51
|
Abstract
Eukaryotic cells possess an exquisitely interwoven and fine-tuned series of signal transduction mechanisms with which to sense and respond to the ubiquitous fermentable carbon source glucose. The budding yeast Saccharomyces cerevisiae has proven to be a fertile model system with which to identify glucose signaling factors, determine the relevant functional and physical interrelationships, and characterize the corresponding metabolic, transcriptomic, and proteomic readouts. The early events in glucose signaling appear to require both extracellular sensing by transmembrane proteins and intracellular sensing by G proteins. Intermediate steps involve cAMP-dependent stimulation of protein kinase A (PKA) as well as one or more redundant PKA-independent pathways. The final steps are mediated by a relatively small collection of transcriptional regulators that collaborate closely to maximize the cellular rates of energy generation and growth. Understanding the nuclear events in this process may necessitate the further elaboration of a new model for eukaryotic gene regulation, called "reverse recruitment." An essential feature of this idea is that fine-structure mapping of nuclear architecture will be required to understand the reception of regulatory signals that emanate from the plasma membrane and cytoplasm. Completion of this task should result in a much improved understanding of eukaryotic growth, differentiation, and carcinogenesis.
Collapse
Affiliation(s)
- George M Santangelo
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406-5018, USA.
| |
Collapse
|
52
|
Yao R, Wang Y, Lu Y, Lemon WJ, End DW, Grubbs CJ, Lubet RA, You M. Efficacy of the farnesyltransferase inhibitor R115777 in a rat mammary tumor model: role of Ha-ras mutations and use of microarray analysis in identifying potential targets. Carcinogenesis 2006; 27:1420-31. [PMID: 16403772 DOI: 10.1093/carcin/bgi341] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rats treated with the alkylating agent methylnitrosourea (MNU) develop multiple, hormonally dependent mammary tumors. Roughly 50% of the tumors have Ha-ras mutation, whereas 50% do not. The MNU-induced rat mammary tumor model was employed to examine the therapeutic efficacy of the farnesyltransferase inhibitor (FTI), R115777, and to examine the use of genomics in identifying susceptible tumors as well as identifying genes whose expression are modulated by FTI treatment. In animals bearing palpable mammary tumors (< 7 mm diameter), we performed a surgical biopsy, and 3 days following the biopsy, rats were treated with R115777 (50 mg/kg body wt/day) by gavage. Tumors with Ha-ras mutations underwent profound regression, with nearly 90% showing complete regressions within 4 weeks. In contrast, the non-Ha-ras mutation-bearing tumors yielded a more variable response, although roughly half of the non-Ha-ras mutation tumors underwent significant regression. These results show that although all tumors appear to respond to the FTI inhibitor the tumors with Ha-ras mutations were exquisitely sensitive. We employed a microarray approach to define potential targets and the mechanism of action of R115777 in Ha-ras mutant or wildtype tumors following treatment with FTI. In addition, we determined whether gene expression prior to FTI treatment can be used to differentiate highly sensitive tumors (Ha-ras mutant) and tumors with variable sensitivity (Ha-ras wildtype). Untreated or FTI-treated (4 days at 50 mg/kg body wt) tumors (Ha-ras mutant or wildtype) were examined using oligonucleotide arrays. A significant number of genes were differentially expressed in control rat mammary tumors with or without an activated Ha-ras mutation, suggesting that a microarray analysis might differentiate highly sensitive and variably sensitive tumors. Most of the genes whose expressions were modulated by FTI in tumors were independent of Ha-ras status and were presumably modulated by effects on farnesylation of proteins other than Ha-ras. However, treatment of Ha-ras-mutated mammary tumors with R155777 results in preferential modulation of genes involved in ras-MAP kinase signal transduction pathway and in decreased expression of many genes involved with cell proliferation. In contrast, several classes of genes are altered in rat mammary tumors without a mutated Ha-ras, suggesting that non-ras targets are involved. Ras pathway related genes, p53, WT1 and PCNA, were preferentially modulated in Ha-ras-mutated tumors, whereas modulation of genes in the G-protein pathway, various cytochrome p450s and RB1 are involved in Ha-ras wildtype tumors. Elucidation of gene expression changes in FTI-treated or control rat mammary adenocarcinomas will help in identifying potential pharmacodynamic markers of FTI treatment as well as potential molecular targets of R115777 and other FTIs.
Collapse
Affiliation(s)
- Ruisheng Yao
- Department of Surgery and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Reese BE, Krissinger D, Yun JK, Billingsley ML. Elucidation of stannin function using microarray analysis: implications for cell cycle control. Gene Expr 2006; 13:41-52. [PMID: 16572589 PMCID: PMC6032450 DOI: 10.3727/000000006783991944] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Stannin (Snn) is a highly conserved, vertebrate protein whose cellular function is unclear. We have recently demonstrated in human umbilical vein endothelial cells (HUVECs) that Snn gene expression is significantly induced by tumor necrosis factor-alpha (TNF-alpha) in a protein kinase C-epsilon (PKC-epsilon)-dependent manner. In HUVEC, TNF-alpha stimulation of HUVECs results in altered gene expression, and a slowing or halting of cell growth. An initial set of experiments established that Snn knockdown via siRNA, prior to TNF-alpha treatment, resulted in a significant inhibition of HUVEC growth compared to TNF-alpha treatment alone. In order to assess how Snn may be involved in TNF-alpha signaling in HUVEC growth arrest, we performed microarray analysis of TNF-alpha-stimulated HUVECs with and without Snn knockdown via siRNA. The primary comparison made was between TNF-alpha-stimulated HUVECs and TNF-alpha-exposed HUVECs that had Snn knocked down via Snn-specific siRNAs. Ninety-six genes were differentially expressed between these two conditions. Of particular interest was the significant upregulation of several genes associated with control of cell growth and/or the cell cycle, including interleukin-4, p29, WT1/PRKC, HRas-like suppressor, and MDM4. These genes act upon cyclin D1 and/or p53, both of which are key regulators of the G1 phase of the cell cycle. Functional studies further supported the role of Snn in cell growth, as cell cycle analysis using flow cytometry shows a significant increase of G1 cell cycle arrest in HUVECs with Snn knockdown in response to TNF-alpha treatment. Together these studies suggest a functional role of Snn in regulation of TNF-alpha-induced signaling associated with HUVEC growth arrest.
Collapse
Affiliation(s)
- Brian E Reese
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
54
|
Cervoni L, Egistelli L, Eufemi M, Scotto d'Abusco A, Altieri F, Lascu I, Turano C, Giartosio A. DNA sequences acting as binding sites for NM23/NDPK proteins in melanoma M14 cells. J Cell Biochem 2006; 98:421-8. [PMID: 16440314 DOI: 10.1002/jcb.20808] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We isolated and analyzed by chromatin immunoprecipitation (ChIP) in viable M14 cells DNA sequences bound to the antimetastatic protein nucleoside diphosphate kinase (NM23/NDPK) to shed some light on the nuclear functions of this protein and on the mechanism by which it acts in development and cancer. We assessed the presence of selected sequences from promoters of platelet-derived growth factor A (PDGF-A), c-myc, myeloperoxidase (MPO), CD11b, p53, WT1, CCR5, ING1, and NM23-H1 genes in the cross-linked complexes. Quantitative PCR (Q-PCR) showed a substantial enrichment of the correlated oncosuppressor genes p53, WT1, ING1, and NM23-H1 in the immunoprecipitated (IP) DNA. This suggests that NM23/NDPK binding is involved in the transcription regulation of these genes. These results reveal new interactions that should help us to disclose the antimetastatic mechanism of NM23.
Collapse
Affiliation(s)
- Laura Cervoni
- Dipartimento di Scienze Biochimiche A. Rossi Fanelli, Università di Roma La Sapienza, Italy
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Abstract
Wilms' tumour, or nephroblastoma, is a common childhood tumour that is intimately linked to early kidney development and is often associated with persistent embryonic renal tissue and other kidney abnormalities. WT1, the first gene found to be inactivated in Wilms' tumour, encodes a transcription factor that functions as both a tumour suppressor and a critical regulator of renal organogenesis. Our understanding of the roles of WT1 in tumour formation and organogenesis have advanced in parallel, providing a striking example of the intersection between tumour biology, cellular differentiation and normal organogenesis.
Collapse
Affiliation(s)
- Miguel N Rivera
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
56
|
Granja NM, Begnami MD, Bortolan J, Filho AL, Schmitt FC. Desmoplastic small round cell tumour: Cytological and immunocytochemical features. Cytojournal 2005; 2:6. [PMID: 15777480 PMCID: PMC555739 DOI: 10.1186/1742-6413-2-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Accepted: 03/18/2005] [Indexed: 11/25/2022] Open
Abstract
Background Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive neoplasm. The cytological diagnosis of these tumors can be difficult because they show morphological features quite similar to other small round blue cells tumors. We described four cases of DSRCT with cytological sampling: one obtained by fine needle aspiration biopsy (FNAB) and three from serous effusions. The corresponding immunocytochemical panel was also reviewed. Methods Papanicolaou stained samples from FNAB and effusions were morphologically described. Immunoreaction with WT1 antibody was performed in all cytological samples. An immunohistochemical panel including the following antibodies was performed in the corresponding biopsies: 34BE12, AE1/AE3, Chromogranin A, CK20, CK7, CK8, Desmin, EMA, NSE, Vimentin and WT1. Results The smears showed high cellularity with minor size alteration. Nuclei were round to oval, some of them with inconspicuous nucleoli. Tumor cells are clustered, showing rosette-like feature. Tumor cells in effusions and FNA were positive to WT1 in 3 of 4 cytology specimens (2 out 3 effusions and one FNA). Immunohistochemical reactions for vimentin, NSE, AE1/AE3 and WT1 were positive in all cases in tissue sections. Conclusion The use of an adjunct immunocytochemical panel coupled with the cytomorphological characteristics allows the diagnosis of DSRCT in cytological specimens.
Collapse
Affiliation(s)
- Nara M Granja
- Pathology Department, School of Medicine, São Paulo University, São Paulo, Brazil
| | - Maria D Begnami
- Department of Pathology and Treatment and Research Center of A.C. Camargo Hospital, São Paulo, Brazil
| | - Jeni Bortolan
- Department of Pathology and Treatment and Research Center of A.C. Camargo Hospital, São Paulo, Brazil
| | - Adhemar Longatto Filho
- Life and Health Sciences Research Institute, Health Sciences School, University of Minho, Braga, Portugal
- Pathology Division, Adolfo Lutz Institute, São Paulo, Brazil
| | - Fernando C Schmitt
- Medical Faculty, Department of Pathology, University of Porto, Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of University of Porto, Porto, Portugal
| |
Collapse
|
57
|
Zhan Q. Gadd45a, a p53- and BRCA1-regulated stress protein, in cellular response to DNA damage. Mutat Res 2005; 569:133-43. [PMID: 15603758 DOI: 10.1016/j.mrfmmm.2004.06.055] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 06/01/2004] [Accepted: 06/10/2004] [Indexed: 01/08/2023]
Abstract
Mammalian cells exhibit complex, but intricate cellular responses to genotoxic stress, including cell cycle checkpoints, DNA repair and apoptosis. Inactivation of these important biological events may result in genomic instability and cell transformation, as well as alterations of therapeutic sensitivity. Gadd45a, a p53- and BRCA1-regulated stress-inducible gene, has been characterized as one of the important players that participate in cellular response to a variety of DNA damage agents. Interestingly, the signaling machinery that regulates Gadd45a induction by genotoxic stress involves both p53-dependent and -independent pathways; the later may employ BRCA1-related or MAP kinase-mediated signals. Gadd45a protein has been reported to interact with multiple important cellular proteins, including Cdc2 protein kinase, proliferating cell nuclear antigen (PCNA), p21Waf1/Cip1 protein, core histone protein and MTK/MEKK4, an up-stream activator of the JNK/SAPK pathway, indicating that Gadd45a may play important roles in the control of cell cycle checkpoint, DNA repair process, and signaling transduction. The importance of Gadd45a in maintaining genomic integrity is well manifested by the demonstration that disruption of endogenous Gadd45a in mice results in genomic instability and increased carcinogenesis. Therefore, Gadd45a appears to be an important component in the cellular defense network that is required for maintenance of genomic stability.
Collapse
Affiliation(s)
- Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing 100021, China.
| |
Collapse
|
58
|
Rolaki A, Drakakis P, Millingos S, Loutradis D, Makrigiannakis A. Novel trends in follicular development, atresia and corpus luteum regression: a role for apoptosis. Reprod Biomed Online 2005; 11:93-103. [PMID: 16102296 DOI: 10.1016/s1472-6483(10)61304-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During ovarian follicular development in humans, only a limited number of follicles mature and ovulate. The vast majority of follicles stop developing after the formation of an antrum and then undergo atresia. The few that are selected to become ovulatory follicles are transformed into corpora lutea following ovulation. The lifespan of the corpus luteum is also limited. In each oestrus/menstrual cycle, corpora lutea regress and are eliminated by a progress called luteolysis. During atresia and luteolysis, granulosa and lutein cells undergo apoptosis. It is believed that there are many signal transduction pathways that control apoptosis in order to suppress full maturation of too many follicles and to protect the dominant follicle from the apoptotic process prior the ovulation. Such interplay between different factors, some of them produced in the ovary, may modulate apoptosis of corpus luteum cells, in order to preserve the function of the corpus luteum during pregnancy or to eliminate the old corpora lutea of the previous cycle. The present review reports a number of factors that regulate follicular atresia and corpus luteum regression, via apoptotic pathways. Elucidation of apoptotic mechanisms may lead to prevention of female infertility or other pathological conditions.
Collapse
Affiliation(s)
- Alexandra Rolaki
- Department of Obstetrics and Gynaecology, Medical School, University of Crete, Heraklion 71110, Greece
| | | | | | | | | |
Collapse
|
59
|
Stanhope-Baker P, Kessler PM, Li W, Agarwal ML, Williams BRG. The Wilms Tumor Suppressor-1 Target Gene Podocalyxin Is Transcriptionally Repressed by p53. J Biol Chem 2004; 279:33575-85. [PMID: 15155752 DOI: 10.1074/jbc.m404787200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Wilms tumors are a heterogeneous class of tumors in which Wilms tumor suppressor-1 (WT1) and the p53 tumor suppressor may be variously inactivated by mutation, reduced in expression, or even overexpressed in the wild-type state. The downstream transcriptional targets of WT1 and p53 that are critical for mediating their roles in Wilms tumorigenesis are not well defined. The WiT49 cell line is characteristic of anaplastic Wilms tumors that are refractory to treatment and expresses wild-type WT1 and mutant p53. We have used the small molecule compound CP-31398 (Pfizer) to restore wild-type p53 function to the codon 248 mutant p53 present in WiT49 cells. In these cells, CP-31398 activated transcription of p53-regulated promoters and enhanced UV light-induced apoptosis without altering the overall p53 protein level. These phenotypes were accompanied by restored binding of the p53 protein to promoter sequences in vivo. Gene expression profiling of CP-31398-treated WiT49 cells revealed subsets of putative p53 target genes that were up- or down-regulated. A preferred target of p53-mediated repression in this system is the podocalyxin (PODXL) gene. PODXL is also transcriptionally regulated by WT1 and has roles in cell adhesion and anti-adhesion. Our results show that PODXL is a bona fide target of p53-mediated transcriptional repression while being positively regulated by WT1. We propose that inappropriate expression of PODXL due to changes in WT1 and/or p53 activity may contribute to Wilms tumorigenesis.
Collapse
Affiliation(s)
- Patricia Stanhope-Baker
- Departments of Cancer Biology and Molecular Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
60
|
Dupont J, Wang X, Marshall DS, Leitao M, Hedvat CV, Hummer A, Thaler H, O'Reilly RJ, Soslow RA. Wilms Tumor Gene (WT1) and p53 expression in endometrial carcinomas: a study of 130 cases using a tissue microarray. Gynecol Oncol 2004; 94:449-55. [PMID: 15297187 DOI: 10.1016/j.ygyno.2004.05.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2003] [Indexed: 01/10/2023]
Abstract
OBJECTIVE With the exception of ovarian serous carcinoma, Wilms tumor suppressor gene (WT1) expression in common gynecologic carcinomas has not been described in detail. We studied a large number of endometrial carcinomas to determine the range of tumors that express WT1; this could have prognostic and therapeutic significance. METHODS We studied the immunohistochemical expression of WT1 and p53 in 130 primary human endometrial carcinomas of various histological subtypes, grades, and stages using a tissue microarray. The clinical data were retrieved from the medical records. RESULTS WT1 was expressed in a wide variety of endometrial cancers and was most marked in malignant mixed Mullerian tumors (MMMTs) (70% positive). WT1 expression was significantly correlated with high histological grade, and there was a trend toward a worse clinical outcome for patients whose tumors expressed WT1. An association between expression of WT1 and p53 and between these and outcome was noted in a univariate analysis, but only stage and p53 status remained prognostically significant independent variables. CONCLUSION WT1 is expressed in appreciable numbers of endometrial cancers, particularly MMMTs. These findings support further investigation of WT1 as a possible therapeutic target in gynecologic malignancies.
Collapse
Affiliation(s)
- Jakob Dupont
- Developmental Chemotherapy Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Acs G, Pasha T, Zhang PJ. WT1 is Differentially Expressed in Serous, Endometrioid, Clear Cell, and Mucinous Carcinomas of the Peritoneum, Fallopian Tube, Ovary, and Endometrium. Int J Gynecol Pathol 2004; 23:110-8. [PMID: 15084838 DOI: 10.1097/00004347-200404000-00004] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Wilms' tumor gene WT1 plays complex roles in the development of the organs of the genitourinary tract and mesothelium, as well as Wilms' tumors. Although its biologic role is still unclear, most serous carcinomas of the ovary and peritoneum, mesotheliomas, and Wilms' tumor have been shown to express WT1. A recent study, however, found no WT1 expression in serous carcinomas of the endometrium, suggesting that WT1 could be useful in identifying the primary site of serous carcinomas. We examined the expression of WT1 and p53 by immunohistochemistry in 69 cases of endometrial carcinoma (35 endometrioid, 18 clear cell, 16 serous), 68 cases of ovarian carcinoma (28 serous, 11 endometrioid, 18 clear cell, and 11 mucinous), 14 fallopian tube carcinomas (12 serous, 2 endometrioid), and 20 primary peritoneal serous carcinomas. WT1 nuclear reactivity of any extent and intensity was considered positive. Immunohistochemical stains were evaluated semiquantitatively using a four-tiered scale. Among endometrial carcinomas, WT1 immunoreactivity was seen in 10 of 16 serous, but in none of 35 endometrioid or 18 clear cell carcinomas. Among ovarian tumors, WT1 expression was seen in 24 of 28 serous and 4 of 18 clear cell carcinomas, but in none of 11 endometrioid and 11 mucinous tumors. All 12 serous carcinomas but none of 2 endometrioid carcinomas of the fallopian tube were positive for WT1. WT1 expression was seen in 19 of 20 serous primary peritoneal carcinomas. The difference in WT1 expression was highly significant between serous and other types of tumors in all sites (p<0.0001, chi-square test), although the level of WT1 expression was significantly different among serous carcinomas arising at different sites (p<0.0001, Kruskal-Wallis test). A significant positive correlation was found between the level of p53 and WT1 expression in all carcinomas combined (r = 0.3935, p<0.0001, Spearman test), but when only serous carcinomas were analyzed, the correlation between p53 and WT1 expression levels did not reach statistical significance. Our results suggest that WT1 expression in epithelial tumors of the female genital tract may be related to cell differentiation and histologic subtypes of carcinomas, rather than to primary site of origin.
Collapse
Affiliation(s)
- Geza Acs
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
62
|
Shichishima T, Noji H. A new aspect of the molecular pathogenesis of paroxysmal nocturnal hemoglobinuria. ACTA ACUST UNITED AC 2004; 7:211-27. [PMID: 14972783 DOI: 10.1080/1024533021000024094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematologic disorder which is manifest by complement-mediated hemolysis, venous thrombosis, and bone marrow failure. Complement-mediated hemolysis in PNH is explained by the deficiency of glycosylphosphatidylinositol (GPI)-anchored proteins, CD55 and CD59 on erythrocyte surfaces. All the PNH patients had phosphatidylinositol glycan-class A (PIG-A) gene abnormalities in various cell types, indicating that PIG-A gene mutations cause the defects in GPI-anchored proteins that are essential for the pathogenesis of PNH. In addition, a PIG-A gene abnormality results in a PNH clone. Bone marrow failure causes cytopenias associated with a proliferative decrease of its hematopoietic stem cells and appears to be related to a pre-leukemic state. Although it is unclear how a PNH clone expands in bone marrow, it is considered that the most important hypothesis implicates negative selection of a PNH clone, but it does not explain the changes in the clinical features at the terminal stage of PNH. Recently, it has been suggested that an immune mechanism, in an HLA-restricted manner, plays an important role in the occurrence or selection of a PNH clone and GPI may be a target for cytotoxic-T lymphocytes. Also, it has been indicated that the Wilms' tumor gene (WT1) product is related to a PNH clone, but the significance of WT1 expression is not clear because of the functional diversity of the gene. To elucidate this problem, it is important to know the pathophysiology of bone marrow failure in detail and how bone marrow failure affects hematopoietic stem cells and immune mechanisms in bone marrow failure syndromes.
Collapse
Affiliation(s)
- Tsutomu Shichishima
- First Department of Internal Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima 960-1295, Japan
| | | |
Collapse
|
63
|
Nakade K, Zheng H, Ganguli G, Buchwalter G, Gross C, Wasylyk B. The tumor suppressor p53 inhibits Net, an effector of Ras/extracellular signal-regulated kinase signaling. Mol Cell Biol 2004; 24:1132-42. [PMID: 14729959 PMCID: PMC321436 DOI: 10.1128/mcb.24.3.1132-1142.2004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The tumor suppressor function of p53 is linked to its ability to repress gene expression, but the mechanisms of specific gene repression are poorly understood. We report that wild-type p53 inhibits an effector of the Ras oncogene/mitogen-activated protein (MAP) kinase pathway, the transcription factor Net. Tumor-associated mutant p53s are less efficient inhibitors. p53 inhibits by preventing phosphorylation of Net by MAP kinases. Loss of p53 in vivo leads to increased Net phosphorylation in response to wound healing and UV irradiation of skin. Our results show that p53 can repress specific gene expression by inhibiting Net, a factor implicated in cell cycle entry.
Collapse
Affiliation(s)
- Koji Nakade
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, 67404 Illkirch cedex, France
| | | | | | | | | | | |
Collapse
|
64
|
Discenza MT, Pelletier J. Insights into the physiological role of WT1 from studies of genetically modified mice. Physiol Genomics 2004; 16:287-300. [PMID: 14966251 DOI: 10.1152/physiolgenomics.00164.2003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Discenza, Maria Teresa, and Jerry Pelletier. Insights into the physiological role of WT1 from studies of genetically modified mice. Physiol Genomics 16: 287-300, 2004; 10.1152/physiolgenomics.00164.2003.—The identification of WT1 gene mutations in children with WAGR and Denys-Drash syndromes pointed toward a role for WT1 in genitourinary system development. Biochemical analysis of the different WT1 protein isoforms showed that WT1 is a transcription factor and also has the ability to bind RNA. Analysis of WT1 complexes identified several target genes and protein partners capable of interacting with WT1. Some of these studies placed WT1, its downstream targets, and protein partners in a transcriptional regulatory network that controls urogenital system development. We review herein studies on WT1 knockout and transgenic models that have been instrumental in defining a physiological role for WT1 in normal and abnormal urogenital development.
Collapse
|
65
|
Vajjhala PR, Macmillan E, Gonda T, Little M. The Wilms' tumour suppressor protein, WT1, undergoes CRM1-independent nucleocytoplasmic shuttling. FEBS Lett 2003; 554:143-8. [PMID: 14596929 DOI: 10.1016/s0014-5793(03)01144-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Wilms' tumour suppressor gene (WT1) encodes a zinc finger-containing nuclear protein essential for kidney and urogenital development. Initially considered a transcription factor, there is mounting evidence that WT1 has a role in post-transcriptional processing. Using the interspecies heterokaryon assay, we have demonstrated that WT1 can undergo nucleocytoplasmic shuttling. We have also mapped the region responsible for nuclear export to residues 182-324. Our data add further complexity to the role of WT1 in transcriptional and post-transcriptional regulation.
Collapse
Affiliation(s)
- P R Vajjhala
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Qld 4072, Australia.
| | | | | | | |
Collapse
|
66
|
Alami J, Williams BR, Yeger H. Differential expression of E-cadherin and beta catenin in primary and metastatic Wilms's tumours. Mol Pathol 2003; 56:218-25. [PMID: 12890743 PMCID: PMC1187324 DOI: 10.1136/mp.56.4.218] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The E-cadherin-catenin adhesion complex is crucial for intercellular adhesiveness and maintenance of tissue architecture. Its impairment is associated with poorly differentiated phenotype and increased invasiveness of carcinomas. AIMS To evaluate E-cadherin, beta catenin, gamma catenin, and ezrin expression and its relation to histopathological features of primary and metastatic Wilms's tumours. METHODS Immunohistochemistry was used to determine the expression and cellular distribution of E-cadherin, beta catenin, gamma catenin, and ezrin in primary and metastatic Wilms's tumours. Western blotting was used to determine polypeptide size and expression of E-cadherin and beta catenin in Wilms's tumours compared with normal kidney. RESULTS Moderate expression of E-cadherin was found mainly in cytoplasm and occasionally cell membranes of dysplastic tubules, whereas low expression was seen in cytoplasm of blastemal cells. Primary and metastatic tumours showed moderate to high beta catenin expression in blastemal and epithelial cells, with predominantly membranous and cytoplasmic staining. Occasional nuclear staining was noted in metastatic tumours. Low to high gamma catenin and ezrin expression was seen in cytoplasm of blastemal and epithelial cells of primary and metastatic tumours. Higher amounts of 92 kDa beta catenin were detected in tumours than in normal kidney. Low expression of 120 kDa E-cadherin was seen in moderately differentiated tumours, whereas expression was lacking in poorly differentiated tumours. CONCLUSIONS Compared with primary tumours, metastatic tumours showed lower expression of E-cadherin and gamma catenin, with nuclear staining for beta catenin. Low E-cadherin was associated with poorly differentiated tumours. These results suggest that abnormal expression of adhesion proteins correlates with the invasive and metastatic phenotype in Wilms's tumours.
Collapse
Affiliation(s)
- J Alami
- Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
67
|
Heinlein CA, Chang C. Induction and repression of peroxisome proliferator-activated receptor alpha transcription by coregulator ARA70. Endocrine 2003; 21:139-46. [PMID: 12897377 DOI: 10.1385/endo:21:2:139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2002] [Revised: 02/10/2003] [Accepted: 02/12/2003] [Indexed: 11/11/2022]
Abstract
In an effort to understand transcriptional regulation by the peroxisome proliferator-activated receptor alpha (PPARalpha), we investigated the ability of a number of transcriptional coactivators to enhance PPARalpha:retinoic acid receptor (RXR) mediated transcription. We identified ARA70, a coactivator of the androgen receptor and PPARgamma, as a ligand-enhanced coactivator of PPARalpha in the prostate cancer cell line DU145. In prostate cancer cells, ARA70 demonstrated the strongest enhancement of PPARalpha transcription among the coactivators examined. Mutation of the N-terminal of the PPARalpha ligandbinding domain dramatically reduced the ability of ARA70 to enhance PPARalpha:RXR transcription. ARA70 was able to physically interact with both the wild-type and mutant PPARalpha as determined by coimmunoprecipitation. However, in the adrenal cell line Y1, ARA70 behaved as a repressor of PPARalpha while still able to coactivate PPARgamma.
Collapse
Affiliation(s)
- Cynthia A Heinlein
- George Whipple Laboratory for Cancer Research, Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
68
|
Johannesen J, Karlsen AE, Pociot F, Roenn SG, Nerup J. Strain dependent rat iNOS promoter activity--correlation to identified WT1 transcription factor binding site. Autoimmunity 2003; 36:167-75. [PMID: 12911284 DOI: 10.1080/0891693031000101288] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The free radical nitric oxide (NO) has been implicated in cytokine mediated destruction of rat beta-cells in islets of Langerhans. Cytokine mediated NO production is associated with increased expression of the inducible nitric oxide synthase (iNOS). We have previously shown a strain dependent difference between Wistar Kyoto (WKY) and Brown Norway (BN) rats of IL-1beta mediated destruction of islets of Langerhans to be related to expression levels of iNOS and NO production. The aim of the present study was to clone and screen the iNOS gene promoter region from WKY and BN rats for polymorphisms and to functionally test such nucleotide differences. Within the total 2077 bp sequenced from both rat strains we identified three polymorphisms in two separate areas: (i) a GT-repeat polymorphism linked to (ii) a C/T polymorphisms, leading to a WT1 binding site approximately 1650bp upstream the BN iNOS promoter and (iii) a G/A SNP in exon 1. Apart from these polymorphisms the homology between all published rat iNOS sequences including the presently described are about 96%. Promoter activity was detected for both genes in a luciferase assay followed cloning of 2012 bp fragments and transient transfection into RIN cells. For both strains IL-1beta induced dose-dependent activity and strain dependent iNOS promoter activity was demonstrated when WT1 was co-expressed. To our knowledge, this is the first demonstration of functional WT1/iNOS promoter interaction. We conclude that the iNOS promoter is strain-dependently regulated which may relate to quantitatively as well as qualitatively strain dependent differences in transcription factor expression, in this study exemplified by WT1.
Collapse
Affiliation(s)
- Jesper Johannesen
- Steno Diabetes Center, Niels Steensensvej 2, DK-2820 Gentofte, Denmark
| | | | | | | | | |
Collapse
|
69
|
Abstract
In the present study we have demonstrated that WT1 (Wilms tumor suppressor gene) enhances the expression of TauT (taurine transporter gene) in human embryonic kidney 293 cells in a dose-dependent manner. TauT promoter activity was increased five-fold by cotransfection of a full-length TauT promoter-reporter construct with WT1. Electrophoretic mobility shift assays (EMSAs) using nuclear extracts from WT1-overexpressing 293 cells showed a putative WT1-binding site in the basal promoter region of TauT, which bound to WT1 in EMSAs. Mutation of this WT1 consensus sequence abolished binding of WT1. These results demonstrate that TauT may represent a downstream target gene of WT1 during renal development.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Pediatrics, University of Tennessee Health Science Center, and the Children's Foundation Research Center at Le Bonheur Children's Medical Center, 50 North Dunlap, Memphis, TN 38103-4909, USA
| | | |
Collapse
|
70
|
Idelman G, Glaser T, Roberts CT, Werner H. WT1-p53 interactions in insulin-like growth factor-I receptor gene regulation. J Biol Chem 2003; 278:3474-82. [PMID: 12444079 DOI: 10.1074/jbc.m211606200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The insulin-like growth factor-I receptor (IGF-IR) plays a critical role in transformation. The expression of the IGF-IR gene is negatively regulated by a number of transcription factors, including the WT1 and p53 tumor suppressors. Previous studies have suggested both physical and functional interactions between the WT1 and p53 proteins. The potential functional interactions between WT1 and p53 in control of IGF-IR promoter activity were addressed by transient coexpression of vectors encoding different isoforms of WT1, together with IGF-IR promoter-luciferase reporter constructs, in p53-null osteosarcoma-derived Saos-2 cells, wild-type p53-expressing kidney tumor-derived G401 cells, and mutant p53-expressing, rhabdomyosarcoma-derived RD cells. Similar studies were also performed to compare p53-expressing Balb/c-3T3 and clonally derived p53-null, (10)1 fibroblasts and the colorectal cancer cell line HCT116 +/+, which expresses a wild-type p53 gene, and its HCT116 -/- derivative, in which the p53 gene has been disrupted by homologous recombination. WT1 splice variants lacking a KTS insert between zinc fingers 3 and 4 suppressed IGF-IR promoter activity in the absence of p53 or in the presence of wild-type p53. WT1 variants that contain the KTS insert are impaired in their ability to bind to the IGF-IR promoter and are unable to suppress IGF-IR promoter. In the presence of mutant p53, WT1 cannot repress the IGF-IR promoter. Coimmunoprecipitation experiments showed that p53 and WT1 physically interact, whereas electrophoretic mobility shift assay studies revealed that p53 modulates the ability of WT1 to bind to the IGF-IR promoter. In summary, the transcriptional activity of WT1 proteins and their ability to function as tumor suppressors or oncogenes depends on the cellular status of p53.
Collapse
Affiliation(s)
- Gila Idelman
- Department of Clinical Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
71
|
Sandberg AA, Bridge JA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors. desmoplastic small round-cell tumors. CANCER GENETICS AND CYTOGENETICS 2002; 138:1-10. [PMID: 12419577 DOI: 10.1016/s0165-4608(02)00680-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Avery A Sandberg
- Department of DNA Diagnostics, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ 85013, USA.
| | | |
Collapse
|
72
|
Du X, Hublitz P, Günther T, Wilhelm D, Englert C, Schüle R. The LIM-only coactivator FHL2 modulates WT1 transcriptional activity during gonadal differentiation. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1577:93-101. [PMID: 12151099 DOI: 10.1016/s0167-4781(02)00414-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An essential step during sex determination is the maintenance of the Müllerian duct in females and its regression in males caused by the expression of Müllerian inhibiting substance (MIS). In testes, the Wilms' tumor suppressor and the orphan nuclear receptor SF1 cooperatively bind to the promoter and activate transcription of MIS. In the ovaries, on the other hand, the orphan nuclear receptor DAX1 binds to SF1, inhibits transactivation by WT1/SF1 and thereby suppresses the induction of MIS expression. In addition, WT1 itself is responsible for the upregulation of DAX1 transcription. So far, little is known on which protein-protein interactions or cofactors elicit the spatiotemporal control of WT1-mediated transcription. Here we demonstrate coexpression of the LIM-only coactivator FHL2 and WT1. FHL2 and WT1 functionally interact both in vitro and in vivo. The importance of this interaction is revealed by the ability of FHL2 to potentiate the synergistic induction of MIS gene expression by WT1/SF1. Moreover, FHL2 coactivates transactivation of the DAX1 promoter by WT1. Hence, we present FHL2 as a novel transcriptional coactivator of WT1. The ability to modulate both DAX1 and MIS expression might allow FHL2 to act in the molecular fine tuning of WT1-dependent control mechanisms in the reproductive organs.
Collapse
Affiliation(s)
- Xiaojuan Du
- Universitäts-Frauenklinik und Zentrum für Klinische Forschung, Klinikum der Universität Freiburg, Breisacherstrasse 66, 79106 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
73
|
Baudry D, Faussillon M, Cabanis MO, Rigolet M, Zucker JM, Patte C, Sarnacki S, Boccon-Gibod L, Junien C, Jeanpierre C. Changes in WT1 splicing are associated with a specific gene expression profile in Wilms' tumour. Oncogene 2002; 21:5566-73. [PMID: 12165855 DOI: 10.1038/sj.onc.1205752] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2002] [Revised: 05/28/2002] [Accepted: 06/07/2002] [Indexed: 11/08/2022]
Abstract
Wilms' tumour (WT) or nephroblastoma is the most frequent kidney cancer in children. In a previous study, we reported alterations to WT1 transcription in 90% of WT tested, with decreased exon 5 +/- isoform ratio being the most frequent alteration (56% of WT). We now report an approach based on cDNA profiling of tumour pools to identify genes likely to be dysregulated in association with a decreased WT1 exon 5 +/- ratio. We compared the expression profiles of pools of tumours classified according to whether this isoform imbalance was present (five tumours) or not (four tumours), using Atlas Cancer cDNA expression arrays. Fourteen of 588 genes tested displayed specific up-regulation (CCND2, PCNA, N-MYC, E2F3, TOP2A, PAK1, DCC and PCDH2) or down-regulation (VEGF, IGFBP5, TIMP3, ARHB, C-FOS and CD9) in the pool of tumours with decreased exon 5 +/- ratio. These results were validated by RT-PCR analysis of four genes (CCND2, PCNA, VEGF and IGFBP5). We extended the analysis of VEGF expression to 51 tumours by real-time RT-PCR and ascertained differential expression of this gene associated with WT1 expression pattern. Moreover, our results suggest that the VEGF expression level may be of prognosis relevance for relapsed patients.
Collapse
Affiliation(s)
- Dominique Baudry
- INSERM U383, Hôpital Necker-Enfants Malades, Université René Descartes, 75743 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Abstract
The Wilms tumor gene (WT1) is expressed in blasts of patients with acute leukemia, irrespective of lineage, and WT1 nuclear protein is detectable in the majority of such blasts. Only very few physiologic hematopoietic progenitors express WT1, but the WT1 expression level of these progenitors and that of leukemic blasts are comparable. Although not specific for acute hematologic malignant diseases, continuous WT1 expression in almost all leukemic blasts strikingly contrasts to its rather transient expression in very few physiologic hematopoietic progenitors. Quantitative and semiquantitative WT1 reverse transcriptase polymerase chain reaction (RT-PCR) protocols have limitations in discriminating physiologic from pathologic overall WT1 expression levels in mononuclear cell preparations. Because of these limitations, reports conflict on the usefulness of long-term monitoring of WT1 expression in patients with acute leukemia. Real-time quantitative WT1 RT-PCR protocols, however, have been developed and tested in small series of patients with acute leukemia. Such protocols hold promise to enable evaluation of the individual treatment response (short-term monitoring) and early diagnosis of imminent relapse through the detection and long-term monitoring of minimal residual disease in patients with acute leukemia. These protocols also should facilitate the notoriously difficult distinction between eosinophilic leukemia and hypereosinophilic syndromes. Data on WT1 expression in leukemic blasts and their physiologic counterparts are discussed in light of clinical relevance.
Collapse
Affiliation(s)
- Hans D Menssen
- Medical Clinic III, Hematology, Oncology, and Transfusion Medicine, University Clinic Benjamin Franklin, Free University, Berlin, Germany.
| | | | | |
Collapse
|
75
|
Algar E. A review of the Wilms' tumor 1 gene (WT1) and its role in hematopoiesis and leukemia. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:589-99. [PMID: 12201948 DOI: 10.1089/15258160260194749] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
One of the first clones of the Wilms tumor 1 (WT1) gene, WT33, was isolated from a B cell leukemia cell line in 1990. Now, 12 years on, WT1 has emerged as a potentially important target for antileukemic therapies. Our understanding of the role that WT1 plays during normal hematopoiesis is still limited, and there is a large amount of conflicting data concerning the precise manner in which WT1 gene expression contributes to leukemogenesis. However, interest in this field has intensified in the past 5 years. This review surveys the progress made in this area.
Collapse
Affiliation(s)
- Elizabeth Algar
- Department of Clinical Haematology and Oncology and the Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia, 3052.
| |
Collapse
|
76
|
Abstract
WT1 encodes a zinc finger transcription factor implicated in normal development and tumorigenesis. Germline mutation or deletion of WT1 results in a spectrum of abnormal kidney development, male-to-female intersex disorders, and predisposition to pediatric nephroblastoma, Wilms tumor. Initially thought to encode a transcriptional repressor, WT1-dependent functions are now more clearly linked to its property as a transcriptional activator of genes involved in renal development and sex determination. WT1 is expressed in 4 isoforms as a result of 2 alternative messenger RNA splicing events, the more significant of which encodes the 3 amino acids lysine, threonine, and serine (KTS) between zinc fingers 3 and 4. Although WT1 isoforms lacking KTS act as sequence-specific DNA binding factors, a large body of evidence now implicates the KTS-containing isoforms in RNA processing. In keeping with distinct biochemical mechanisms for these isoforms, genetic data from humans and mice point to separate but partially overlapping roles for WT1 (+KTS) and (-KTS) during genitourinary development. Recently, a hematopoietic model system has been used to study functional properties of WT1 in vitro. WT1 expression in primary hematopoietic cells leads to stage-specific effects that may be relevant to WT1-mediated tumor suppression.
Collapse
Affiliation(s)
- Leif W Ellisen
- Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA.
| |
Collapse
|
77
|
Shichishima T, Okamoto M, Ikeda K, Kaneshige T, Sugiyama H, Terasawa T, Osumi K, Maruyama Y. HLA class II haplotype and quantitation of WT1 RNA in Japanese patients with paroxysmal nocturnal hemoglobinuria. Blood 2002; 100:22-8. [PMID: 12070003 DOI: 10.1182/blood.v100.1.22] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is unclear how a paroxysmal nocturnal hemoglobinuria (PNH) clone expands in bone marrow, although immune mechanisms involving cytotoxic T lymphocytes, autosomal proliferation, and apoptosis resistance have been hypothesized. To clarify aspects of immune mechanisms and proliferation of PNH cells, we investigated HLA-DRB1, -DQA1, and -DQB1 alleles by polymerase chain reaction (PCR)-based genotyping and expression of the Wilms' tumor gene, WT1, by real-time reverse transcriptase-PCR (RT-PCR) in 21 PNH and 21 aplastic anemia (AA) patients. HLA genotyping indicated that the frequency of DRB1*1501, DQA1*0102, and DQB1*0602 alleles in PNH patients and of DQB1*0602 allele in AA patients was significantly higher than in 916 Japanese controls, and that the HLA-DRB1*1501-DQA1*0102-DQB1*0602 haplotype, found in 13 of 21 PNH patients, 5 of 7 AA-PNH syndrome patients, and 7 of 21 AA patients showed significant differences compared with healthy individuals. RT-PCR analysis showed that the mean values of WT1 RNA were 3413, 712, and 334 copies/microg RNA in PNH, AA, and healthy individuals, respectively. The values for PNH patients were significantly higher than for AA patients and healthy volunteers and were correlated with the proportion of CD16b(-) granulocytes. The high frequency of HLA-DRB1*1501-DQA1*0102-DQB1*0602 haplotype in PNH, including AA-PNH syndrome, and AA patients suggests that linkage exists between the disorders and that immune mechanisms in an HLA-restricted manner play an important role in the pathogenesis of these disorders. In addition, high expression of WT1 RNA in PNH patients is related to a PNH clone, but it remains unclear whether this causes expansion of a PNH clone.
Collapse
Affiliation(s)
- Tsutomu Shichishima
- First Department of Internal Medicine, Fukushima Medical University, Fukushima, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Valerius MT, Patterson LT, Witte DP, Potter SS. Microarray analysis of novel cell lines representing two stages of metanephric mesenchyme differentiation. Mech Dev 2002; 112:219-32. [PMID: 11850199 DOI: 10.1016/s0925-4773(02)00008-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Clonal cell lines representing different developmental stages of the metanephric mesenchyme were made from transgenic mice with the Simian Virus 40 T-antigen (SV40 Tag) gene driven by the Hoxa 11 promoter. The resulting mK3 cell line represented early metanephric mesenchyme, prior to induction by the ureteric bud. These cells showed a spindle-shaped, fibroblast morphology. They expressed genes characteristic of early mesenchyme, including Hoxa 11, Hoxd 11, collagen I, and vimentin. Moreover, the mK3 cells displayed early metanephric mesenchyme biological function. In organ co-culture experiments they were able to induce growth and branching of the ureteric bud. Another cell line, mK4, represented later, induced metanephric mesenchyme undergoing epithelial conversion. These cells were more polygonal, or epithelial in shape, and expressed genes diagnostic of late mesenchyme, including Pax-2, Pax-8, Wnt-4, Cadherin-6, Collagen IV, and LFB3. To better define the gene expression patterns of kidney metanephric mesenchyme cells at these two stages of development, RNAs from the mK3 and mK4 cells were hybridized to Affymetrix GeneChip probe arrays. Over 4000 expressed genes were identified and thereby implicated in kidney formation. Comparison of the mK3 and mK4 gene expression profiles revealed 121 genes showing greater than a ten-fold difference in expression level. Several are known to be expressed during metanephric mesenchyme differentiation, but most had not been previously associated with this process. In situ hybridizations were used to confirm that selected novel genes were expressed in the developing kidney.
Collapse
Affiliation(s)
- M Todd Valerius
- Division of Developmental Biology, Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
79
|
Valerius MT, Patterson LT, Witte DP, Potter SS. Microarray analysis of novel cell lines representing two stages of metanephric mesenchyme differentiation. Mech Dev 2002; 110:151-64. [PMID: 11744376 DOI: 10.1016/s0925-4773(01)00581-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Clonal cell lines representing different developmental stages of the metanephric mesenchyme were made from transgenic mice with the Simian Virus 40 T-antigen (SV40 Tag) gene driven by the Hoxa 11 promoter. The resulting mK3 cell line represented early metanephric mesenchyme, prior to induction by the ureteric bud. These cells showed a spindle-shaped, fibroblast morphology. They expressed genes characteristic of early mesenchyme, including Hoxa 11, Hoxd 11, collagen I, and vimentin. Moreover, the mK3 cells displayed early metanephric mesenchyme biological function. In organ co-culture experiments they were able to induce growth and branching of the ureteric bud. Another cell line, mK4, represented later, induced metanephric mesenchyme undergoing epithelial conversion. These cells were more polygonal, or epithelial in shape, and expressed genes diagnostic of late mesenchyme, including Pax-2, Pax-8, Wnt-4, Cadherin-6, Collagen IV, and LFB3. To better define the gene expression patterns of kidney metanephric mesenchyme cells at these two stages of development, RNAs from the mK3 and mK4 cells were hybridized to Affymetrix GeneChip probe arrays. Over 4000 expressed genes were identified and thereby implicated in kidney formation. Comparison of the mK3 and mK4 gene expression profiles revealed 121 genes showing greater than a ten-fold difference in expression level. Several are known to be expressed during metanephric mesenchyme differentiation, but most had not been previously associated with this process. In situ hybridizations were used to confirm that selected novel genes were expressed in the developing kidney.
Collapse
Affiliation(s)
- M Todd Valerius
- Division of Developmental Biology, Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
80
|
Lee TH, Pelletier J. Functional characterization of WT1 binding sites within the human vitamin D receptor gene promoter. Physiol Genomics 2001; 7:187-200. [PMID: 11773605 DOI: 10.1152/physiolgenomics.00046.2001] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Wilms' tumor suppressor gene, wt1, encodes a zinc finger transcription factor that can regulate gene expression. It plays an essential role in tumorigenesis, kidney differentiation, and urogenital development. To identify WT1 downstream targets, gene expression profiling was conducted using a cDNA array hybridization approach. We confirm herein that the human vitamin D receptor (VDR), a ligand-activated transcription factor, is a WT1 downstream target. Nuclear run on experiments demonstrated that the effect of WT1 on VDR expression is at the transcriptional level. Transient transfection assays, deletion mutagenesis, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays suggest that, although WT1 is presented with a possibility of three binding sites within the VDR promoter, activation of the human VDR gene appears to occur through a single site. This site differs from a previously identified WT1-responsive site in the murine VDR promoter (Maurer U, Jehan F, Englert C, Hübinger G, Weidmann E, DeLucas HF, and Bergmann L. J Biol Chem 276: 3727-3732, 2001). We also show that the products of a Denys-Drash syndrome allele of wt1 inhibit WT1-mediated transactivation of the human VDR promoter. Our results indicate that the human VDR gene is a downstream target of WT1 and may be regulated differently than its murine counterpart.
Collapse
Affiliation(s)
- T H Lee
- Department of Biochemistry, McGill Cancer Center, McGill University, Montreal, Quebec, Canada H3G 1Y6
| | | |
Collapse
|
81
|
Hübinger G, Schmid M, Linortner S, Manegold A, Bergmann L, Maurer U. Ribozyme-mediated cleavage of wt1 transcripts suppresses growth of leukemia cells. Exp Hematol 2001; 29:1226-35. [PMID: 11602325 DOI: 10.1016/s0301-472x(01)00719-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The Wilms' tumor gene product (WT1) was identified as a tumor suppressor in pediatric kidney tumors. Conversely, acute leukemias express WT1 at a high frequency, and leukemias with high levels of WT1 expressed by leukemic blast cells have a significantly worse prognosis, suggesting an oncogenic function of WT1 in leukemic cells. To address this issue, we developed five hammerhead ribozymes (RZ1-RZ5) designed to cleave various wt1-mRNA GUC-recognition sites and thus suppress wt1 expression. METHODS Using in vitro transcribed ribozymes and truncated wt1 target RNAs as substrates, we performed in vitro cleavage assays. The sequence of two ribozymes was then cloned into the pCDNA3 expression vector containing a self-processing ribozyme cassette. Downregulation of wt1 due to ribozyme expression was analyzed in the human 293 embryonic kidney and the K562 chronic myeloid leukemia cell line by Western blotting and RT-PCR. Growth of stable transfected K562 cells was determined by proliferation analysis and 3H-thymidine incorporation. RESULTS In vitro, the anti-wt1 ribozymes were able to recognize and cleave the target RNA in a highly sequence-specific and time-dependent manner. The ribozymes showed different catalytic activity. Coexpression of wt1 and the self-processing ribozymes pRZ3 and pRZ5, respectively, resulted in a significantly downregulated WT1 protein level when transiently transfected in 293 cells. Furthermore, stable transfection of pRZ3 and pRZ5 resulted in considerably reduced expression of endogenous wt1 in K562 cells, correlating with the inhibition of cell proliferation and the induction of cell death. CONCLUSION Our data suggest that anti-wt1 ribozymes are a potent inhibitor of wt1 expression with possible implications for the inhibition of cell proliferation in leukemic cells.
Collapse
Affiliation(s)
- G Hübinger
- Department of Internal Medicine III, University of Ulm, Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
82
|
Abstract
The Wilms' tumor gene WT1 is a gene responsible for the childhood renal tumor. Wilms' tumor, and is defined as a tumor suppressor gene. However, the wild-type WT1 gene is highly expressed in leukemic blast cells of myeloid and lymphoid origin, and thus, WT1 messenger RNA provides a novel tumor marker for detection of minimal residual disease of leukemias and for monitoring disease progression of myelodysplastic syndromes. The WT1 gene exerts an oncogenic function rather than a tumor-suppressor gene function in solid tumors as well as leukemias, and the WT1 gene product is an attractive tumor antigen capable of eliciting cytotoxic T lymphocytes against WT1-expressing tumors.
Collapse
Affiliation(s)
- H Sugiyama
- Department of Clinical Laboratory Science, Osaka University Medical School, Japan.
| |
Collapse
|
83
|
Abstract
The Wilms' tumor 1 gene (WT1) has been identified as a tumor suppressor gene involved in the etiology of Wilms' tumor. Approximately 10% of all Wilms' tumors carry mutations in the WT1 gene. Alterations in the WT1 gene have also been observed in other tumor types, such as leukemia, mesothelioma and desmoplastic small round cell tumor. Dependent on the tumor type, WT1 proteins might either function as tumor suppressor proteins or as survival factors. Mutations in the WT1 gene can also result in congenital abnormalities as observed in Denys-Drash and Frasier syndrome patients. Mouse models have proven the critical importance of WT1 expression for the development of several organs, including the kidneys, the gonads and the spleen. The WT1 proteins seem to perform two main functions. They regulate the transcription of a variety of target genes and may be involved in post-transcriptional processing of RNA. The WT1 gene encodes at least 24 protein forms. These isoforms have partially distinct biological functions and effects, which in many cases are also specific for the model system in which WT1 is studied. This review discusses the molecular mechanisms by which the various WT1 isoforms exert their functions in normal development and how alterations in WT1 may lead to developmental abnormalities and tumor growth.
Collapse
Affiliation(s)
- V Scharnhorst
- Department of Molecular and Cellular Biology and Center for Biomedical Genetics, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL, The, Leiden, Netherlands
| | | | | |
Collapse
|
84
|
Wagner KD, Wagner N, Sukhatme VP, Scholz H. Activation of Vitamin D Receptor by the Wilms' Tumor GeneProduct Mediates Apoptosis of Renal Cells. J Am Soc Nephrol 2001; 12:1188-1196. [PMID: 11373341 DOI: 10.1681/asn.v1261188] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Abstract. The Wilms' tumor transcription factor WT1 is required for kidney development, but little is known about WT1 downstream signaling in renal cells. This study reported an approximately fivefold upregulation of vitamin D receptor (VDR) mRNA and protein in human embryonic kidney (HEK) 293 cells that stably expressed WT1 at a level comparable to the developing kidney in vivo. Co-transfection of HEK 293 cells with expression plasmids encoding four different WT1 splicing variants stimulated mouse vdr promoter activity more than fourfold. A 201-bp fragment was identified in the proximal vdr promoter that was required for transactivation by WT1. This critical sequence contained a predicted WT1 consensus site, which bound to recombinant WT1 protein. Temporal changes of vdr and wt1 mRNA levels in developing rat kidneys were correlated closely. The active metabolite 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) strongly inhibited the proliferation of wt1-transfected HEK 293 cells. Exposure to 1,25-(OH)2D3 caused apoptosis of cultured wt1 immunopositive cells from mouse embryonic kidney cortex. These findings suggest that transcriptional activation of the VDR by WT1 can mediate programmed death of renal embryonic cells in response to 1,25-(OH)2D3. The results provide the first evidence for a role of the vitamin D endocrine system in renal cell growth and differentiation during development.
Collapse
Affiliation(s)
- Kay-Dietrich Wagner
- Johannes-Müller-Institut für Physiologie, Humboldt-Universität, Charité, Berlin, Germany
| | - Nicole Wagner
- Klinik für Innere Medizin I, Humboldt-Universität, Charité, Berlin, Germany
| | - Vikas P Sukhatme
- Beth Israel Deaconess Medical Center, Renal Division, Boston, Massachusetts
| | - Holger Scholz
- Johannes-Müller-Institut für Physiologie, Humboldt-Universität, Charité, Berlin, Germany
| |
Collapse
|
85
|
Sandberg AA, Bridge JA. Updates on the cytogenetics and molecular genetics of bone and soft tissue tumors. Mesothelioma. ACTA ACUST UNITED AC 2001. [DOI: 10.1016/s0165-4608(01)00432-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
86
|
Dohn M, Zhang S, Chen X. p63alpha and DeltaNp63alpha can induce cell cycle arrest and apoptosis and differentially regulate p53 target genes. Oncogene 2001; 20:3193-205. [PMID: 11423969 DOI: 10.1038/sj.onc.1204427] [Citation(s) in RCA: 228] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2000] [Revised: 02/16/2001] [Accepted: 02/26/2001] [Indexed: 12/14/2022]
Abstract
The p53 tumor suppressor protein plays a critical role in the regulation of the cell cycle and apoptosis. The importance of p53's functions is underscored by the high incidence of p53 mutations in human cancers. Recently, two p53-related proteins, p73 and p63, were identified as members of the p53 gene family. Multiple isoforms of p73 have been found, including DeltaN variants in which the N-termini are truncated. p63 is expressed as three major forms, p63alpha, p63beta and p63gamma, each of which differ in their C-termini. All three forms can be alternatively transcribed from a cryptic promoter located within intron 3, producing DeltaNp63alpha, DeltaNp63beta and DeltaNp63gamma. The high degree of similarity of p73 and p63 to evolutionarily conserved regions of p53 suggests that these proteins play an important and potentially redundant role in regulating cell cycle arrest and apoptosis. Here we describe the characterization of cell lines generated to inducibly express p63alpha and DeltaNp63alpha. We have found that p63alpha and DeltaNp63alpha can differentially regulate endogenous p53 target genes and induce cell cycle arrest and apoptosis. Deletion of the N-terminal 26 amino acids of DeltaNp63alpha abolished its ability to transactivate p53 target genes and induce cell cycle arrest and apoptosis. This indicates that a putative transactivation domain exists within the N-terminus of the DeltaN variants of p63. Furthermore, the differential regulation of p53 target genes by p63alpha and DeltaNp63alpha suggests that p63 and p53 utilize both similar and different signaling pathways to execute their cellular functions.
Collapse
Affiliation(s)
- M Dohn
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, GA 30912, USA
| | | | | |
Collapse
|
87
|
Wang W, Lee SB, Palmer R, Ellisen LW, Haber DA. A functional interaction with CBP contributes to transcriptional activation by the Wilms tumor suppressor WT1. J Biol Chem 2001; 276:16810-6. [PMID: 11278547 DOI: 10.1074/jbc.m009687200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Wilms tumor gene WT1 encodes a zinc finger transcription factor that is required for normal kidney development. WT1 was identified as a transcriptional repressor, based on its suppression of promoter reporters, but analysis of native transcripts using high density microarrays has uncovered transcriptional activation, rather than repression, of potential target genes. We report here that WT1 binds to the transcriptional coactivator CBP, leading to synergistic activation of a physiologically relevant promoter. The physical interaction between WT1 and CBP is evident in vitro and in vivo, and the two proteins are co-immunoprecipitated from embryonic rat kidney cells. The WT1-CBP association requires the first two zinc fingers of WT1 and the adenovirus 5 E1A-binding domain of CBP. Overexpression of this domain of CBP is sufficient to inhibit WT1-mediated transcriptional activation of a promoter reporter, as is co-transfection of E1A. Retrovirally driven expression of either the CBP fragment or of E1A in human hematopoietic cells suppresses the induction by WT1 of its endogenous target gene, p21(Cip1). These observations support a model of WT1 as a transcriptional activator of genes required for cellular differentiation.
Collapse
Affiliation(s)
- W Wang
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | |
Collapse
|
88
|
Abstract
Considering the characteristics of RA synovial tissues such as marked proliferation and invasion to adjacent tissues, comparisons with transformed or neoplastic tissue are natural. RA synovial tissues or cells are not truly malignant, but they have many features of transformation, denoted as "partial transformation" in this article. These features include anchorage-independent growth, loss of contact inhibition, oncogene activation, monoclonal or oligoclonal expansion, detectable telomerase activity, and somatic gene mutations. Although it is not possible to conclude whether most of these cells are permanently changed in association with some genetic alterations or are passively changed by virtue of environmental factors (i.e., cytokine-mediated imprinting), the presence of p53 mutations in RA synovial tissues is especially persuasive. A number of transcription factors play a critical role in the activation, differentiation, and proliferation of RA synovial cells. In particular, the roles of AP-1, MAPKs, and NF-kappa B have been investigated carefully because of their ability to regulate numerous inflammation-related genes. These transcription factors also control expression and activation of matrix-degrading enzymes, including MMPs, aggrecanase, and cysteine proteases, which are the primary enzymes responsible for joint destruction. Elucidation of gene mutations and detailed signal transduction pathways that are specific to RA as well as mechanisms of action of matrix-degrading enzymes may lead to development of a novel therapy for RA. Careful mapping of cytokine networks a decade ago led to groundbreaking advances in therapy. Similarly, methodical evaluation and prioritization of intracellular targets might provide the basis for therapeutic interventions.
Collapse
Affiliation(s)
- Y Yamanishi
- Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | | |
Collapse
|
89
|
Rauscher FJ. Chromosome translocation-mediated conversion of a tumor suppressor gene into a dominant oncogene: fusion of EWS1 to WT1 in desmoplastic small round cell tumors. Curr Top Microbiol Immunol 2001; 220:151-62. [PMID: 9103681 DOI: 10.1007/978-3-642-60479-9_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
MESH Headings
- Adolescent
- Cell Transformation, Neoplastic/genetics
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Wilms Tumor
- Heterogeneous-Nuclear Ribonucleoproteins
- Humans
- Male
- Models, Genetic
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Oncogenes
- Ribonucleoproteins/genetics
- Ribonucleoproteins/physiology
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/pathology
- Sarcoma, Small Cell/genetics
- Sarcoma, Small Cell/pathology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Translocation, Genetic
- WT1 Proteins
- Zinc Fingers/genetics
- Zinc Fingers/physiology
Collapse
|
90
|
Abstract
Wilms tumor or nephroblastoma is a pediatric kidney cancer arising from pluripotent embryonic renal precursors. Multiple genetic loci have been linked to Wilms tumorigenesis; positional cloning strategies have led to the identification of the WT1 tumor suppressor gene at chromosome 11p13. WT1 encodes a zinc finger transcription factor that is inactivated in the germline of children with genetic predisposition to Wilms tumor and in a subset of sporadic cancers. When present in the germline, specific heterozygous dominant-negative mutations are associated with severe abnormalities of renal and sexual differentiation, pointing to the essential role of WT1 for normal genitourinary development. The role of this tumor suppressor in normal organ-specific differentiation is also supported by the highly restricted temporal and spatial expression of WT1 in glomerular precursors of the developing kidney and by the failure of kidney development in wt1-null mice. Of two major alternative splicing products encoded by WT1, the (-KTS) isoform appears to mediate transcriptional activation of genes implicated in cellular differentiation, possibly also repressing proliferation-associated genes. The (+KTS) isoform, whose DNA-binding domain is disrupted by the insertion of three amino acids, may be involved in some aspect of mRNA processing. In addition to its function in genitourinary development, a role for WT1 in hematopoiesis is suggested by its aberrant expression and/or mutation in a subset of acute human leukemias. WT1 is also expressed in mesothelial cells; a specific oncogenic chromosomal translocation fusing the N-terminal domain of the Ewing sarcoma gene EWS to the three C-terminal zinc fingers of WT1 underlies desmoplastic small round cell tumor, an abdominal tumor thought to arise from the peritoneal lining. Understanding the distinct functional properties of WT1 isoforms and tumor-associated variants will provide unique insight into the link between normal organ-specific differentiation and malignancy.
Collapse
Affiliation(s)
- S B Lee
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachussetts 02129, USA
| | | |
Collapse
|
91
|
Abstract
WT1, a tumor suppressor gene responsible for the development of childhood kidney tumors, is now also thought to be involved in the occurrence of human leukemia. First, evidence has shown that WT1 functions during hematopoiesis and regulates the proliferation and differentiation of blood cells. Second, specific expression patterns of this gene correlate with the malignant phenotype of leukemia compared with the physiological situation. Third, mutations of WT1 can be detected, though not frequently, in human leukemia but not in normal hematopoietic cells. Thus, a possible role of WT1 in human leukemogenesis has been proposed. Because the expression of this gene is relatively high during the so-called myelodysplastic stages and in all subtypes of human leukemia compared with normal blood cells, the notion has been raised that WT1 can be used as a "panleukemic marker" for the diagnosis of leukemia at the molecular level. The expression level of WT1 may have significance in predicting prognosis and monitoring relapse. Moreover, with a deeper understanding of its role in leukemogenesis, WT1 may serve as a target molecule in the strategy of gene therapy for leukemia.
Collapse
Affiliation(s)
- Z Chen
- Jiangsu Institute of Hematology, First Affiliated Hospital, Suzhou University, China.
| |
Collapse
|
92
|
Kushner L, Chiu PY, Pinto P, Weis GH. Expression of c-met and WT-1. METHODS IN MOLECULAR MEDICINE 2001; 53:175-191. [PMID: 21318796 DOI: 10.1385/1-59259-144-2:175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Protooncogenes and tumor-suppressor genes are two types of genes associated with cancer development.
Collapse
Affiliation(s)
- L Kushner
- Department of Urology, Long Island Jewish Medical Center, New Hyde Park, NY
| | | | | | | |
Collapse
|
93
|
Stanhope-Baker P, Williams BR. Identification of connective tissue growth factor as a target of WT1 transcriptional regulation. J Biol Chem 2000; 275:38139-50. [PMID: 10973960 DOI: 10.1074/jbc.m004901200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Wilms tumor suppressor WT1 has transcription-activating and -suppressing capabilities. WT1-responsive promoters have been described; however, in large part, it remains unclear which potential downstream genes are physiologically relevant and mediate the function of WT1 in tumorigenesis and development. To identify genes regulated by WT1 in vivo, we used a dominant-negative version of WT1 to modulate WT1 activity in a Wilms tumor cell line. Screening oligonucleotide arrays with RNA from these cells uncovered a number of genes whose expression was altered by abrogation of WT1 function. Several of the genes encode members of the CCN family of growth regulators. The promoter of one of these genes, connective tissue growth factor (CTGF), is suppressed by WT1 both in its endogenous location and in reporter constructs. WT1 regulation of CTGF expression is not mediated by previously identified WT1 recognition elements and may therefore involve a novel mechanism. Our results indicate that CTGF is a bona fide target of WT1 transcriptional suppression and likely plays a role in Wilms tumorigenesis and associated disease syndromes.
Collapse
Affiliation(s)
- P Stanhope-Baker
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
94
|
Carrington D, Algar E. Overexpression of murine WT1 + / + and - / - isoforms has no effect on chemoresistance but delays differentiation in the K562 leukemia cell line. Leuk Res 2000; 24:927-36. [PMID: 11086176 DOI: 10.1016/s0145-2126(00)00064-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The Wilms' tumor gene (WT1) encodes a zinc-finger transcription factor that is expressed as four distinct isoforms designated as, + / +, + / -, - / + and - / -. It is expressed in leukemic cells, and is proposed to play a role in their proliferation and differentiation. In this study we have shown that cell lines of the erythroleukemia, K562, overexpressing the murine + / + and - / - WT1 isoforms grow normally and do not exhibit altered responses to the induction of apoptosis by the reagents cisplatin and adriamycin, or to serum withdrawal. However, differentiation of K562 cells with 12-O-tetradecanoylphorbol 13-acetate, modeling aspects of megakaryopoiesis, was partially inhibited by the persistent expression of both the murine + / + and - / - WT1 isoforms. This finding suggests that WT1 plays a role in the regulation of hematopoietic differentiation and is consistent with an oncogenic role for WT1 in leukemogenesis.
Collapse
Affiliation(s)
- D Carrington
- Department of Haematology and Oncology, Royal Children's Hospital, Parkville, Australia
| | | |
Collapse
|
95
|
Abstract
The Wilms' tumour suppressor gene WT1 is essential for the normal development of the genitourinary system. It appears to play a role in both transcriptional and post-transcriptional regulation of certain cellular genes. However, the mechanisms behind WT1 function are not clearly understood despite the identification of numerous potential target genes and the isolation of several WT1-binding proteins. This study therefore sets out to identify other WT1-associating proteins to help to unravel how WT1 interacts with the cellular machinery. We report the identification of a novel human WT1-associating protein, WTAP, which was isolated using the yeast two-hybrid system. Both in vitro and in vivo assays have shown that the interaction between WTAP and WT1 is specific and occurs endogenously in cells. The mouse homologue of WTAP was isolated and found to be >90% conserved at the nucleotide and protein levels. The human and mouse genes were mapped using fluorescence in situ hybridization to regions in chromosomes 6 (which is thought to harbour a tumour suppressor gene) and 17, respectively. The expression pattern of WTAP was investigated and shown to be ubiquitous, perhaps reflecting a housekeeping role. WTAP is a nuclear protein, which like WT1 localizes throughout the nucleoplasm as well as in speckles and partially co-localizes with splicing factors. Although the significance of this interaction is not yet known, WTAP promises to be an interesting WT1-binding partner.
Collapse
Affiliation(s)
- N A Little
- MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | | | | |
Collapse
|
96
|
Abstract
The last few years have provided dramatic breakthroughs in understanding the genetic factors involved in Wilms' tumorigenesis and normal kidney development. The implications of these findings for the clinical management of children with Wilms' tumor are only now becoming apparent. Over 80% of patients with Wilms' tumor can be cured using contemporary multimodality therapy. As a consequence, the current NWTSG is attempting to intensify treatment for patients with poor prognostic features while decreasing therapy, and thereby adverse late effects, for patients with favorable prognosticators.
Collapse
Affiliation(s)
- M J Coppes
- Southern Alberta Children's Cancer Program, University of Calgary, Alberta, Canada.
| | | |
Collapse
|
97
|
Spinsanti P, de Grazia U, Faggioni A, Frati L, Calogero A, Ragona G. Wilms' tumor gene expression by normal and malignant human B lymphocytes. Leuk Lymphoma 2000; 38:611-9. [PMID: 10953983 DOI: 10.3109/10428190009059281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Very little is known about Wilms' tumor gene (WT1) expression in B cells and its importance for growth regulation and differentiation. We have investigated WT1 expression in fresh B lymphocytes and in a panel of B-cell lines of normal and malignant origin, including both Epstein-Barr virus (EBV) genome negative and EBV carrying cell lines. WT1 is constitutively activated in all lymphoblastoid cell lines (LCL) derived from EBV immortalization of lymphocytes from normal donors in vitro. These cell lines are distinguished for the presence of activated B-cell markers and an unrestricted expression of viral latent genes. In contrast, WT1 expression is abrogated in normal B lymphocytes and in all Burkitt tumor derived cell lines, irrespective of the EBV genome carrying status and their phenotype pattern. A single step RT-PCR for simultaneous detection of the four spliced transcript isoforms has been applied to confirm their expression. Analysis of variant relative proportions suggested the maintenance of a balanced expression of the isoforms in LCL, as reported in non tumorous tissues. These data, together with the evidence that the replication in vitro of lymphoblastoid cells is not affected by WT1 activation following viral immortalization, support the hypothesis that gene inactivation, in addition to disrupted alternate splicing, may play a role in growth control derangements.
Collapse
Affiliation(s)
- P Spinsanti
- Dipartimento di Medicina Sperimentale e Patologia, Università di Roma La Sapienza, Rome, Italy
| | | | | | | | | | | |
Collapse
|
98
|
Affiliation(s)
- K J Turner
- Imperial Cancer Research Fund, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
99
|
Abstract
Mutations in the WT1 gene were anticipated to explain the genetic basis of the childhood kidney cancer, Wilms tumour (WT). Six years on, we review 100 reports of intragenic WT1 mutations and examine the accompanying clinical phenotypes. While only 5% of sporadic Wilms' tumours have intragenic WT1 mutations, > 90% of patients with the Denys-Drash syndrome (renal nephropathy, gonadal anomaly, predisposition to WT) carry constitutional intragenic WT1 mutations. WT1 mutations have also been reported in juvenile granulosa cell tumour, non-asbestos related mesothelioma, desmoplastic small round cell tumour and, most recently, acute myeloid leukemia.
Collapse
Affiliation(s)
- M Little
- Centre for Molecular and Cellular Biology, University of Queensland, St. Lucia, Australia
| | | |
Collapse
|
100
|
Hosono S, Gross I, English MA, Hajra KM, Fearon ER, Licht JD. E-cadherin is a WT1 target gene. J Biol Chem 2000; 275:10943-53. [PMID: 10753894 DOI: 10.1074/jbc.275.15.10943] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The WT1 tumor suppressor gene encodes a transcription factor that can activate and repress gene expression. Transcriptional targets relevant for the growth suppression functions of WT1 are poorly understood. We found that mesenchymal NIH 3T3 fibroblasts stably expressing WT1 exhibit growth suppression and features of epithelial differentiation including up-regulation of E-cadherin mRNA. Acute expression of WT1 in NIH 3T3 fibroblasts after retroviral infection induced murine E-cadherin expression. In transient transfection experiments, the human and murine E-cadherin promoters were activated by co-expression of WT1. E-cadherin promoter activity was increased in cells overexpressing WT1 and was blocked by a dominant negative form of WT1. WT1 activated the murine E-cadherin promoter through a conserved GC-rich sequence similar to an EGR-1 binding site as well as through a CAAT box sequence. WT1 produced in vitro or derived from nuclear extracts bound to the WT1-response element within the murine E-cadherin promoter, but not the CAAT box. E-cadherin, a gene important in epithelial differentiation and neoplastic transformation, represents a downstream target gene that links the roles of the WT1 in differentiation and growth control.
Collapse
Affiliation(s)
- S Hosono
- Derald H. Ruttenberg Cancer Center, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | |
Collapse
|