1
|
López Riquelme I, Martínez García S, Serrano Ordónez A, Martínez Pilar L. Germline mutations predisposing to melanoma and associated malignancies and syndromes: a narrative review. Int J Dermatol 2024. [PMID: 39651613 DOI: 10.1111/ijd.17602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
The pathogenesis of melanoma is influenced by a complex combination of environmental factors and individual genetic susceptibility. Familial melanoma refers to cases where there are two first-degree relatives with a melanoma diagnosis. Less strict definitions include second-degree relatives or even three or more of any degree from the same family, although this is not clearly defined in the literature. The term hereditary melanoma is reserved for sporadic or familial melanomas linked to high-risk genes with high penetrance. The first genes related to melanoma were CDKN2A and CDK4, but recently, other genes, mostly tumor suppressor genes, have been described. Internal malignancies, particularly pancreatic cancer, have also been associated with melanoma. Recent studies suggest that there could be a link between melanoma and other neoplasms and tumor predisposition syndromes. This review presents an updated overview of familial melanoma criteria and genes involved in melanoma pathogenesis, emphasizing their clinicopathological aspects and other associated malignancies.
Collapse
Affiliation(s)
- Irene López Riquelme
- Dermatology Department, Hospital Regional Universitario de Málaga, Malaga, Spain
| | | | - Ana Serrano Ordónez
- Dermatology Department, Hospital Regional Universitario de Málaga, Malaga, Spain
| | | |
Collapse
|
2
|
Faja F, Esteves S, Pallotti F, Cicolani G, Di Chiano S, Delli Paoli E, Lenzi A, Lombardo F, Paoli D. Environmental disruptors and testicular cancer. Endocrine 2022; 78:429-435. [PMID: 36036879 PMCID: PMC9637059 DOI: 10.1007/s12020-022-03171-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 01/01/2023]
Abstract
PURPOSE Testicular cancer (TC) is the most common malignancy among young adult males. The etiology is multifactorial, and both environmental and genetic factors play an essential role in the origin and development of this tumor. In particular, exposure to environmental endocrine disruptors (EEDs), resulting from industrialization and urbanization, seems crucial both in pre-and postnatal life. However, the lack of long-term studies on a wide caseload and the difficulty in evaluating their toxic effects in vivo make it challenging to establish a causal link. This review aims to discuss the main human epidemiological studies currently available in the literature to define a possible association between these chemicals and TC. METHODS A comprehensive Medline/PubMed and Embase search was performed, selecting all relevant, peer-reviewed papers in English published from 2002 to January 2022. Other relevant papers were selected from the reference lists. RESULTS To date, literature evidence is limited due to the scarcity and heterogeneity of human studies and shows controversial data, highlighting the complexity of the topic. However, most human epidemiological studies seem to point toward a correlation between EEDs exposure and TC. CONCLUSION Although the molecular mechanisms are not yet fully understood, the role of EEDs in TC onset is plausible, but several factors, such as the individual genetic background, the exposure time, and the complex mechanism of action of these chemicals, do not allow defining the causal link with certainty and make further studies necessary to investigate this complex topic.
Collapse
Affiliation(s)
- Fabiana Faja
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Sandro Esteves
- Andrology and Human Reproduction Clinic, Av. Dr. Heitor Penteado, 1464, Campinas, Brazil
- Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Francesco Pallotti
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Gaia Cicolani
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Silvia Di Chiano
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Enrico Delli Paoli
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Andrea Lenzi
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Francesco Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy
| | - Donatella Paoli
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" Università di Roma, Rome, Italy.
| |
Collapse
|
3
|
Zarkower D, Murphy MW. DMRT1: An Ancient Sexual Regulator Required for Human Gonadogenesis. Sex Dev 2022; 16:112-125. [PMID: 34515237 PMCID: PMC8885888 DOI: 10.1159/000518272] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/25/2021] [Indexed: 01/03/2023] Open
Abstract
Transcriptional regulators related to the invertebrate sexual regulators doublesex and mab-3 occur throughout metazoans and control sex in most animal groups. Seven of these DMRT genes are found in mammals, and mouse genetics has shown that one, Dmrt1, plays a crucial role in testis differentiation, both in germ cells and somatic cells. Deletions and, more recently, point mutations affecting human DMRT1 have demonstrated that its heterozygosity is associated with 46,XY complete gonadal dysgenesis. Most of our detailed knowledge of DMRT1 function in the testis, the focus of this review, derives from mouse studies, which have revealed that DMRT1 is essential for male somatic and germ cell differentiation and maintenance of male somatic cell fate after differentiation. Moreover, ectopic DMRT1 can reprogram differentiated female granulosa cells into male Sertoli-like cells. The ability of DMRT1 to control sexual cell fate likely derives from at least 3 properties. First, DMRT1 functionally collaborates with another key male sex regulator, SOX9, and possibly other proteins to maintain and reprogram sexual cell fate. Second, and related, DMRT1 appears to function as a pioneer transcription factor, binding "closed" inaccessible chromatin and promoting its opening to allow binding by other regulators including SOX9. Third, DMRT1 binds DNA by a highly unusual form of interaction and can bind with different stoichiometries.
Collapse
Affiliation(s)
- David Zarkower
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Mark W. Murphy
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Familial Melanoma and Susceptibility Genes: A Review of the Most Common Clinical and Dermoscopic Phenotypic Aspect, Associated Malignancies and Practical Tips for Management. J Clin Med 2021; 10:jcm10163760. [PMID: 34442055 PMCID: PMC8397216 DOI: 10.3390/jcm10163760] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
A family history of melanoma greatly increases the risk of developing cutaneous melanoma, a highly aggressive skin cancer whose incidence has been steadily increasing worldwide. Familial melanomas account for about 10% of all malignant melanomas and display an inheritance pattern consistent with the presence of pathogenic germline mutations, among which those involving CDKN2A are the best characterized. In recent years, a growing number of genes, such as MC1R, MITF, CDK4, POT1, TERT, ACD, TERF2IP, and BAP1, have been implicated in familial melanoma. The fact that individuals harboring these germline mutations along with their close blood relatives have a higher risk of developing multiple primary melanomas as well as other internal organ malignancies, especially pancreatic cancer, makes cascade genetic testing and surveillance of these families of the utmost importance. Unfortunately, due to a polygenic inheritance mechanism involving multiple low-risk alleles, genetic modifiers, and environmental factors, it is still very difficult to predict the presence of these mutations. It is, however, known that germline mutation carriers can sometimes develop specific clinical traits, such as high atypical nevus counts and specific dermoscopic features, which could theoretically help clinicians predict the presence of these mutations in prone families. In this review, we provide a comprehensive overview of the high- and intermediate-penetrance genes primarily linked to familial melanoma, highlighting their most frequently associated non-cutaneous malignancies and clinical/dermoscopic phenotypes.
Collapse
|
5
|
Hellesnes R, Myklebust TÅ, Bremnes RM, Karlsdottir Á, Kvammen Ø, Negaard HFS, Tandstad T, Wilsgaard T, Fosså SD, Haugnes HS. Metachronous Contralateral Testicular Cancer in the Cisplatin Era: A Population-Based Cohort Study. J Clin Oncol 2020; 39:308-318. [PMID: 33356420 DOI: 10.1200/jco.20.02713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE It is hypothesized that cisplatin-based chemotherapy (CBCT) reduces the occurrence of metachronous contralateral (second) germ cell testicular cancer (TC). However, studies including treatment details are lacking. The aim of this study was to assess the second TC risk, emphasizing the impact of previous TC treatment. PATIENTS AND METHODS Based on the Cancer Registry of Norway, 5,620 men were diagnosed with first TC between 1980 and 2009. Treatment data regarding TC were retrieved from medical records. Cumulative incidences of second TC were estimated, and standardized incidence ratios were calculated. The effect of treatment intensity was investigated using Cox proportional hazard regression. RESULTS Median follow-up was 18.0 years, during which 218 men were diagnosed with a second TC after median 6.2 years. Overall, the 20-year crude cumulative incidence was 4.0% (95% CI, 3.5 to 4.6), with lower incidence after chemotherapy (CT) (3.2%; 95% CI, 2.5 to 4.0) than after surgery only (5.4%; 95% CI, 4.2 to 6.8). The second TC incidence was also lower for those age ≥ 30 years (2.8%; 95% CI, 2.3 to 3.4) at first TC diagnosis than those age < 30 years (6.0%; 95% CI, 5.0 to 7.1). Overall, the second TC risk was 13-fold higher compared with the risk of developing TC in the general male population (standardized incidence ratio, 13.1; 95% CI, 11.5 to 15.0). With surgery only as reference, treatment with CT significantly reduced the second TC risk (hazard ratio [HR], 0.55). For each additional CBCT cycle administered, the second TC risk decreased significantly after three, four, and more than four cycles (HRs, 0.53, 0.41, and 0.21, respectively). CONCLUSION Age at first TC diagnosis and treatment intensity influenced the second TC risk, with significantly reduced risks after more than two CBCT cycles.
Collapse
Affiliation(s)
- Ragnhild Hellesnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Tor Åge Myklebust
- Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway.,Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Ása Karlsdottir
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Øivind Kvammen
- Department of Oncology, Ålesund Hospital, Ålesund, Norway
| | | | - Torgrim Tandstad
- The Cancer Clinic, St. Olav's University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom Wilsgaard
- Department of Community Medicine, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Sophie D Fosså
- Department of Registration, Cancer Registry of Norway, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hege S Haugnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT, The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
Xie Y, Wei BH, Ni FD, Yang WX. Conversion from spermatogonia to spermatocytes: Extracellular cues and downstream transcription network. Gene 2020; 764:145080. [PMID: 32858178 DOI: 10.1016/j.gene.2020.145080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
Spermatocyte (spc) formation from spermatogonia (spg) differentiation is the first step of spermatogenesis which produces prodigious spermatozoa for a lifetime. After decades of studies, several factors involved in the functioning of a mouse were discovered both inside and outside spg. Considering the peculiar expression and working pattern of each factor, this review divides the whole conversion of spg to spc into four consecutive development processes with a focus on extracellular cues and downstream transcription network in each one. Potential coordination among Dmrt1, Sohlh1/2 and BMP families mediates Ngn3 upregulation, which marks progenitor spg, with other changes. After that, retinoic acid (RA), as a master regulator, promotes A1 spg formation with its helpers and Sall4. A1-to-B spg transition is under the control of Kitl and impulsive RA signaling together with early and late transcription factors Stra8 and Dmrt6. Finally, RA and its responsive effectors conduct the entry into meiosis. The systematic transcription network from outside to inside still needs research to supplement or settle the controversials in each process. As a step further ahead, this review provides possible drug targets for infertility therapy by cross-linking humans and mouse model.
Collapse
Affiliation(s)
- Yi Xie
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bang-Hong Wei
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei-Da Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Wang J, Li S, Lin S, Fu S, Qiu L, Ding K, Liang K, Du H. B-cell lymphoma 2 family genes show a molecular pattern of spatiotemporal heterogeneity in gynaecologic and breast cancer. Cell Prolif 2020; 53:e12826. [PMID: 32419250 PMCID: PMC7309952 DOI: 10.1111/cpr.12826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/20/2022] Open
Abstract
Objectives BCL2 family proteins have been widely studied over the past decade due to their essential roles in apoptosis, oncogenesis and anti‐cancer therapy. However, the similarities and differences in the spatial pattern of the BCL2 gene family within the context of chromatin have not been well characterized. We sought to fill this knowledge gap by assessing correlations between gene alteration, gene expression, chromatin accessibility, and clinical outcomes in gynaecologic and breast cancer. Materials and methods In this study, the molecular characteristics of the BCL2 gene family in gynaecologic cancer were systematically analysed by integrating multi‐omics datasets, including transcriptomics, chromatin accessibility, copy number variation, methylomics and clinical outcome. Results We evaluated spatiotemporal associations between long‐range regulation peaks and tumour heterogeneity. Differential expression of the BCL2 family was coupled with widespread chromatin accessibility changes in gynaecologic cancer, accompanied by highly heterogeneous distal non‐coding accessibility surrounding the BCL2L1 gene loci. A relationship was also identified between gene expression, gene amplification, enhancer signatures, DNA methylation and overall patient survival. Prognostic analysis implied clinical correlations with BAD, BIK and BAK1. A shared protein regulatory network was established in which the co‐mutation signature of TP53 and PIK3CA was linked to the BCL2L1 gene. Conclusions Our results provide the first systematic identification of the molecular features of the BCL2 family under the spatial pattern of chromatin in gynaecologic and breast cancer. These findings broaden the therapeutic scope of the BCL2 family to the non‐coding region by including a significantly conserved distal region overlaying an enhancer.
Collapse
Affiliation(s)
- Jiajian Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Sidi Li
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shudai Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Li Qiu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Ke Ding
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Keying Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Hoshi S, Bilim V, Hoshi K, Sasagawa I, Otake K, Chiba D, Suenaga S, Konno M, Katsumata Y, Morozumi K, Takemoto J, Numahata K, Ito K. Familial testicular germ cell tumors in two brothers. IJU Case Rep 2020; 3:100-102. [PMID: 32743483 PMCID: PMC7292156 DOI: 10.1002/iju5.12156] [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: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Two percent of testicular germ cell tumors occur in family clusters. Here, we report metachronous testicular germ cell tumors in two brothers. CASE PRESENTATION An elder brother was diagnosed at the age of 30 years old and the pathological diagnosis was mixed testicular germ cell tumor. A tumor in the younger brother was suspected during testicular self-examination. It was confirmed by ultrasound examination at the age of 30 years old, 3 years and 6 months after the diagnosis of the testicular tumor in elder brother. The pathological diagnosis was pure seminoma. Both brothers had stage 1 testicular germ cell tumors and no recurrence was observed during the follow-up period of 4 years and 4 months and 10 months, respectively. CONCLUSION Various histological types of tumor can occur in members of one family. Besides genetic predisposition, shared diet, environmental exposure and other factors can contribute to the familial testicular cancer. Testicular self-examination is recommended for family members of a person with testicular germ cell tumor.
Collapse
Affiliation(s)
- Senji Hoshi
- Department of UrologyYamagata Tokushukai HospitalYamagataJapan
| | - Vladimir Bilim
- Department of UrologyKameda Daiichi HospitalNiigataJapan
| | - Kiyotsugu Hoshi
- Department of UrologyYamagata Tokushukai HospitalYamagataJapan
| | - Isoji Sasagawa
- Department of UrologyYamagata Tokushukai HospitalYamagataJapan
| | - Kotarou Otake
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Daigo Chiba
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Shinta Suenaga
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Masahito Konno
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Yuki Katsumata
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Kento Morozumi
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Jun Takemoto
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | - Kenji Numahata
- Department of UrologyYamagata Prefectural Central HospitalYamagataJapan
| | | |
Collapse
|
9
|
Qin J, Yang Y, Zhuang X, Xing J. Association Between BAK1 Gene rs210138 Polymorphisms and Testicular Germ Cell Tumors: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2020; 11:2. [PMID: 32038496 PMCID: PMC6989409 DOI: 10.3389/fendo.2020.00002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Several studies including some genome-wide association studies (GWAS) had shown that BAK1 gene rs210138 polymorphisms might be associated with testicular germ cell tumors (TGCT). Here we tried to sum up the association through a systematic review and meta-analysis. Methods: Studies associated with BAK1 rs210138 and TGCT was systematically searched in databases. The effect size was pooled according to ORs and 95% CIs. Results: Our systematic review and meta-analysis comprised 14 articles. Significantly increased risk of TGCT was found in eligible GWAS and follow-up studies, in overall group and its Caucasian subgroup. Conclusions: Compared with adenine (A), BAK1 rs210138 guanine (G) is associated with increased risk of TGCT. Well-planned studies with larger sample size and more subgroups are needed to verify the risk identified in our systematic review and meta-analysis.
Collapse
Affiliation(s)
- Jiaxuan Qin
- Department of Urology Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Center of Diagnosis and Treatment of Urinary System Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, China
- The Key Laboratory of Urinary Tract Tumors and Calculi of Xiamen City, The First Affiliated Hospital of Xiamen University, Xiamen, China
- *Correspondence: Jiaxuan Qin
| | - Yufeng Yang
- Department of Urology Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Center of Diagnosis and Treatment of Urinary System Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, China
- The Key Laboratory of Urinary Tract Tumors and Calculi of Xiamen City, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xuan Zhuang
- Department of Urology Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Center of Diagnosis and Treatment of Urinary System Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, China
- The Key Laboratory of Urinary Tract Tumors and Calculi of Xiamen City, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jinchun Xing
- Department of Urology Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Center of Diagnosis and Treatment of Urinary System Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, China
- The Key Laboratory of Urinary Tract Tumors and Calculi of Xiamen City, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Jinchun Xing
| |
Collapse
|
10
|
Lobo J, Gillis AJM, Jerónimo C, Henrique R, Looijenga LHJ. Human Germ Cell Tumors are Developmental Cancers: Impact of Epigenetics on Pathobiology and Clinic. Int J Mol Sci 2019; 20:E258. [PMID: 30634670 PMCID: PMC6359418 DOI: 10.3390/ijms20020258] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/25/2018] [Accepted: 01/07/2019] [Indexed: 02/03/2023] Open
Abstract
Current (high throughput omics-based) data support the model that human (malignant) germ cell tumors are not initiated by somatic mutations, but, instead through a defined locked epigenetic status, representative of their cell of origin. This elegantly explains the role of both genetic susceptibility as well as environmental factors in the pathogenesis, referred to as 'genvironment'. Moreover, it could also explain various epidemiological findings, including the rising incidence of this type of cancer in Western societies. In addition, it allows for identification of clinically relevant and informative biomarkers both for diagnosis and follow-up of individual patients. The current status of these findings will be discussed, including the use of high throughput DNA methylation profiling for determination of differentially methylated regions (DMRs) as well as chromosomal copy number variation (CNV). Finally, the potential value of methylation-specific tumor DNA fragments (i.e., XIST promotor) as well as embryonic microRNAs as molecular biomarkers for cancer detection in liquid biopsies will be presented.
Collapse
Affiliation(s)
- João Lobo
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal;.
| | - Ad J M Gillis
- Laboratory of Experimental Patho-Oncology (LEPO), Josephine Nefkens Building, Erasmus MC, Department of Pathology, University Medical Center, Cancer Institute, Be-432A, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands.
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal;.
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal;.
| | - Leendert H J Looijenga
- Laboratory of Experimental Patho-Oncology (LEPO), Josephine Nefkens Building, Erasmus MC, Department of Pathology, University Medical Center, Cancer Institute, Be-432A, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands.
| |
Collapse
|
11
|
Semaan A, Haddad FG, Eid R, Kourie HR, Nemr E. Immunotherapy: last bullet in platinum refractory germ cell testicular cancer. Future Oncol 2019; 15:533-541. [PMID: 30624089 DOI: 10.2217/fon-2018-0571] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Testicular germ cell tumors are chemosensitive with very high cure-rates even in the metastatic setting. However, patients with platinum-refractory and relapsing tumors after autologous stem cell transplant have very poor outcomes despite salvage treatments, and with no effective alternative therapies. Immunotherapy, notably with PD-1 inhibitors, has proven to be very effective in treating various solid tumors. This review summarizes the experience with anti-PD-1 agents (pembrolizumab, nivolumab) in the treatment of testicular germ cell tumor relapsing after multiple lines of treatment, and exposes future trials evaluating newer checkpoint inhibitors in this setting.
Collapse
Affiliation(s)
- Albert Semaan
- Urology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Fady Gh Haddad
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Roland Eid
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Hampig Raphael Kourie
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Elie Nemr
- Urology Department, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| |
Collapse
|
12
|
Markulin D, Vojta A, Samaržija I, Gamulin M, Bečeheli I, Jukić I, Maglov Č, Zoldoš V, Fučić A. Association Between RASSF1A Promoter Methylation and Testicular Germ Cell Tumor: A Meta-analysis and a Cohort Study. Cancer Genomics Proteomics 2018; 14:363-372. [PMID: 28871003 DOI: 10.21873/cgp.20046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/05/2017] [Accepted: 08/08/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The RAS association domain family protein 1a (RASSF1A) is a prominent tumor suppressor gene showing altered promoter methylation in testicular germ cell tumors (TGCT). RASSF1A promoter hypermethylation might represent an early event in TGCT tumorigenesis. We investigated whether the RASSF1A promoter methylation in peripheral blood of TGCT patients can be associated with testicular cancer risk. MATERIALS AND METHODS Following a meta-analysis, we performed a cohort study including 32 testicular cancer patients and 32 healthy controls. Promoter methylation of the RASSF1A and O6-methylguanine-DNA-methyltransferase (MGMT) genes was analyzed using bisulfite pyrosequencing of DNA from peripheral blood. RESULTS Meta-analysis showed an odds ratio (OR) of 7.69 for RASSF1A promoter methylation as a risk factor for TGCT. Cohort study found altered methylation of the RASSF1A promoter in blood of TGCT patients. Methylation was higher in TGCT patients before BEP chemotherapy. CONCLUSION The meta-analysis indicates a role of the RASSF1A promoter hypermethylation from peripheral blood in TCGT. We confirmed that finding in our cohort study, which represents the first report of changed RASSF1A promoter methylation in peripheral blood TGCT.
Collapse
Affiliation(s)
- Dora Markulin
- University of Zagreb, Faculty of Science, Department of Biology, Division of Molecular Biology, Zagreb, Croatia
| | - Aleksandar Vojta
- University of Zagreb, Faculty of Science, Department of Biology, Division of Molecular Biology, Zagreb, Croatia
| | - Ivana Samaržija
- University of Zagreb, Faculty of Science, Department of Biology, Division of Molecular Biology, Zagreb, Croatia
| | - Marija Gamulin
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
| | | | - Irena Jukić
- Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Čedomir Maglov
- Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Vlatka Zoldoš
- University of Zagreb, Faculty of Science, Department of Biology, Division of Molecular Biology, Zagreb, Croatia
| | - Aleksandra Fučić
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| |
Collapse
|
13
|
Xing JS, Bai ZM. Is testicular dysgenesis syndrome a genetic, endocrine, or environmental disease, or an unexplained reproductive disorder? Life Sci 2018; 194:120-129. [DOI: 10.1016/j.lfs.2017.11.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/14/2017] [Accepted: 11/24/2017] [Indexed: 11/29/2022]
|
14
|
Salz HK, Dawson EP, Heaney JD. Germ cell tumors: Insights from the Drosophila ovary and the mouse testis. Mol Reprod Dev 2017; 84:200-211. [PMID: 28079292 DOI: 10.1002/mrd.22779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022]
Abstract
Ovarian and testicular germ cell tumors of young adults are thought to arise from defects in germ cell development, but the molecular mechanisms underlying malignant transformation are poorly understood. In this review, we focus on the biology of germ cell tumor formation in the Drosophila ovary and the mouse testis, for which evidence supports common underlying mechanisms, such as blocking initiation into the differentiation pathway, impaired lineage progression, and sexual identity instability. We then discuss how these concepts inform our understanding of the disease in humans. Mol. Reprod. Dev. 84: 200-211, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Helen K Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Emily P Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
15
|
Facchini G, Rossetti S, Cavaliere C, D'Aniello C, Di Franco R, Iovane G, Grimaldi G, Piscitelli R, Muto P, Botti G, Perdonà S, Veneziani BM, Berretta M, Montanari M. Exploring the molecular aspects associated with testicular germ cell tumors: a review. Oncotarget 2017; 9:1365-1379. [PMID: 29416701 PMCID: PMC5787445 DOI: 10.18632/oncotarget.22373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/05/2017] [Indexed: 11/25/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most common solid tumors affecting young men. They constitute a distinct entity because of their embryonic origin and their unique biological behavior. Recent preclinical data regarding biological signaling machinery as well as genetic and epigenetic mechanisms associated with molecular patterns of tumors have contribute to explain the pathogenesis and the differentiation of TGCTs and to understand the mechanisms responsible for the development of resistance to treatment. In this review, we discuss the main genetic and epigenetic events associated with TGCTs development in order to better define their role in the pathogenesis of these tumors and in cisplatin-acquired resistance.
Collapse
Affiliation(s)
- Gaetano Facchini
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Carla Cavaliere
- Medical Oncology Unit, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Naples, Italy
| | - Rossella Di Franco
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gelsomina Iovane
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Giovanni Grimaldi
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy.,Scientific Management, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sisto Perdonà
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, Aviano, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy.,Department of Biology, College of Science and Technology, Temple University, Philadelphia, USA
| |
Collapse
|
16
|
Aoun F, Kourie HR, Albisinni S, Roumeguère T. Will Testicular Germ Cell Tumors Remain Untargetable? Target Oncol 2017; 11:711-721. [PMID: 27184492 DOI: 10.1007/s11523-016-0439-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Testicular Germ cell tumors (TGCT) represent the most common solid tumors affecting young men. They constitute a distinct entity because of their embryonic origin and their unique biological behavior. Recently, new preclinical data on genetic and epigenetic susceptibility profiles, biological signaling machinery as well as on molecular patterns of tumors and pathways of pathogenesis helped to elucidate the pathogenesis and the differentiation of TGCTs and to understand the mechanisms behind the development of resistance to treatment. In the present work, we have reviewed new clues to the development, differentiation and progression of TGCTs. We focus on the most important epigenetic and molecular biomarkers, and discussed their diagnostic and prognostic accuracy compared to the currently used biomarkers. The mechanisms underlying the development of resistance to cisplatin and commonly used chemotherapeutic agents are also discussed in detail. Finally, we summarize failed and ongoing clinical trials using targeted therapies in resistant TGCTs, and analyze the potential of new targeted therapies.
Collapse
Affiliation(s)
- Fouad Aoun
- Department of Urology, Hôtel Dieu de France, Beyrouth, Lebanon.
| | - Hampig Raphael Kourie
- Department of Oncology, Jules Bordet Institute, 1 Héger Bordet Street, 1000, Brussels, Belgium
| | - Simone Albisinni
- Department of Urology, Erasme Hospital, Route de Lennik 808, 1070, Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, Erasme Hospital, Route de Lennik 808, 1070, Brussels, Belgium
| |
Collapse
|
17
|
Zhang T, Zarkower D. DMRT proteins and coordination of mammalian spermatogenesis. Stem Cell Res 2017; 24:195-202. [PMID: 28774758 DOI: 10.1016/j.scr.2017.07.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 10/19/2022] Open
Abstract
DMRT genes encode a deeply conserved family of transcription factors that share a unique DNA binding motif, the DM domain. DMRTs regulate development in a broad variety of metazoans and they appear to have controlled sexual differentiation for hundreds of millions of years. In mice, starting during embryonic development, three Dmrt genes act sequentially to help establish and maintain spermatogenesis. Dmrt1 has notably diverse functions that include repressing pluripotency genes and promoting mitotic arrest in embryonic germ cells, reactivating prospermatogonia perinatally, establishing and maintaining spermatogonial stem cells (SSCs), promoting spermatogonial differentiation, and controlling the mitosis/meiosis switch. Dmrt6 acts in differentiating spermatogonia to coordinate an orderly exit from the mitotic/spermatogonial program and allow proper timing of entry to the meiotic/spermatocyte program. Finally, Dmrt7 takes over during the first meiotic prophase to help choreograph a transition in histone modifications that maintains transcriptional silencing of the sex chromosomes. The combined action of these three Dmrt genes helps ensure robust and sustainable spermatogenesis.
Collapse
Affiliation(s)
- Teng Zhang
- Department of Genetics, Cell Biology, and Development, and Developmental Biology Center, University of Minnesota Medical School, 6-160 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA.
| | - David Zarkower
- Department of Genetics, Cell Biology, and Development, and Developmental Biology Center, University of Minnesota Medical School, 6-160 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Martinot E, Sèdes L, Baptissart M, Holota H, Rouaisnel B, Damon-Soubeyrand C, De Haze A, Saru JP, Thibault-Carpentier C, Keime C, Lobaccaro JMA, Baron S, Benoit G, Caira F, Beaudoin C, Volle DH. The Bile Acid Nuclear Receptor FXRα Is a Critical Regulator of Mouse Germ Cell Fate. Stem Cell Reports 2017; 9:315-328. [PMID: 28669602 PMCID: PMC5511114 DOI: 10.1016/j.stemcr.2017.05.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is the process by which spermatozoa are generated from spermatogonia. This cell population is heterogeneous, with self-renewing spermatogonial stem cells (SSCs) and progenitor spermatogonia that will continue on a path of differentiation. Only SSCs have the ability to regenerate and sustain spermatogenesis. This makes the testis a good model to investigate stem cell biology. The Farnesoid X Receptor alpha (FXRα) was recently shown to be expressed in the testis. However, its global impact on germ cell homeostasis has not yet been studied. Here, using a phenotyping approach in Fxrα−/− mice, we describe unexpected roles of FXRα on germ cell physiology independent of its effects on somatic cells. FXRα helps establish and maintain an undifferentiated germ cell pool and in turn influences male fertility. FXRα regulates the expression of several pluripotency factors. Among these, in vitro approaches show that FXRα controls the expression of the pluripotency marker Lin28 in the germ cells. FXRα regulated germ cell apoptotis independently of androgen homeostasis FXRα controls germ cell differentiation FXRα regulates the establishment and maintenance of undifferentiated germ cells In germ cells, FXRα controls the expression of pluripotency markers such as Lin28
Collapse
Affiliation(s)
- Emmanuelle Martinot
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Lauriane Sèdes
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Marine Baptissart
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Hélène Holota
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Betty Rouaisnel
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Christelle Damon-Soubeyrand
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Angélique De Haze
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Jean-Paul Saru
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | | | - Céline Keime
- IGBMC - CNRS UMR 7104 - Inserm U 964, 1 BP 10142, 67404 Illkirch Cedex, France
| | - Jean-Marc A Lobaccaro
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Silvère Baron
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, 63000 Clermont-Ferrand, France
| | - Gérard Benoit
- Laboratoire de Biologie Moléculaire de la Cellule, Ecole normale supérieure de Lyon, UMR5239 CNRS/ENS Lyon/UCBL/HCL, 46, allée d'Italie, 69364 Lyon Cedex 07, France
| | - Françoise Caira
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Claude Beaudoin
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - David H Volle
- INSERM U 1103, Université Clermont Auvergne, CNRS UMR 6293, Laboratoire GReD, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
19
|
Webster KA, Schach U, Ordaz A, Steinfeld JS, Draper BW, Siegfried KR. Dmrt1 is necessary for male sexual development in zebrafish. Dev Biol 2017; 422:33-46. [PMID: 27940159 PMCID: PMC5777149 DOI: 10.1016/j.ydbio.2016.12.008] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 10/20/2022]
Abstract
The dmrt1 (doublesex and mab-3 related transcription factor 1) gene is a key regulator of sex determination and/or gonadal sex differentiation across metazoan animals. This is unusual given that sex determination genes are typically not well conserved. The mechanisms by which zebrafish sex is determined have remained elusive due to the lack of sex chromosomes and the complex polygenic nature of sex determination in domesticated strains. To investigate the role of dmrt1 in zebrafish sex determination and gonad development, we isolated mutations disrupting this gene. We found that the majority of dmrt1 mutant fish develop as fertile females suggesting a complete male-to-female sex reversal in mutant animals that would have otherwise developed as males. A small percentage of mutant animals became males, but were sterile and displayed testicular dysgenesis. Therefore zebrafish dmrt1 functions in male sex determination and testis development. Mutant males had aberrant gonadal development at the onset of gonadal sex-differentiation, displaying reduced oocyte apoptosis followed by development of intersex gonads and failed testis morphogenesis and spermatogenesis. By contrast, female ovaries developed normally. We found that Dmrt1 is necessary for normal transcriptional regulation of the amh (anti-Müllerian hormone) and foxl2 (forkhead box L2) genes, which are thought to be important for male or female sexual development respectively. Interestingly, we identified one dmrt1 mutant allele that co-operates with a linked segregation distorter locus to generate an apparent XY sex determination mechanism. We conclude that dmrt1 is dispensable for ovary development but necessary for testis development in zebrafish, and that dmrt1 promotes male development by transcriptionally regulating male and female genes as has been described in other animals. Furthermore, the strong sex-ratio bias caused by dmrt1 reduction-of-function points to potential mechanisms through which sex chromosomes may evolve.
Collapse
Affiliation(s)
- Kaitlyn A Webster
- University of Massachusetts Boston, Biology Department, 100 Morrissey Blvd., Boston, MA 02125, USA
| | - Ursula Schach
- Max Planck Institute for Developmental Biology, Department Genetics, Spemanstrasse 35, 72076 Tübingen, Germany
| | - Angel Ordaz
- University of California Davis, Department of Molecular and Cellular Biology, One Shields Ave., Davis, CA 95616, USA
| | - Jocelyn S Steinfeld
- University of Massachusetts Boston, Biology Department, 100 Morrissey Blvd., Boston, MA 02125, USA
| | - Bruce W Draper
- University of California Davis, Department of Molecular and Cellular Biology, One Shields Ave., Davis, CA 95616, USA
| | - Kellee R Siegfried
- University of Massachusetts Boston, Biology Department, 100 Morrissey Blvd., Boston, MA 02125, USA.
| |
Collapse
|
20
|
Nemtsova MV, Ivkin EV, Simonova OA, Rudenko VV, Chernykh VB, Mikhaylenko DS, Loran OB. Polymorphisms of KITLG, SPRY4, and BAK1 genes in patients with testicular germ cell tumors and individuals with infertility associated with AZFc deletion of the Y chromosome. Mol Biol 2016. [DOI: 10.1134/s0026893316050137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Parent-of-origin effects of A1CF and AGO2 on testicular germ-cell tumors, testicular abnormalities, and fertilization bias. Proc Natl Acad Sci U S A 2016; 113:E5425-33. [PMID: 27582469 DOI: 10.1073/pnas.1604773113] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Testicular tumors, the most common cancer in young men, arise from abnormalities in germ cells during fetal development. Unconventional inheritance for testicular germ cell tumor (TGCT) risk both in humans and mice implicates epigenetic mechanisms. Apolipoprotein B mRNA-editing enzyme complex 1 (APOBEC1) cytidine deaminase and Deadend-1, which are involved in C-to-U RNA editing and microRNA-dependent mRNA silencing, respectively, are potent epigenetic modifiers of TGCT susceptibility in the genetically predisposed 129/Sv inbred mouse strain. Here, we show that partial loss of either APOBEC1 complementation factor (A1CF), the RNA-binding cofactor of APOBEC1 in RNA editing, or Argonaute 2 (AGO2), a key factor in the biogenesis of certain noncoding RNAs, modulates risk for TGCTs and testicular abnormalities in both parent-of-origin and conventional genetic manners. In addition, non-Mendelian inheritance was found among progeny of A1cf and Ago2 mutant intercrosses but not in backcrosses and without fetal loss. Together these findings suggest nonrandom union of gametes rather than meiotic drive or preferential lethality. Finally, this survey also suggested that A1CF contributes to long-term reproductive performance. These results directly implicate the RNA-binding proteins A1CF and AGO2 in the epigenetic control of germ-cell fate, urogenital development, and gamete functions.
Collapse
|
22
|
Skakkebaek NE, Rajpert-De Meyts E, Buck Louis GM, Toppari J, Andersson AM, Eisenberg ML, Jensen TK, Jørgensen N, Swan SH, Sapra KJ, Ziebe S, Priskorn L, Juul A. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol Rev 2016; 96:55-97. [PMID: 26582516 DOI: 10.1152/physrev.00017.2015] [Citation(s) in RCA: 618] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
It is predicted that Japan and European Union will soon experience appreciable decreases in their populations due to persistently low total fertility rates (TFR) below replacement level (2.1 child per woman). In the United States, where TFR has also declined, there are ethnic differences. Caucasians have rates below replacement, while TFRs among African-Americans and Hispanics are higher. We review possible links between TFR and trends in a range of male reproductive problems, including testicular cancer, disorders of sex development, cryptorchidism, hypospadias, low testosterone levels, poor semen quality, childlessness, changed sex ratio, and increasing demand for assisted reproductive techniques. We present evidence that several adult male reproductive problems arise in utero and are signs of testicular dysgenesis syndrome (TDS). Although TDS might result from genetic mutations, recent evidence suggests that it most often is related to environmental exposures of the fetal testis. However, environmental factors can also affect the adult endocrine system. Based on our review of genetic and environmental factors, we conclude that environmental exposures arising from modern lifestyle, rather than genetics, are the most important factors in the observed trends. These environmental factors might act either directly or via epigenetic mechanisms. In the latter case, the effects of exposures might have an impact for several generations post-exposure. In conclusion, there is an urgent need to prioritize research in reproductive physiology and pathophysiology, particularly in highly industrialized countries facing decreasing populations. We highlight a number of topics that need attention by researchers in human physiology, pathophysiology, environmental health sciences, and demography.
Collapse
Affiliation(s)
- Niels E Skakkebaek
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Germaine M Buck Louis
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Jorma Toppari
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Michael L Eisenberg
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Tina Kold Jensen
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Shanna H Swan
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Katherine J Sapra
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Søren Ziebe
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Lærke Priskorn
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
23
|
Sanchez A, Amatruda JF. Zebrafish Germ Cell Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:479-94. [PMID: 27165367 DOI: 10.1007/978-3-319-30654-4_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Germ cell tumors (GCTs) are malignant cancers that arise from embryonic precursors known as Primordial Germ Cells. GCTs occur in neonates, children, adolescents and young adults and can occur in the testis, the ovary or extragonadal sites. Because GCTs arise from pluripotent cells, the tumors can exhibit a wide range of different histologies. Current cisplatin-based combination therapies cures most patients, however at the cost of significant toxicity to normal tissues. While GWAS studies and genomic analysis of human GCTs have uncovered somatic mutations and loci that might confer tumor susceptibility, little is still known about the exact mechanisms that drive tumor development, and animal models that faithfully recapitulate all the different GCT subtypes are lacking. Here, we summarize current understanding of germline development in humans and zebrafish, describe the biology of human germ cell tumors, and discuss progress and prospects for zebrafish GCT models that may contribute to better understanding of human GCTs.
Collapse
Affiliation(s)
- Angelica Sanchez
- Departments of Pediatrics and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - James F Amatruda
- Departments of Pediatrics, Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
24
|
Krahling T, Balassa K, Kiss KP, Bors A, Batai A, Halm G, Egyed M, Fekete S, Remenyi P, Masszi T, Tordai A, Andrikovics H. Co-occurrence of Myeloproliferative Neoplasms and Solid Tumors Is Attributed to a Synergism Between Cytoreductive Therapy and the Common TERT Polymorphism rs2736100. Cancer Epidemiol Biomarkers Prev 2015; 25:98-104. [DOI: 10.1158/1055-9965.epi-15-0805] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 10/16/2015] [Indexed: 11/16/2022] Open
|
25
|
Pathak A, Adams CD, Loud JT, Nichols K, Stewart DR, Greene MH. Prospectively Identified Incident Testicular Cancer Risk in a Familial Testicular Cancer Cohort. Cancer Epidemiol Biomarkers Prev 2015; 24:1614-21. [PMID: 26265202 DOI: 10.1158/1055-9965.epi-14-1240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/28/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Human testicular germ cell tumors (TGCT) have a strong genetic component and a high familial relative risk. However, linkage analyses have not identified a rare, highly penetrant familial TGCT (FTGCT) susceptibility locus. Currently, multiple low-penetrance genes are hypothesized to underlie the familial multiple-case phenotype. The observation that two is the most common number of affected individuals per family presents an impediment to FTGCT gene discovery. Clinically, the prospective TGCT risk in the multiple-case family context is unknown. METHODS We performed a prospective analysis of TGCT incidence in a cohort of multiple-affected-person families and sporadic-bilateral-case families; 1,260 men from 140 families (10,207 person-years of follow-up) met our inclusion criteria. Age-, gender-, and calendar time-specific standardized incidence ratios (SIR) for TGCT relative to the general population were calculated using SEER*Stat. RESULTS Eight incident TGCTs occurred during prospective FTGCT cohort follow-up (versus 0.67 expected; SIR = 11.9; 95% CI, 5.1-23.4; excess absolute risk = 7.2/10,000). We demonstrate that the incidence rate of TGCT is greater among bloodline male relatives from multiple-case testicular cancer families than that expected in the general population, a pattern characteristic of adult-onset Mendelian cancer susceptibility disorders. Two of these incident TGCTs occurred in relatives of sporadic-bilateral cases (0.15 expected; SIR = 13.4; 95% CI, 1.6-48.6). CONCLUSIONS Our data are the first to indicate that despite relatively low numbers of affected individuals per family, members of both multiple-affected-person FTGCT families and sporadic-bilateral TGCT families comprise high-risk groups for incident testicular cancer. IMPACT Men at high TGCT risk might benefit from tailored risk stratification and surveillance strategies.
Collapse
Affiliation(s)
- Anand Pathak
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Charleen D Adams
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jennifer T Loud
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
26
|
McMaster ML, Heimdal KR, Loud JT, Bracci JS, Rosenberg PS, Greene MH. Nontesticular cancers in relatives of testicular germ cell tumor (TGCT) patients from multiple-case TGCT families. Cancer Med 2015; 4:1069-78. [PMID: 25882629 PMCID: PMC4529345 DOI: 10.1002/cam4.450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 12/20/2022] Open
Abstract
Testicular germ cell tumors (TGCT) exhibit striking familial aggregation that remains incompletely explained. To improve the phenotypic definition of familial TGCT (FTGCT), we studied an international cohort of multiple-case TGCT families to determine whether first-degree relatives of FTGCT cases are at increased risk of other types of cancer. We identified 1041 first-degree relatives of TGCT cases in 66 multiple-case TGCT families from Norway and 64 from the United States (combined follow-up of 31,556 person-years). We collected data on all cancers (except nonmelanoma skin cancers) reported by the family informant in these relatives, and we attempted to verify all reported cancer diagnoses through medical or cancer registry records. We calculated observed-to-expected (O/E) standardized incidence ratios, together with 95% confidence intervals (CI), for invasive cancers other than TGCT. We found no increase in risk of cancer overall (Norway O/E = 0.8; 95% CI: 0.6–1.1 and United States O/E = 0.9; 95% CI: 0.7–1.3). Site-specific analyses pooled across the two countries revealed a leukemia excess (O/E = 6.5; 95% CI: 3.0–12.3), deficit of female breast cancer (O/E = 0.0; 95% CI: 0.0–0.6) and increased risk of soft tissue sarcoma (O/E = 7.2; 95% CI: 2.0–18.4); in all instances, these results were based on small case numbers and statistically significant only in Norway. While limited by sample size and potential issues relating to completeness of cancer reporting, this study in multiple-case TGCT families does not support the hypothesis that cancers other than testis cancer contribute to the FTGCT phenotype.
Collapse
Affiliation(s)
- Mary L McMaster
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892-9769.,Commissioned Corps of the U.S. Public Health Service, U.S. Department of Health and Human Services, Washington, District of Columbia
| | - Ketil R Heimdal
- Section for Clinical Genetics, Department of Medical Genetics, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jennifer T Loud
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892-9769
| | | | - Philip S Rosenberg
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892-9769
| | - Mark H Greene
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892-9769
| |
Collapse
|
27
|
Rijlaarsdam MA, Tax DMJ, Gillis AJM, Dorssers LCJ, Koestler DC, de Ridder J, Looijenga LHJ. Genome wide DNA methylation profiles provide clues to the origin and pathogenesis of germ cell tumors. PLoS One 2015; 10:e0122146. [PMID: 25859847 PMCID: PMC4479500 DOI: 10.1371/journal.pone.0122146] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/07/2015] [Indexed: 12/18/2022] Open
Abstract
The cell of origin of the five subtypes (I-V) of germ cell tumors (GCTs) are assumed to be germ cells from different maturation stages. This is (potentially) reflected in their methylation status as fetal maturing primordial germ cells are globally demethylated during migration from the yolk sac to the gonad. Imprinted regions are erased in the gonad and later become uniparentally imprinted according to fetal sex. Here, 91 GCTs (type I-IV) and four cell lines were profiled (Illumina’s HumanMethylation450BeadChip). Data was pre-processed controlling for cross hybridization, SNPs, detection rate, probe-type bias and batch effects. The annotation was extended, covering snRNAs/microRNAs, repeat elements and imprinted regions. A Hidden Markov Model-based genome segmentation was devised to identify differentially methylated genomic regions. Methylation profiles allowed for separation of clusters of non-seminomas (type II), seminomas/dysgerminomas (type II), spermatocytic seminomas (type III) and teratomas/dermoid cysts (type I/IV). The seminomas, dysgerminomas and spermatocytic seminomas were globally hypomethylated, in line with previous reports and their demethylated precursor. Differential methylation and imprinting status between subtypes reflected their presumed cell of origin. Ovarian type I teratomas and dermoid cysts showed (partial) sex specific uniparental maternal imprinting. The spermatocytic seminomas showed uniparental paternal imprinting while testicular teratomas exhibited partial imprinting erasure. Somatic imprinting in type II GCTs might indicate a cell of origin after global demethylation but before imprinting erasure. This is earlier than previously described, but agrees with the totipotent/embryonic stem cell like potential of type II GCTs and their rare extra-gonadal localization. The results support the common origin of the type I teratomas and show strong similarity between ovarian type I teratomas and dermoid cysts. In conclusion, we identified specific and global methylation differences between GCT subtypes, providing insight into their developmental timing and underlying developmental biology. Data and extended annotation are deposited at GEO (GSE58538 and GPL18809).
Collapse
Affiliation(s)
- Martin A. Rijlaarsdam
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - David M. J. Tax
- Faculty of Electrical Engineering, Mathematics and Computer Science Intelligent Systems—Delft Bioinformatics Lab, Technical University of Delft, Delft, The Netherlands
| | - Ad J. M. Gillis
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lambert C. J. Dorssers
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Devin C. Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jeroen de Ridder
- Faculty of Electrical Engineering, Mathematics and Computer Science Intelligent Systems—Delft Bioinformatics Lab, Technical University of Delft, Delft, The Netherlands
| | - Leendert H. J. Looijenga
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
28
|
Brabrand S, Johannessen B, Axcrona U, Kraggerud SM, Berg KG, Bakken AC, Bruun J, Fosså SD, Lothe RA, Lehne G, Skotheim RI. Exome sequencing of bilateral testicular germ cell tumors suggests independent development lineages. Neoplasia 2015; 17:167-74. [PMID: 25748235 PMCID: PMC4351294 DOI: 10.1016/j.neo.2014.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/08/2014] [Indexed: 12/17/2022]
Abstract
Intratubular germ cell neoplasia, the precursor of testicular germ cell tumors (TGCTs), is hypothesized to arise during embryogenesis from developmentally arrested primordial germ cells (PGCs) or gonocytes. In early embryonal life, the PGCs migrate from the yolk sac to the dorsal body wall where the cell population separates before colonizing the genital ridges. However, whether the malignant transformation takes place before or after this separation is controversial. We have explored the somatic exome-wide mutational spectra of bilateral TGCT to provide novel insight into the in utero critical time frame of malignant transformation and TGCT pathogenesis. Exome sequencing was performed in five patients with bilateral TGCT (eight tumors), of these three patients in whom both tumors were available (six tumors) and two patients each with only one available tumor (two tumors). Selected loci were explored by Sanger sequencing in 71 patients with bilateral TGCT. From the exome-wide mutational spectra, no identical mutations in any of the three bilateral tumor pairs were identified. Exome sequencing of all eight tumors revealed 87 somatic non-synonymous mutations (median 10 per tumor; range 5-21), some in already known cancer genes such as CIITA, NEB, platelet-derived growth factor receptor α (PDGFRA), and WHSC1. SUPT6H was found recurrently mutated in two tumors. We suggest independent development lineages of bilateral TGCT. Thus, malignant transformation into intratubular germ cell neoplasia is likely to occur after the migration of PGCs. We reveal possible drivers of TGCT pathogenesis, such as mutated PDGFRA, potentially with therapeutic implications for TGCT patients.
Collapse
Affiliation(s)
- Sigmund Brabrand
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ulrika Axcrona
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sigrid M Kraggerud
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaja G Berg
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne C Bakken
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jarle Bruun
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sophie D Fosså
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gustav Lehne
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
29
|
Litchfield K, Shipley J, Turnbull C. Common variants identified in genome-wide association studies of testicular germ cell tumour: an update, biological insights and clinical application. Andrology 2015; 3:34-46. [DOI: 10.1111/andr.304] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 01/13/2023]
Affiliation(s)
- K. Litchfield
- Division of Genetics and Epidemiology; The Institute of Cancer Research; London UK
| | - J. Shipley
- Divisions of Molecular Pathology and Cancer Therapeutics; The Institute of Cancer Research; London UK
| | - C. Turnbull
- Division of Genetics and Epidemiology; The Institute of Cancer Research; London UK
- Royal Marsden NHS Foundation Trust; London UK
| |
Collapse
|
30
|
|
31
|
Koster R, Mitra N, D'Andrea K, Vardhanabhuti S, Chung CC, Wang Z, Loren Erickson R, Vaughn DJ, Litchfield K, Rahman N, Greene MH, McGlynn KA, Turnbull C, Chanock SJ, Nathanson KL, Kanetsky PA. Pathway-based analysis of GWAs data identifies association of sex determination genes with susceptibility to testicular germ cell tumors. Hum Mol Genet 2014; 23:6061-8. [PMID: 24943593 PMCID: PMC4204765 DOI: 10.1093/hmg/ddu305] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 05/28/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association (GWA) studies of testicular germ cell tumor (TGCT) have identified 18 susceptibility loci, some containing genes encoding proteins important in male germ cell development. Deletions of one of these genes, DMRT1, lead to male-to-female sex reversal and are associated with development of gonadoblastoma. To further explore genetic association with TGCT, we undertook a pathway-based analysis of SNP marker associations in the Penn GWAs (349 TGCT cases and 919 controls). We analyzed a custom-built sex determination gene set consisting of 32 genes using three different methods of pathway-based analysis. The sex determination gene set ranked highly compared with canonical gene sets, and it was associated with TGCT (FDRG = 2.28 × 10(-5), FDRM = 0.014 and FDRI = 0.008 for Gene Set Analysis-SNP (GSA-SNP), Meta-Analysis Gene Set Enrichment of Variant Associations (MAGENTA) and Improved Gene Set Enrichment Analysis for Genome-wide Association Study (i-GSEA4GWAS) analysis, respectively). The association remained after removal of DMRT1 from the gene set (FDRG = 0.0002, FDRM = 0.055 and FDRI = 0.009). Using data from the NCI GWA scan (582 TGCT cases and 1056 controls) and UK scan (986 TGCT cases and 4946 controls), we replicated these findings (NCI: FDRG = 0.006, FDRM = 0.014, FDRI = 0.033, and UK: FDRG = 1.04 × 10(-6), FDRM = 0.016, FDRI = 0.025). After removal of DMRT1 from the gene set, the sex determination gene set remains associated with TGCT in the NCI (FDRG = 0.039, FDRM = 0.050 and FDRI = 0.055) and UK scans (FDRG = 3.00 × 10(-5), FDRM = 0.056 and FDRI = 0.044). With the exception of DMRT1, genes in the sex determination gene set have not previously been identified as TGCT susceptibility loci in these GWA scans, demonstrating the complementary nature of a pathway-based approach for genome-wide analysis of TGCT.
Collapse
Affiliation(s)
- Roelof Koster
- Translational Medicine and Human Genetics, Department of Medicine
| | | | - Kurt D'Andrea
- Translational Medicine and Human Genetics, Department of Medicine
| | | | - Charles C Chung
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services,National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services,National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, USA
| | - R Loren Erickson
- Walter Reed Army Institute of Research, Silver Spring, MD, USA and
| | - David J Vaughn
- Division of Hematology-Oncology, Department of Medicine and, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Litchfield
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, Surrey, UK
| | - Nazneen Rahman
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, Surrey, UK
| | - Mark H Greene
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services,National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services,National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clare Turnbull
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, Surrey, UK
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services,National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katherine L Nathanson
- Translational Medicine and Human Genetics, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter A Kanetsky
- Department of Biostatistics and Epidemiology, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,
| |
Collapse
|
32
|
Kratz CP, Edelman DC, Wang Y, Meltzer PS, Greene MH. Genetic and epigenetic analysis of monozygotic twins discordant for testicular cancer. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2014; 5:135-139. [PMID: 25379132 PMCID: PMC4214260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/10/2014] [Indexed: 06/04/2023]
Abstract
Despite the notion that monozygotic (identical) twins share 100% identical genetic information, genetic differences among monozygotic twin pairs do occur and can be explained by mechanisms occurring during post-zygotic events. Despite such twins being fundamentally "identical", these post-zygotic genetic changes may give rise to phenotypic differences and genetic diseases. Consequently, studies of monozygotic twin pairs discordant for specific genetic diseases represent an important tool for the identification of disease genes. We used array comparative genomic hybridization (aCGH) and methylation arrays to search for genetic and epigenetic differences in blood drawn from four monozygotic twin pairs discordant for testicular germ cell tumors. No consistent differences were identified. A larger twin study would be required to achieve confident discovery of very subtle differences between monozygotic twins discordant for testicular germ cell tumors.
Collapse
Affiliation(s)
- Christian P Kratz
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human ServicesRockville, Maryland, USA
- Department of Pediatric Hematology/Oncology, Hannover Medical SchoolHannover, Germany
| | - Daniel C Edelman
- Genetics Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human ServicesBethesda, MD, USA
| | - Yonghong Wang
- Genetics Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human ServicesBethesda, MD, USA
| | - Paul S Meltzer
- Genetics Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human ServicesBethesda, MD, USA
| | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human ServicesRockville, Maryland, USA
| |
Collapse
|
33
|
Greene MH, Mai PL, Loud JT, Pathak A, Peters JA, Mirabello L, McMaster ML, Rosenberg P, Stewart DR. Familial testicular germ cell tumors (FTGCT) - overview of a multidisciplinary etiologic study. Andrology 2014; 3:47-58. [PMID: 25303766 DOI: 10.1111/andr.294] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/08/2014] [Accepted: 09/15/2014] [Indexed: 02/06/2023]
Abstract
This Review summarizes the cumulative results of the National Cancer Institute Clinical Genetics Branch Multidisciplinary Etiologic Study of Familial Testicular Germ Cell Tumors (FTGCT). Initiated 12 years ago, this protocol enrolled 724 subjects from 147 unrelated families with either ≥2 affected men (n = 90) with TGCT or a proband with bilateral TGCT and a negative family history for this cancer (n = 57). Data were collected directly from 162 subjects evaluated at the NIH Clinical Center, and 562 subjects provided information from their home communities (Field Cohort). The primary study aims included (i) ascertaining, enrolling eligible FTGCT kindred, (ii) characterizing the clinical phenotype of multiple-case families, (iii) identifying the underlying genetic mechanism for TGCT susceptibility in families, (iv) evaluating counseling, psychosocial, and behavioral issues resulting from membership in an FTGCT family, and (v) creating an annotated biospecimen repository to permit subsequent translational research studies. Noteworthy findings include (i) documenting the epidemiologic similarities between familial and sporadic TGCT, (ii) demonstrating significantly younger age-at-diagnosis for familial vs. sporadic TGCT, (iii) absence of a dysmorphic phenotype in affected family members, (iv) shifting the focus of gene discovery from a search for rare, highly penetrant susceptibility variants to the hypothesis that multiple, more common, lower penetrance genes underlie TGCT genetic risk, (v) implicating testicular microlithiasis in FTGCT risk, and (vi) observing that aberrant methylation may contribute to FTGCT risk. A clinically based, biospecimen-intensive, multidisciplinary research strategy has provided novel, valuable insights into the etiology of FTGCT, and created a research resource which will support FTGCT clinical and laboratory studies for years to come.
Collapse
Affiliation(s)
- M H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Béranger R, Pérol O, Bujan L, Faure E, Blain J, Cornet CL, Flechon A, Charbotel B, Philip T, Schüz J, Fervers B. Studying the impact of early life exposures to pesticides on the risk of testicular germ cell tumors during adulthood (TESTIS project): study protocol. BMC Cancer 2014; 14:563. [PMID: 25095793 PMCID: PMC4129121 DOI: 10.1186/1471-2407-14-563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/22/2014] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The incidence of testicular germ cell tumors (TGCT), the most common cancer in men aged 15 to 45 years, has doubled over the last 30 years in developed countries. Reasons remain unclear but a role of environmental factors, especially during critical periods of development, is strongly suspected. Reliable data on environmental exposure during this critical time period are sparse. Little is known on whether it could be a combined effect of early and later-life exposures. METHODS/DESIGN Our research aims to study the association between TGCT risk and pesticide exposures (domestic, occupational and environmental) during critical time periods of development and combined early and later-life exposures. The study design, developed during a 2-year pilot study, is a multicenter case-control study of 500 cases (ascertained through histology) and 1000 fertile/fecund controls recruited through 21 French 'Centres d'Etude et de Conservation des Œufs et de Sperme humain' (CECOS). Trained professional interviewers interview the subjects and their mothers by phone. Using a geographic information system developed and tested for application in this study design, environmental pesticides exposure assessment is based on life-time residential history. Occupational pesticides exposures are assessed by an industrial hygienist based on parents' occupations and tasks. Exposures during the prenatal period, early childhood and puberty are focused. A blood sample is collected from each participant to assess genetic polymorphisms known to be associated with TGCT risk, as well as to explore gene-environment interactions. DISCUSSION The results of our study will contribute to better understanding the causes of TGCT and the rapid increase of its incidence. We explore the effect of combined early and later-life pesticides exposure from multiple sources, as well as potential gene-environment interactions that have until now been rarely studied for TGCT. Our design allows future pooled studies and the bio-bank allows additional genetic or toxicological analyses.
Collapse
Affiliation(s)
- Rémi Béranger
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
- />Section of Environment and Radiation, International Agency for Research on Cancer (IARC), Lyon, France
- />EAM 4128 “Santé Individu Société”, Université Claude Bernard – Lyon 1, Villeurbanne, France
| | - Olivia Pérol
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
| | - Louis Bujan
- />Hôpital Paule de Viguier; Fédération Française des CECOS, CECOS, CHU, Toulouse, France
- />Université de Toulouse; UPS; Groupe de recherche en Fertilité Humaine (EA 3694, Human Fertility Research Group), Toulouse, France
| | - Elodie Faure
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
| | - Jeffrey Blain
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
| | - Charlotte Le Cornet
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
- />Section of Environment and Radiation, International Agency for Research on Cancer (IARC), Lyon, France
| | - Aude Flechon
- />Centre de Lutte Contre le Cancer, Centre Léon Bérard, Lyon, France
| | - Barbara Charbotel
- />Université de Lyon, F-69003 Lyon, France
- />Université Lyon 1, UMRESTTE (Unité mixte IFSTTAR/UCBL), Domaine Rockefeller, 69373 Lyon, France
| | - Thierry Philip
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
| | - Joachim Schüz
- />Section of Environment and Radiation, International Agency for Research on Cancer (IARC), Lyon, France
| | - Béatrice Fervers
- />Unité Cancer et Environnement, Centre Léon Bérard, 28 rue Laennec, 69373 Lyon, 08 Cedex, France
- />EAM 4128 “Santé Individu Société”, Université Claude Bernard – Lyon 1, Villeurbanne, France
| |
Collapse
|
35
|
Rijlaarsdam MA, Looijenga LHJ. An oncofetal and developmental perspective on testicular germ cell cancer. Semin Cancer Biol 2014; 29:59-74. [PMID: 25066859 DOI: 10.1016/j.semcancer.2014.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 07/17/2014] [Indexed: 12/19/2022]
Abstract
Germ cell tumors (GCTs) represent a diverse group of tumors presumably originating from (early fetal) developing germ cells. Most frequent are the testicular germ cell cancers (TGCC). Overall, TGCC is the most frequent malignancy in Caucasian males (20-40 years) and remains an important cause of (treatment related) mortality in these young men. The strong association between the phenotype of TGCC stem cell components and their totipotent ancestor (fetal primordial germ cell or gonocyte) makes these tumors highly relevant from an onco-fetal point of view. This review subsequently discusses the evidence for the early embryonic origin of TGCCs, followed by an overview of the crucial association between TGCC pathogenesis, genetics, environmental exposure and the (fetal) testicular micro-environment (genvironment). This culminates in an evaluation of three genvironmentally modulated hallmarks of TGCC directly related to the oncofetal pathogenesis of TGCC: (1) maintenance of pluripotency, (2) cell cycle control/cisplatin sensitivity and (3) regulation of proliferation/migration/apoptosis by KIT-KITL mediated receptor tyrosine kinase signaling. Briefly, TGCC exhibit identifiable stem cell components (seminoma and embryonal carcinoma) and progenitors that show large and consistent similarities to primordial/embryonic germ cells, their presumed totipotent cells of origin. TGCC pathogenesis depends crucially on a complex interaction of genetic and (micro-)environmental, i.e. genvironmental risk factors that have only been partly elucidated despite significant effort. TGCC stem cell components also show a high degree of similarity with embryonic stem/germ cells (ES) in the regulation of pluripotency and cell cycle control, directly related to their exquisite sensitivity to DNA damaging agents (e.g. cisplatin). Of note, (ES specific) micro-RNAs play a pivotal role in the crossover between cell cycle control, pluripotency and chemosensitivity. Moreover, multiple consistent observations reported TGCC to be associated with KIT-KITL mediated receptor tyrosine kinase signaling, a pathway crucially implicated in proliferation, migration and survival during embryogenesis including germ cell development. In conclusion, TGCCs are a fascinating model for onco-fetal developmental processes especially with regard to studying cell cycle control, pluripotency maintenance and KIT-KITL signaling. The knowledge presented here contributes to better understanding of the molecular characteristics of TGCC pathogenesis, translating to identification of at risk individuals and enhanced quality of care for TGCC patients (diagnosis, treatment and follow-up).
Collapse
Affiliation(s)
- Martin A Rijlaarsdam
- Department of Pathology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Leendert H J Looijenga
- Department of Pathology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Tumor loci and their interactions on mouse chromosome 19 that contribute to testicular germ cell tumors. BMC Genet 2014; 15:65. [PMID: 24886204 PMCID: PMC4053281 DOI: 10.1186/1471-2156-15-65] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Complex genetic factors underlie testicular germ cell tumor (TGCT) development. One experimental approach to dissect the genetics of TGCT predisposition is to use chromosome substitution strains, such as the 129.MOLF-Chr 19 (M19). M19 carries chromosome (Chr) 19 from the MOLF whereas all other chromosomes are from the 129 strain. 71% of M19 males develop TGCTs in contrast to 5% in 129 strain. To identify and map tumor loci from M19 we generated congenic strains harboring MOLF chromosome 19 segments on 129 strain background and monitored their TGCT incidence. RESULTS We found 3 congenic strains that each harbored tumor promoting loci that had high (14%-32%) whereas 2 other congenics had low (4%) TGCT incidences. To determine how multiple loci influence TGCT development, we created double and triple congenic strains. We found additive interactions were predominant when 2 loci were combined in double congenic strains. Surprisingly, we found an example where 2 loci, both which do not contribute significantly to TGCT, when combined in a double congenic strain resulted in greater than expected TGCT incidence (positive interaction). In an opposite example, when 2 loci with high TGCT incidences were combined, males of the double congenic showed lower than expected TGCT incidence (negative interaction). For the triple congenic strain, depending on the analysis, the overall TGCT incidence could be additive or could also be due to a positive interaction of one region with others. Additionally, we identified loci that promote bilateral tumors or testicular abnormalities. CONCLUSIONS The congenic strains each with their characteristic TGCT incidences, laterality of tumors and incidence of testicular abnormalities, are useful for identification of TGCT susceptibility modifier genes that map to Chr 19 and also for studies on the genetic and environmental causes of TGCT development. TGCTs are a consequence of aberrant germ cell and testis development. By defining predisposing loci and some of the locus interactions from M19, this study further advances our understanding of the complex genetics of TGCTs, which is the most common cancer in young human males.
Collapse
|
37
|
|
38
|
Mueller CM, Korde LA, McMaster ML, Peters JA, Bratslavsky G, Watkins RJ, Ling A, Kratz CP, Wulfsberg EA, Rosenberg PS, Greene MH. Familial testicular germ cell tumor: no associated syndromic pattern identified. Hered Cancer Clin Pract 2014; 12:3. [PMID: 24559313 PMCID: PMC3937045 DOI: 10.1186/1897-4287-12-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/12/2014] [Indexed: 11/10/2022] Open
Abstract
Background Testicular germ cell tumor (TGCT) is the most common malignancy in young men. Familial clustering, epidemiologic evidence of increased risk with family or personal history, and the association of TGCT with genitourinary (GU) tract anomalies have suggested an underlying genetic predisposition. Linkage data have not identified a rare, highly-penetrant, single gene in familial TGCT (FTGCT) cases. Based on its association with congenital GU tract anomalies and suggestions that there is an intrauterine origin to TGCT, we hypothesized the existence of unrecognized dysmorphic features in FTGCT. Methods We evaluated 38 FTGCT individuals and 41 first-degree relatives from 22 multiple-case families with detailed dysmorphology examinations, physician-based medical history and physical examination, laboratory testing, and genitourinary imaging studies. Results The prevalence of major abnormalities and minor variants did not significantly differ between either FTGCT individuals or their first-degree relatives when compared with normal population controls, except for tall stature, macrocephaly, flat midface, and retro-/micrognathia. However, these four traits were not manifest as a constellation of features in any one individual or family. We did detect an excess prevalence of the genitourinary anomalies cryptorchidism and congenital inguinal hernia in our population, as previously described in sporadic TGCT, but no congenital renal, retroperitoneal or mediastinal anomalies were detected. Conclusions Overall, our study did not identify a constellation of dysmorphic features in FTGCT individuals, which is consistent with results of genetic studies suggesting that multiple low-penetrance genes are likely responsible for FTGCT susceptibility.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Testicular cancer: biology and biomarkers. Virchows Arch 2014; 464:301-13. [DOI: 10.1007/s00428-013-1522-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022]
|
40
|
Feng CW, Bowles J, Koopman P. Control of mammalian germ cell entry into meiosis. Mol Cell Endocrinol 2014; 382:488-497. [PMID: 24076097 DOI: 10.1016/j.mce.2013.09.026] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 08/24/2013] [Accepted: 09/20/2013] [Indexed: 11/21/2022]
Abstract
Germ cells are unique in undergoing meiosis to generate oocytes and sperm. In mammals, meiosis onset is before birth in females, or at puberty in males, and recent studies have uncovered several regulatory steps involved in initiating meiosis in each sex. Evidence suggests that retinoic acid (RA) induces expression of the critical pre-meiosis gene Stra8 in germ cells of the fetal ovary, pubertal testis and adult testis. In the fetal testis, CYP26B1 degrades RA, while FGF9 further antagonises RA signalling to suppress meiosis. Failsafe mechanisms involving Nanos2 may further suppress meiosis in the fetal testis. Here, we draw together the growing knowledge relating to these meiotic control mechanisms, and present evidence that they are co-ordinately regulated and that additional factors remain to be identified. Understanding this regulatory network will illuminate not only how the foundations of mammalian reproduction are laid, but also how mis-regulation of these steps can result in infertility or germline tumours.
Collapse
Affiliation(s)
- Chun-Wei Feng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Josephine Bowles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
41
|
Bellefroid EJ, Leclère L, Saulnier A, Keruzore M, Sirakov M, Vervoort M, De Clercq S. Expanding roles for the evolutionarily conserved Dmrt sex transcriptional regulators during embryogenesis. Cell Mol Life Sci 2013; 70:3829-45. [PMID: 23463235 PMCID: PMC11113232 DOI: 10.1007/s00018-013-1288-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/18/2013] [Accepted: 01/31/2013] [Indexed: 01/20/2023]
Abstract
Dmrt genes encode a large family of transcription factors characterized by the presence of a DM domain, an unusual zinc finger DNA binding domain. While Dmrt genes are well known for their important role in sexual development in arthropodes, nematodes and vertebrates, several new findings indicate emerging functions of this gene family in other developmental processes. Here, we provide an overview of the evolution, structure and mechanisms of action of Dmrt genes. We summarize recent findings on their function in sexual regulation and discuss more extensively the role played by these proteins in somitogenesis and neural development.
Collapse
Affiliation(s)
- Eric J Bellefroid
- Laboratoire de Génétique du Développement, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, rue des Profs. Jeener et Brachet 12, 6041, Gosselies, Belgium,
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen HS, Pfeiffer RM, Zhang S. A powerful method for combining P-values in genomic studies. Genet Epidemiol 2013; 37:814-9. [PMID: 23959976 DOI: 10.1002/gepi.21755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 06/25/2013] [Accepted: 07/11/2013] [Indexed: 11/10/2022]
Abstract
After genetic regions have been identified in genomewide association studies (GWAS), investigators often follow up with more targeted investigations of specific regions. These investigations typically are based on single nucleotide polymorphisms (SNPs) with dense coverage of a region. Methods are thus needed to test the hypothesis of any association in given genetic regions. Several approaches for combining P-values obtained from testing individual SNP hypothesis tests are available. We recently proposed a sequential procedure for testing the global null hypothesis of no association in a region. When this global null hypothesis is rejected, this method provides a list of significant hypotheses and has weak control of the family-wise error rate. In this paper, we devise a permutation-based version of the test that accounts for correlations of tests based on SNPs in the same genetic region. Based on simulated data, the method has correct control of the type I error rate and higher or comparable power to other tests.
Collapse
Affiliation(s)
- Huann-Sheng Chen
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
43
|
Azevedo MF, Horvath A, Bornstein ER, Almeida MQ, Xekouki P, Faucz FR, Gourgari E, Nadella K, Remmers EF, Quezado M, de Alexandre RB, Kratz CP, Nesterova M, Greene MH, Stratakis CA. Cyclic AMP and c-KIT signaling in familial testicular germ cell tumor predisposition. J Clin Endocrinol Metab 2013; 98:E1393-400. [PMID: 23771924 PMCID: PMC3733859 DOI: 10.1210/jc.2012-2838] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Familial testicular germ cell tumors (FTGCTs) are hypothesized to result from the combined interaction of multiple low-penetrance genes. We reported inactivating germline mutations of the cAMP-binding phosphodiesterase 11A (PDE11A) as modifiers of FTGCT risk. Recent genome-wide association studies have identified single-nucleotide polymorphisms in the KITLG gene, the ligand for the cKIT tyrosine kinase receptor, as strong modifiers of susceptibility to both familial and sporadic testicular germ cell tumors. DESIGN We studied 94 patients with FTGCTs and 50 at-risk male relatives from 63 unrelated kindreds, in whom the PDE11A gene had been sequenced by investigating the association between KITLG genome-wide association study single-nucleotide polymorphisms rs3782179 and rs4474514 and FTGCT risk in these patients and in 692 controls. We also examined cAMP and c-KIT signaling in testicular tissues and cell lines and extended the studies to 2 sporadic cases, one with a PDE11A defect and one without, as a comparison. RESULTS We found a higher frequency of the KITLG risk alleles in FTGCT patients who also had a PDE11A sequence variant, compared with those with a wild-type PDE11A sequence. In NTERA-2 and Tcam-2 cells transfected with the mutated forms of PDE11A (R52T, F258Y, Y727C, R804H, V820M, R867G, and M878V), cAMP levels were significantly higher, and the relative phosphodiesterase activity was lower than in the wild-type cells. KITLG expression was consistently increased in the presence of PDE11A-inactivating defects, both at the RNA and protein levels, in familial testicular germ cell tumors. The 2 sporadic cases that were studied, one with a PDE11A defect and another without, agreed with the data in FTGTCT and in the cell lines. CONCLUSIONS Patients with FTGCT and PDE11A defects also carry KITLG risk alleles more frequently. There may be an interaction between cAMP and c-KIT signaling in predisposition to testicular germ cell tumors.
Collapse
Affiliation(s)
- Monalisa F Azevedo
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Quinonez SC, Park JM, Rabah R, Owens KM, Yashar BM, Glover TW, Keegan CE. 9p partial monosomy and disorders of sex development: Review and postulation of a pathogenetic mechanism. Am J Med Genet A 2013; 161A:1882-96. [DOI: 10.1002/ajmg.a.36018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/27/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Shane C. Quinonez
- Department of Pediatrics, Division of Genetics; University of Michigan; Ann Arbor; Michigan
| | - John M. Park
- Department of Urology; University of Michigan; Ann Arbor; Michigan
| | - Raja Rabah
- Department of Pathology; University of Michigan; Ann Arbor; Michigan
| | - Kailey M. Owens
- Department of Pediatrics, Division of Genetics; University of Michigan; Ann Arbor; Michigan
| | - Beverly M. Yashar
- Department of Human Genetics; University of Michigan; Ann Arbor; Michigan
| | | | | |
Collapse
|
45
|
Zechel JL, Doerner SK, Lager A, Tesar PJ, Heaney JD, Nadeau JH. Contrasting effects of Deadend1 (Dnd1) gain and loss of function mutations on allelic inheritance, testicular cancer, and intestinal polyposis. BMC Genet 2013; 14:54. [PMID: 23773267 PMCID: PMC3693958 DOI: 10.1186/1471-2156-14-54] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 06/07/2013] [Indexed: 11/10/2022] Open
Abstract
Background Certain mutations in the Deadend1 (Dnd1) gene are the most potent modifiers of testicular germ cell tumor (TGCT) susceptibility in mice and rats. In the 129 family of mice, the Dnd1Ter mutation significantly increases occurrence of TGCT-affected males. To test the hypothesis that he Dnd1Ter allele is a loss-of-function mutation; we characterized the consequences of a genetically-engineered loss-of-function mutation in mice, and compared these results with those for Dnd1Ter. Results We found that intercrossing Dnd1+/KO heterozygotes to generate a complete loss-of-function led to absence of Dnd1KO/KO homozygotes and significantly reduced numbers of Dnd1+/KO heterozygotes. Further crosses showed that Dnd1Ter partially rescues loss of Dnd1KO mice. We also found that loss of a single copy of Dnd1 in Dnd1KO/+ heterozygotes did not affect baseline occurrence of TGCT-affected males and that Dnd1Ter increased TGCT risk regardless whether the alternative allele was loss-of-function (Dnd1KO) or wild-type (Dnd1+). Finally, we found that the action of Dnd1Ter was not limited to testicular cancer, but also significantly increased polyp number and burden in the Apc+/Min model of intestinal polyposis. Conclusion These results show that Dnd1 is essential for normal allelic inheritance and that Dnd1Ter has a novel combination of functions that significantly increase risk for both testicular and intestinal cancer.
Collapse
Affiliation(s)
- Jennifer L Zechel
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland OH 44106, USA
| | | | | | | | | | | |
Collapse
|
46
|
Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, Skotheim RI, Abeler VM, Rajpert-De Meyts E, Lothe RA. Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr Rev 2013; 34:339-76. [PMID: 23575763 PMCID: PMC3787935 DOI: 10.1210/er.2012-1045] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review focuses on the molecular characteristics and development of rare malignant ovarian germ cell tumors (mOGCTs). We provide an overview of the genomic aberrations assessed by ploidy, cytogenetic banding, and comparative genomic hybridization. We summarize and discuss the transcriptome profiles of mRNA and microRNA (miRNA), and biomarkers (DNA methylation, gene mutation, individual protein expression) for each mOGCT histological subtype. Parallels between the origin of mOGCT and their male counterpart testicular GCT (TGCT) are discussed from the perspective of germ cell development, endocrinological influences, and pathogenesis, as is the GCT origin in patients with disorders of sex development. Integrated molecular profiles of the 3 main histological subtypes, dysgerminoma (DG), yolk sac tumor (YST), and immature teratoma (IT), are presented. DGs show genomic aberrations comparable to TGCT. In contrast, the genome profiles of YST and IT are different both from each other and from DG/TGCT. Differences between DG and YST are underlined by their miRNA/mRNA expression patterns, suggesting preferential involvement of the WNT/β-catenin and TGF-β/bone morphogenetic protein signaling pathways among YSTs. Characteristic protein expression patterns are observed in DG, YST and IT. We propose that mOGCT develop through different developmental pathways, including one that is likely shared with TGCT and involves insufficient sexual differentiation of the germ cell niche. The molecular features of the mOGCTs underline their similarity to pluripotent precursor cells (primordial germ cells, PGCs) and other stem cells. This similarity combined with the process of ovary development, explain why mOGCTs present so early in life, and with greater histological complexity, than most somatic solid tumors.
Collapse
Affiliation(s)
- Sigrid Marie Kraggerud
- Department of Cancer Prevention, Institute for Cancer Research, Oslo University Hospital, N-0310 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
47
|
Sharpe RM, Mitchell RT. The downside of 'inappropriate messaging': new insight into the development of testicular germ cell tumours in young men? J Pathol 2013; 229:497-501. [PMID: 23335366 DOI: 10.1002/path.4167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 12/30/2012] [Accepted: 01/01/2013] [Indexed: 12/19/2022]
Abstract
How invasive testicular germ cell tumours (TGCTs) develop from precursor carcinoma in situ/intratubular germ cell neoplasia unclassified (CIS/IGCNU) cells, and only after puberty, is unknown. In the current issue of The Journal of Pathology, Jørgensen and colleagues have compared the protein expression profile of CIS before and after puberty and in pre-invasive versus invasive TGCT and show that the mitosis-meiosis controller DMRT1 switches off in CIS cells postpubertally and is associated with invasiveness. They also show that CIS cells express a 'confusing' mix of pro- and anti-meiotic proteins; this may predispose CIS cells to accumulate extra chromosomal material which ultimately leads to tumourigenesis.
Collapse
Affiliation(s)
- Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| | | |
Collapse
|
48
|
Jørgensen A, Nielsen JE, Almstrup K, Toft BG, Petersen BL, Rajpert-De Meyts E. Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J Pathol 2013; 229:588-98. [PMID: 23303528 DOI: 10.1002/path.4154] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/15/2012] [Accepted: 11/27/2012] [Indexed: 12/17/2022]
Abstract
Testicular germ cell tumours (TGCT) of young adults arise from the intratubular precursor, carcinoma in situ (CIS). CIS cells are thought to be developmentally arrested and transformed fetal germ cells that survive through childhood and gain invasive capacity after puberty. Given that germ cell neoplasms arise frequently in undervirilized and dysgenetic gonads and the striking physiological difference between meiotic entry in ovaries (fetal life) versus testes (at puberty), this study aimed to investigate whether errors in regulation of meiosis may be implicated in the pathogenesis of CIS or its invasive progression to TGCT. The main focus was on a key sex differentiation and meiosis regulator, DMRT1, which has also been linked to TGCT risk in recent genetic association studies. Expression patterns of DMRT1 and other meiosis regulators (SCP3, DMC1, STRA8, CYP26B1, NANOS2, NANOS3) were investigated in pre- and post-pubertal CIS samples and TGCT by quantitative RT-PCR and immunohistochemistry. The results demonstrated that meiosis markers and meiosis inhibitors were simultaneously expressed in CIS cells, in both pre- and post-pubertal testis samples. DMRT1 was present in a restricted subset of CIS cells, which was relatively greater in pre-pubertal (27%) compared to adult (2.6%) samples. In contrast to the majority of CIS cells, DMRT1-positive CIS cells in adult testes were not proliferating. DMRT1 and most of the other meiosis regulators were absent or expressed at low levels in invasive TGCT, except in spermatocytic seminoma (not derived from CIS). In conclusion, this study indicates that meiosis signalling is dysregulated in CIS cells and that a key regulator of the mitosis-meiosis switch, DMRT1, is expressed in 'early-stage' CIS cells but is down-regulated with further invasive transformation. Whether this mixed meiosis signalling in CIS cells is caused by insufficient virilization of the fetal somatic niche or a partial post-pubertal maturation remains uncertain and requires further study.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital, Denmark
| | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Ni Z, Tao K, Chen G, Chen Q, Tang J, Luo X, Yin P, Tang J, Wang X. CLPTM1L is overexpressed in lung cancer and associated with apoptosis. PLoS One 2012; 7:e52598. [PMID: 23300716 PMCID: PMC3530437 DOI: 10.1371/journal.pone.0052598] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 11/16/2012] [Indexed: 11/18/2022] Open
Abstract
CLPTM1L is believed to be associated with lung cancer. However, there is little information regarding its expression and function. Here using immunohistochemistry, we found that CLPTM1L expression was markedly increased in lung cancer tissues relative to normal tissues, especially in lung adenocarcinoma. CLPTM1L expression was not found to be associated with stages, smoking status, lymph node metastasis, or T lymphocyte infiltration but with differentiation stage. We found CLPTM1L to be enriched in the mitochondrial compared with plasma membrane protein extracts. CLPTM1L-EGFP transfection showed that the molecule product was expressed in cytoplasm and indicated the mitochondrial localization stained with mitochondrial marker MitoTracker. CLPTM1L transferred lung cancer cell line 95-D showed no growth inhibition or cell apoptosis, but it did show inhibited sensitivity to cis-diamminedichloroplatinum(II) (cisplatin, CDDP). Knockdown of CLPTM1L by RNAi did not interfere with cell proliferation but it did increase cell sensitivity to CDDP and activation of caspase-9 and caspase-3/7. These data indicate CLPTM1L is a mitochondria protein and that it may be associated with anti-apoptotic mechanism which affects drug-resistance in turn.
Collapse
Affiliation(s)
- Zhenhua Ni
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kun Tao
- Department of Pathology, Shanghai Changning Central Hospital, Shanghai, China
| | - Guo Chen
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingge Chen
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianmin Tang
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuming Luo
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peihao Yin
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jihong Tang
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiongbiao Wang
- Central Lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|