(-KTS) has more obvious effects. In conclusion, WT1 can decrease progesterone secretion, likely due in part to the inhibition of CYP11A1 and 3ß-HSD.


Subject(s)
Progesterone/biosynthesis , Theca Cells/metabolism , WT1 Proteins/physiology , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cattle , Cell Proliferation/physiology , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/antagonists & inhibitors , Female , Gene Expression/physiology , Gene Knockdown Techniques , Progesterone/genetics , RNA, Small Interfering/genetics , Transfection , WT1 Proteins/genetics
5.
Biol Reprod ; 103(1): 60-69, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32301970

ABSTRACT

Wt1 gene encodes a nuclear transcription factor which is specifically expressed in ovarian granulosa cells and testicular Sertoli cells. Our previous studies demonstrated that Wt1 is required for the lineage specification of supporting cells and inactivation of Wt1 results in Sertoli cells to Leydig-like cells transformation. To test whether Wt1 is also involved in lineage maintenance of granulosa cells during ovary development, Wt1 was specifically deleted in pre-granulosa cells using Foxl2-cre. We found that the female Wt1-/flox; Foxl2-cre mice were infertile with atrophic ovaries and no growing follicles with multiple layers of granulosa cells were observed. A large number of 3ß-HSD-positive steroidogenic cells were detected in ovaries of Wt1-/flox; Foxl2-cre mice during embryonic stage and these cells were derived from Foxl2-expressing pre-granulosa cells. The quantitative results showed the expression of granulosa cell marker genes (Foxl2, Follistatin) was downregulated and steroidogenic cell marker genes (3ß-HSD, Cyp11a1, Star and Sf1) was dramatically increased in Wt1-/flox; Foxl2-cre ovaries. We also found that the meiosis of germ cells in Wt1-/flox; Foxl2-cre ovaries was delayed but not arrested. This study demonstrates that Wt1 is required for lineage maintenance of granulosa cells and inactivation of Wt1 results in pre-granulosa cells to steroidogenic cells transformation which in turn causes the defect of ovary development.


Subject(s)
Cell Differentiation/physiology , Granulosa Cells/physiology , Ovary/growth & development , Steroids/biosynthesis , WT1 Proteins/deficiency , WT1 Proteins/physiology , 3-Hydroxysteroid Dehydrogenases/analysis , Animals , Cellular Reprogramming , Crosses, Genetic , Female , Forkhead Box Protein L2/genetics , Forkhead Box Protein L2/physiology , Granulosa Cells/enzymology , Infertility, Female/etiology , Male , Meiosis/physiology , Mice , Mice, Knockout , Mice, Transgenic , Ovarian Follicle/growth & development , Ovary/enzymology , Sex Differentiation/physiology , WT1 Proteins/genetics
6.
Theriogenology ; 138: 84-93, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31302435

ABSTRACT

Sertoli cells (SCs) are polarized epithelial cells and provide a microenvironment for the development of germ cells (GCs). The Wilms' tumor suppressor gene WT1 which support spermatogenesis is expressed explicitly in SCs. This study investigated the effect of WT1 on the polarity and blood-testis barrier (BTB) formation of bovine SCs in order to provide theoretical and practical bases for the spermatogenic process in mammals. In this study, newborn calf SCs were used as research material, and the RNAi technique was used to knockdown the endogenous WT1. The results show that WT1 knockdown did not affect the proliferation ability of SCs, but down-regulated the expression of polarity-associated proteins (Par3, Par6b, and E-cadherin), junction-associated protein (occludin) and inhibits transcription of downstream genes (WNT4, JNK, αPKC, and CDC42) in non-canonical WNT signaling pathway. WT1 also altered ZO-1 and occludin protein distribution. Overexpression of WNT1 did not affect the expression of Par6b, E-cadherin, and occludin, whereas the non-canonical WNT signaling pathway inhibitors wnt-c59, CCG-1423, and GO-6983 down-regulated the expression of Par6b, E-cadherin, and occludin respectively. This study indicates that WT1 mediates the regulation of several proteins involved in bovine SCs polarity maintenance and intercellular tight junctions (TJ) by non-canonical WNT signaling pathway.


Subject(s)
Cell Polarity/genetics , Sertoli Cells/physiology , Tight Junctions/genetics , WT1 Proteins/physiology , Wnt Signaling Pathway/physiology , Animals , Animals, Newborn , Blood-Testis Barrier/metabolism , Cattle , Cells, Cultured , Male , Spermatogenesis/genetics , Tight Junctions/metabolism , WT1 Proteins/genetics , Wnt Signaling Pathway/genetics
7.
Cancer Lett ; 451: 48-57, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30851419

ABSTRACT

WT1 associated protein (WTAP), playing an important role in several malignancies owing to its complex function in transcriptional and post-transcriptional regulation, is an independent prognostic indicator for pancreatic cancer (PC). However, its specific role and underlying mechanism in PC remain unclear. In the present study, we found that WTAP could promote migration/invasion and suppress chemo-sensitivity to gemcitabine in PC. Further mechanical investigation revealed that WTAP could bind to and stabilize Fak mRNA which in turn activated the Fak-PI3K-AKT and Fak-Src-GRB2-Erk1/2 signaling pathways. In addition, GSK2256098, a specific Fak inhibitor, could reverse WTAP-mediated chemo-resistance to gemcitabine and metastasis in PC. Taken together, Fak inhibitor might be a promising therapeutic option for PC patients with WTAP overexpression.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Focal Adhesion Protein-Tyrosine Kinases/genetics , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , WT1 Proteins/physiology , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Signal Transduction , Gemcitabine
8.
Br J Cancer ; 119(12): 1508-1517, 2018 12.
Article in English | MEDLINE | ID: mdl-30374123

ABSTRACT

BACKGROUND: The Wilms' tumour protein (WT1), which influences tumour development and angiogenesis, is a promising therapeutic target in breast cancer. We hypothesised that WT1 expression would vary in endothelial cells in distinct sub-classifications of breast cancer. METHODS: WT1 expression and vascular density were quantified by immunohistochemical analysis of human (n = 57) and murine breast cancers. Human tumours were sub-classified by histopathological grade, ER status and HER2 enrichment. RESULTS: WT1 was identified in endothelial (and epithelial and smooth muscle) cells in tumours and tumour-free tissues (controls) from patients and mice with breast cancer. WT1 expression was higher in tumours than in controls, but this was not due to increased endothelial WT1. Vascular WT1 in cancers decreased as histopathological grade increased. WT1 was higher in ER-positive versus ER-negative cancers. Strikingly, reduced WT1 expression in controls correlated with an increased Nottingham Prognostic Index score. CONCLUSIONS: Expression of WT1 is increased in breast cancers but this is not limited to the vascular compartment. The association between reduced WT1 in tumour-free tissue and poor prognosis suggests a protective role for WT1 in the healthy breast.


Subject(s)
Breast Neoplasms/pathology , WT1 Proteins/analysis , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Female , Humans , Mice , Neoplasm Grading , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , WT1 Proteins/physiology
9.
Haematologica ; 103(2): 266-277, 2018 02.
Article in English | MEDLINE | ID: mdl-29170254

ABSTRACT

Loss-of-function mutations and deletions in Wilms tumor 1 (WT1) gene are present in approximately 10% of T-cell acute lymphoblastic leukemia. Clinically, WT1 mutations are enriched in relapsed series and are associated to inferior relapse-free survival in thymic T-cell acute lymphoblastic leukemia cases. Here we demonstrate that WT1 plays a critical role in the response to DNA damage in T-cell leukemia. WT1 loss conferred resistance to DNA damaging agents and attenuated the transcriptional activation of important apoptotic regulators downstream of TP53 in TP53-competent MOLT4 T-leukemia cells but not in TP53-mutant T-cell acute lymphoblastic leukemia cell lines. Notably, WT1 loss positively affected the expression of the X-linked inhibitor of apoptosis protein, XIAP, and genetic or chemical inhibition with embelin (a XIAP inhibitor) significantly restored sensitivity to γ-radiation in both T-cell acute lymphoblastic leukemia cell lines and patient-derived xenografts. These results reveal an important role for the WT1 tumor suppressor gene in the response to DNA damage, and support the view that anti-XIAP targeted therapies could have a role in the treatment of WT1-mutant T-cell leukemia.


Subject(s)
DNA Damage/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Protein p53/drug effects , WT1 Proteins/deficiency , Animals , Cell Line, Tumor , Dose-Response Relationship, Radiation , Gamma Rays , Heterografts , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/physiology , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Tumor Suppressor Protein p53/physiology , WT1 Proteins/physiology
10.
J Nephrol ; 30(1): 11-18, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27059325

ABSTRACT

The study of kidney development at the cellular and molecular levels remains an active area of nephrology research. The functional integrity of the kidney depends on normal development as well as on physiological cell turnover. Apoptosis induction is essential for these mechanisms. A route to cell death revealed in the past decade shows that heat shock proteins (HSPs) and their cofactors are responsible for regulating the apoptotic pathway. Specifically, heat shock protein 70 (Hsp70), the most ubiquitous and highly conserved HSP, helps proteins adopt native conformation or regain function after misfolding. Hsp70 is an important cofactor for the function of Wilms' tumour 1 (WT1) and suggests a potential role for this chaperone during kidney differentiation. In addition, we have demonstrated that WT1 expression is modulated by nitric oxide (NO) availability and Hsp70 interaction after neonatal unilateral ureteral obstruction. NO has been identified as playing an important role in the developing kidney. These findings suggest that Hsp70 and NO may play a critical and fundamental role in the capacity to modulate both apoptotic pathway and oxidative stress during kidney development. Furthermore, the design of experimental protocols that assess renal epithelial functionality in this context, could contribute to the understanding of renal development and alterations.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Kidney/embryology , Nitric Oxide/physiology , WT1 Proteins/physiology , Adult , Animals , Apoptosis , Epithelial-Mesenchymal Transition , HSP72 Heat-Shock Proteins/physiology , Humans , Oxidative Stress
11.
Eur J Gynaecol Oncol ; 37(6): 852-857, 2016.
Article in English | MEDLINE | ID: mdl-29943935

ABSTRACT

In this study, the authors investigated the expression and significance of WTl in xenotransplanted ovarian carcinoma cell SKOV3 of nude mice treated with paclitaxel. Xenotransplanted ovarian carcinoma was established in nude mice using the SKOV3 cell line. The mice were randomized into the treatment group with paclitaxel and control group with normal sodium. The sizes of the xenotransplanted tumors were measured and the tumor specimens were confirmed by routine hemotoxylin-eosin (H&E) staining. The apoptosis index was then assayed using flow cytometry. WTl and bcl-2 expression were detected with immunohistochemistry, and WT1 mRNA expression was determined by reverse transcriptase polymerase chain reaction (RT-PCR). The authors found that the growth of the xenotransplanted tumor was inhibited by paclitaxel therapy. Compared to the control group, the apoptosis rate was significantly increased in the treatment group (p < 0.05). At the same time, the expression of WTl, bcl-2 and WTI, mRNA were significantly decreased in the paclitaxel therapy group (p < 0.05). The authors conclude that the WTl gene may play an important role during apoptosis of ovarian carcinoma and the mechanism may be closely related to bcl-2.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , WT1 Proteins/physiology , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/analysis , WT1 Proteins/analysis , Xenograft Model Antitumor Assays
12.
Nat Rev Nephrol ; 11(9): 535-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26055352

ABSTRACT

Podocytes are unique, highly specialized, terminally differentiated cells that are integral components of the kidney glomerular filtration barrier. Podocytes are vulnerable to a variety of injuries and in response they undergo a series of changes ranging from hypertrophy, autophagy, dedifferentiation, mesenchymal transition and detachment to apoptosis, depending on the nature and extent of the insult. Emerging evidence indicates that Wnt/ß-catenin signalling has a central role in mediating podocyte dysfunction and proteinuria. Wnts are induced and ß-catenin is activated in podocytes in various proteinuric kidney diseases. Genetic or pharmacologic activation of ß-catenin is sufficient to impair podocyte integrity and causes proteinuria in healthy mice, whereas podocyte-specific ablation of ß-catenin protects against proteinuria after kidney injury. Mechanistically, Wnt/ß-catenin controls the expression of several key mediators implicated in podocytopathies, including Snail1, the renin-angiotensin system and matrix metalloproteinase 7. Wnt/ß-catenin also negatively regulates Wilms tumour protein, a crucial transcription factor that safeguards podocyte integrity. Targeted inhibition of Wnt/ß-catenin signalling preserves podocyte integrity and ameliorates proteinuria in animal models. This Review highlights advances in our understanding of the pathomechanisms of Wnt/ß-catenin signalling in mediating podocyte injury, and describes the therapeutic potential of targeting this pathway for the treatment of proteinuric kidney disease.


Subject(s)
Kidney Diseases/physiopathology , Podocytes , Proteinuria/physiopathology , Wnt Signaling Pathway/physiology , Animals , Humans , WT1 Proteins/physiology
13.
Mol Cell ; 57(4): 573-574, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25699704

ABSTRACT

In this issue, Wang et al., 2015 describes that WT1 recruits TET2 to the DNA, an important feature of a new regulatory pathway linked to the development of acute myeloid leukemia (AML). This pathway consists of WT1, IDH1/2, and TET2 (WIT) genes, with exclusive mutations of the three genes inducing myeloid cell proliferation.


Subject(s)
DNA-Binding Proteins/physiology , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/physiology , WT1 Proteins/physiology , Dioxygenases , Humans
14.
Mol Cell ; 57(4): 662-673, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25601757

ABSTRACT

The TET2 DNA dioxygenase regulates cell identity and suppresses tumorigenesis by modulating DNA methylation and expression of a large number of genes. How TET2, like most other chromatin-modifying enzymes, is recruited to specific genomic sites is unknown. Here we report that WT1, a sequence-specific transcription factor, is mutated in a mutually exclusive manner with TET2, IDH1, and IDH2 in acute myeloid leukemia (AML). WT1 physically interacts with and recruits TET2 to its target genes to activate their expression. The interaction between WT1 and TET2 is disrupted by multiple AML-derived TET2 mutations. TET2 suppresses leukemia cell proliferation and colony formation in a manner dependent on WT1. These results provide a mechanism for targeting TET2 to a specific DNA sequence in the genome. Our results also provide an explanation for the mutual exclusivity of WT1 and TET2 mutations in AML, and suggest an IDH1/2-TET2-WT1 pathway in suppressing AML.


Subject(s)
DNA-Binding Proteins/physiology , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/physiology , WT1 Proteins/physiology , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Gene Expression Regulation, Neoplastic , HEK293 Cells , HL-60 Cells , Humans , Inhibitor of Differentiation Protein 2/genetics , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
15.
J Ovarian Res ; 7: 118, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25491731

ABSTRACT

BACKGROUND: The important role of WT1 in early folliculogenesis was evident from its restricted expression pattern in immature follicles and from its involvement in transcriptional control of inhibin-α and FSH receptor. There is also considerable evidence that WT1 is a potent inhibitor of apoptotic cell death in the developing kidney and male germ cells, suggesting that it could play a role in the regulation of follicle survival. Therefore, we evaluated if WT1 involves in cell survival of granulosa cells (GCs) during the FSH-independent stage. METHODS: GCs were obtained from small preantral follicles of immature rat ovary. Bax and bcl-2 mRNA and protein levels in GCs transfected with WT1 (-KTS) or WT1 (+KTS) were analyzed by Real-time RT-PCR and immune-blotting analysis. Cell viability was measured with MTT assays and apoptosis was analyzed with caspase 3/7 activity and TUNEL assay. The mechanism by which WT1 regulates Bax expression was investigated using Bax promoter-luciferase reporter assay and ChIP assays from GCs. RESULTS: Here, we showed that WT1 (-KTS) suppressed endogenous Bax transcript and protein expression, and this inhibition resulted from direct binding of WT1 in the Bax promoter region in vivo. In addition, anti-apoptotic effects of WT1 (-KTS) were demonstrated based on MTT assays, a sensitive bioluminescence caspase 3/7 assay and TUNEL assays. On the other hand, WT1 has no role on bcl-2 expression in GCs. CONCLUSION: These findings suggest that activation of WT1 is necessary for maintenance of GC survival during early stage of follicles and WT1 can play a role in protecting apoptosis through the regulation of upstream activator (Bax), as well as through regulation of downstream effecter (caspases 3 and 7).


Subject(s)
Apoptosis , Granulosa Cells/metabolism , WT1 Proteins/physiology , bcl-2-Associated X Protein/genetics , Animals , Base Sequence , Cell Survival , Cells, Cultured , Down-Regulation , Female , Gene Expression Regulation , Promoter Regions, Genetic , Rats, Sprague-Dawley , Transcription, Genetic , bcl-2-Associated X Protein/metabolism
16.
Am J Physiol Endocrinol Metab ; 307(12): E1131-43, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25336526

ABSTRACT

Wilms' tumor 1 (Wt1) is a tumor suppressor gene encoding ∼24 zinc finger transcription factors. In the mammalian testis, Wt1 is expressed mostly by Sertoli cells (SCs) involved in testis development, spermatogenesis, and adult Leydig cell (ALC) steroidogenesis. Global knockout (KO) of Wt1 is lethal in mice due to defects in embryogenesis. Herein, we showed that Wt1 is involved in regulating fetal Leydig cell (FLC) degeneration and ALC differentiation during testicular development. Using Wt1(-/flox);Amh-Cre mice that specifically deleted Wt1 in the SC vs. age-matched wild-type (WT) controls, FLC-like-clusters were found in Wt1-deficient testes that remained mitotically active from postnatal day 1 (P1) to P56, and no ALC was detected at these ages. Leydig cells in mutant adult testes displayed morphological features of FLC. Also, FLC-like cells in adult mutant testes had reduced expression in ALC-associated genes Ptgds, Sult1e1, Vcam1, Hsd11b1, Hsd3b6, and Hsd17b3 but high expression of FLC-associated genes Thbs2 and Hsd3b1. Whereas serum LH and testosterone level in mutant mice were not different from controls, intratesticular testosterone level was significantly reduced. Deletion of Wt1 gene also perturbed the expression of steroidogenic enzymes Star, P450c17, Hsd3b6, Hsd3b1, Hsd17b1, and Hsd17b3. FLCs in adult mutant testes failed to convert androstenedione to testosterone due to a lack of Hsd17b3, and this defect was rescued by coculturing with fetal SCs. In summary, FLC-like cells in mutant testes are putative FLCs that remain mitotically active in adult mice, illustrating that Wt1 dictates the fate of FLC and ALC during postnatal testis development.


Subject(s)
Cell Differentiation/genetics , Leydig Cells/physiology , Testis/embryology , Testis/growth & development , WT1 Proteins/physiology , Animals , Embryo, Mammalian , Fetus/embryology , Fetus/physiology , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sertoli Cells/physiology , Testis/cytology
17.
Biochim Biophys Acta ; 1846(1): 180-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25018051

ABSTRACT

Wilms tumor recapitulates the development of the kidney and represents a unique opportunity to understand the relationship between normal and tumor development. This has been illustrated by the findings that mutations of Wnt/ß-catenin pathway-related WT1, ß-catenin, and WTX together account for about one-third of Wilms tumor cases. While intense efforts are being made to explore the genetic basis of the other two-thirds of tumor cases, it is worth noting that, epigenetic changes, particularly the loss of imprinting of the DNA region encoding the major fetal growth factor IGF2, which results in its biallelic over-expression, are closely associated with the development of many Wilms tumors. Recent investigations also revealed that mutations of Drosha and Dicer, the RNases required for miRNA generation, and Dis3L2, the 3'-5' exonuclease that normally degrades miRNAs and mRNAs, could cause predisposition to Wilms tumors, demonstrating that miRNA can play a pivotal role in Wilms tumor development. Interestingly, Lin28, a direct target of miRNA let-7 and potent regulator of stem cell self-renewal and differentiation, is significantly elevated in some Wilms tumors, and enforced expression of Lin28 during kidney development could induce Wilms tumor. With the success in establishing mice nephroblastoma models through over-expressing IGF2 and deleting WT1, and advances in understanding the ENU-induced rat model, we are now able to explore the molecular and cellular mechanisms induced by these genetic, epigenetic, and miRNA alterations in animal models to understand the development of Wilms tumor. These animal models may also serve as valuable systems to assess new treatment targets and strategies for Wilms tumor.


Subject(s)
Kidney Neoplasms/genetics , MicroRNAs/physiology , WT1 Proteins/physiology , Wilms Tumor/genetics , Animals , Cell Transformation, Neoplastic/genetics , Genes, Wilms Tumor , Humans , Kidney Neoplasms/pathology , Mice , Neoplasms, Experimental , Rats , Wilms Tumor/pathology
18.
Biochem J ; 461(1): 15-32, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24927120

ABSTRACT

The WT1 (Wilms' tumour 1) gene encodes a zinc finger transcription factor and RNA-binding protein that direct the development of several organs and tissues. WT1 manifests both tumour suppressor and oncogenic activities, but the reasons behind these opposing functions are still not clear. As a transcriptional regulator, WT1 can either activate or repress numerous target genes resulting in disparate biological effects such as growth, differentiation and apoptosis. The complex nature of WT1 is exemplified by a plethora of isoforms, post-translational modifications and multiple binding partners. How WT1 achieves specificity to regulate a large number of target genes involved in diverse physiological processes is the focus of the present review. We discuss the wealth of the growing molecular information that defines our current understanding of the versatility and utility of WT1 as a master regulator of organ development, a tumour suppressor and an oncogene.


Subject(s)
Gene Expression Regulation, Neoplastic , WT1 Proteins/physiology , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Genes, Tumor Suppressor , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Isoforms/genetics , Zinc Fingers/genetics
19.
J Am Soc Nephrol ; 25(11): 2539-45, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24722440

ABSTRACT

Odd-skipped related 1 (Osr1) encodes a zinc finger transcription factor required for kidney development. Osr1 deficiency in mice results in metanephric kidney agenesis, whereas knockdown or mutation studies in zebrafish revealed that pronephric nephrons require osr1 for proximal tubule and podocyte development. osr1-deficient pronephric podocyte progenitors express the Wilms' tumor suppressor wt1a but do not undergo glomerular morphogenesis or express the foot process junctional markers nephrin and podocin. The function of osr1 in podocyte differentiation remains unclear, however. Here, we found by double fluorescence in situ hybridization that podocyte progenitors coexpress osr1 and wt1a. Knockdown of wt1a disrupted podocyte differentiation and prevented expression of osr1. Blocking retinoic acid signaling, which regulates wt1a, also prevented osr1 expression in podocyte progenitors. Furthermore, unlike the osr1-deficient proximal tubule phenotype, which can be rescued by manipulation of endoderm development, podocyte differentiation was not affected by altered endoderm development, as assessed by nephrin and podocin expression in double osr1/sox32-deficient embryos. These results suggest a different, possibly cell- autonomous requirement for osr1 in podocyte differentiation downstream of wt1a. Indeed, osr1-deficient embryos did not exhibit podocyte progenitor expression of the transcription factor lhx1a, and forced expression of activated forms of the lhx1a gene product rescued nephrin expression in osr1-deficient podocytes. Our results place osr1 in a framework of transcriptional regulators that control the expression of podocin and nephrin and thereby mediate podocyte differentiation.


Subject(s)
Podocytes/physiology , Transcription Factors/physiology , WT1 Proteins/physiology , Zebrafish Proteins/physiology , Animals , Cell Differentiation/physiology , Female , Gene Expression Regulation, Developmental , Kidney Tubules/cytology , Kidney Tubules/embryology , Kidney Tubules/physiology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/physiology , Male , Podocytes/cytology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics , Transcription, Genetic/physiology , WT1 Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
20.
Hum Mol Genet ; 23(2): 333-41, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24009315

ABSTRACT

The Wt1 gene encodes a nuclear transcription factor that is specifically expressed in ovarian granulosa cells. However, the physiological significance of Wt1 in ovarian follicle development remains elusive. In this study, we found that Wt1(+/R394W) mice were grossly normal, however, the females displayed severe reproductive defects. Only ∼15% of the Wt1(+/R394W) females became pregnant after mating with wild-type males, compared with 88.2% of control females. Further study revealed that the subfertility of Wt1(+/R394W) females was caused by aberrant ovarian follicle development. Compared with control females, the ovary size and the number of developing follicles was significantly decreased in Wt1 mutant ovaries which was very similar to premature ovarian failure (POF) in human patients. The results of in vitro studies demonstrated that the expression of follicle stimulating hormone receptor (FSHR), 3ß-hydroxysteroid dehydrogenase and Aromatase was inhibited by Wt1 in granulosa cells, and mutation of Wt1 resulted in the upregulation of these genes and in the premature differentiation of granulosa cells. We also found that Wt1 was likely involved in granulosa cell development via the regulation of E-cadherin and Par6b expression. Mutation in Wt1 caused defects in polarity establishment in granulosa cells, which also likely contributed to the observed aberrant follicle development. The results of this study provide new mechanisms for understanding the regulation of ovarian follicle development and potential pathological cause of POF in human patients.


Subject(s)
Granulosa Cells/cytology , Granulosa Cells/metabolism , Ovarian Follicle/physiology , WT1 Proteins/physiology , 3-Hydroxysteroid Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Aromatase/metabolism , Cadherins/metabolism , Cell Differentiation , Cell Polarity , Female , Fertility , Gene Expression Regulation , Granulosa Cells/pathology , Humans , Male , Mice , Ovarian Follicle/pathology , Ovulation , Pregnancy , Primary Ovarian Insufficiency/pathology , Receptors, FSH/metabolism , WT1 Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...