1
|
Immunohistochemical Expression of Wilms’ Tumor 1 Protein in Human Tissues: From Ontogenesis to Neoplastic Tissues. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app10010040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The human Wilms’ tumor gene (WT1) was originally isolated in a Wilms’ tumor of the kidney as a tumor suppressor gene. Numerous isoforms of WT1, by combination of alternative translational start sites, alternative RNA splicing and RNA editing, have been well documented. During human ontogenesis, according to the antibodies used, anti-C or N-terminus WT1 protein, nuclear expression can be frequently obtained in numerous tissues, including metanephric and mesonephric glomeruli, and mesothelial and sub-mesothelial cells, while cytoplasmic staining is usually found in developing smooth and skeletal cells, myocardium, glial cells, neuroblasts, adrenal cortical cells and the endothelial cells of blood vessels. WT1 has been originally described as a tumor suppressor gene in renal Wilms’ tumor, but more recent studies emphasized its potential oncogenic role in several neoplasia with a variable immunostaining pattern that can be exclusively nuclear, cytoplasmic or both, according to the antibodies used (anti-C or N-terminus WT1 protein). With the present review we focus on the immunohistochemical expression of WT1 in some tumors, emphasizing its potential diagnostic role and usefulness in differential diagnosis. In addition, we analyze the WT1 protein expression profile in human embryonal/fetal tissues in order to suggest a possible role in the development of organs and tissues and to establish whether expression in some tumors replicates that observed during the development of tissues from which these tumors arise.
Collapse
|
2
|
Wang D, Horton JR, Zheng Y, Blumenthal RM, Zhang X, Cheng X. Role for first zinc finger of WT1 in DNA sequence specificity: Denys-Drash syndrome-associated WT1 mutant in ZF1 enhances affinity for a subset of WT1 binding sites. Nucleic Acids Res 2019; 46:3864-3877. [PMID: 29294058 PMCID: PMC5934627 DOI: 10.1093/nar/gkx1274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/22/2017] [Indexed: 12/14/2022] Open
Abstract
Wilms tumor protein (WT1) is a Cys2-His2 zinc-finger transcription factor vital for embryonic development of the genitourinary system. The protein contains a C-terminal DNA binding domain with four tandem zinc-fingers (ZF1-4). An alternative splicing of Wt1 can add three additional amino acids-lysine (K), threonine (T) and serine (S)-between ZF3 and ZF4. In the -KTS isoform, ZF2-4 determine the sequence-specificity of DNA binding, whereas the function of ZF1 remains elusive. Three X-ray structures are described here for wild-type -KTS isoform ZF1-4 in complex with its cognate DNA sequence. We observed four unique ZF1 conformations. First, like ZF2-4, ZF1 can be positioned continuously in the DNA major groove forming a 'near-cognate' complex. Second, while ZF2-4 make base-specific interactions with one DNA molecule, ZF1 can interact with a second DNA molecule (or, presumably, two regions of the same DNA molecule). Third, ZF1 can intercalate at the joint of two tail-to-head DNA molecules. If such intercalation occurs on a continuous DNA molecule, it would kink the DNA at the ZF1 binding site. Fourth, two ZF1 units can dimerize. Furthermore, we examined a Denys-Drash syndrome-associated ZF1 mutation (methionine at position 342 is replaced by arginine). This mutation enhances WT1 affinity for a guanine base. X-ray crystallography of the mutant in complex with its preferred sequence revealed the interactions responsible for this affinity change. These results provide insight into the mechanisms of action of WT1, and clarify the fact that ZF1 plays a role in determining sequence specificity of this critical transcription factor.
Collapse
Affiliation(s)
- Dongxue Wang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John R Horton
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Zheng
- RGENE, Inc., 953 Indiana Street, San Francisco, CA 94107, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Xing Zhang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaodong Cheng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3
|
Timbergen MJ, Janssen ML, Verhoef C, Grünhagen DJ, Chibon F, Smid M, Sleijfer S, Wiemer EA. Wnt targets genes are not differentially expressed in desmoid tumors bearing different activating β-catenin mutations. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2019; 45:691-698. [PMID: 30528042 DOI: 10.1016/j.ejso.2018.09.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/17/2018] [Accepted: 09/24/2018] [Indexed: 11/21/2022]
|
4
|
Sallum LF, Andrade L, Ramalho S, Ferracini AC, de Andrade Natal R, Brito ABC, Sarian LO, Derchain S. WT1, p53 and p16 expression in the diagnosis of low- and high-grade serous ovarian carcinomas and their relation to prognosis. Oncotarget 2018. [PMID: 29662608 DOI: 10.18632/oncotarget.24530] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective To evaluate the diagnostic and prognostic value of the immunohistochemical expression of WT1, p53 and p16 in low- (LGSOCs) and high-grade serous ovarian carcinomas (HGSOCs). Results HGSOC had a significantly higher proportion of advanced stage disease, higher CA125 levels, higher proportion of post-surgery residual disease and higher recurrence or disease progression. WT1 was expressed in 71.4% of LGSOCs and in 57.1% of HGSOCs (p = 0.32). Focal and/or complete absence of p53 expression with negative p16 expression was found in 90.5% of LGSOCs, in contrast to the 88.1% of HGSOCs with diffuse or complete absence of p53 expression with positive p16 expression (<0.001). The IHC p53/p16 index and the morphological classification were closely matched (k = 0.68). In the univariate analysis, FIGO stage, post-surgery residual disease and histological grade were significantly associated with progression-free survival (PFS) and overall survival (OS). The IHC p53/p16 index was associated only with PFS. WT1 was not associated with PFS or OS. According to the multivariate analysis, advanced FIGO stage and presence of post-surgery residual disease remained independent prognostic factors for worst PFS, however these features had only a trend association with OS. Methods 21 LGSOC and 85 HGSOC stage I-IV cases were included. The morphological classification was assessed according to the World Health Organization (WHO) criteria. Immunohistochemistry (IHC) was performed in tissue microarray slides. IHC p53/p16 index was compared with the morphological classification. Conclusions The IHC p53/p16 index was a good marker for the differentiation of LGSOC and HGSOC, but the morphologic classification showed a better association with survival. FIGO stage and post-surgery residual disease remained the only independent prognostic factors for survival.
Collapse
Affiliation(s)
- Luis Felipe Sallum
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Liliana Andrade
- Department of Pathology, University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Susana Ramalho
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Amanda Canato Ferracini
- Program in Medical Sciences, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Rodrigo de Andrade Natal
- Laboratory of Investigative and Molecular Pathology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | | | - Luis Otávio Sarian
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Sophie Derchain
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| |
Collapse
|
5
|
Sallum LF, Andrade L, Ramalho S, Ferracini AC, de Andrade Natal R, Brito ABC, Sarian LO, Derchain S. WT1, p53 and p16 expression in the diagnosis of low- and high-grade serous ovarian carcinomas and their relation to prognosis. Oncotarget 2018; 9:15818-15827. [PMID: 29662608 PMCID: PMC5882299 DOI: 10.18632/oncotarget.24530] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
Objective To evaluate the diagnostic and prognostic value of the immunohistochemical expression of WT1, p53 and p16 in low- (LGSOCs) and high-grade serous ovarian carcinomas (HGSOCs). Results HGSOC had a significantly higher proportion of advanced stage disease, higher CA125 levels, higher proportion of post-surgery residual disease and higher recurrence or disease progression. WT1 was expressed in 71.4% of LGSOCs and in 57.1% of HGSOCs (p = 0.32). Focal and/or complete absence of p53 expression with negative p16 expression was found in 90.5% of LGSOCs, in contrast to the 88.1% of HGSOCs with diffuse or complete absence of p53 expression with positive p16 expression (<0.001). The IHC p53/p16 index and the morphological classification were closely matched (k = 0.68). In the univariate analysis, FIGO stage, post-surgery residual disease and histological grade were significantly associated with progression-free survival (PFS) and overall survival (OS). The IHC p53/p16 index was associated only with PFS. WT1 was not associated with PFS or OS. According to the multivariate analysis, advanced FIGO stage and presence of post-surgery residual disease remained independent prognostic factors for worst PFS, however these features had only a trend association with OS. Methods 21 LGSOC and 85 HGSOC stage I–IV cases were included. The morphological classification was assessed according to the World Health Organization (WHO) criteria. Immunohistochemistry (IHC) was performed in tissue microarray slides. IHC p53/p16 index was compared with the morphological classification. Conclusions The IHC p53/p16 index was a good marker for the differentiation of LGSOC and HGSOC, but the morphologic classification showed a better association with survival. FIGO stage and post-surgery residual disease remained the only independent prognostic factors for survival.
Collapse
Affiliation(s)
- Luis Felipe Sallum
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Liliana Andrade
- Department of Pathology, University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Susana Ramalho
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Amanda Canato Ferracini
- Program in Medical Sciences, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Rodrigo de Andrade Natal
- Laboratory of Investigative and Molecular Pathology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | | | - Luis Otávio Sarian
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| | - Sophie Derchain
- Department of Obstetrics and Gynecology, State University of Campinas, Campinas, Faculty of Medical Sciences, Campinas, São Paulo, Brazil
| |
Collapse
|
6
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
7
|
Hong L, Zhao X, Shao X, Zhu H. miR-590 regulates WT1 during proliferation of G401 cells. Mol Med Rep 2017; 16:247-253. [PMID: 28498419 PMCID: PMC5482064 DOI: 10.3892/mmr.2017.6561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/22/2017] [Indexed: 01/25/2023] Open
Abstract
Nephroblastoma (Wilms' tumor) is frequently associated with mortality in children. MicroRNAs (miRNAs) are important for tumor development serving as oncogenes or tumor suppressors. In the present study, miRNA-590 (miR-590) was identified to be upregulated in Wilms' tumor tissues compared with the normal adjacent tissues. Additionally, the levels of miR-590 were consistent with their clinical stage. Wilms' tumor 1 (WT1) was considered to be a tumor suppressor in certain tumor types, and it has been detected at low expression levels in various types of cancer with high cell proliferation and aggressive behavior. The expression levels of miR-590 were quantified using reverse transcription-quantitative polymerase chain reaction. Cell proliferation was measured using 5-ethynyl-20-deoxyuridine assays. The protein expression levels of WT1 were investigated by western blot analysis. To the best of our knowledge, the present study was the first to determine that WT1 was a target gene of miR-590 as miR-590 was able to negatively regulate WT1 expression level by binding to the specific target site within the 3′-untranslated region (3′-UTR) of WT1 in G401 cells. Additionally, overexpression of miR-590 promoted G401 cell proliferation which was consistent with the effect of small interfering RNA-WT1. Subsequently, the present study determined that the cell phenotype altered by miR-590 overexpression may be reversed by upregulation of WT1 in G401 cells. In conclusion, the observations indicated that miR-590 may function as an oncogene via targeting WT1 to induce G401 cell proliferation. These results may contribute to current understanding of the function of miR-590 in nephroblastoma.
Collapse
Affiliation(s)
- Liyi Hong
- Clinical Medical Laboratory, Children's Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xu Zhao
- Clinical Medical Laboratory, Children's Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xuejun Shao
- Clinical Medical Laboratory, Children's Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Hong Zhu
- Clinical Medical Laboratory, Children's Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
8
|
Li BQ, Huang S, Shao QQ, Sun J, Zhou L, You L, Zhang TP, Liao Q, Guo JC, Zhao YP. WT1-associated protein is a novel prognostic factor in pancreatic ductal adenocarcinoma. Oncol Lett 2017; 13:2531-2538. [PMID: 28454430 PMCID: PMC5403317 DOI: 10.3892/ol.2017.5784] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
Although Wilms tumor 1 (WT1)-associated protein (WTAP) was initially found to be a specific WT1-binding protein, it has increasingly attracted attention because of its oncogenic role in various types of malignancies, including cholangiocarcinoma, glioblastoma and acute myeloid leukemia. However, the clinical impact of WTAP on pancreatic ductal adenocarcinoma (PDAC) is still unknown. A total of 145 patients who underwent surgical treatment from 2004 to 2008 were enrolled in the present study. The cytoplasmic and nuclear expression of WTAP in tumor and adjacent normal tissues was examined by immunohistochemical analysis in order to investigate the relationship between WTAP and the clinicopathological factors and prognosis of patients with PDAC. The nuclear and cytoplasmic expression of WTAP in tumor tissues was significantly higher compared with non-tumor tissues (P<0.001). High expression of WTAP in the nucleus was significantly associated with gender (P=0.010) and tumor stage (P=0.020), while high expression of WTAP in the cytoplasm was significantly associated with gender (P=0.018), histological grade (P=0.047) and perineural invasion (P=0.028). In addition, a univariate analysis revealed that high nuclear expression of WTAP in tumor tissues was significantly associated with poor overall survival (P<0.001), as well as several clinicopathological variables, including gender and N stage. In a multivariate Cox regression analysis, nuclear WTAP expression was identified as an independent prognostic indicator for PDAC (relative risk, 1.855; 95% confidence interval, 1.033–3.333; P=0.039). The results of the present study indicated that high nuclear expression of WTAP is a valuable molecular biomarker of a poor prognosis among patients with PDAC.
Collapse
Affiliation(s)
- Bing-Qi Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Qian-Qian Shao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
9
|
Graziano ACE, Cardile V, Avola R, Vicario N, Parenti C, Salvatorelli L, Magro G, Parenti R. Wilms' tumor gene 1 silencing inhibits proliferation of human osteosarcoma MG-63 cell line by cell cycle arrest and apoptosis activation. Oncotarget 2017; 8:13917-13931. [PMID: 28107196 PMCID: PMC5355150 DOI: 10.18632/oncotarget.14715] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/06/2017] [Indexed: 12/13/2022] Open
Abstract
Wilms' tumor gene 1 (WT1) plays complex roles in tumorigenesis, acting as tumor suppressor gene or an oncogene depending on the cellular context. A high WT1 expression level was described in various types of human bone and soft-tissue sarcomas, including osteosarcoma (OS), but its function in carcinogenesis is not yet well understood. This study investigated WT1 both in human OS tissues and in human OS MG-63 cell line in which WT1 gene is up-regulated. The results demonstrated that WT1 is expressed in 50% of human OS cases. WT1-silenced MG-63 cells showed deregulation of proteins of cell cycle and down-regulation of PI3K/AKT pathway. Induction of apoptotic programme was also established by activation of caspase-3 and increase of Bax/Bcl2 ratio and p53 protein. This study provided new findings on role of WT1 and indicated an association between WT1 expression, cell cycle and apoptotic machinery. In conclusion, WT1 acts as a tumour promoter in osteosarcoma and it could be a potential therapeutic target.
Collapse
Affiliation(s)
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Rosanna Avola
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, 95125 Catania, Italy
| | - Lucia Salvatorelli
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele” Anatomic Pathology, University of Catania, 95125 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele” Anatomic Pathology, University of Catania, 95125 Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| |
Collapse
|
10
|
Hashimoto H, Zhang X, Zheng Y, Wilson GG, Cheng X. Denys-Drash syndrome associated WT1 glutamine 369 mutants have altered sequence-preferences and altered responses to epigenetic modifications. Nucleic Acids Res 2016; 44:10165-10176. [PMID: 27596598 PMCID: PMC5137435 DOI: 10.1093/nar/gkw766] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 01/10/2023] Open
Abstract
Mutations in human zinc-finger transcription factor WT1 result in abnormal development of the kidneys and genitalia and an array of pediatric problems including nephropathy, blastoma, gonadal dysgenesis and genital discordance. Several overlapping phenotypes are associated with WT1 mutations, including Wilms tumors, Denys-Drash syndrome (DDS), Frasier syndrome (FS) and WAGR syndrome (Wilms tumor, aniridia, genitourinary malformations, and mental retardation). These conditions vary in severity from individual to individual; they can be fatal in early childhood, or relatively benign into adulthood. DDS mutations cluster predominantly in zinc fingers (ZF) 2 and 3 at the C-terminus of WT1, which together with ZF4 determine the sequence-specificity of DNA binding. We examined three DDS associated mutations in ZF2 of human WT1 where the normal glutamine at position 369 is replaced by arginine (Q369R), lysine (Q369K) or histidine (Q369H). These mutations alter the sequence-specificity of ZF2, we find, changing its affinity for certain bases and certain epigenetic forms of cytosine. X-ray crystallography of the DNA binding domains of normal WT1, Q369R and Q369H in complex with preferred sequences revealed the molecular interactions responsible for these affinity changes. DDS is inherited in an autosomal dominant fashion, implying a gain of function by mutant WT1 proteins. This gain, we speculate, might derive from the ability of the mutant proteins to sequester WT1 into unproductive oligomers, or to erroneously bind to variant target sequences.
Collapse
Affiliation(s)
- Hideharu Hashimoto
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xing Zhang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yu Zheng
- RGENE, Inc., 953 Indiana Street, San Francisco, CA 94107, USA
| | | | - Xiaodong Cheng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Kim HS, Kim J, Nam KH, Kim WH. Clinical significance of midkine expression in sporadic desmoid tumors. Oncol Lett 2016; 11:1677-1684. [PMID: 26998061 PMCID: PMC4774436 DOI: 10.3892/ol.2016.4129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 12/23/2015] [Indexed: 11/17/2022] Open
Abstract
The aim of the present study was to identify the prognostic factors for the propensity for recurrence in sporadic desmoid tumors. The catenin (cadherin-associated protein) β1 (CTNNB1) genotypes and expression of Wnt pathway proteins and midkine (also termed neurite growth-promoting factor 2) were investigated in 159 patients with sporadic desmoid tumors. Formalin-fixed paraffin-embedded tissues of the surgically resected desmoid tumors were examined by direct sequencing of CTNNB1 exon 3, and immunostained for the expression of β-catenin, T-cell factor 4 (TCF-4), phosphorylated protein kinase B (pAkt), midkine and menin using a tissue microarray method. Among the samples, 70% (111/159) exhibited point mutations of the CTNNB1 gene, including T41A (56%), S45F (8%), S45P (2%), S45N (2%) and T42A (1%). In addition, 100, 57, 24, 15 and 92% of the tumors expressed β-catenin, TCF-4, midkine, pAkt and menin, respectively. Positive midkine expression was significantly associated with the recurrence of tumors (P=0.001). The multivariate analysis of recurrence demonstrated that an extra-abdominal tumor site [hazard ratio (HR), 2.625; P=0.001] and midkine expression (HR, 2.077; P<0.009) were independent prognostic factors of tumor recurrence. In conclusion, the present results suggest that the tumor site and midkine expression may be predictive markers for the recurrence of sporadic desmoid tumors.
Collapse
Affiliation(s)
- Hee Sung Kim
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 156-755, Republic of Korea
| | - Jin Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Kyung Han Nam
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan 612-896, Republic of Korea
| | - Woo Ho Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; Department of Pathology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| |
Collapse
|
12
|
Taube ET, Denkert C, Sehouli J, Kunze CA, Dietel M, Braicu I, Letsch A, Darb-Esfahani S. Wilms tumor protein 1 (WT1)-- not only a diagnostic but also a prognostic marker in high-grade serous ovarian carcinoma. Gynecol Oncol 2015; 140:494-502. [PMID: 26721227 DOI: 10.1016/j.ygyno.2015.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/15/2015] [Accepted: 12/21/2015] [Indexed: 10/22/2022]
Abstract
AIMS Wilms tumor protein 1 (WT1) expression is used in gynecological pathology as a diagnostic marker of serous differentiation, and is frequently co-expressed with ER-α. Early phase studies on WT1 vaccine in gynecological cancers are ongoing. In this study we aimed to determine the prognostic value of WT1 in high-grade serous ovarian carcinoma. METHODS WT1 protein expression was determined by immunohistochemistry in a cohort of 207 primary high-grade serous ovarian carcinomas. WT1 mRNA expression was evaluated in a cohort of 1137 ovarian carcinomas from publically available gene expression datasets. RESULTS High WT1 expression was a significant positive prognostic factor in primary high-grade serous ovarian carcinoma regarding overall survival (OS, p=0.008) and progression free survival (PFS, p=0.015), which was independent of age, stage, and residual tumor (OS: p=0.024, PFS: p=0.047). The prognostic significance of immunohistochemical WT1 expression could be reproduced in an independent cohort of 72 patients. On the mRNA level the prognostic significance was validated in silico in publically available gene expression datasets including TCGA data (OS: p=0.002, PFS: p=0.011). WT1 expression was significantly linked to ER-α expression (p=0.001), and tumors that co-expressed both markers (WT1+/ER-α+) had a longer survival time than tumors of all other marker combinations (OS: p=0.002, PFS: p=0.013). CONCLUSION We present WT1 as a robust prognostic marker in high-grade serous ovarian carcinoma, which adds prognostic information to ER-α. This should be kept in mind when WT1 is used as a biomarker in the context of WT1-targeting therapies.
Collapse
Affiliation(s)
| | - Carsten Denkert
- Institute of Pathology, Charité University Hospital, Berlin, Germany; Tumor Bank Ovarian Cancer Network (TOC), Berlin, Germany
| | - Jalid Sehouli
- Tumor Bank Ovarian Cancer Network (TOC), Berlin, Germany; Department of Gynecology, Charité University Hospital, Berlin, Germany
| | | | - Manfred Dietel
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Ioana Braicu
- Tumor Bank Ovarian Cancer Network (TOC), Berlin, Germany; Department of Gynecology, Charité University Hospital, Berlin, Germany
| | - Anne Letsch
- Department of Hematology and Oncology, Charité University Hospital, Berlin, Germany
| | - Silvia Darb-Esfahani
- Institute of Pathology, Charité University Hospital, Berlin, Germany; Tumor Bank Ovarian Cancer Network (TOC), Berlin, Germany
| |
Collapse
|
13
|
Parenti R, Cardile V, Graziano ACE, Parenti C, Venuti A, Bertuccio MP, Furno DL, Magro G. Wilms' tumor gene 1 (WT1) silencing inhibits proliferation of malignant peripheral nerve sheath tumor sNF96.2 cell line. PLoS One 2014; 9:e114333. [PMID: 25474318 PMCID: PMC4256418 DOI: 10.1371/journal.pone.0114333] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/06/2014] [Indexed: 12/25/2022] Open
Abstract
Wilms' tumor gene 1 (WT1) plays complex roles in tumorigenesis, acting as tumor suppressor gene or an oncogene depending on the cellular context. WT1 expression has been variably reported in both benign and malignant peripheral nerve sheath tumors (MPNSTs) by means of immunohistochemistry. The aim of the present study was to characterize its potential pathogenetic role in these relatively uncommon malignant tumors. Firstly, immunohistochemical analyses in MPNST sNF96.2 cell line showed strong WT1 staining in nuclear and perinuclear areas of neoplastic cells. Thus, we investigated the effects of silencing WT1 by RNA interference. Through Western Blot analysis and proliferation assay we found that WT1 knockdown leads to the reduction of cell growth in a time- and dose-dependent manner. siWT1 inhibited proliferation of sNF96.2 cell lines likely by influencing cell cycle progression through a decrease in the protein levels of cyclin D1 and inhibition of Akt phosphorylation compared to the control cells. These results indicate that WT1 knockdown attenuates the biological behavior of MPNST cells by decreasing Akt activity, demonstrating that WT1 is involved in the development and progression of MPNSTs. Thus, WT1 is suggested to serve as a potential therapeutic target for MPNSTs.
Collapse
Affiliation(s)
- Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | | | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, 95125 Catania, Italy
| | - Assunta Venuti
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Maria Paola Bertuccio
- Business Unit Oncology, Nerviano Medical Sciences S.r.l., 20014 Nerviano Milano, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95125 Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria “Policlinico-Vittorio Emanuele” Anatomic Pathology, University of Catania, 95125 Catania, Italy
| |
Collapse
|
14
|
Abstract
The WT1 (Wilms' tumour 1) gene encodes a zinc finger transcription factor and RNA-binding protein that direct the development of several organs and tissues. WT1 manifests both tumour suppressor and oncogenic activities, but the reasons behind these opposing functions are still not clear. As a transcriptional regulator, WT1 can either activate or repress numerous target genes resulting in disparate biological effects such as growth, differentiation and apoptosis. The complex nature of WT1 is exemplified by a plethora of isoforms, post-translational modifications and multiple binding partners. How WT1 achieves specificity to regulate a large number of target genes involved in diverse physiological processes is the focus of the present review. We discuss the wealth of the growing molecular information that defines our current understanding of the versatility and utility of WT1 as a master regulator of organ development, a tumour suppressor and an oncogene.
Collapse
|
15
|
Morphologic and immunophenotypic analysis of desmoid-type fibromatosis after radiation therapy. Hum Pathol 2012; 43:1418-24. [PMID: 22404950 DOI: 10.1016/j.humpath.2011.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 10/31/2011] [Accepted: 11/02/2011] [Indexed: 11/24/2022]
|
16
|
Van Driessche A, Berneman ZN, Van Tendeloo VFI. Active specific immunotherapy targeting the Wilms' tumor protein 1 (WT1) for patients with hematological malignancies and solid tumors: lessons from early clinical trials. Oncologist 2012; 17:250-9. [PMID: 22291091 DOI: 10.1634/theoncologist.2011-0240] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
There is a growing body of evidence that Wilms' tumor protein 1 (WT1) is a promising tumor antigen for the development of a novel class of universal cancer vaccines. Recently, in a National Cancer Institute prioritization project, WT1 was ranked first in a list of 75 cancer antigens. In this light, we exhaustively reviewed all published cancer vaccine trials reporting on WT1-targeted active specific immunotherapy in patients with hematological malignancies and solid tumors. In all clinical trials, vaccine-induced immunological responses could be detected. Importantly, objective clinical responses (including stable disease) were observed in 46% and 64% of evaluable vaccinated patients with solid tumors and hematological malignancies, respectively. Immunogenicity of WT1-based cancer vaccines was demonstrated by the detection of a specific immunological response in 35% and 68% of evaluable patients with solid tumors and hematological malignancies, respectively. In order to become part of the armamentarium of the modern oncologist, it will be important to design WT1-based immunotherapies applicable to a large patient population, to standardize vaccination protocols enabling systematic review, and to further optimize the immunostimulatory capacity of the vaccine components. Moreover, improved immunomonitoring tools that reveal clinically relevant T-cell responses will further shape the ideal WT1 immunotherapy strategy. In conclusion, the clinical results obtained so far in WT1-targeted cancer vaccine trials reveal an untapped potential for inducing cancer immunity with minimal side effects and hold promise for a new adjuvant treatment against residual disease and against cancer relapse.
Collapse
Affiliation(s)
- Ann Van Driessche
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VaxInfectio), Faculty of Medicine, University of Antwerp, Antwerp, Belgium.
| | | | | |
Collapse
|
17
|
Colombo C, Creighton CJ, Ghadimi MP, Bolshakov S, Warneke CL, Zhang Y, Lusby K, Zhu S, Lazar AJ, West RB, van de Rijn M, Lev D. Increased midkine expression correlates with desmoid tumour recurrence: a potential biomarker and therapeutic target. J Pathol 2011; 225:574-82. [PMID: 21826666 DOI: 10.1002/path.2951] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/17/2011] [Accepted: 06/05/2011] [Indexed: 12/18/2022]
Abstract
Desmoid tumours (DTs) are soft tissue monoclonal neoplasms exhibiting a unique phenotype, consisting of aggressive local invasiveness without metastatic capacity. While DTs can infrequently occur as part of familial adenomatosis polyposis, most cases arise sporadically. Sporadic DTs harbour a high prevalence of CTNNB1 mutations and hence increased β-catenin signalling. However, β-catenin downstream transcriptional targets and other molecular deregulations operative in DT inception and progression are currently not well defined, contributing to the lack of sensitive molecular prognosticators and efficacious targeted therapeutic strategies. We compared the gene expression profiles of 14 sporadic DTs to those of five corresponding normal tissues and six solitary fibrous tumour specimens. A DT expression signature consisting of 636 up- and 119 down-regulated genes highly enriched for extracellular matrix, cell adhesion and wound healing-related proteins was generated. Furthermore, 98 (15%) of the over-expressed genes were demonstrated to contain a TCF/LEF consensus binding site in their promoters, possibly heralding direct β-catenin downstream targets relevant to DT. The protein products of three of the up-regulated DT genes: ADAM12, MMP2 and midkine, were found to be commonly expressed in a large cohort of human DT samples assembled on a tissue microarray. Interestingly, enhanced midkine expression significantly correlated with a higher propensity and decreased time for primary DT recurrence (log-rank p = 0.0025). Finally, midkine was found to enhance the migration and invasion of primary DT cell cultures. Taken together, these studies provide insights into potential DT molecular aberrations and novel β-catenin transcriptional targets. Further studies to confirm the utility of midkine as a clinical DT molecular prognosticator and a potential therapeutic target are therefore warranted. Raw gene array data can be found at: http://smd.stanford.edu/
Collapse
Affiliation(s)
- Chiara Colombo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center (MDACC), Houston, TX 77054, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Carragher LAS, Snell KR, Giblett SM, Aldridge VSS, Patel B, Cook SJ, Winton DJ, Marais R, Pritchard CA. V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol Med 2011; 2:458-71. [PMID: 20941790 PMCID: PMC3394506 DOI: 10.1002/emmm.201000099] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The majority of human colorectal cancers (CRCs) are initiated by mutations arising in the adenomatous polyposis coli (APC) tumour suppressor gene. However, a new class of non-APC mutated CRCs has been defined that have a serrated histopathology and carry the V600EBRAF oncogene. Here we have investigated the pathogenesis of serrated CRCs by expressing V600EBraf in the proliferative cells of the mouse gastrointestinal tract. We show that the oncogene drives an initial burst of Mek-dependent proliferation, leading to the formation of hyperplastic crypts. This is associated with β-catenin nuclear localization by a mechanism involving Mapk/Erk kinase (Mek)-dependent, Akt-independent phosphorylation of Gsk3β. However, hyperplastic crypts remain dormant for prolonged periods due to the induction of crypt senescence accompanied by upregulation of senescence-associated β-galactosidase and p16Ink4a. We show that tumour progression is associated with down-regulation of p16Ink4a through enhanced CpG methylation of exon 1 and knockout of Cdkn2a confirms this gene is a barrier to tumour progression. Our studies identify V600EBRAF as an early genetic driver mutation in serrated CRCs and indicate that, unlike APC-mutated cancers, this subtype arises by the bypassing of a V600EBraf driven oncogene-induced senescence programme.
Collapse
|
19
|
Abstract
BACKGROUND/PURPOSE The oncogenic properties of the Wilms' tumor gene (WT1) have recently been reported in various malignancies. However, the role of WT1 in pediatric tumors is unclear. To elucidate the role of WT1 in the development of neuroblastoma (NB), we examined the WT1 expression in NB and the effect of WT1 suppression on NB cell proliferation. METHODS We examined the expression of the WT1 protein in 20 NBs and 5 ganglioneuromas (GNs) by performing immunohistochemical analysis. We determined WT1 messenger RNA expression in 22 NBs, 5 GNs, and 4 NB cell lines by real-time reverse transcription polymerase chain reaction. We studied the effects of WT1 suppression on cell proliferation using small interfering RNA against WT1. RESULTS Expression of WT1 was higher in mature ganglionic cells, and in the immunohistochemical analysis, the WT1 positivity for GNs was significantly higher than that for NBs (P < .01). The level of WT1 messenger RNA expression did not correlate with histologic grade, clinical stage, and prognosis of the tumor. Knockdown of WT1 gene promoted the proliferation of NB69 cells (P < .01). CONCLUSIONS The WT1 may govern cell differentiation and suppress cell proliferation in NB. The WT1 does not act as an oncogene, but it may participate in the maturation of NB.
Collapse
|
20
|
Joyner DE, Trang SH, Damron TA, Aboulafia AJ, Cummings JE, Randall RL. Desmoid cell motility is induced in vitro by rhEGF. J Orthop Res 2009; 27:1258-62. [PMID: 19242968 DOI: 10.1002/jor.20865] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Desmoid tumors are benign but locally invasive myofibroblastic lesions that arise predominantly in the abdominal wall or shoulder and are prone to aggressive local recurrences. A perceived association between desmoid activity and the expression of growth factors during pregnancy or following trauma suggests a cause-and-effect relationship between growth factor stimulation and desmoid invasiveness. We used Boyden Chambers to quantify cell motility in order to determine the effect of growth factor stimulation on desmoid cell migration. Desmoid cell cultures were treated under serum-free conditions with epidermal growth factor (rhEGF) or transforming growth factor alpha (rhTGFalpha). Additional cell cultures were pretreated under serum-free conditions with the EGF receptor (EGFR) inhibitor AG1478, alone or in combination with the TGFbeta1 receptor inhibitor SB431542, and then stimulated with growth factor prior to being assayed for cell motility. The experiments demonstrated a direct dose-dependent relationship between rhEGF stimulation and desmoid motility. In contrast, rhTGFalpha was less effective at inducing cell migration. rhEGF-induced cell migration could be diminished, but not reduced to control levels, by inhibiting EGFR. When EGF and TGFbeta1 receptors were inhibited simultaneously, the level of rhEGF-induced cell migration was reduced significantly beyond the level of cell migration generated by inhibition of EGFR alone.
Collapse
Affiliation(s)
- David E Joyner
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah 84112, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Garrido-Ruiz MC, Rodriguez-Pinilla SM, Pérez-Gómez B, Rodriguez-Peralto JL. WT 1 expression in nevi and melanomas: a marker of melanocytic invasion into the dermis. J Cutan Pathol 2009; 37:542-8. [PMID: 19638168 DOI: 10.1111/j.1600-0560.2009.01379.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND WT1, first recognized as a tumor suppressor gene involved in the development of Wilms' tumor, may have apparently contradictory findings and functions. As WT1 has been identified as a molecular target for cancer immunotherapy, immunodetection of WT1 in tumor cells has become an essential step in cancer studies. METHODS We compare the expression of this protein among different types of melanocytic nevi and among stages in primary melanoma progression. Tissue microarrays containing normal tissues and 271 primary melanocytic lesion samples (163 primary melanomas and 108 nevi) were studied by immunohistochemistry using monoclonal antibody against WT1. RESULTS AND DISCUSSION The present study shows these: 1. WT1 protein is predominantly expressed in the cytoplasm of the neoplastic cells. 2. A higher rate of WT1 staining in melanocytic nevi against melanomas has been observed. 3. WT1 expression is increased in advanced stages of melanoma progression: a significant (p < 0.05) increase of expression of WT1 was detected in vertical cases 46.5% vs. radial cases 16.0%, in high levels of Clark (IV, V) 57.4% vs. low levels (I, II, III) 33.0% and when comparing depth of invasion within thickness subgroups. 4. Finally, this study establishes an association of WT1 protein expression with shorter overall survival in melanoma.
Collapse
Affiliation(s)
- M C Garrido-Ruiz
- Dpto. de Anatomía Patológica, Hospital Universitario12 de Octubre, Avda de Córdoba s/n, 28041 Madrid, Spain
| | | | | | | |
Collapse
|
22
|
Abstract
The Wilms' tumour-suppressor gene (WT1), encodes a zinc-finger transcription factor that is critical for the development of several organs, including the kidneys, gonads and spleen. Despite its identification as a tumour suppressor that plays a crucial role in the formation of a paediatric malignancy of the kidneys (Wilms' tumour), it has also emerged as an oncogenic factor influencing proliferation and apoptosis in a large variety of adult cancers. This review focuses on new insights into WT1's role in early development and its potential oncogenic role in adult cancer.
Collapse
|
23
|
Schittenhelm J, Beschorner R, Simon P, Tabatabai G, Herrmann C, Schlaszus H, Capper D, Weller M, Meyermann R, Mittelbronn M. Diagnostic value of WT1 in neuroepithelial tumours. Neuropathol Appl Neurobiol 2008; 35:69-81. [PMID: 18466223 DOI: 10.1111/j.1365-2990.2008.00957.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS Currently, clinical trials using WT1 (Wilms tumour gene) peptide vaccines are conducted in haematopoietic malignancies and solid cancers. Single reports showed that the Wilms tumour gene product WT1 is also expressed in astrocytic neoplasms. Our aim was to investigate WT1 expression in a large cohort of various neuroepithelial tumours of different World Health Organization (WHO) grades and in normal central nervous system (CNS) tissue specimens to test its potential value as a diagnostic marker. METHODS Specimens were assessed by RT-PCR, Western blotting and immunohistochemistry. The samples investigated in our study consisted of 334 human neuroepithelial tumours, among those 33 oligodendrogliomas, 219 astrocytomas (including 105 glioblastomas) and 47 ependymomas. RESULTS Our results showed a de novo WT1 expression in neuroepithelial tumours. In diffuse astrocytomas and ependymomas, WT1 expression increased significantly with the grade of malignancy. In contrast, no significant difference was seen between WHO grade-II and -III oligodendrogliomas. Controlling for WHO grade, the comparison of oligodendrogliomas with ependymal and astrocytic tumours showed higher expression values for the latter. CONCLUSIONS Our study shows that WT1 is expressed de novo in numerous neuroepithelial tumours and increases with the grade of malignancy. These results suggest an important role of WT1 in tumourigenesis and progression in human brain tumours.
Collapse
Affiliation(s)
- J Schittenhelm
- Institute of Brain Research, University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Riggi N, Cironi L, Suvà ML, Stamenkovic I. Sarcomas: genetics, signalling, and cellular origins. Part 1: The fellowship of TET. J Pathol 2007; 213:4-20. [PMID: 17691072 DOI: 10.1002/path.2209] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Sarcomas comprise some of the most aggressive solid tumours that, for the most part, respond poorly to chemo- and radiation therapy and are associated with a sombre prognosis when surgical removal cannot be performed or is incomplete. Partly because of their lower frequency, sarcomas have not been studied as intensively as carcinomas and haematopoietic malignancies, and the molecular mechanisms that underlie their pathogenesis are only beginning to be understood. Even more enigmatic is the identity of the primary cells from which these tumours originate. Over the past 25 years, however, several non-random chromosomal translocations have been found to be associated with defined sarcomas. Each of these translocations generates a fusion gene believed to be directly related to the pathogenesis of the sarcoma in which it is expressed. The corresponding fusion proteins provide a unique tool not only to study the process of sarcoma development, but also to identify cells that are permissive for their putative oncogenic properties. This is the first of two reviews that cover the mechanisms whereby specific fusion/mutant gene products participate in sarcoma development and the cellular context that may provide the necessary permissiveness for their expression and oncogenicity. Part 1 of the review focuses on sarcomas that express fusion genes containing TET gene family products, including EWSR1, TLS/FUS, and TAFII68. Part 2 (J Pathol 2007; DOI: 10.1002/path.2008) summarizes our current understanding of the genetic and cellular origins of sarcomas expressing fusion genes exclusive of TET family members; it also covers soft tissue malignancies harbouring specific mutations in RTK-encoding genes, the prototype of which are gastrointestinal stromal tumours (GIST).
Collapse
Affiliation(s)
- N Riggi
- Division of Experimental Pathology, Institute of Pathology, University of Lausanne, Lausanne, Switzerland
| | | | | | | |
Collapse
|
25
|
Durno C, Monga N, Bapat B, Berk T, Cohen Z, Gallinger S. Does early colectomy increase desmoid risk in familial adenomatous polyposis? Clin Gastroenterol Hepatol 2007; 5:1190-4. [PMID: 17916546 DOI: 10.1016/j.cgh.2007.06.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Desmoid tumors are non-metastasizing fibromatoses that occur in 10%-20% of subjects with familial adenomatous polyposis (FAP). Intra-abdominal desmoid tumors are a major cause of mortality in FAP. FAP-associated desmoid tumors are linked to trauma, particularly abdominal surgery, family history of desmoids, hormonal factors, and the location of the APC mutation. We hypothesized that prophylactic colectomy at an early age might increase the risk of developing desmoids. The aim of this study was to determine whether colectomy earlier in life is a risk factor for the development of desmoid tumors. METHODS An analysis was made of the association between development of desmoid and age at colectomy, family history of desmoids, gender, and APC mutation in FAP patients in the Registry (1980-2005) at Mount Sinai Hospital, Toronto, Ontario, Canada. RESULTS FAP patients (n = 930) from 365 kindreds were identified. Desmoid prevalence was 14% (n = 121). Female patients were more likely to develop desmoids than male patients (17% vs 11%, P = .03). Female patients who had an early colectomy were more than 2 times more likely to develop a desmoid, compared with women who had a colectomy at >18 years (P = .01). Early colectomy did not increase risk of developing a desmoid in male patients (P = .42). Female patients who had an early colectomy (</=18 years) were 2.5 times more likely to develop desmoids, compared with male patients who had a late colectomy (P = .05). The prevalence of desmoids in the 5' and 3' groups was 13% and 38%, respectively (P = .0005). Patients with a mutation after codon 1399 were found to have 4 times greater chance of developing a desmoid. CONCLUSIONS Female patients with FAP are more likely to develop desmoids than male patients. Female patients who had an early colectomy are at significantly greater risk of developing a desmoid compared with female patients who had a colectomy in adulthood. Patients with APC mutations beyond codon 1399 are more likely to develop desmoids. These results suggest that delayed colectomy might be considered in young female patients with FAP to decrease the chances of developing desmoids.
Collapse
Affiliation(s)
- Carol Durno
- Familial Gastrointestinal Cancer Registry and Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Coosemans A, Nik SA, Caluwaerts S, Lambin S, Verbist G, Van Bree R, Schelfhout V, de Jonge E, Dalle I, Jacomen G, Cassiman JJ, Moerman P, Vergote I, Amant F. Upregulation of Wilms’ tumour gene 1 (WT1) in uterine sarcomas. Eur J Cancer 2007; 43:1630-7. [PMID: 17531467 DOI: 10.1016/j.ejca.2007.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 04/03/2007] [Accepted: 04/05/2007] [Indexed: 11/27/2022]
Abstract
AIM Overexpression of Wilms' tumour gene (WT1) has been proven in several tumours. Previous research of our group on the cell cycle of uterine leiomyosarcoma (LMS) and carcinosarcoma (CS) suggested a possible role for WT1. We therefore intended to further explore the expression pattern of WT1 in uterine sarcomas. METHODS 27 CS, 38 LMS, 15 endometrial stromal sarcomas (ESS) and seven undifferentiated sarcomas (US) were collected. WT1 expression was evaluated by immunohistochemistry (IHC) in 87 samples, by RT-PCR (m-RNA expression) in 23 random selected samples and by Western blotting in 12 samples, separating cytoplasmic and nuclear proteins. A pilot study to detect mutations (exons 7-10) was performed on eight samples. RESULTS IHC showed WT1 positivity in 12/27 CS, 29/38 LMS, 7/15 ESS and 4/7 US. All-but-one sample had a positive RT-PCR. All Western blottings were positive with more cytoplasmic expression in 9/12 cases. No mutations were found. CONCLUSIONS WT1 is overexpressed in uterine sarcomas. Since increased levels of mRNA determine the biological role, WT1 might contribute to uterine sarcoma tumour biology.
Collapse
Affiliation(s)
- A Coosemans
- Leuven Cancer Institute (LKI), UZ Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Seinfeld J, Kleinschmidt-DeMasters BK, Tayal S, Lillehei KO. Desmoid-type fibromatosis involving the brachial plexus. Neurosurg Focus 2007; 22:E22. [PMID: 17613214 DOI: 10.3171/foc.2007.22.6.23] [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: 11/06/2022]
Abstract
✓Desmoid-type fibromatosis involving the brachial plexus is a rare and challenging disease. Due to involvement of crucial neurovascular structures, wide local excision of the associated fibromas is rarely feasible and recurrence is common. The authors describe their experience in four surgically treated patients with desmoid-type fibromatosis involving the brachial plexus and review the relevant neurosurgical literature.
All tumors were assessed for c-KIT oncogene mutations in hopes of establishing a biological basis for using the tyrosine kinase inhibitor imatimib mesylate as an adjuvant therapy. Three patients experienced tumor recurrence requiring reoperation. Fractionated radiotherapy achieved local control in three patients, and the disease in one patient progressed beyond the treatment field. Single base pair changes at exon 10 of the c-KIT oncogene were identified in three tumors. One tumor with this mutation did not respond to treatment with imatimib mesylate. A review of the literature revealed 17 additional patients in two different case series. Analysis of these cases emphasizes the need for careful resection in patients with desmoid-type fibromatosis and supports the conclusion that without adjuvant radiotherapy a high local recurrence rate can be anticipated. For optimal local disease control, the authors recommend post-surgical radiation therapy regardless of the extent of resection achieved. The mutational status of the c-KIT oncogene remains an intriguing biological marker that in the future may predict which lesions will be responsive to imatimib mesylate; larger series will be necessary to test this hypothesis.
Collapse
Affiliation(s)
- Joshua Seinfeld
- Department of Neurosurgery, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | |
Collapse
|
28
|
Yang L, Han Y, Suarez Saiz F, Saurez Saiz F, Minden MD. A tumor suppressor and oncogene: the WT1 story. Leukemia 2007; 21:868-76. [PMID: 17361230 DOI: 10.1038/sj.leu.2404624] [Citation(s) in RCA: 329] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Wilms' tumor 1 (WT1) gene encodes a transcription factor important for normal cellular development and cell survival. The initial discovery of WT1 as the causative gene in an autosomal-recessive condition identified it as a tumor suppressor gene whose mutations are associated with urogenital disease and the development of kidney tumors. However, this view is not in keeping with the frequent finding of wild-type, full-length WT1 in human leukemia, breast cancer and several other cancers including the majority of Wilms' tumors. Rather, these observations suggest that in those conditions, WT1 has an oncogenic role in tumor formation. In this review, we explore the literature supporting both views of WT1 in human cancer and in particular human leukemias. To understand the mechanism by which WT1 can do this, we will also examine its functional activity as a transcription factor and the influence of protein partners on its dual behavior.
Collapse
Affiliation(s)
- L Yang
- Department of Cellular and Molecular Biology, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
29
|
Abstract
When connective tissue undergoes malignant transformation, glioblastomas and sarcomas arise. However, the ancient biochemical mechanisms, which are now operational in sarcomas distorted by mutations and gene fusions in misaligned chromosomes, were originally acquired by those cells that emerged during the Cambrian explosion. Preserved throughout evolution up to the genus Homo, these mechanisms dictate the apoptosis- and senescence-resistant immortality of malignant cells. A 'retroviral paradox' distinguishes human sarcomas from those of the animal world. In contrast to the retrovirally induced sarcomatous transformation of animal (avian, murine, feline and simian) cells, human sarcomas have so far failed to yield a causative retroviral isolate. However, the proto-oncogenes/oncogenes transduced from their host cells by retroviruses of animals are the same that are active in human sarcomas. Since the encoded oncoproteins arise after birth, they are recognized frequently by the immune system of the host. Immune lymphocytes that kill autologous sarcoma cells in vitro commonly fail to do so in vivo. Sarcoma vaccines generate immune T- and natural killer cell reactions; even when vaccinated patients do not show a clinical response, their tumors become more sensitive to chemotherapy. The aim of this review is to lay a solid molecular biological foundation for the conclusion that targeting the sarcoma oncogenes will result in regression of the disease.
Collapse
Affiliation(s)
- Joseph G Sinkovics
- Cancer Institute of St. Joseph's Hospital Affiliated with the HL Moffitt Cancer Center, The University of South Florida College of Medicine, Department of Medical Microbiology and Immunology, Tampa, Florida, USA.
| |
Collapse
|
30
|
Abstract
Over the years, many apparently contradictory findings and functions have been ascribed to the protein product of the WT1 tumour suppressor gene. These include being a transcriptional activator or repressor, a function in transcription versus RNA metabolism, and these days even a function as oncogene or tumour suppressor gene. To fully understand the role of WT1 in different diseases and normal development, we will need to understand these contradictions. In this review, we will discuss the present state of knowledge and suggest that a role for WT1 in influencing the mesenchymal-epithelial state of cells might be a common function that could explain many of the previously described findings.
Collapse
Affiliation(s)
- Peter Hohenstein
- MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | | |
Collapse
|
31
|
Seinfeld J, Kleinschmidt-Demasters BK, Tayal S, Lillehei KO. Desmoid-type fibromatoses involving the brachial plexus: treatment options and assessment of c-KIT mutational status. J Neurosurg 2006; 104:749-56. [PMID: 16703880 DOI: 10.3171/jns.2006.104.5.749] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Desmoid-type fibromatoses are a locally invasive soft-tissue lesion that is most commonly encountered in abdominal sites. The tumor also affects head and neck areas, particularly the supraclavicular region, where it may encase and distort the brachial plexus and compromise neurovascular structures. Neurosurgeons may be called on to treat desmoid-type fibromatoses in these sites. The authors describe their experience in treating four patients with desmoid-type fibromatoses involving the brachial plexus and report the results of immunohistochemical analysis of the tumors. METHODS Gross-total excision with nerve sparing was the first-line therapy of choice, although the surgery was challenging. Intraoperative identification of the site of tumor origin from musculoaponeurotic tissues by the neurosurgeon was necessary in two of the four cases to achieve a correct frozen section or final pathological diagnosis. Immunostaining for c-KIT (CD117) was undertaken in all cases in light of a previous report of positive CD117 immunoreactivity in abdominal desmoid-type fibromatoses. All four tumors manifested weak focal immunostaining for c-KIT. One of the patients was given adjuvant imatinib mesylate therapy, with limited success. Subsequent polymerase chain reaction testing revealed that three of the four tumors manifested a single base pair change in exon 10 of the c-KIT gene (A to C in two cases and A to G in one case). There was local recurrence in three patients, despite gross-total excision. With the combination of surgery and radiation therapy, local disease control was achieved in three of the four patients. CONCLUSIONS This represents the first report of c-KIT sequencing in desmoid-type fibromatoses and suggests a possible biological basis for continuing to explore the use of adjuvant imatinib mesylate therapy.
Collapse
Affiliation(s)
- Joshua Seinfeld
- Department of Neurosurgery, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | |
Collapse
|
32
|
Bacac M, Migliavacca E, Stehle JC, McKee T, Delorenzi M, Coindre JM, Guillou L, Stamenkovic I. A gene expression signature that distinguishes desmoid tumours from nodular fasciitis. J Pathol 2006; 208:543-53. [PMID: 16440290 DOI: 10.1002/path.1915] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nodular fasciitis (NF) is a rapidly growing cellular mass composed of fibroblasts/myofibroblasts, usually localized in subcutaneous tissues, that typically undergoes fibrosis and almost never recurs. Desmoid tumours (DTs) are rare forms of fibroblastic/myofibroblastic growth that arise in deep soft tissues, display a propensity for local infiltration and recurrence, but fail to metastasize. Given that both entities are primarily fibroblastic/myofibroblastic lesions with overlapping histological features, their gene expression profiles were compared to identify differentially expressed genes that may provide not only potential diagnostic markers, but also clues as to the pathogenesis of each disorder. Differentially expressed transcripts (89 clones displaying increased expression in DTs and 246 clones displaying increased expression in NF) included genes encoding several receptor and non-receptor tyrosine kinases (EPHB3, PTPRF, GNAZ, SYK, LYN, EPHA4, BIRC3), transcription factors (TWIST1, PITX2, EYA2, OAS1, MITF, TCF20), and members of the Wnt signalling pathway (AXIN2, WISP1, SFRP). Remarkably, almost one-quarter of the differentially expressed genes encode proteins associated with inflammation and tissue remodelling, including members of the interferon (IFN), tumour necrosis factor (TNF), and transforming growth factor beta (TGF-beta) signalling pathways as well as metalloproteinases (MMP1, 9, 13, 23), urokinase plasminogen activator (PLAU), and cathepsins. The observations provide the first comparative molecular characterization of desmoid tumours and nodular fasciitis and suggest that selected tyrosine kinases, transcription factors, and members of the Wnt, TGF-beta, IFN, and TNF signalling pathways may be implicated in influencing and distinguishing their fate.
Collapse
Affiliation(s)
- M Bacac
- Division of Experimental Pathology, University Institute of Pathology, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Desmoid tumors (also called deep fibromatoses) are rare benign tumors associated with pregnancy and Gardner syndrome. These tumors are characterized by bland-appearing fibroblasts, indistinct margins, and an ability to cause pathology by local invasion and recurrence. They arise in the abdominal cavity, in the abdominal wall, or in the extremities/trunk, each with a slightly different biologic behavior. Though they are not cancer and do not metastasize, desmoids can cause significant morbidity and occasionally death through local/regional invasion of critical structures. Treatment primarily is surgical, although radiation or systemic therapy can be beneficial to the patient when surgery is not feasible. This article highlights the biology and clinical features of desmoid tumors.
Collapse
Affiliation(s)
- Marcus Schlemmer
- Medical Clinic and Polyclinic III, Clinic Grosshadern Munich, Ludwig-Maximilian-University Munich, Marchioninistrasse 15, Muenchen D-81377, Germany.
| |
Collapse
|