1
|
Sekimoto A, Takaso Y, Saruyama H, Ookawa M, Yamamoto M, Toyohara T, Saigusa D, Fukuuchi T, Otsuka M, Fushiki Y, Yamakoshi S, Tanaka K, Ikeda T, Tanaka T, Takahashi N, Mishima E, Sato E. Impacts of low birthweight on kidney development and intergenerational growth of the offspring. iScience 2024; 27:111159. [PMID: 39524353 PMCID: PMC11546680 DOI: 10.1016/j.isci.2024.111159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/10/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Low birthweight (LBW) increases the risk of adult-onset diseases, including kidney diseases, with intergenerational consequences; however, the underlying mechanisms and effective interventions are unclear. To examine the cross-generational effects of LBW, we established an LBW mouse model through reduced uterine perfusion pressure (RUPP) and investigated the therapeutic potential of tadalafil, a phosphodiesterase 5 inhibitor, on LBW-associated consequences. RUPP-pups (R1) had lower fetal and birth weights, delayed renal development, and fewer glomeruli than Sham-pups. In adulthood, R1 mice exhibited persistently fewer glomeruli and elevated blood pressure, while Tadalafil-R1 mice showed reduced hypertension in both sexes and improved renal pathological changes in males. Additionally, pregnant R1 mice displayed inadequate gestational liver hypertrophy, impaired hepatic purine metabolism, and diminished placental angiogenesis, resulting in fetal growth restriction in the subsequent generation. These findings underscore the lasting impact of LBW on adult health and future generations and suggest tadalafil's potential to mitigate LBW-associated risks.
Collapse
Affiliation(s)
- Akiyo Sekimoto
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
- Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoko Takaso
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Haruka Saruyama
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Masataka Ookawa
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Mari Yamamoto
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Takafumi Toyohara
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 983-8536, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Tomoko Fukuuchi
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Mayu Otsuka
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Yui Fushiki
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Seiko Yamakoshi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
| | - Kayo Tanaka
- Department of Obstetrics and Gynecology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Tetsuhiro Tanaka
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 983-8536, Japan
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 983-8536, Japan
| | - Eikan Mishima
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 983-8536, Japan
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai 980-8578, Japan
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 983-8536, Japan
| |
Collapse
|
2
|
Fu W, Deng L, Yan X, Hua RX, Zhang J, Zhou H, Deng C, Li S, Cheng J, Ruan J, He J, Liu G. LMO family gene polymorphisms and Wilms tumor susceptibility in Chinese children: a five-center case-control study. BMC Cancer 2024; 24:772. [PMID: 38937681 PMCID: PMC11209997 DOI: 10.1186/s12885-024-12557-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/24/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Wilms tumor is the most prevalent embryonal kidney malignancy in children worldwide. Previous genome-wide association study (GWAS) identified that LIM domain only 1 (LMO1) gene polymorphisms affected the susceptibility to develop certain tumor types. Apart from LMO1, the LMO gene family members also include LMO2-4, each of which has oncogenic potential. METHODS We conducted this five-center case‒control study to assess the correlations between single nucleotide polymorphisms in LMO family genes and Wilms tumor susceptibility. Odds ratios and 95% confidence intervals were calculated to evaluate the strength of the association. RESULTS We found LMO1 rs2168101 G > T and rs11603024 C > T as well as LMO2 rs7933499 G > A were significantly associated with Wilms tumor risk. Stratified analysis demonstrated a protective role of rs2168101 GT/TT genotypes against Wilms tumor in the subgroups of age ≤ 18 months, males and clinical stages I/II compared to the rs2168101 GG genotype. Nevertheless, carriers with the rs11603024 TT genotype were more likely to have an increased risk of Wilms tumor than those with rs11603024 CC/CT genotypes in age > 18 months. And the rs11603024 was identified as a protective polymorphism for reducing the risk of Wilms tumor in the sex- and gender- subgroup. Likewise, carriers with the rs7933499 GA/AA genotypes were at significantly elevated risk of Wilms tumor in age ≤ 18 months and clinical stages I/II. CONCLUSION Overall, our study identified the importance of LMO family gene polymorphisms on Wilms tumor susceptibility in Chinese children. Further investigations are needed to validate our conclusions.
Collapse
Affiliation(s)
- Wen Fu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China.
| | - Linqing Deng
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Xiaosong Yan
- Department of Pathology, The Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710003, China
| | - Rui-Xi Hua
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Jiao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Haixia Zhou
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Changmi Deng
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, Shannxi, 030013, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jichen Ruan
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China.
| | - Guochang Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| |
Collapse
|
3
|
Duhme C, Busch M, Heine E, de Torres C, Mora J, Royer-Pokora B. WT1-Mutant Wilms Tumor Progression Is Associated With Diverting Clonal Mutations of CTNNB1. J Pediatr Hematol Oncol 2021; 43:e180-e183. [PMID: 31876779 DOI: 10.1097/mph.0000000000001697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 11/15/2019] [Indexed: 11/26/2022]
Abstract
WT1-mutant Wilms tumors exhibit a high rate of concomitant CTNNB1 mutations, associated with activated Wnt signaling. Here, we show by laser and manual microdissection of different histologic cell types from 6 WT1-mutant tumor samples that 1 patient's tumor can contain up to 4 distinct mutations in CTNNB1 and/or WTX. Consecutive sections may also harbor different CTNNB1 mutations. The variability of activating CTNNB1 mutations demonstrates the multifocal nature of WT1-mutant Wilms tumors. As multiple independent tumors can occur in patients with constitutional WT1 mutations, they need to be surveyed more closely for tumor development.
Collapse
Affiliation(s)
- Constanze Duhme
- Institute of Human Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Maike Busch
- Institute of Human Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Eva Heine
- Institute of Human Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Carmen de Torres
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Deu, Barcelona, Spain
| | - Jaume Mora
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Deu, Barcelona, Spain
| | | |
Collapse
|
4
|
Drake KA, Chaney CP, Das A, Roy P, Kwartler CS, Rakheja D, Carroll TJ. Stromal β-catenin activation impacts nephron progenitor differentiation in the developing kidney and may contribute to Wilms tumor. Development 2020; 147:dev189597. [PMID: 32541007 PMCID: PMC7406317 DOI: 10.1242/dev.189597] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/02/2020] [Indexed: 02/03/2023]
Abstract
Wilms' tumor (WT) morphologically resembles the embryonic kidney, consisting of blastema, epithelial and stromal components, suggesting tumors arise from the dysregulation of normal development. β-Catenin activation is observed in a significant proportion of WTs; however, much remains to be understood about how it contributes to tumorigenesis. Although activating β-catenin mutations are observed in both blastema and stromal components of WT, current models assume that activation in the blastemal lineage is causal. Paradoxically, studies performed in mice suggest that activation of β-catenin in the nephrogenic lineage results in loss of nephron progenitor cell (NPC) renewal, a phenotype opposite to WT. Here, we show that activation of β-catenin in the stromal lineage non-autonomously prevents the differentiation of NPCs. Comparisons of the transcriptomes of kidneys expressing an activated allele of β-catenin in the stromal or nephron progenitor cells reveals that human WT more closely resembles the stromal-lineage mutants. These findings suggest that stromal β-catenin activation results in histological and molecular features of human WT, providing insights into how alterations in the stromal microenvironment may play an active role in tumorigenesis.
Collapse
Affiliation(s)
- Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher P Chaney
- Department of Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amrita Das
- Amgen, Inc., San Francisco, CA 94080, USA
| | - Priti Roy
- Department of Ophthalmology and Visual Sciences, Chicago, IL 60612, USA
| | - Callie S Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas J Carroll
- Department of Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
5
|
Chong WC, Cain JE. Lessons learned from the developmental origins of childhood renal cancer. Anat Rec (Hoboken) 2019; 303:2561-2577. [DOI: 10.1002/ar.24315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/14/2019] [Accepted: 10/05/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Wai Chin Chong
- Centre for Cancer ResearchHudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health SciencesMonash University Clayton Victoria Australia
| | - Jason E. Cain
- Centre for Cancer ResearchHudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health SciencesMonash University Clayton Victoria Australia
| |
Collapse
|
6
|
Marakhonov AV, Vasilyeva TA, Voskresenskaya AA, Sukhanova NV, Kadyshev VV, Kutsev SI, Zinchenko RA. LMO2 gene deletions significantly worsen the prognosis of Wilms’ tumor development in patients with WAGR syndrome. Hum Mol Genet 2019; 28:3323-3326. [DOI: 10.1093/hmg/ddz168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/23/2022] Open
Abstract
AbstractWAGR syndrome (OMIM #194072) is a rare genetic disorder that consists of development of Wilms’ tumor (nephroblastoma), aniridia, genitourinary anomalies and intellectual disability (mental retardation). It is associated with WAGR-region deletions in the 11p13 chromosome region. Our previous study of congenital aniridia patients revealed a noticeable number of aniridia patients with WAGR-region deletions but without Wilms’ tumor in their medical history. We assessed the involvement of other neighboring genes from affected chromosome regions in the patients with and without Wilms’ tumor. Reliable confidence was obtained for the LMO2 gene, which is significantly more often deleted in patients with nephroblastoma. Thus, our study presents genetic evidence that the development of Wilms tumors in WAGR syndrome patients should be attributed to the deletion of WT1 and LMO2 rather than WT1 only.
Collapse
Affiliation(s)
- Andrey V Marakhonov
- Research Center for Medical Genetics, Moscow 115522, Russia
- Far Eastern Federal University, Vladivostok 690090, Russia
| | | | - Anna A Voskresenskaya
- Cheboksary Branch of the S. Fyodorov Eye Microsurgery Federal State Institution, Cheboksary 428028, Russia
| | - Natella V Sukhanova
- National Medical Research Center for Children’s Health, Moscow 119296, Russia
| | | | - Sergey I Kutsev
- Research Center for Medical Genetics, Moscow 115522, Russia
- Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Rena A Zinchenko
- Research Center for Medical Genetics, Moscow 115522, Russia
- Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
7
|
Murphy AJ, Davidoff AM. Bilateral Wilms Tumor: A Surgical Perspective. CHILDREN-BASEL 2018; 5:children5100134. [PMID: 30250006 PMCID: PMC6210093 DOI: 10.3390/children5100134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/13/2018] [Accepted: 09/21/2018] [Indexed: 12/27/2022]
Abstract
Historically, the management of bilateral Wilms tumor (BWT) was non-standardized and suffered from instances of prolonged chemotherapy and inconsistent surgical management which resulted in suboptimal renal and oncologic outcomes. Because of the risk of end-stage renal disease associated with the management of BWT, neoadjuvant chemotherapy and nephron-sparing surgery have been adopted as the guiding management principles. This management strategy balances acceptable oncologic outcomes against the risk of end-stage renal disease. A recent multi-institutional Children’s Oncology Group study (AREN0534) has confirmed the benefits of standardized 3-drug neoadjuvant chemotherapy and the utilization of nephron-sparing surgery in BWT patients; however, less than 50% of patients underwent bilateral nephron-sparing surgery. The coordination of neoadjuvant chemotherapy and the timing and implementation of bilateral nephron-sparing surgery are features of BWT management that require collaboration between oncologists and surgeons. This review discusses the surgical management strategy in the context of BWT disease biology, with an emphasis on timepoints during therapy at which surgical decision making can greatly impact this disease and minimize long-term toxicities.
Collapse
Affiliation(s)
- Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38105, USA.
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38105, USA.
| |
Collapse
|
8
|
Polosukhina D, Love HD, Moses HL, Lee E, Zent R, Clark PE. Pharmacologic Inhibition of β-Catenin With Pyrvinium Inhibits Murine and Human Models of Wilms Tumor. Oncol Res 2017; 25:1653-1664. [PMID: 28695795 PMCID: PMC5670010 DOI: 10.3727/096504017x14992942781895] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Wilms tumor (WT) is the most common renal malignancy in children and the fourth most common pediatric solid malignancy in the US. Although the mechanisms underlying the WT biology are complex, these tumors most often demonstrate activation of the canonical Wnt/β-catenin pathway. We and others have shown that constitutive activation of β-catenin restricted to the renal epithelium is sufficient to induce primitive renal epithelial tumors, which resemble human WT. Here we demonstrate that pharmacologic inhibition of β-catenin gene transcription with pyrvinium inhibits tumor growth and metastatic progression in a murine model of WT. Cellular invasion is significantly inhibited in both murine WT-like and human WT cells and is accompanied by downregulation of the oncogenes Myc and Birc5 (survivin). Our studies provide proof of the concept that the canonical Wnt/β-catenin pathway may be a novel therapeutic target in the management of WT.
Collapse
|
9
|
Alexandrescu S, Akhavanfard S, Harris MH, Vargas SO. Clinical, Pathologic, and Genetic Features of Wilms Tumors With WTX Gene Mutation. Pediatr Dev Pathol 2017; 20:105-111. [PMID: 28326956 DOI: 10.1177/1093526616683881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Clinical and pathologic features of patients with WTX-mutated Wilms tumor (WT) have not been studied in detail. We characterize the clinical and pathologic findings in WT with WTX abnormalities and provide comparison with WT without WTX mutation. Clinical, gross, and microscopic features in 35 patients with WT were examined. Karyotype was examined in a subset of cases. All cases had been previously analyzed for WTX, WT1, and CTNNB1 aberrations via array comparative genomic hybridization; OncoMap 4 high throughput genotyping was performed on 18 cases. Eleven tumors had WTX abnormality. No significant differences were identified between patients with mutated versus nonmutated WTX with respect to gender (45% versus 33% male), age (mean 3.9 versus 4.1 years), tumor size (mean 12.7 cm versus 12.8 cm), anaplasia (9% versus 12%), rhabdomyoblastic differentiation (18% versus 8%), cartilage differentiation (9% versus 4%), mucinous epithelial differentiation (9% versus 4%), nephrogenic rests (28% versus 21%), or relapse rate (11% versus 25%). Mutations in KRAS, MYC, and PIK3R1 were restricted to WTX-mutated WT, mutations in AKT, CKDN2A, EFGR, HRAS, MET, and RET were restricted to WT without WTX mutation, and mutations in BRAF, CTTNB1, NRAS, PDGFRA, and STK11 were seen in both groups. Our study revealed no clinical or pathologic distinctions between WT with and without WTX abnormality. This similarity lends support to the concept of a common tumorigenic pathway between WT with aberrant WTX and those without.
Collapse
Affiliation(s)
- Sanda Alexandrescu
- 1 Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA.,2 Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Akhavanfard
- 3 Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marian H Harris
- 1 Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA.,2 Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara O Vargas
- 1 Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA.,2 Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Deng C, Dai R, Li X, Liu F. Genetic variation frequencies in Wilms' tumor: A meta-analysis and systematic review. Cancer Sci 2016; 107:690-9. [PMID: 26892980 PMCID: PMC4970837 DOI: 10.1111/cas.12910] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022] Open
Abstract
Over the last few decades, numerous biomarkers in Wilms' tumor have been confirmed and shown variations in prevalence. Most of these studies were based on small sample sizes. We carried out a meta-analysis of the research published from 1992 to 2015 to obtain more precise and comprehensive outcomes for genetic tests. In the present study, 70 out of 5175 published reports were eligible for the meta-analysis, which was carried out using Stata 12.0 software. Pooled prevalence for gene mutations WT1, WTX, CTNNB1, TP53, MYCN, DROSHA, and DGCR8 was 0.141 (0.104, 0.178), 0.147 (0.110, 0.184), 0.140 (0.100, 0.190), 0.410 (0.214, 0.605), 0.071 (0.041, 0.100), 0.082 (0.048, 0.116), and 0.036 (0.026, 0.046), respectively. Pooled prevalence of loss of heterozygosity at 1p, 11p, 11q, 16q, and 22q was 0.109 (0.084, 0.133), 0.334 (0.295, 0.373), 0.199 (0.146, 0.252), 0.151 (0.129, 0.172), and 0.148 (0.108, 0.189), respectively. Pooled prevalence of 1q and chromosome 12 gain was 0.218 (0.161, 0.275) and 0.273 (0.195, 0.350), respectively. The limited prevalence of currently known genetic alterations in Wilms' tumors indicates that significant drivers of initiation and progression remain to be discovered. Subgroup analyses indicated that ethnicity may be one of the sources of heterogeneity. However, in meta-regression analyses, no study-level characteristics of indicators were found to be significant. In addition, the findings of our sensitivity analysis and possible publication bias remind us to interpret results with caution.
Collapse
Affiliation(s)
- Changkai Deng
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China.,Chengdu Women and Children's Central Hospital, Chengdu, China
| | - Rong Dai
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Xuliang Li
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Feng Liu
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| |
Collapse
|
11
|
Adli M, Parlak M, Li Y, El-Dahr SS. Epigenetic States of nephron progenitors and epithelial differentiation. J Cell Biochem 2016; 116:893-902. [PMID: 25560433 DOI: 10.1002/jcb.25048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 12/26/2022]
Abstract
In mammals, formation of new nephrons ends perinatally due to consumption of mesenchymal progenitor cells. Premature depletion of progenitors due to prematurity or postnatal loss of nephrons due to injury causes chronic kidney disease and hypertension. Intensive efforts are currently invested in designing regenerative strategies to form new nephron progenitors from pluripotent cells, which upon further differentiation provide a potential source of new nephrons. To know if reprogramed renal cells can maintain their identity and fate requires knowledge of the epigenetic states of native nephron progenitors and their progeny. In this article, we summarize current knowledge and gaps in the epigenomic landscape of the developing kidney. We now know that Pax2/PTIP/H3K4 methyltransferase activity provides the initial epigenetic specification signal to the metanephric mesenchyme. During nephrogenesis, the cap mesenchyme housing nephron progenitors is enriched in bivalent chromatin marks; as tubulogenesis proceeds, the tubular epithelium acquires H3K79me2. The latter mark is uniquely induced during epithelial differentiation. Analysis of histone landscapes in clonal metanephric mesenchyme cell lines and in Wilms tumor and normal fetal kidney has revealed that promoters of poised nephrogenesis genes carry bivalent histone signatures in progenitors. Differentiation or stimulation of Wnt signaling promotes resolution of bivalency; this does not occur in Wilms tumor cells consistent with their developmental arrest. The use of small cell number ChIP-Seq should facilitate the characterization of the chromatin landscape of the metanephric mesenchyme and various nephron compartments during nephrogenesis. Only then we will know if stem and somatic cell reprogramming into kidney progenitors recapitulates normal development.
Collapse
Affiliation(s)
- Mazhar Adli
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virgina
| | | | | | | |
Collapse
|
12
|
Abstract
Wilms' tumor, or nephroblastoma, is the most common pediatric renal cancer. The tumors morphologically resemble embryonic kidneys with a disrupted architecture and are associated with undifferentiated metanephric precursors. Here, we discuss genetic and epigenetic findings in Wilms' tumor in the context of renal development. Many of the genes implicated in Wilms' tumorigenesis are involved in the control of nephron progenitors or the microRNA (miRNA) processing pathway. Whereas the first group of genes has been extensively studied in normal development, the second finding suggests important roles for miRNAs in general-and specific miRNAs in particular-in normal kidney development that still await further analysis. The recent identification of Wilms' tumor cancer stem cells could provide a framework to integrate these pathways and translate them into new or improved therapeutic interventions.
Collapse
Affiliation(s)
- Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom;
| | - Kathy Pritchard-Jones
- UCL Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Jocelyn Charlton
- UCL Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| |
Collapse
|
13
|
Yoshizawa K, Kinoshita Y, Emoto Y, Kimura A, Uehara N, Yuri T, Shikata N, Tsubura A. N -Methyl- N -nitrosourea-induced Renal Tumors in Rats: Immunohistochemical Comparison to Human Wilms Tumors. J Toxicol Pathol 2013; 26:141-8. [PMID: 23914056 PMCID: PMC3695336 DOI: 10.1293/tox.26.141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/11/2013] [Indexed: 12/27/2022] Open
Abstract
N-Methyl-N-nitrosourea (MNU)-induced renal tumors in rats and Wilms tumors in humans were compared. Renal mesenchymal tumors (RMTs) and nephroblastomas (blastemal and epithelial components) in female Lewis rats treated with a single intraperitoneal injection of 50 mg/kg MNU at birth and Wilms tumors (blastemal, epithelial and mesenchymal components) in humans were analyzed for the expression of pancytokeratin (CK), vimentin, p63, α-smooth muscle actin (SMA), desmin, S-100, CD57, CD117/c-kit, Wilms tumor 1 protein (WT1) and β-catenin. The mesenchymal components of rat RMTs and human Wilms tumors expressed vimentin, SMA and β-catenin. The blastemal components of rat nephroblastomas and human Wilms tumors expressed vimentin, CD117/c-kit and β-catenin. The epithelial components of rat nephroblastomas and human Wilms tumors expressed vimentin and β-catenin. WT1 was expressed in different cellular components of rat tumors as compared with human Wilms tumors; the expression was seen in mesenchymal tumors and blastemal components of nephroblastomas in rats and epithelial components in human Wilms tumors. CK, p63 and CD57 were not expressed in rat RMTs or nephroblastomas, while CK and WT1 were expressed in epithelial components and CD57 was expressed in blastemal and epithelial components of human Wilms tumors. Rat and human tumors were universally negative for the expression of desmin and S-100. The immunohistochemical characteristics of rat renal tumors and human Wilms tumors may provide valuable information on the differences in renal oncogenesis and biology between the two species.
Collapse
Affiliation(s)
- Katsuhiko Yoshizawa
- Department of Pathology II, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Sigamani E, Wari MN, Iyer VK, Agarwala S, Sharma A, Bakhshi S, Dinda A. Loss of heterozygosity at 11p13 and 11p15 in Wilms tumor: a study of 22 cases from India. Pediatr Surg Int 2013; 29:223-7. [PMID: 23292539 DOI: 10.1007/s00383-012-3254-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2012] [Indexed: 11/29/2022]
Abstract
PURPOSE 11p13 and 11p15 loss of heterozygosity (LOH) in Wilms tumor (WT), the commonest molecular pathogenetic event in WT, shows variation in different parts of the world. The present study looked for the presence of 11p13 and 11p15 LOH as well as nephrogenic rests in WT occurring in India. METHOD Twenty-two cases of WT were subjected to thorough pathological examination for presence of nephrogenic rests. Fresh frozen tissue was evaluated for LOH at 11p13 and 11p15, using PCR for microsatellite markers. RESULTS Among twenty-two consecutive cases of WT, 20 were unilateral and 2 were bilateral. 6/22 showed LOH at 11p13 (27.7 %) and 1/22 showed LOH at 11p15 (4.54 %). 2/22 cases showed presence of nephrogenic rests. One of the cases with LOH at 11p13 had intralobar nephrogenic rest in the adjacent kidney. One specimen had perilobar nephrogenic rest in the adjacent kidney but did not show LOH for either 11p13 or 11p15 in the tumor. CONCLUSION LOH at 11p13 is seen in 27.27 % of WT in India, which is similar to reports in the English language literature. LOH at 11p15 was seen in 4.54 % of WT, which is lower than that reported from Western subjects.
Collapse
Affiliation(s)
- Elanthenral Sigamani
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | | | | | | | | | | |
Collapse
|
15
|
Multicentre study of Wilm’s tumours treated by different therapeutic strategies in two different countries. ANNALS OF PEDIATRIC SURGERY 2013. [DOI: 10.1097/01.xps.0000422612.69446.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
16
|
James RG, Davidson KC, Bosch KA, Biechele TL, Robin NC, Taylor RJ, Major MB, Camp ND, Fowler K, Martins TJ, Moon RT. WIKI4, a novel inhibitor of tankyrase and Wnt/ß-catenin signaling. PLoS One 2012; 7:e50457. [PMID: 23227175 PMCID: PMC3515623 DOI: 10.1371/journal.pone.0050457] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/22/2012] [Indexed: 12/21/2022] Open
Abstract
The Wnt/ß-catenin signaling pathway controls important cellular events during development and often contributes to disease when dysregulated. Using high throughput screening we have identified a new small molecule inhibitor of Wnt/ß-catenin signaling, WIKI4. WIKI4 inhibits expression of ß-catenin target genes and cellular responses to Wnt/ß-catenin signaling in cancer cell lines as well as in human embryonic stem cells. Furthermore, we demonstrate that WIKI4 mediates its effects on Wnt/ß-catenin signaling by inhibiting the enzymatic activity of TNKS2, a regulator of AXIN ubiquitylation and degradation. While TNKS has previously been shown to be the target of small molecule inhibitors of Wnt/ß-catenin signaling, WIKI4 is structurally distinct from previously identified TNKS inhibitors.
Collapse
Affiliation(s)
- Richard G. James
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kathryn C. Davidson
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Katherine A. Bosch
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Travis L. Biechele
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nicholas C. Robin
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Russell J. Taylor
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Michael B. Major
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | - Nathan D. Camp
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kerry Fowler
- KWF Consulting, Seattle, Washington, United States of America
| | - Timothy J. Martins
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Quellos High Throughput Screening Core, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Randall T. Moon
- Department of Pharmacology, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
17
|
Huss S, Nehles J, Binot E, Wardelmann E, Mittler J, Kleine MA, Künstlinger H, Hartmann W, Hohenberger P, Merkelbach-Bruse S, Buettner R, Schildhaus HU. β-Catenin (CTNNB1) mutations and clinicopathological features of mesenteric desmoid-type fibromatosis. Histopathology 2012; 62:294-304. [DOI: 10.1111/j.1365-2559.2012.04355.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Murphy AJ, Pierce J, de Caestecker C, Taylor C, Anderson JR, Perantoni AO, de Caestecker MP, Lovvorn HN. SIX2 and CITED1, markers of nephronic progenitor self-renewal, remain active in primitive elements of Wilms' tumor. J Pediatr Surg 2012; 47:1239-49. [PMID: 22703800 PMCID: PMC3377935 DOI: 10.1016/j.jpedsurg.2012.03.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 12/15/2022]
Abstract
PURPOSE SIX2 and CITED1 are transcriptional regulators that specify self-renewing nephronic progenitor cells of the embryonic kidney. We hypothesized that SIX2, which promotes and maintains this stem cell population, and CITED1 remain active in Wilms' tumor (WT). METHODS To evaluate expression domains and the pathogenic significance of SIX2 and CITED1 across WT, the Children's Oncology Group provided 40 WT specimens of stages I to IV (n = 10 per stage), which were enriched for unfavorable histology (n = 20) and treatment failure (relapse or death, n = 20). SIX2 and CITED1 protein expression was evaluated qualitatively (immunohistochemistry) and quantitatively (Western blot, or WB). Gene transcription was estimated using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS SIX2 was visualized by immunohistochemistry in 36 (94.7%) of 38 specimens. Protein and messenger RNA expression of SIX2 were quantitatively similar across all stages of disease (P = .48 WB; P = 0.38 qPCR), in favorable or unfavorable histology (P = 0.51 WB; P = 0.58 qPCR), and in treatment failure or success (P = 0.86 WB; P = 0.49 qPCR). Although CITED1 expression paralleled SIX2 qualitatively, no quantitative correlation between SIX2 and CITED1 expression was observed (Spearman correlation coefficient, 0.28; P = 0.08). As in the fetal kidney, overlapping, but also distinct, WT cellular expression domains were observed between SIX2 and CITED1. CONCLUSION SIX2 and CITED1 remain active across all disease characteristics of WT. Activity of these genes in WT potentially identifies a population of self-renewing cancer cells that exhibit an embryonic, stemlike phenotype. Taken together, these transcriptional regulators may be fundamental to WT cellular self-renewal and may represent targets for novel therapies that promote terminal differentiation.
Collapse
Affiliation(s)
- Andrew J. Murphy
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN
| | - Janene Pierce
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN
| | - Christian de Caestecker
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN
| | - Chase Taylor
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN
| | - James R. Anderson
- Department of Biostatistics, Children’s Oncology Group and University of Nebraska Medical Center, Omaha, NE
| | - Alan O. Perantoni
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Mark P. de Caestecker
- Departments of Medicine, Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Harold N. Lovvorn
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN
| |
Collapse
|
19
|
Murphy AJ, Axt JR, de Caestecker C, Pierce J, Correa H, Seeley EH, Caprioli RM, Newton MW, de Caestecker MP, Lovvorn HN. Molecular characterization of Wilms' tumor from a resource-constrained region of sub-Saharan Africa. Int J Cancer 2012; 131:E983-94. [PMID: 22437966 DOI: 10.1002/ijc.27544] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 03/05/2012] [Indexed: 11/10/2022]
Abstract
Sub-Saharan African children have an increased incidence of Wilms' tumor (WT) and experience alarmingly poor outcomes. Although these outcomes are largely due to inadequate therapy, we hypothesized that WT from this region exhibits features of biological aggressiveness that may warrant broader implementation of high-risk therapeutic protocols. We evaluated 15 Kenyan WT (KWT) for features of aggressive disease (blastemal predominance and Ki67/cellular proliferation) and treatment resistance (anaplasia and p53 immunopositivity). To explore the additional biological features of KWT, we determined the mutational status of the CTNNB1/β-catenin and WT1 genes and performed immunostaining for markers of Wnt pathway activation (β-catenin) and nephronic progenitor cell self-renewal (WT1, CITED1 and SIX2). We characterized the proteome of KWT using imaging mass spectrometry (IMS). The results were compared to histology- and age-matched North American WT (NAWT) controls. For patients with KWT, blastemal predominance was noted in 53.3% and anaplasia in 13%. We detected increased loss to follow-up (p = 0.028), disease relapse (p = 0.044), mortality (p = 0.001) and nuclear unrest (p = 0.001) in patients with KWT compared to controls. KWT and NAWT showed similar Ki67/cellular proliferation. We detected an increased proportion of epithelial nuclear β-catenin in KWT (p = 0.013). All 15 KWT specimens were found to harbor wild-type CTNNB1/β-catenin, and one contained a WT1 nonsense mutation. WT1 was detected by immunostaining in 100% of KWT, CITED1 in 80% and SIX2 in 80%. IMS revealed a molecular signature unique to KWT that was distinct from NAWT. The African WT specimens appear to express markers of adverse clinical behavior and treatment resistance and may require alternative therapies or implementation of high-risk treatment protocols.
Collapse
Affiliation(s)
- Andrew J Murphy
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Activation of beta-catenin is a late event in the pathogenesis of nephroblastomas and rarely correlated with genetic changes of the APC gene. Pathology 2012; 43:702-6. [PMID: 22081130 DOI: 10.1097/pat.0b013e32834bf65c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIMS Activation of β-catenin has been identified as a possible mechanism for the development of nephroblastomas. In our study we investigated whether this activation occurs already in precursor lesions of nephroblastomas, called nephrogenic rests (NRs). Inactivation of the adenomatous polyposis coli (APC) protein is an important regulatory mechanism of activating β-catenin. We clarified the role of APC by assessing loss of heterozygosity (LOH) and possible mutations within the genomic region. METHODS Activation of β-catenin was examined by immunohistochemistry identifying nuclear translocation. Two polymorphic loci of the APC gene were investigated for LOH and sequence analysis was performed for the mutation cluster region of the APC gene on formalin fixed, paraffin embedded samples. RESULTS Four of the 18 nephroblastomas available for immunohistochemistry exhibited nuclear staining of β-catenin, but none of the NRs. Analysis of LOH revealed 14 homozygous samples, 10 heterozygous tumours and six tumours exhibiting LOH of the APC gene. One blastema-type nephroblastoma showed nuclear localisation of β-catenin in conjunction with LOH of the APC gene. Analysis of 12 nephroblastomas revealed no sequence aberration. CONCLUSION Our results indicate that nuclear activation of β-catenin is a late event in the tumorigenesis of nephroblastomas coinciding in some tumours with LOH of the APC gene.
Collapse
|
21
|
Maschietto M, Trapé AP, Piccoli FS, Ricca TI, Dias AAM, Coudry RA, Galante PA, Torres C, Fahhan L, Lourenço S, Grundy PE, de Camargo B, de Souza S, Neves EJ, Soares FA, Brentani H, Carraro DM. Temporal blastemal cell gene expression analysis in the kidney reveals new Wnt and related signaling pathway genes to be essential for Wilms' tumor onset. Cell Death Dis 2011; 2:e224. [PMID: 22048167 PMCID: PMC3223691 DOI: 10.1038/cddis.2011.105] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Wilms' tumors (WTs) originate from metanephric blastema cells that are unable to complete differentiation, resulting in triphasic tumors composed of epithelial, stromal and blastemal cells, with the latter harboring molecular characteristics similar to those of the earliest kidney development stages. Precise regulation of Wnt and related signaling pathways has been shown to be crucial for correct kidney differentiation. In this study, the gene expression profile of Wnt and related pathways was assessed in laser-microdissected blastemal cells in WTs and differentiated kidneys, in human and in four temporal kidney differentiation stages (i.e. E15.5, E17.5, P1.5 and P7.5) in mice, using an orthologous cDNA microarray platform. A signaling pathway-based gene signature was shared between cells of WT and of earliest kidney differentiation stages, revealing genes involved in the interruption of blastemal cell differentiation in WT. Reverse transcription-quantitative PCR showed high robustness of the microarray data demonstrating 75 and 56% agreement in the initial and independent sample sets, respectively. The protein expression of CRABP2, IGF2, GRK7, TESK1, HDGF, WNT5B, FZD2 and TIMP3 was characterized in WTs and in a panel of human fetal kidneys displaying remarkable aspects of differentiation, which was recapitulated in the tumor. Taken together, this study reveals new genes candidate for triggering WT onset and for therapeutic treatment targets.
Collapse
Affiliation(s)
- M Maschietto
- Laboratory of Genomics and Molecular Biology, CIPE-AC Camargo Hospital, São Paulo, SP, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Clark PE, Polosukhina D, Love H, Correa H, Coffin C, Perlman EJ, de Caestecker M, Moses HL, Zent R. β-Catenin and K-RAS synergize to form primitive renal epithelial tumors with features of epithelial Wilms' tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:3045-55. [PMID: 21983638 DOI: 10.1016/j.ajpath.2011.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/27/2011] [Accepted: 08/10/2011] [Indexed: 12/19/2022]
Abstract
Wilms' tumor (WT) is the most common childhood renal cancer. Although mutations in known tumor-associated genes (WT1, WTX, and CATNB) occur only in a third of tumors, many tumors show evidence of activated β-catenin-dependent Wnt signaling, but the molecular mechanism by which this occurs is unknown. A key obstacle to understanding the pathogenesis of WT is the paucity of mouse models that recapitulate its features in humans. Herein, we describe a transgenic mouse model of primitive renal epithelial neoplasms that have high penetrance and mimic the epithelial component of human WT. Introduction of a stabilizing β-catenin mutation restricted to the kidney is sufficient to induce primitive renal epithelial tumors; however, when compounded with activation of K-RAS, the mice develop large, bilateral, metastatic, multifocal primitive renal epithelial tumors that have the histologic and staining characteristics of the epithelial component of human WT. These highly malignant tumors have increased activation of the phosphatidylinositol 3-kinase-AKT and extracellular signal-regulated kinase pathways, increased expression of total and nuclear β-catenin, and increased downstream targets of this pathway, such as c-Myc and survivin. Thus, we developed a novel mouse model in which activated K-RAS synergizes with canonical Wnt/β-catenin signaling to form metastatic primitive renal epithelial tumors that mimic the epithelial component of human WT.
Collapse
Affiliation(s)
- Peter E Clark
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2765, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Wilms' tumour (WT) is an embryonal cancer of childhood and is thought to be derived from embryonic kidney precursor cells. The Knudson two hit model was initially thought to occur in WT, but findings emerging from genetic and cytogenetic studies in the past two decades have implicated several genetic events. Recent techniques in genetic analysis have improved our ability to characterise changes in genes involved in WT which include WT1, CTNNB1, IGF2 and WTX. These genetic events have not only provided insight into the pathobiology of this malignancy, but the recognition of these candidate genes may offer potential targets for novel therapies. In this review, we will provide an overview of the pathological, genetic and cytogenetic characteristics of WT.
Collapse
|
24
|
Karner CM, Das A, Ma Z, Self M, Chen C, Lum L, Oliver G, Carroll TJ. Canonical Wnt9b signaling balances progenitor cell expansion and differentiation during kidney development. Development 2011; 138:1247-57. [PMID: 21350016 DOI: 10.1242/dev.057646] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mammalian kidney is composed of thousands of individual epithelial tubules known as nephrons. Deficits in nephron number are associated with myriad diseases ranging from complete organ failure to congenital hypertension. A balance between differentiation and maintenance of a mesenchymal progenitor cell population determines the final number of nephrons. How this balance is struck is poorly understood. Previous studies have suggested that Wnt9b/β-catenin signaling induced differentiation (mesenchymal-to-epithelial transition) in a subset of the progenitors but needed to be repressed in the remaining progenitors to keep them in the undifferentiated state. Here, we report that Wnt9b/β-catenin signaling is active in the progenitors and is required for their renewal/proliferation. Using a combination of approaches, we have revealed a mechanism through which cells receiving the same Wnt9b/β-catenin signal can respond in distinct ways (proliferate versus differentiate) depending on the cellular environment in which the signal is received. Interpretation of the signal is dependent, at least in part, on the activity of the transcription factor Six2. Six2-positive cells that receive the Wnt9b signal are maintained as progenitors whereas cells with reduced levels of Six2 are induced to differentiate by Wnt9b. Using this simple mechanism, the kidney is able to balance progenitor cell expansion and differentiation insuring proper nephron endowment. These findings provide novel insights into the molecular mechanisms that regulate progenitor cell differentiation during normal and pathological conditions.
Collapse
Affiliation(s)
- Courtney M Karner
- Department of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Wilms' tumour (WT) is the most common malignant renal tumour of childhood. During the past two decades or so, molecular studies carried out on biopsy specimens and tumour-derived cell lines have identified a multitude of chromosomal and epigenetic alterations in WT. In addition, a significant amount of evidence has been gathered to identify the genes and signalling pathways that play a defining role in its genesis, growth, survival and treatment responsiveness. As such, these molecules and mechanisms constitute potential targets for novel therapeutic strategies for refractory WT. In this report we aim to review some of the many candidate genes and intersecting pathways that underlie the complexities of WT biology.
Collapse
|
26
|
Loss of PTEN/MMAC1 activity is a rare and late event in the pathogenesis of nephroblastomas. Hum Pathol 2010; 41:1172-7. [DOI: 10.1016/j.humpath.2009.10.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/08/2009] [Indexed: 10/19/2022]
|
27
|
Ehrlich D, Bruder E, Thome MA, Gutt CN, von Knebel Doeberitz M, Niggli F, Perantoni AO, Koesters R. Nuclear accumulation of beta-catenin protein indicates activation of wnt signaling in chemically induced rat nephroblastomas. Pediatr Dev Pathol 2010; 13:1-8. [PMID: 19348510 PMCID: PMC2990985 DOI: 10.2350/08-03-0443.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aberrant wnt signaling caused by mutations in CTNNB1 occurs in about 15% of Wilms tumors, and these mutations appear to be dependent on the concomitant mutational inactivation of the zinc-finger protein WT1. Nuclear beta-catenin protein, a substitute marker of active wnt signaling, has been detected in an even higher proportion (>50%) of Wilms tumors, suggesting alternative genetic pathways leading to beta-catenin activation. Thus, targeting wnt signaling may become an important future therapeutic strategy in Wilms tumor patients. Currently, chemically induced rat nephroblastomas provide the only available rodent model for this tumor. To determine the contribution of active wnt signaling in this model, we investigated 24 chemically induced rat nephroblastomas for beta-catenin protein expression and for Ctnnb1 and WT1 mutations. Immunohistochemistry showed focal strong nuclear accumulation of beta-catenin protein in 18 of 24 tumors, although in a heterogenous pattern. Blastemal and mesenchymal compartments displayed nuclear-positive cells more frequently than areas of epithelial differentiation. Interestingly, we found no mutation of exon 3 of Ctnnb1 and no mutation within the zinc-finger region of WT1 in any of the 24 tumors analyzed. In conclusion, our findings suggest activation of wnt signaling in the majority (63%) of chemically induced rat nephroblastomas. Nuclear expression of beta-catenin in the absence of Ctnnb1 mutations implies, however, alternate mutational targets in rat nephroblastomas.
Collapse
Affiliation(s)
- David Ehrlich
- Division of Applied Tumor Biology, Institute of Pathology, University Hospital of Heidelberg, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Yoo NJ, Kim S, Lee SH. Mutational analysis of WTX gene in Wnt/ beta-catenin pathway in gastric, colorectal, and hepatocellular carcinomas. Dig Dis Sci 2009; 54:1011-4. [PMID: 18720004 DOI: 10.1007/s10620-008-0458-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 07/16/2008] [Indexed: 02/06/2023]
Abstract
A recent study of Wilms' tumors discovered a new X chromosome gene, Wilms' tumor gene on the X chromosome (WTX), which was found to harbor small deletions and point mutations. WTX protein negatively regulates Wnt/ beta-catenin signaling, and is considered a tumor-suppressor gene. One of the questions about the WTX gene is whether the genetic alterations of the WTX gene are specific to only Wilms' tumors. To see whether somatic point mutations of WTX occur in other malignancies, we analyzed the WTX gene for the detection of mutations in 141 cancer tissues by a single-strand conformation polymorphism assay. The cancer tissues consisted of 47 gastric adenocarcinomas, 47 colorectal adenocarcinomas, and 47 hepatocellular carcinomas. Overall, we detected one WTX mutation in the colorectal carcinomas (1/47; 2.1%), but there was no WTX mutation in other cancers analyzed. The detected mutation was a missense mutation (c. 1117G > A (p.Ala373Thr)). Although the WTX mutation is common in Wilms' tumors, our data indicate that it is rare in colorectal, gastric, and hepatocellular carcinomas. The data also suggest that deregulation of Wnt/ beta-catenin signaling by WTX gene mutation may be a rare event in the pathogenesis of colorectal, gastric, and hepatocellular carcinomas.
Collapse
Affiliation(s)
- Nam J Yoo
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul 137-701, Korea
| | | | | |
Collapse
|
29
|
Diwan BA, Timofeeva O, Rice JM, Yang Y, Sharma N, Fortini ME, Wang H, Perantoni AO. Inheritance of susceptibility to induction of nephroblastomas in the Noble rat. Differentiation 2009; 77:424-32. [PMID: 19281789 PMCID: PMC2696123 DOI: 10.1016/j.diff.2008.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 12/18/2008] [Accepted: 12/19/2008] [Indexed: 11/25/2022]
Abstract
Noble (Nb) strain rats are susceptible to nephroblastoma induction with transplacental exposure to direct-acting alkylating agent N-nitrosoethylurea (ENU), while F344 strain rats are highly resistant. To study the inheritance of susceptibility to induction of these embryonal renal tumors, fetal Nb and F344 rats and F1, F2 and reciprocal backcross hybrids were exposed transplacentally to ENU once on day 18 of gestation. Nephroblastomas developed in 53% of Nb offspring with no apparent gender difference, while no nephroblastomas developed in inbred F344 offspring. F1 and F2 hybrid offspring had intermediate responses, 28% and 30%, respectively. Nephroblastoma incidence in the offspring of F1 hybrids backcrossed to the susceptible strain Nb was 46%, while that in F1 hybrids backcrossed to resistant strain F344 was much lower (16%). Carcinogenic susceptibility is therefore consistent with the involvement of one major autosomal locus; the operation of a gene dosage effect; and a lack of simple Mendelian dominance for either susceptibility or resistance. Since established Wilms tumor-associated suppressor genes, Wt1 and Wtx, were not mutated in normal or neoplastic tissues, genomic profiling was performed on isolated Nb and F344 metanephric progenitors to identify possible predisposing factors to nephroblastoma induction. Genes preferentially elevated in expression in Nb rat progenitors included Wnt target genes Epidermal growth factor receptor, Inhibitor of DNA binding 2, and Jagged1, which were further increased in nephroblastomas. These studies demonstrate the value of this model for genetic analysis of nephroblastoma development and implicate both the Wnt and Notch pathways in its pathogenesis.
Collapse
Affiliation(s)
- Bhalchandra A. Diwan
- Basic Research Program, Science Applications International Corporation – Frederick, Inc., National Cancer Institute, Frederick, MD 21702
| | - Olga Timofeeva
- Laboratory of Comparative Carcinogenesis, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Jerry M. Rice
- Laboratory of Comparative Carcinogenesis, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Yili Yang
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Nirmala Sharma
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Mark E. Fortini
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Honghe Wang
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Alan O. Perantoni
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| |
Collapse
|
30
|
Abstract
Wilms tumours (WTs) have two distinct types of histology with or without ectopic mesenchymal elements, suggesting that WTs arise from either the mesenchymal or epithelial nephrogenic lineages. Regardless of the presence or absence of CTNNB1 mutations, nuclear accumulation of beta-catenin is often observed in WTs with ectopic mesenchymal elements. Here, we addressed the relationship between the WNT-signalling pathway and lineage in WTs by examining CTNNB1 and WT1 mutations, nuclear accumulation of beta-catenin, tumour histology and gene expression profiles. In addition, we screened for mutations in WTX, which has been proposed to be a negative regulator of the canonical WNT-signalling pathway. Unsupervised clustering analysis identified two classes of tumours: mesenchymal lineage WNT-dependent tumours, and epithelial lineage WNT-independent tumours. In contrast to the mesenchymal lineage specificity of CTNNB1 mutations, WTX mutations were surprisingly observed in both lineages. WTX-mutant WTs with ectopic mesenchymal elements had nuclear accumulation of beta-catenin, upregulation of WNT target genes and an association with CTNNB1 mutations in exon 7 or 8. However, epithelial lineage WTs with WTX mutations had no indications of active WNT signalling, suggesting that the involvement of WTX in the WNT-signalling pathway may be lineage dependent, and that WTX may have an alternative function to its role in the canonical WNT-signalling pathway.
Collapse
|
31
|
Abstract
Wilms' tumor is the most common pediatric renal neoplasm, but its occurrence in adults is very rare. In contrast to pediatric Wilms' tumor (PWT), very little is known about the pathogenesis of adult Wilms' tumor (AWT). Despite there currently being no morphological difference between AWT and PWT, a cytogenetic study has suggested that the pathogenesis of AWT might be different from that of PWT. Although dysregulation of the Wnt pathway has been implicated in PWT, its role in AWT has never been investigated. To investigate the role of dysregulation of the Wnt pathway in AWT, tumor samples from 4 AWTs and 19 PWTs were surveyed for subcellular localization of beta-catenin by immunohistochemistry and potential mutation of the beta-catenin gene by sequencing. Nuclear translocation of beta-catenin was found in one out of four cases of AWT, but none of them carried mutation of the beta-catenin gene. By comparison, nuclear translocation for beta-catenin and mutation of the beta-catenin gene were present in 53% (10/19) and 15.8% (3/19) of PWTs, respectively. Of the three mutations identified, we found a novel mutation combining a silent mutation (TCT to TCC, Ser37Ser) and an in-frame six-base-pair deletion (del GGTGCC, del Gly38Ala39). This report suggests that dysregulation of the Wnt pathway might also play a role in the pathogenesis of AWT.
Collapse
Affiliation(s)
- Min-Cheng Su
- Department of Pathology, Min-Sheng General Hospital, Taoyuan, Taiwan
| | | | | |
Collapse
|
32
|
Uccini S, Perotti D, Colarossi C, Stoppacciaro A, Sardella M, Mannarino O, Collini P, Casieri P, Cozzi D, Amoroso L, Spreafico F, Radice P, Dominici C. Molecular evidence of the independent origin of multiple Wilms tumors in a case of WAGR syndrome. Pediatr Blood Cancer 2008; 51:344-8. [PMID: 18293378 DOI: 10.1002/pbc.21507] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND This study investigated the genetic events leading to tumorigenesis in a patient affected with WAGR syndrome who developed multiple distinct Wilms tumors (WTs). PROCEDURE AND RESULTS At 1 year of age, the child developed two synchronous bilateral WTs that were resected by partial nephrectomy. Histologically, these tumors were fetal rhabdomyomatous nephroblastomas. Immunohistochemical study revealed the absence of nuclear expression of WT1 protein, while beta-catenin protein was expressed at nuclear level by the large majority of tumor cells. Molecular investigations of WT1 gene and exon 3 of beta-catenin (CTNNB1) gene detected no mutations. At 4 years of age, 28 months after the chemotherapy completion, a third WT was diagnosed in the left kidney, and surgically removed before any further chemotherapy. Nine months after surgery, a metastasis was detected in the left lung. Both the third renal tumor and the lung metastasis showed a blastema-predominant morphology. Immunohistochemistry confirmed the lack of expression of WT1 protein, while beta-catenin protein was expressed at nuclear level by the large majority of tumor cells. Molecular analysis of the third renal tumor and the lung metastasis revealed a 4 bp deletion in exon 7 of WT1 gene, leading to a frameshift of the reading frame and to a premature stop of the translation (c.925_928delACTC, p.T309LfsX71); no mutations in the exon 3 of the beta-catenin gene were documented. CONCLUSIONS These data demonstrate that multiple WTs can arise as a consequence of different genetic events in a patient with genetic predisposition, such as WAGR syndrome.
Collapse
Affiliation(s)
- Stefania Uccini
- Department of Experimental Medicine and Pathology, La Sapienza University, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Perotti D, Gamba B, Sardella M, Spreafico F, Terenziani M, Collini P, Pession A, Nantron M, Fossati-Bellani F, Radice P. Functional inactivation of the WTX gene is not a frequent event in Wilms' tumors. Oncogene 2008; 27:4625-32. [PMID: 18391980 DOI: 10.1038/onc.2008.93] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For many years the precise genetic etiology of the majority of Wilms' tumors has remained unexplained. Recently, the WTX gene, mapped to chromosome Xq11.1, has been reported to be lost or mutated in approximately one-third of Wilms' tumors. Moreover, in female cases, the somatically inactivated alleles were found to invariantly derive from the active chromosome X. Consequently, WTX has been proposed as a 'one-hit' tumor suppressor gene. To provide further insights on the contribution of WTX to the development of the disease, we have examined 102 Wilms' tumors, obtained from 43 male and 57 female patients. Quantitative PCR analyses detected WTX deletions in 5 of 45 (11%) tumors from males, whereas loss of heterozygosity at WTX-linked microsatellites was observed in 9 tumors from 50 informative females (19%). However, in the latter group, using a combination of HUMARA assay and bisulfite-modified DNA sequencing, we found that the deletion affected the active chromosome X only in two cases (4%). Sequence analyses detected an inactivating somatic mutation of WTX in a single tumor, in which a strongly reduced expression of the mutant allele respect to the wild-type allele was observed, a finding not consistent with its localization on the active chromosome X. Overall, a functional somatic nullizygosity of the WTX gene was ascertained only in seven of the Wilms' tumors included in the study (approximately 7%). Our findings indicate that previously reported estimates on the proportion of Wilms' tumors due to WTX alterations should be reconsidered.
Collapse
Affiliation(s)
- D Perotti
- Department of Experimental Oncology and Laboratories, Genetic Susceptibility to Cancer Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Basta-Jovanovic G, Gvozdenovic E, Dimitrijevic I, Brasanac D, Jovanovic M, Kalezic N, Baralic I, Radojevic-Skodric S, Arsic D. Immunohistochemical analysis of gamma catenin in Wilms' tumors. Fetal Pediatr Pathol 2008; 27:63-70. [PMID: 18568994 DOI: 10.1080/15513810802028704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of our study was to investigate the expression of gamma-catenin in normal kidney and in Wilms' tumor by immunohistochemistry and to correlate the results with tumor stage, histological type, and prognostic group. We investigated 28 cases of Wilms' tumor, 2 Wilms' tumor metastases in lungs, and 1 specimen of normal renal tissue. Expression of gamma-catenin was detected in 14 cases. There was a weak inverse relationship between gamma-catenin expression and tumor stage. Expression of gamma-catenin was detected in various histologic types of Wilms' tumor, but there was no statistically significant correlation, except in cases with diffuse anaplasia that were negative. In 2 metastatic cases and in the case of bilateral Wilms' tumor gamma-catenin immunostaining was not observed Our findings suggest an absence of strong correlation between the loss of gamma-catenin and unfavorable outcome.
Collapse
|
35
|
Lovvorn HN, Westrup J, Opperman S, Boyle S, Shi G, Anderson J, Perlman EJ, Perantoni AO, Wills M, de Caestecker M. CITED1 expression in Wilms' tumor and embryonic kidney. Neoplasia 2007; 9:589-600. [PMID: 17710162 PMCID: PMC1941694 DOI: 10.1593/neo.07358] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Accepted: 06/30/2005] [Indexed: 11/18/2022] Open
Abstract
Wilms' tumors, or nephroblastomas, are thought to arise from abnormal postnatal retention and dysregulated differentiation of nephrogenic progenitor cells that originate as a condensed metanephric mesenchyme within embryonic kidneys. We have previously shown that the transcriptional regulator CITED1 (CBP/p300-interacting transactivators with glutamic acid [E]/aspartic acid [D]-rich C-terminal domain) is expressed exclusively in these nephrogenic progenitor cells and is downregulated as they differentiate to form nephronic epithelia. In the current study, we show that CITED1 expression persists in blastemal cell populations of both experimental rat nephroblastomas and human Wilms' tumors, and that primary human Wilms' tumors presenting with disseminated disease show the highest level of CITED1 expression. Unlike the predominantly cytoplasmic subcellular localization of CITED1 in the normal developing kidney, CITED1 is clearly detectable in the nuclear compartment of Wilms' tumor blastema. These findings indicate that CITED1 is a marker of primitive blastema in Wilms' tumors and suggest that persistent expression and/or altered subcellular localization of CITED1 in the condensed metanephric mesenchyme could play a role in Wilms' tumor initiation and pathogenesis.
Collapse
Affiliation(s)
- Harold N Lovvorn
- The Department of Pediatric Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Major MB, Camp ND, Berndt JD, Yi X, Goldenberg SJ, Hubbert C, Biechele TL, Gingras AC, Zheng N, Maccoss MJ, Angers S, Moon RT. Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science 2007; 316:1043-6. [PMID: 17510365 DOI: 10.1126/science/1141515] [Citation(s) in RCA: 318] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aberrant WNT signal transduction is involved in many diseases. In colorectal cancer and melanoma, mutational disruption of proteins involved in the degradation of beta-catenin, the key effector of the WNT signaling pathway, results in stabilization of beta-catenin and, in turn, activation of transcription. We have used tandem-affinity protein purification and mass spectrometry to define the protein interaction network of the beta-catenin destruction complex. This assay revealed that WTX, a protein encoded by a gene mutated in Wilms tumors, forms a complex with beta-catenin, AXIN1, beta-TrCP2 (beta-transducin repeat-containing protein 2), and APC (adenomatous polyposis coli). Functional analyses in cultured cells, Xenopus, and zebrafish demonstrate that WTX promotes beta-catenin ubiquitination and degradation, which antagonize WNT/beta-catenin signaling. These data provide a possible mechanistic explanation for the tumor suppressor activity of WTX.
Collapse
Affiliation(s)
- Michael B Major
- Howard Hughes Medical Institute, University of Washington School of Medicine, Box 357370, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Roel Nusse
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University, School of Medicine, Stanford, CA 94305-5323, USA.
| |
Collapse
|
38
|
Jilong Y, Jian W, Xiaoyan Z, Xiaoqiu L, Xiongzeng Z. Analysis of APC/beta-catenin genes mutations and Wnt signalling pathway in desmoid-type fibromatosis. Pathology 2007; 39:319-25. [PMID: 17558858 DOI: 10.1080/00313020701329823] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The abnormalities of the Wnt signalling pathway in desmoid-type fibromatosis were analysed, with the purpose of exploring the mechanism of tumorigenesis and progression. METHODS The clinical and histopathological features of 96 cases were analysed. Beta-catenin, cyclin-D1, c-myc, and Ki-67 proteins were detected in 69 cases using formalin-fixed, paraffin-embedded tissues. Using the same materials, apoptosis of the tumour cells was investigated by terminal deoxynucleotidyl transferase mediated dUTP nick end-labelling (TUNEL) testing. Polymerase chain reaction (PCR), denaturing high performance liquid chromatography (DHPLC) assay, and sequencing were performed to detect abnormalities of the adenomatous polyposis coli (APC) and beta-catenin genes. RESULTS APC gene mutations were found in 18 cases (26.1%, 18/69). Somatic mutations of codon 41 in exon 3 of beta-catenin were detected in 13 cases (18.8%, 13/69). No correlation of beta-catenin abnormal expression with the mutations of APC gene or beta-catenin gene was identified (p>0.05). The cases with abnormal beta-catenin expression showed a higher level of c-myc protein expression (69.7%, 23/33) than those without (22.2%, 8/36, p = 0.001). The apoptotic indices (AIs) were significantly lower in cyclin-D1 positive cases and c-myc positive cases (p = 0.015, p = 0.007). CONCLUSIONS There are somatic mutations of the APC and beta-catenin gene in desmoid-type fibromatosis, and there are abnormalities in the Wnt signalling pathway. These abnormalities may result in aberrant cell proliferation and apoptosis, which are likely to be important factors in tumorigenesis and progression.
Collapse
Affiliation(s)
- Yang Jilong
- Department of Bone and Soft Tissue Tumor, Cancer Hospital, Tianjin Medical University. Tianjin, China.
| | | | | | | | | |
Collapse
|
39
|
Schaub R, Burger A, Bausch D, Niggli FK, Schäfer BW, Betts DR. Array comparative genomic hybridization reveals unbalanced gain of the MYCN region in Wilms tumors. ACTA ACUST UNITED AC 2007; 172:61-5. [PMID: 17175381 DOI: 10.1016/j.cancergencyto.2006.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 08/22/2006] [Accepted: 08/29/2006] [Indexed: 11/24/2022]
Abstract
Wilms tumor (WT) is one of the most common solid tumors in childhood. It is characterized by a nonrandom pattern of chromosomal aberrations whose clinical significance is still uncertain. To gain further insight into different genetic events and their corresponding biological role, conventional cytogenetics and array comparative genomic hybridization (array CGH) were performed on 13 tumor samples. In two of these, array CGH revealed, together with other aberrations, a low-level amplification and an unbalanced gain in the region of chromosome bands 2p23 approximately p24 that encompass the MYCN gene. Both events were confirmed with a MYCN-specific fluorescence in situ hybridization probe, which showed a signal pattern consistent with a small homogenous staining region in one case. In addition, mRNA expression levels for MYCN were determined by quantitative reverse-transcriptase polymerase chain reaction and revealed that gain of chromosomal material was reflected in enhanced levels of MYCN mRNA in both tumors, whereby also additional tumors showed increased MYCN expression. Therefore, our findings suggest that WT is an additional childhood tumor where MYCN gain might play an important role in tumor development.
Collapse
Affiliation(s)
- Rahel Schaub
- Department of Oncology, University Children's Hospital, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Natrajan R, Reis-Filho JS, Little SE, Messahel B, Brundler MA, Dome JS, Grundy PE, Vujanic GM, Pritchard-Jones K, Jones C. Blastemal expression of type I insulin-like growth factor receptor in Wilms' tumors is driven by increased copy number and correlates with relapse. Cancer Res 2006; 66:11148-55. [PMID: 17145858 DOI: 10.1158/0008-5472.can-06-1931] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most Wilms' tumors are of low stage, favorable histology, and have a high likelihood of cure with current multimodal therapy. Despite this, there remains a group of patients whose tumors recur for whom intensive salvage regimens result in survival of only 50%. Fitting a Cox proportional hazards model to microarray-based comparative genomic hybridization (aCGH) data on 68 Wilms' tumor samples, we identified a significant correlation between increased copy number at chromosome 15q26.3 insulin-like growth factor I receptor (IGFIR) and tumor relapse (adjusted P = 0.014). Wilms' tumors (13%) exhibited a low-level gain corresponding to three to four copies of the gene by aCGH analysis, 9 of 10 of which exhibited high IGFIR mRNA levels. Although IGFIR protein expression was restricted to the epithelial cells of fetal kidney and Wilms' tumors in most cases, 12% of tumors were also found to express IGFIR in the blastemal compartment. Blastemal IGFIR protein expression was associated with an increased copy number and a shorter relapse-free survival time (P = 0.027, log-rank test). In addition to the membrane localization, IGFIR was localized to the perinuclear region of the blastemal cells in 6% of Wilms' tumors. These data provide evidence that an increase in IGFIR gene copy number results in aberrant expression in the blastemal compartment of some Wilms' tumors and is associated with an adverse outcome in these patients. These findings suggest the possibility of use of targeted agents in the therapy of these children.
Collapse
Affiliation(s)
- Rachael Natrajan
- Paediatric Oncology, Institute of Cancer Research/Royal Marsden National Health Service Trust, Sutton, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sansom OJ, Griffiths DFR, Reed KR, Winton DJ, Clarke AR. Apc deficiency predisposes to renal carcinoma in the mouse. Oncogene 2006; 24:8205-10. [PMID: 16116480 DOI: 10.1038/sj.onc.1208956] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Deregulation of Wnt signalling has recently been implicated in human renal cancer. Here, we directly test this association by using a Cre-LoxP strategy to inactivate the Adenomatous Polyposis Coli (Apc) gene in the murine renal epithelium. Mice homozygous for a conditional Apc allele were intercrossed with mice transgenic for Cre recombinase under control of the Cyp1A promoter, which delivers constitutive recombination within a proportion of cells in the renal epithelium. Inactivation of Apc leads to the accumulation of nuclear beta-catenin and the rapid development of multiple dysplastic foci. Renal carcinoma was observed with an earliest onset of 4 months. This predisposition was accelerated by p53 deficiency, reducing the earliest onset to 2 months. Compared to other murine models of kidney neoplasia, this represents particularly rapid onset of disease, and so implicates an important role for Apc in suppressing renal carcinoma.
Collapse
Affiliation(s)
- Owen J Sansom
- Cardiff School of Biosciences, Cardiff University, Wales CF10 3US, UK
| | | | | | | | | |
Collapse
|
43
|
Shan J, Shi DL, Wang J, Zheng J. Identification of a specific inhibitor of the dishevelled PDZ domain. Biochemistry 2006; 44:15495-503. [PMID: 16300398 DOI: 10.1021/bi0512602] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Wnt signaling pathways are involved in embryo development as well as in tumorigenesis. Dishevelled (Dvl) transduces Wnt signals from the receptor Frizzled (Fz) to downstream components in canonical and noncanonical Wnt signaling pathways. The Dvl PDZ domain is thought to play an essential role in both pathways, and we recently demonstrated that the Dvl PDZ domain binds directly to Fz receptors. In this study, using structure-based virtual ligand screening, we identified an organic molecule (NSC668036) from the National Cancer Institute small-molecule library that can bind to the Dvl PDZ domain. We then used molecular dynamics simulation to analyze the binding between the PDZ domain and NSC668036 in detail. In addition, we showed that, in Xenopus, as expected, NSC668036 inhibited the signaling induced by Wnt3A. This compound provides a basis for rational design of high-affinity inhibitors of the PDZ domain, which can block Wnt signaling by interrupting the Fz-Dvl interaction.
Collapse
Affiliation(s)
- Jufang Shan
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | |
Collapse
|
44
|
Ramburan A, Hadley GP, Govender D. Expression of E-cadherin, cadherin-11, α-, β- and γ-catenins in nephroblastomas: relationship with clinicopathological parameters, prognostic factors and outcome. Pathology 2006; 38:39-44. [PMID: 16484007 DOI: 10.1080/00313020500462056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AIM This study was undertaken to determine the expression of cell adhesion molecules E-cadherin, cadherin-11, and alpha-, beta- and gamma-catenins in nephroblastomas and to correlate this expression with pathological features and known prognostic factors. METHODS Immunohistochemistry was performed on 140 cases of nephroblastoma following heat-induced epitope retrieval and using the streptavidin-biotin technique. RESULTS E-cadherin was expressed in 75 cases (54%), cadherin-11 in 128 cases (91%), alpha-catenin in 93 cases (66%), beta-catenin in 133 cases (95%) and gamma-catenin in 22 cases (16%). Nuclear localisation of beta-catenin was not demonstrated. There was a statistically significant relationship between the administration of preoperative chemotherapy and the expression of E-cadherin, alpha- and gamma-catenin, respectively. These proteins were more frequently expressed in tumours treated with preoperative chemotherapy. Those tumours that expressed all four proteins (E-cadherin, alpha-, beta- and gamma-catenin) showed a statistically significant association with the administration of preoperative chemotherapy, in contrast to tumours that did not express all four proteins. CONCLUSION Nephroblastomas show a heterogeneous distribution of staining for E-cadherin, cadherin-11, alpha-, beta- and gamma-catenins. Tumours treated with preoperative chemotherapy are more likely to express these molecules. The expression status of E-cadherin, cadherin-11 and the catenins in this cohort does not appear to be of prognostic value.
Collapse
Affiliation(s)
- Amsha Ramburan
- Department of Paediatric Surgery, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
45
|
Kundu JK, Choi KY, Surh YJ. beta-Catenin-mediated signaling: a novel molecular target for chemoprevention with anti-inflammatory substances. Biochim Biophys Acta Rev Cancer 2005; 1765:14-24. [PMID: 16226380 DOI: 10.1016/j.bbcan.2005.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2004] [Revised: 08/16/2005] [Accepted: 08/23/2005] [Indexed: 01/12/2023]
Abstract
Inflammation is thought to play a role in the pathophysiology of cancer. Accumulating evidence from clinical and laboratory-based studies suggests that substances with anti-inflammatory activities are potential candidates for chemoprevention. Recent advances in cellular and molecular biology of cancer shed light on components of intracellular signaling cascades that can be potential molecular targets of chemoprevention with various anti-inflammatory substances. Although cyclooxygenase-2, a primary enzyme that mediates inflammatory responses, has been well recognized as a molecular target for chemoprevention by both synthetic and natural anti-inflammatory agents, the cellular signaling mechanisms that associate inflammation and cancer are not still clearly illustrated. Recent studies suggest that beta-catenin-mediated signaling, which regulates developmental processes, may act as a potential link between inflammation and cancer. This review aims to focus on beta-catenin-mediated signaling pathways, particularly in relation to its contribution to carcinogenesis, and the modulation of inappropriately activated beta-catenin-mediated signaling by nonsteroidal anti-inflammatory drugs and chemopreventive phytochemicals possessing anti-inflammatory properties.
Collapse
Affiliation(s)
- Joydeb Kumar Kundu
- National Research Laboratory of Molecular Carcinogenesis and Chemoprevention, College of Pharmacy, Seoul National University, Shinlim-dong, Kwanak-ku, Seoul 151-742, South Korea
| | | | | |
Collapse
|
46
|
Abstract
Abnormalities in the Wnt signalling pathway are found in a wide range of cancers. The diverse origin of these malignancies implies that the contribution that disrupted Wnt signalling makes to tumourigenesis is not limited to specific tissue types and thus can be regarded as a step which is 'generic' to the process of carcinogenesis. In recent years, rapid progress has been made in the understanding of the Wnt signalling pathway, giving an insight into how inappropriate activation of this pathway may facilitate the neoplastic conversion of a normal cell. Furthermore, elucidation of the mechanisms that regulate Wnt signalling has led to the possibility of manipulating these mechanisms in order to down-regulate Wnt signalling in established tumours. In this review, the Wnt signalling pathway is described. The role of aberrant Wnt signalling in tumour development is discussed together with its clinical implications for anti-tumour therapy.
Collapse
Affiliation(s)
- Mohammad Ilyas
- Academic Unit of Pathology, Leeds General Infirmary, Leeds LS1 3EX, UK.
| |
Collapse
|
47
|
Zirn B, Wittmann S, Graf N, Gessler M. Chibby, a novel antagonist of the Wnt pathway, is not involved in Wilms tumor development. Cancer Lett 2005; 220:115-20. [PMID: 15737694 DOI: 10.1016/j.canlet.2004.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Revised: 08/02/2004] [Accepted: 08/02/2004] [Indexed: 11/23/2022]
Abstract
Deregulation of the Wnt signalling pathway is a key event in the development of a broad spectrum of human malignancies and mutations in beta-catenin (CTNNB1), a central component of the Wnt pathway, have been detected in 10-15% of Wilms tumors (nephroblastoma). Furthermore, nuclear immunoreactivity for beta-catenin has been described even in the absence of detectable beta-catenin mutations. This suggests that other components of the Wnt pathway may be involved in the pathogenesis of a subgroup of Wilms tumors. Chibby (C22ORF2) is a recently identified antagonistic component of the Wnt pathway that inhibits the transcriptional activity of beta-catenin. Our study addresses the question whether mutation or down-regulation of Chibby is involved in Wilms tumorigenesis. We analysed the expression of Chibby by real time RT-PCR in 142 Wilms tumors, but there was no significant expression difference in any group of tumors stratified according to clinical, histological and mutational criteria. Mutation analysis of a smaller cohort did not reveal any mutations of the coding sequence. We only detected a constitutive splice variant leading to the absence of exon 4 in all Wilms tumors as well as in normal tissues. In addition, we detected a frequent silent polymorphism in the Chibby exon 4 sequence (435T/C). These data strongly suggest that despite its attractive function as a modulator of beta-catenin activity, Chibby is not involved in Wilms tumorigenesis.
Collapse
Affiliation(s)
- Birgit Zirn
- Theodor-Boveri-Institut fuer Biowissenschaften, Physiologische Chemie I, Am Hubland, D-97074 Wuerzburg, Germany
| | | | | | | |
Collapse
|
48
|
Ramburan A, Oladiran F, Smith C, Hadley GP, Govender D. Microsatellite analysis of the adenomatous polyposis coli (APC) gene and immunoexpression of beta catenin in nephroblastoma: a study including 83 cases treated with preoperative chemotherapy. J Clin Pathol 2005; 58:44-50. [PMID: 15623481 PMCID: PMC1770552 DOI: 10.1136/jcp.2004.019752] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS To determine whether microsatellite mutations of the adenomatous polyposis coli (APC) gene have pathological or prognostic significance in nephroblastomas and to correlate APC alterations with beta catenin immunoexpression. METHODS One hundred nephroblastomas were analysed, 83 of which received preoperative chemotherapy. Normal and tumour DNA was isolated using standard proteinase K digestion and phenol/chloroform extraction from paraffin wax embedded tissue. Polymerase chain reaction using four APC microsatellite markers-D5S210, D5S299, D5S82, and D5S346-was performed and the products analysed. Immunohistochemistry was performed using the LSAB kit with diaminobenzidine as chromogen. Results were correlated with clinicopathological data using the chi(2) test. RESULTS Allelic imbalance/loss of heterozygosity was more frequent than microsatellite instability, with 30% of cases showing allelic imbalance/ loss of heterozygosity and 16% showing microsatellite instability. Although there was a significant correlation between the results for individual markers and the clinicopathological data, the overall results do not support a prognostic role for APC in nephroblastoma. Expression of beta catenin was seen in 93% of cases. Staining was predominantly membranous, with epithelium, blastema, and stroma being immunoreactive. Cytoplasmic redistribution was seen in 58% of cases, but no nuclear staining was detected. No significant associations between beta catenin expression and the clinicopathological parameters were found. Kaplan-Meier survival plots showed that patients with loss of membranous staining and pronounced cytoplasmic staining (score, 3) had a significantly shorter survival (p = 0.04; median survival, 5.87 months). CONCLUSION Microsatellite analysis of APC and immunoexpression of beta catenin did not provide significant pathological or prognostic information in this cohort of nephroblastomas.
Collapse
Affiliation(s)
- A Ramburan
- Molecular Biology Research Facility, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Congella, 7925 Durban, South Africa
| | | | | | | | | |
Collapse
|
49
|
Li CM, Kim CE, Margolin AA, Guo M, Zhu J, Mason JM, Hensle TW, Murty VVVS, Grundy PE, Fearon ER, D'Agati V, Licht JD, Tycko B. CTNNB1 mutations and overexpression of Wnt/beta-catenin target genes in WT1-mutant Wilms' tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 165:1943-53. [PMID: 15579438 PMCID: PMC1618727 DOI: 10.1016/s0002-9440(10)63246-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gain-of-function mutations in exon 3 of beta-catenin (CTNNB1) are specific for Wilms' tumors that have lost WT1, but 50% of WT1-mutant cases lack such "hot spot" mutations. To ask whether stabilization of beta-catenin might be essential after WT1 loss, and to identify downstream target genes, we compared expression profiles in WT1-mutant versus WT1 wild-type Wilms' tumors. Supervised and nonsupervised hierarchical clustering of the expression data separated these two classes of Wilms' tumor. The WT1-mutant tumors overexpressed genes encoding myogenic and other transcription factors (MOX2, LBX1, SIM2), signaling molecules (TGFB2, FST, BMP2A), extracellular Wnt inhibitors (WIF1, SFRP4), and known beta-catenin/TCF targets (FST, CSPG2, CMYC). Beta-Catenin/TCF target genes were overexpressed in the WT1-mutant tumors even in the absence of CTNNB1 exon 3 mutations, and complete sequencing revealed gain-of-function mutations elsewhere in the CTNNB1 gene in some of these tumors, increasing the overall mutation frequency to 75%. Lastly, we identified and validated a novel direct beta-catenin target gene, GAD1, among the WT1-mutant signature genes. These data highlight two molecular classes of Wilms' tumor, and indicate strong selection for stabilization of beta-catenin in the WT1-mutant class. Beta-Catenin stabilization can initiate tumorigenesis in other systems, and this mechanism is likely critical in tumor formation after loss of WT1.
Collapse
Affiliation(s)
- Chi-Ming Li
- Institute of Cancer Genetics, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bläker H, Sutter C, Kadmon M, Otto HF, Von Knebel-Doeberitz M, Gebert J, Helmke BM. Analysis of somatic APC mutations in rare extracolonic tumors of patients with familial adenomatous polyposis coli. Genes Chromosomes Cancer 2004; 41:93-8. [PMID: 15287021 DOI: 10.1002/gcc.20071] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Patients with familial adenomatous polyposis coli (FAP) carry heterozygous mutations of the APC gene. At a young age, these patients develop multiple colorectal adenomas that consistently display a second somatic mutation in the remaining APC wild-type allele. Inactivation of APC leads to impaired degradation of beta-catenin, thereby promoting continuous cell-cycle progression. The role of APC inactivation in rare extracolonic tumors of FAP patients has not been characterized sufficiently. Among tissue specimen from 174 patients with known APC germ-line mutations, we identified 8 tumors infrequently seen in FAP. To investigate the pathogenic role of APC pathway deregulation in these lesions, they were analyzed for second-hit somatic mutations in the mutational cluster region of the APC gene. Immunohistochemistry was performed to compare the expression pattern of beta-catenin to the mutational status of the APC gene. Exon 3 of the beta-catenin gene (CTNNB1) was analyzed for activating mutations to investigate alternative mechanisms of elevated beta-catenin concentration. Although CTNNB1 mutations were not observed, second somatic APC mutations were found in 4 of the 8 tumors: a uterine adenocarcinoma, a hepatocellular adenoma, an adrenocortical adenoma, and an epidermal cyst. These tumors showed an elevated concentration of beta-catenin. No APC mutations were seen in focal nodular hyperplasia of the liver, angiofibrolipoma, and seborrheic wart. This is the first study reporting second somatic APC mutations in FAP-associated uterine adenocarcinoma and epidermal cysts. Furthermore, our data strengthen a role for impaired APC function in the pathogenesis of adrenal and hepatic neoplasms in FAP patients.
Collapse
Affiliation(s)
- Hendrik Bläker
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|