Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Sci Rep ; 11(1): 18003, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504167

ABSTRACT

RNA-based therapeutics are emerging as innovative options for cancer treatment, with microRNAs being attractive targets for therapy development. We previously implicated microRNA-642a-5p (miR-642a-5p) as a tumor suppressor in prostate cancer (PCa), and here we characterize its mode of action, using 22Rv1 PCa cells. In an in vivo xenograft tumor model, miR-642a-5p induced a significant decrease in tumor growth, compared to negative control. Using RNA-Sequencing, we identified gene targets of miR-642a-5p which were enriched for gene sets controlling cell cycle; downregulated genes included Wilms Tumor 1 gene (WT1), NUAK1, RASSF3 and SKP2; and upregulated genes included IGFBP3 and GPS2. Analysis of PCa patient datasets showed a higher expression of WT1, NUAK1, RASSF3 and SKP2; and a lower expression of GPS2 and IGFBP3 in PCa tissue compared to non-malignant prostate tissue. We confirmed the prostatic oncogene WT1, as a direct target of miR-642a-5p, and treatment of 22Rv1 and LNCaP PCa cells with WT1 siRNA or a small molecule inhibitor of WT1 reduced cell proliferation. Taken together, these data provide insight into the molecular mechanisms by which miR-642a-5p acts as a tumor suppressor in PCa, an effect partially mediated by regulating genes involved in cell cycle control; and restoration of miR-642-5p in PCa could represent a novel therapeutic approach.


Subject(s)
Cell Cycle/genetics , MicroRNAs/genetics , Prostate/metabolism , Prostatic Neoplasms/genetics , WT1 Proteins/genetics , 3' Untranslated Regions , Animals , Base Pairing , Base Sequence , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, SCID , MicroRNAs/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Survival Analysis , Tumor Burden , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/metabolism , Xenograft Model Antitumor Assays
2.
Cell Rep ; 35(3): 109010, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882316

ABSTRACT

Acute myeloid leukemia (AML) is caused by recurrent mutations in members of the gene regulatory and signaling machinery that control hematopoietic progenitor cell growth and differentiation. Here, we show that the transcription factor WT1 forms a major node in the rewired mutation-specific gene regulatory networks of multiple AML subtypes. WT1 is frequently either mutated or upregulated in AML, and its expression is predictive for relapse. The WT1 protein exists as multiple isoforms. For two main AML subtypes, we demonstrate that these isoforms exhibit differential patterns of binding and support contrasting biological activities, including enhanced proliferation. We also show that WT1 responds to oncogenic signaling and is part of a signaling-responsive transcription factor hub that controls AML growth. WT1 therefore plays a central and widespread role in AML biology.


Subject(s)
Chromatin/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Lung Neoplasms/genetics , WT1 Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chromatin/metabolism , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Signal Transduction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Translocation, Genetic , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
3.
Biochim Biophys Acta Gene Regul Mech ; 1863(12): 194642, 2020 12.
Article in English | MEDLINE | ID: mdl-33017668

ABSTRACT

Dysregulated alternative splicing plays a prominent role in all hallmarks of cancer. The splice factor kinase SRPK1 drives the activity of oncogenic splice factors such as SRSF1. SRSF1 in turn promotes the expression of splice isoforms that favour tumour growth, including proangiogenic VEGF. Knockdown (with siRNA) or chemical inhibition (using SPHINX) of SRPK1 in K562 leukemia and PC3 prostate cancer cell lines reduced cell proliferation, invasion and migration. In glomerular podocytes, the Wilms tumour suppressor zinc-finger transcription factor WT1 represses SRPK1 transcription. Here we show that in cancer cells WT1 activates SRPK1 transcription, unless a canonical WT1 binding site adjacent to the transcription start site is mutated. The ability of WT1 to activate SRPK1 transcription was reversed by the transcriptional corepressor BASP1, and both WT1 and BASP1 co-precipitated with the SRPK1 promoter. BASP1 significantly increased the expression of the antiangiogenic VEGF165b splice isoform. We propose that by upregulating SRPK1 transcription WT1 can direct an alternative splicing landscape that facilitates tumour growth.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , WT1 Proteins/metabolism , Binding Sites , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Male , PC-3 Cells , Promoter Regions, Genetic , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Vascular Endothelial Growth Factor A/metabolism , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
4.
J Biol Chem ; 295(14): 4673-4683, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32098869

ABSTRACT

We previously reported that the transcription factor Wilms tumor 1 (WT1) regulates the expression of insulin-like growth factor-binding protein-1 (IGFBP-1) and prolactin (PRL) during decidualization of human endometrial stromal cells (ESCs). However, other roles of WT1 in decidualization remain to be fully clarified. Here, we investigated how WT1 regulates the physiological functions of human ESCs during decidualization. We incubated ESCs isolated from proliferative-phase endometrium with cAMP to induce decidualization, knocked down WT1 with siRNA, and generated three types of treatments (nontreated cells, cAMP-treated cells, and cAMP-treated + WT1-knockdown cells). To identify WT1-regulated genes, we used gene microarrays and compared the transcriptome data obtained among these three treatments. We observed that WT1 up-regulates 121 genes during decidualization, including several genes involved in lipid transport. The WT1 knockdown inhibited lipid accumulation (LA) in the cAMP-induced ESCs. To examine the mechanisms by which WT1 regulates LA, we focused on very low-density lipoprotein receptor (VLDLR), which is involved in lipoprotein uptake. We found that cAMP up-regulates VLDLR and that the WT1 knockdown inhibits it. Results of ChIP assays revealed that cAMP increases the recruitment of WT1 to the promoter region of the VLDLR gene, indicating that WT1 regulates VLDLR expression. Moreover, VLDLR knockdown inhibited cAMP-induced LA, and VLDLR overexpression reverted the suppression of LA caused by the WT1 knockdown. Taken together, our results indicate that WT1 enhances lipid storage by up-regulating VLDLR expression in human ESCs during decidualization.


Subject(s)
Lipid Metabolism , WT1 Proteins/metabolism , Adult , Cells, Cultured , Cyclic AMP/metabolism , Early Growth Response Protein 1/metabolism , Endometrium/cytology , Female , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Middle Aged , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Receptors, LDL/antagonists & inhibitors , Receptors, LDL/genetics , Receptors, LDL/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
5.
J Cell Mol Med ; 24(2): 1370-1382, 2020 01.
Article in English | MEDLINE | ID: mdl-31845546

ABSTRACT

Wilm's tumour-1 (WT1) is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and enhances metastasis. Deubiquitination stabilizes target proteins, and inhibiting deubiquitination facilitates the degradation of target proteins. However, whether inhibiting deubiquitination of WT1 facilitates its degradation and presents anti-cancer ability in PDAC is unknown. Here, we found that deubiquitinase inhibitor degrasyn rapidly induced the degradation of endogenous and exogenous WT1 through enhancing ubiquitination of WT1 followed by the up-regulation of E-cadherin. Knockdown of WT1 by short hairpin RNAs (shRNAs) inhibited metastasis and overexpression of WT1 partially prevented degrasyn-induced anti-metastasis activity, suggesting that degrasyn presents anti-metastasis activity partially through degrading WT1 protein. We further identified that USP5 deubiquitinated WT1 and stabilized its expression. The higher expressions of USP5 and WT1 are associated with tumour metastasis. More importantly, degrasyn inhibited the activity of USP5 and overexpression of USP5 partially prevented degrasyn-induced degradation of WT1 protein, suggesting that degrasyn degraded WT1 protein through inhibiting the activity of USP5. Finally, degrasyn reduced the tumorigenicity in a xenograft mouse model and reduced the metastasis in vivo. Our results indicate that degrasyn presents strong anti-cancer activity through USP5-WT1-E-cadherin signalling in PDAC. Therefore, degrasyn holds promise as cancer therapeutic agent in PDAC with high expressions of USP5 and WT1.


Subject(s)
Cadherins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Cyanoacrylates/pharmacology , Deubiquitinating Enzymes/antagonists & inhibitors , Endopeptidases/chemistry , Pancreatic Neoplasms/drug therapy , Pyridines/pharmacology , WT1 Proteins/antagonists & inhibitors , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Proliferation , Endopeptidases/genetics , Endopeptidases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Tumor Cells, Cultured , WT1 Proteins/genetics , WT1 Proteins/metabolism , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
7.
Cell Physiol Biochem ; 48(6): 2493-2502, 2018.
Article in English | MEDLINE | ID: mdl-30121641

ABSTRACT

BACKGROUND/AIMS: IL-1ß is an important mediator of "inflammation-cancer" transformation through IL-1ß/NF-κB/COX-2/HIF-1α signaling pathway, whereas certain portion of patients with lung adenocarcinoma (LUAD) still suffer from rapid tumor progression in clinical practice, indicating the occurrence of potential bypass. METHODS: Real-time polymerase chain reaction was applied to examine the expressions of mir-144-3p, WT1, NF-κB, COX2 and HIF-1α at the mRNA level in 127 LUAD samples and corresponding adjacent tissues. miR-144-3p mimic and antagormiR were used to trigger activation and suppression of miR-144-3p in A549 cells, respectively. MTT assay and Western blotting analysis were carried out to evaluate the cell proliferation. Stable clones with over-expression or knockdown of WT1 were generated with plasmid or shRNA by lentiviral vector technology in H1568 and H1650 NSCLC cell lines, respectively. Dual luciferase reporter assay was performed to validate the effect of miR-144-3p on WT1D. Xenograft model was established for in vivo experiment, and TCGA data were extracted for validation. RESULTS: miR-144-3p could suppress the WT1D expression at the post-transcriptional level, hence regulating cell proliferation in LUAD. WT1 and COX-2 were independent prognostic factors of LUAD patients. In addition, inhibition of IL-1ß/miR-144-3p/WT1D and IL-1ß/NF-κB/COX-2/HIF-1α pathways using miR-144-3p mimic and Celecoxib, respectively, displayed synergistic suppressive effect on cell proliferation in LUAD. CONCLUSION: A de novo IL-1ß/miR-144-3p/WT1D axis was involved in proliferative regulation of LUAD. Moreover, simultaneous blockade of both IL-1ß/miR-144-3p/WT1D and IL-1ß/NF-κB/COX-2/ HIF-1α pathways might have synergistic suppressive effect on cell proliferation in LUAD.


Subject(s)
Adenocarcinoma/pathology , Interleukin-1beta/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , Signal Transduction , WT1 Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Aged , Aged, 80 and over , Animals , Antagomirs/metabolism , Celecoxib/pharmacology , Celecoxib/therapeutic use , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
8.
J Am Soc Nephrol ; 29(10): 2529-2545, 2018 10.
Article in English | MEDLINE | ID: mdl-30143559

ABSTRACT

BACKGROUND: Podocyte injury is the hallmark of proteinuric kidney diseases, such as FSGS and minimal change disease, and destabilization of the podocyte's actin cytoskeleton contributes to podocyte dysfunction in many of these conditions. Although agents, such as glucocorticoids and cyclosporin, stabilize the actin cytoskeleton, systemic toxicity hinders chronic use. We previously showed that loss of the kidney-enriched zinc finger transcription factor Krüppel-like factor 15 (KLF15) increases susceptibility to proteinuric kidney disease and attenuates the salutary effects of retinoic acid and glucocorticoids in the podocyte. METHODS: We induced podocyte-specific KLF15 in two proteinuric murine models, HIV-1 transgenic (Tg26) mice and adriamycin (ADR)-induced nephropathy, and used RNA sequencing of isolated glomeruli and subsequent enrichment analysis to investigate pathways mediated by podocyte-specific KLF15 in Tg26 mice. We also explored in cultured human podocytes the potential mediating role of Wilms Tumor 1 (WT1), a transcription factor critical for podocyte differentiation. RESULTS: In Tg26 mice, inducing podocyte-specific KLF15 attenuated podocyte injury, glomerulosclerosis, tubulointerstitial fibrosis, and inflammation, while improving renal function and overall survival; it also attenuated podocyte injury in ADR-treated mice. Enrichment analysis of RNA sequencing from the Tg26 mouse model shows that KLF15 induction activates pathways involved in stabilization of actin cytoskeleton, focal adhesion, and podocyte differentiation. Transcription factor enrichment analysis, with further experimental validation, suggests that KLF15 activity is in part mediated by WT1. CONCLUSIONS: Inducing podocyte-specific KLF15 attenuates kidney injury by directly and indirectly upregulating genes critical for podocyte differentiation, suggesting that KLF15 induction might be a potential strategy for treating proteinuric kidney disease.


Subject(s)
DNA-Binding Proteins/biosynthesis , Kidney Diseases/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Transcription Factors/biosynthesis , Actin Cytoskeleton/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Models, Animal , Focal Adhesions , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Diseases/genetics , Kidney Diseases/pathology , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Nephrosis, Lipoid/genetics , Nephrosis, Lipoid/metabolism , Nephrosis, Lipoid/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Transcription Factors/genetics , Up-Regulation , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , WT1 Proteins/metabolism
9.
Diabetes ; 65(5): 1164-78, 2016 05.
Article in English | MEDLINE | ID: mdl-26936961

ABSTRACT

Increased visceral fat, rather than subcutaneous fat, during the onset of obesity is associated with a higher risk of developing metabolic diseases. The inherent adipogenic properties of human adipose-derived stem cells (ASCs) from visceral depots are compromised compared with those of ASCs from subcutaneous depots, but little is known about the underlying mechanisms. Using ontological analysis of global gene expression studies, we demonstrate that many genes involved in retinoic acid (RA) synthesis or regulated by RA are differentially expressed in human tissues and ASCs from subcutaneous and visceral fat. The endogenous level of RA is higher in visceral ASCs; this is associated with upregulation of the RA synthesis gene through the visceral-specific developmental factor WT1. Excessive RA-mediated activity impedes the adipogenic capability of ASCs at early but not late stages of adipogenesis, which can be reversed by antagonism of RA receptors or knockdown of WT1. Our results reveal the developmental origin of adipocytic properties and the pathophysiological contributions of visceral fat depots.


Subject(s)
Adipogenesis , Down-Regulation , Gene Expression Regulation, Developmental , Intra-Abdominal Fat/metabolism , Receptors, Retinoic Acid/agonists , Signal Transduction , Tretinoin/metabolism , Active Transport, Cell Nucleus/drug effects , Adipogenesis/drug effects , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Bariatric Surgery , Benzoates/pharmacology , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Gene Ontology , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Middle Aged , Naphthalenes/pharmacology , Obesity, Morbid/metabolism , Obesity, Morbid/pathology , Obesity, Morbid/surgery , RNA Interference , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Response Elements/drug effects , Signal Transduction/drug effects , Stilbenes/pharmacology , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Up-Regulation/drug effects , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , WT1 Proteins/metabolism
10.
Tumour Biol ; 37(7): 9967-77, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26815508

ABSTRACT

The WT1 gene is an important oncogene, and its overexpression is considered as an effective target for anticancer therapy. Regulation of its gene transcription is one way for WT1-targeting drug design. Recently, in silico analysis of some oncogene promoters like WT1 showed some guanine-rich regions with the ability to form G-quadruplex structures. Ligands like 5,10,15,20-tetra(N-methyl-4-pyridyl)-porphine (TMPyP4) have predominant effect on G-quadruplex stabilization. The aim of this study was to understand the effect of TMPyP4 on WT1 gene transcription via stabilization of promoter G-quadruplexes. We examined the formation of new G-quadruplex motifs in WT1 promoter in the presence of TMPyP4. In order to understand the nature of its interaction with WT1 promoter quadruplexes, differential pulse voltammetry (DPV), circular dichroism (CD), polyacrylamide gel electrophoresis, electrophoretic mobility shift assay (EMSA), polymerase chain reaction (PCR) stop assays, and quantitative RT-PCR were performed. According to the results, the WT1 promoter can form stable intramolecular parallel G-quadruplexes. In addition, after 48 and 96 h of incubation, 100 µM TMPyP4 reduced the WT1 transcription to 9 and 0.4 %, respectively, compare to control. We report that ligand-mediated stabilization of G-quadruplexes within the WT1 promoter can silence WT1 expression. This study might offer the basis for the reasonable design and improvement of new porphyrin derivatives as effective anti-leukemia agents for cancer therapy.


Subject(s)
DNA, Neoplasm/chemistry , G-Quadruplexes , Gene Expression Regulation, Neoplastic , Leukemia, Erythroblastic, Acute/genetics , Porphyrins/metabolism , WT1 Proteins/antagonists & inhibitors , Cell Proliferation , Circular Dichroism , Electrophoretic Mobility Shift Assay , Humans , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Ligands , Models, Molecular , Porphyrins/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , WT1 Proteins/genetics , WT1 Proteins/metabolism
11.
PLoS One ; 10(6): e0130578, 2015.
Article in English | MEDLINE | ID: mdl-26090994

ABSTRACT

The Wilms' tumor gene WT1 consists of 10 exons and encodes a zinc finger transcription factor. There are four major WT1 isoforms resulting from alternative splicing at two sites, exon 5 (17AA) and exon 9 (KTS). All major WT1 isoforms are overexpressed in leukemia and solid tumors and play oncogenic roles such as inhibition of apoptosis, and promotion of cell proliferation, migration and invasion. In the present study, a novel alternatively spliced WT1 isoform that had an extended exon 4 (designated as exon 4a) with an additional 153 bp (designated as 4a sequence) at the 3' end was identified and designated as an Ex4a(+)WT1 isoform. The insertion of exon 4a resulted in the introduction of premature translational stop codons in the reading frame in exon 4a and production of C-terminal truncated WT1 proteins lacking zinc finger DNA-binding domain. Overexpression of the truncated Ex4a(+)WT1 isoform inhibited the major WT1-mediated transcriptional activation of anti-apoptotic Bcl-xL gene promoter and induced mitochondrial damage and apoptosis. Conversely, suppression of the Ex4a(+)WT1 isoform by Ex4a-specific siRNA attenuated apoptosis. These results indicated that the Ex4a(+)WT1 isoform exerted dominant negative effects on anti-apoptotic function of major WT1 isoforms. Ex4a(+)WT1 isoform was endogenously expressed as a minor isoform in myeloid leukemia and solid tumor cells and increased regardless of decrease in major WT1 isoforms during apoptosis, suggesting the dominant negative effects on anti-apoptotic function of major WT1 isoforms. These results indicated that Ex4a(+)WT1 isoform had an important physiological function that regulated oncogenic function of major WT1 isoforms.


Subject(s)
WT1 Proteins/chemistry , WT1 Proteins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Base Sequence , Cloning, Molecular , Doxorubicin/toxicity , Exons , HL-60 Cells , Haplorhini , Humans , K562 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
12.
PLoS One ; 9(12): e114333, 2014.
Article in English | MEDLINE | ID: mdl-25474318

ABSTRACT

Wilms' tumor gene 1 (WT1) plays complex roles in tumorigenesis, acting as tumor suppressor gene or an oncogene depending on the cellular context. WT1 expression has been variably reported in both benign and malignant peripheral nerve sheath tumors (MPNSTs) by means of immunohistochemistry. The aim of the present study was to characterize its potential pathogenetic role in these relatively uncommon malignant tumors. Firstly, immunohistochemical analyses in MPNST sNF96.2 cell line showed strong WT1 staining in nuclear and perinuclear areas of neoplastic cells. Thus, we investigated the effects of silencing WT1 by RNA interference. Through Western Blot analysis and proliferation assay we found that WT1 knockdown leads to the reduction of cell growth in a time- and dose-dependent manner. siWT1 inhibited proliferation of sNF96.2 cell lines likely by influencing cell cycle progression through a decrease in the protein levels of cyclin D1 and inhibition of Akt phosphorylation compared to the control cells. These results indicate that WT1 knockdown attenuates the biological behavior of MPNST cells by decreasing Akt activity, demonstrating that WT1 is involved in the development and progression of MPNSTs. Thus, WT1 is suggested to serve as a potential therapeutic target for MPNSTs.


Subject(s)
Cell Proliferation/genetics , Neurilemmoma/genetics , WT1 Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cyclin D1/biosynthesis , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neurilemmoma/pathology , RNA Interference , WT1 Proteins/antagonists & inhibitors
13.
Nat Commun ; 5: 5852, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25510679

ABSTRACT

Angiogenesis, activation of metastasis and avoidance of immune destruction are important for cancer progression. These biological capabilities are, apart from cancer cells, mediated by different cell types, including endothelial, haematopoietic progenitor and myeloid-derived suppressor cells. We show here that all these cell types frequently express the Wilms' tumour suppressor Wt1, which transcriptionally controls expression of Pecam-1 (CD31) and c-kit (CD117). Inducible conditional knockout of Wt1 in endothelial, haematopoietic and myeloid-derived suppressor cells is sufficient to cause regression of tumour vascularization and an enhanced immune response, leading to decreased metastasis, regression of established tumours and enhanced survival. Thus, Wt1 is an important regulator of cancer growth via modulation of tumour vascularization, immune response and metastasis formation.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma, Experimental/genetics , Neovascularization, Pathologic/genetics , Skin Neoplasms/genetics , WT1 Proteins/genetics , Animals , Disease Progression , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Gene Knockout Techniques , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Immunity, Innate , Melanoma, Experimental/blood supply , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Skin Neoplasms/blood supply , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Survival Analysis , Tumor Burden , Tumor Cells, Cultured , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/immunology
14.
Anticancer Res ; 34(3): 1333-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24596380

ABSTRACT

BACKGROUND: Wilms' tumor 1 (WT1) gene has different functional properties depending on the isoform type. This gene correlates with cell proliferation in various types of cancer. Here, we investigated the expression of WT1 isoforms in breast cancer tissues, and focused on the oncogenic role through estrogen receptor-alpha (ER-α) and human epidermal growth factor receptor 2 (HER2). MATERIALS AND METHODS: Expression of WT1(17AA+) and (17AA-) was investigated in adjacent normal breast and breast cancer using Reverse transcription-polymerase chain reaction and western blotting. The correlation of WT1 isoforms with HER2 and ER-α was examined using MCF-7 cells stably-overexpressing WT1s and siRNA against WT1 gene. RESULTS: The expression of WT(17AA-) was significantly found in adjacent normal breast tissues. A mixture of WT1(17AA+) and WT1(17AA-) were highly expressed in breast carcinoma tissues. MCF-7 cells overexpressing WT1+/+ and WT1+/- represented strong expression of ER-α and HER2. Moreover, the silencing of WT1+/+ and WT1+/- resulted in a decrease of both ER-α and HER2 and led to a decrease of cell numbers. CONCLUSION: Our results suggest that WT1(17AA+) was exhibited dominantly in breast carcinoma tissues. WT1+/+ and WT1+/- correlated with the high expression of ER-α and HER2, leading to cell proliferation and might be involved in cancer development and progression.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Estrogen Receptor alpha/metabolism , Receptor, ErbB-2/metabolism , WT1 Proteins/metabolism , Adult , Aged , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Proliferation , Estrogen Receptor alpha/genetics , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Grading , Prognosis , Protein Isoforms , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
15.
Anticancer Res ; 34(1): 61-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24403445

ABSTRACT

The prognosis for patients with glioblastoma is very poor, despite intensive treatment, including surgery and chemoradiotherapy. Wilms' tumor 1 (WT1) is expressed in most glioblastoma samples, and immunotherapy targeting WT1 has proven to be effective in recurrent glioblastoma. However, the functional roles of WT1 in glioblastoma are not clear. To examine the functional roles of WT1 in glioblastoma, glioblastoma cell lines with reduced WT1 expression were generated using short hairpin RNA(shRNA)-expressing lentivirus. Proliferation of WT1-knockdown glioblastoma cells was significantly slower than control cells with high WT1 expression. In addition, apoptosis was increased in WT1-knockdown glioblastoma cells. Furthermore, WT1-knockdown glioblastoma cells, and control glioblastoma cells were intra-cranially injected into immunodeficient mice. In vivo tumor growth of WT1-knockdown glioblastoma cells was significantly reduced compared to control glioblastoma cells. These results show that WT1 is involved in glioblastoma cell proliferation and apoptosis and that this protein has oncogenic roles in glioblastoma.


Subject(s)
Apoptosis , Cell Proliferation , Glioblastoma/pathology , WT1 Proteins/metabolism , Animals , Animals, Newborn , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , DNA-Binding Proteins/physiology , Gene Expression Profiling , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Immunoenzyme Techniques , Mice , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics , Survival Rate , Tumor Cells, Cultured , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , Xenograft Model Antitumor Assays
16.
FASEB J ; 28(5): 2020-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24451388

ABSTRACT

Steroidogenic factor 1 (SF1; Ad4BP/NR5A1) plays key roles in gonadal development. Initially, the Sf1 gene is expressed in mouse fetal gonads of both sexes, but later is up-regulated in testes and down-regulated in ovaries. While Sf1 expression is activated and maintained by Wilms tumor 1 (WT1) and LIM homeobox 9 (LHX9), the mechanism of sex-specific regulation remains unclear. We hypothesized that Sf1 is repressed by the transcription factor Forkhead box L2 (FOXL2) during ovarian development. In an in vitro system (TM3 cells), up-regulation of Sf1 by the WT1 splice variant WT1-KTS was antagonized by FOXL2, as determined by quantitative RT-PCR. Using reporter assays, we localized the Sf1 proximal promoter region involved in this antagonism to a 674-bp interval. A conserved FOXL2 binding site was identified in this interval by in vitro chromatin immunoprecipitation. Introducing mutations into this site abolished negative regulation by FOXL2 in reporter assays. Finally, in Foxl2-null mice, Sf1 expression was increased 2-fold relative to wild-type XX fetal gonads. Our results support the hypothesis that FOXL2 negatively regulates Sf1 expression by antagonizing WT1-KTS during early ovarian development in mice.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Ovary/embryology , WT1 Proteins/metabolism , Animals , Binding Sites , Cell Differentiation , Female , Forkhead Box Protein L2 , Genes, Reporter , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred ICR , Mutagenesis , Mutation , Ovary/metabolism , Plasmids/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptional Activation , WT1 Proteins/antagonists & inhibitors
17.
Int J Cancer ; 134(2): 291-300, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23832418

ABSTRACT

The role of Wilms' tumor suppressor 1 (WT1) in leukemogenesis has been investigated mostly in acute (AML) and chronic (CML) myeloid leukemias. So far, its oncogenic role has been controversially discussed because both overexpression and inactivating mutations are found. A recent study on primary samples from patients with acute T-cell leukemia (T-ALL) revealed that most of them do not express WT1 proteins although they express WT1 mRNA. In our study, we investigated WT-1 expression in ten T-ALL cell lines established from leukemia/lymphoma patients. We show that consistent with the finding in primary T-ALL cells, most of the leukemic T-cell lines tested do not overexpress WT1 proteins. We found that leukemic T-cells overexpressing WT1 protein produce higher levels of CD95L and show elevated CD95L-mediated activation-induced cell death (AICD) compared to cells lacking or expressing low levels of WT1. Ectopic expression of WT1 in the WT1-nonexpressing leukemic T-cell line increases CD95L expression and elevates activation-induced apoptosis, whereas silencing WT1 expression in the WT1-overexpressing leukemic T-cell line by siRNA confers reduced CD95L expression and reduction in AICD. Chromatin immunoprecipitation and luciferase-promoter reporter analysis demonstrate that WT1 binds to and enhances CD95L promoter activity through the Egr-binding sites. Our study provides a new role of WT1 in regulation of CD95L-mediated cell death.


Subject(s)
Apoptosis/drug effects , Fas Ligand Protein/metabolism , Leukemia, T-Cell/pathology , Transcription Factors/metabolism , WT1 Proteins/metabolism , Blotting, Western , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Fas Ligand Protein/genetics , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Lymphocyte Activation , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
18.
J Thorac Oncol ; 8(9): 1163-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23945386

ABSTRACT

INTRODUCTION: The Wilms' tumor gene (WT1) has been identified as an oncogene in many malignant diseases, and aberrant WT1 expression has been linked to development, progression, and prognosis of non-small-cell lung cancer (NSCLC). We sought to investigate the underlying mechanism of WT1 and metastasis in NSCLC. METHODS: Real-time polymerase chain reaction was applied to detect WT1 and CDH1 mRNA in 159 NSCLC samples and corresponding adjacent tissues. Stable clones with overexpression and knockdown of WT1 were generated with plasmid and shRNA via lentivirus technology in H1568 and H1650 NSCLC cell lines. Wound-healing assay, transwell assays, and polymerase chain reaction array were carried out for invasion evaluation. Dual luciferase reporter assay was performed to validate the effect of WT1 on CDH1. RESULTS: The level of the WT1 mRNA was negatively correlated with that of E-cadherin (CDH1) and associated with pathological stage, metastasis, and survival rate of 159 NSCLC patients. A series of genes were regulated by WT1, and WT1 could suppress CDH1 transcription via direct binding to its promoter and may enhance the invasive ability of H1568 and H1650 NSCLC cell lines. CONCLUSIONS: WT1 expression was correlated with clinical stage, metastasis, and survival rate in 159 NSCLC patients. Via direct binding to the promoter, WT1 could suppress CDH1 and promote NSCLC invasion.


Subject(s)
Adenocarcinoma/pathology , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , WT1 Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Aged , Aged, 80 and over , Antigens, CD , Apoptosis , Blotting, Western , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , Wound Healing
19.
Exp Cell Res ; 319(5): 612-22, 2013 Mar 10.
Article in English | MEDLINE | ID: mdl-23291318

ABSTRACT

Wilms tumor gene WT1 encodes a zinc finger-containing transcription factor which is required for renal development. Mutations in WT1 are observed in 20% of Wilms tumors (a pediatric kidney cancer), but the in vivo WT1 targets and associated molecular pathways involved in the etiology of Wilms tumor are still elusive. To identify WT1 targets we performed genome-wide comprehensive expression profiling using Affymetrix Gene Chip Mouse Genome 430 2.0 Arrays, comparing E13.5 mouse kidneys in which Wt1 had been somatically ablated with littermate controls. We identified Usp18 as the most differentially expressed gene in mutant kidney. Using tetracycline inducible cells we demonstrated a repressive effect of WT1 on USP18 expression. Conversely, knockdown of WT1 led to the upregulation of Usp18. Furthermore, direct binding of WT1 to the Usp18 promoter was demonstrated by ChIP assay. Overexpression of USP18 in murine and human cell lines resulted in cell proliferation. Additionally, Usp18 upregulation was observed in a mouse model of Wilms tumor. Taken together our data demonstrate that Usp18 is a transcriptional target of WT1 and suggest that increased expression of USP18 following WT1 loss contributes to Wilms tumorigenesis.


Subject(s)
Endopeptidases/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , WT1 Proteins/physiology , Wilms Tumor/genetics , Animals , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Endopeptidases/metabolism , Female , Gene Expression Profiling , Humans , In Situ Hybridization , Kidney/embryology , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/pathology , Luciferases/metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcriptional Activation , Transfection , Ubiquitin Thiolesterase , WT1 Proteins/antagonists & inhibitors , Wilms Tumor/pathology
20.
World J Surg Oncol ; 10: 11, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22244202

ABSTRACT

AIM: Recent studies have reported that double-stranded RNA (dsRNA) can activate gene expression by targeting promoter sequence in a process termed RNA activation. The present study was conducted to evaluate the potential of WT1 induction by small activating RNA targeting the WT1 promoter (dsWT1) in the treatment of hepatocellular carcinoma. METHODS: The human hepatocellular carcinoma cell line HepG2 was transfected with dsRNA by liposomes. The expression of mRNA and protein in cells were investigated using real-time reverse real-time quantitative PCR and Western blot, respectively. Cell viability and clonogenicity were determined by MTT assay and clonogenicity assay, respectively. Cell apoptosis was evaluated by flow-cytometric analysis. RESULTS: Expressions of WT1 mRNA and protein in dsWT1 treated HepG2 cells were significantly elevated. Inhibition of cell viability by dsWT1 was dose-dependent and time-dependent. Reduction of the number and size of colonies formed were found in dsWT1 treated cells. dsWT1 induced significant apoptosis in HepG2 cells. The decreased anti-apoptotic protein Bcl-2 and elevated pro-apoptotic protein Bak expression were detected in dsWT1 treated cells. The level of pro-caspase-3 remarkably decreased and cleaved caspase-3 and PARP fragment were also detected in dsWT1 treated cells. CONCLUSION: These data show that RNAa-mediated overexpression of WT1 may have therapeutic potential in the treatment of hepatocellular carcinoma.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , RNA, Double-Stranded/genetics , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , Blotting, Western , Carcinoma, Hepatocellular/genetics , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tumor Stem Cell Assay , WT1 Proteins/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...