1
|
Rajpert-De Meyts E, Goriely A, Almstrup K. New analysis of atypical spermatocytic tumours reveals extensive heterogeneity and plasticity of germ cell tumours †. J Pathol 2024; 263:1-4. [PMID: 38362619 DOI: 10.1002/path.6262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024]
Abstract
Testicular germ cell tumours (TGCTs) derived from immature (type I) and pluripotent germ cell neoplasia in situ (GCNIS, type II) are characterised by remarkable phenotypic heterogeneity and plasticity. In contrast, the rare spermatocytic tumour (SpT, type III), derived from mature spermatogonia, is considered a homogenous and benign tumour but may occasionally present as an anaplastic or an aggressive sarcomatoid tumour. While various oncogenic processes had been proposed, the precise mechanism driving malignant progression remained elusive until the molecular characterisation of a series of atypical SpTs described in a recent issue of The Journal of Pathology. The emerging picture suggests the presence of two distinct trajectories for SpTs, involving either RAS/mitogen-activated protein kinase pathway mutations or a ploidy shift with secondary TP53 mutations and/or gain of chromosome 12p, the latter known as pathognomonic for type II GCNIS-derived TGCTs. Here, we discuss the implications of these findings, seen from the perspective of germ cell biology and the unique features of different TGCTs. The evolving phenotype of SpTs, induced by genomic and epigenetic changes, illustrates that the concept of plasticity applies to all germ cell tumours, making them inherently heterogenous and capable of significant transformation during progression. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ewa Rajpert-De Meyts
- Department of Growth & Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anne Goriely
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Kristian Almstrup
- Department of Growth & Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Noughabi MK, Matin MM, Farshchian M, Bahrami AR. Immunomodulatory Properties of Mouse Mesenchymal Stromal/Stem Cells Upon Ectopic Expression of Immunoregulator Nanos2. Stem Cell Rev Rep 2022; 19:734-753. [PMID: 36348161 DOI: 10.1007/s12015-022-10451-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) are known for their involvement in modulating the immune system of mammals. This potency could be enhanced by different strategies, including regulation of key proteins, in order to meet desirable therapeutic properties. Nanos2, encoding an RNA-binding protein involved in regulation of key spermatogonial signaling pathways, has been demonstrated to downregulate a range of immune related genes in mouse embryonic fibroblasts (MEFs). Accordingly, it was hypothesized that Nanos2 functions as a potent immunosuppressing factor. This study was aimed to measure the expression profile of the immune-related genes in mouse mesenchymal stromal/stem cells (mMSCs) and assess their functional properties after Nanos2 ectopic expression. METHODS As inflammatory mediators, interferon (IFN-γ) and poly(I:C) were used to provoke an immune response. The interactions between the control and engineered mMSCs overexpressing Nanos2, with mouse peripheral blood mononuclear cells (mPBMCs) were then compared. The sensitivity of these cells to an inflammatory environment was assessed by using a conditioned medium containing high levels of inflammatory cytokines. Finally, the functional properties of the cells were investigated both in vivo and in vitro in presence of tumor and immune cells. RESULTS Deep transcriptome analysis indicated that numerous genes were downregulated as a result of higher Nanos2 expression. Most of the genes subjected to gene expression alteration, were responsible for controlling responses to external stimuli, cell-cell adhesion, and wound healing. In comparison to the control cells, Nanos2-overexpressing cells showed lower expression of several immune-related genes after pretreatment with IFN-γ and poly(I:C). They also exhibited inhibitory effects against mPBMCs proliferation. Tumor growth rate, in B16-F0 administered mice was obviously increased upon their treatment with the Nanos2-mMSCs, while no tumor or very small ones were developed in the control group. In addition, the cytotoxic environment had no significant effects on Nanos2-mMSCs. CONCLUSIONS According to the literature, MSCs are believed to be tuned very precisely by their internal and external conditions to act as either pro-inflammatory or anti-inflammatory agents. We show here that Nanos2 plays a significant role in promoting anti-inflammatory properties when expressed at higher levels by MSCs. This approach could be adopted for controlling the excessive inflammatory conditions in clinical programs, however more experiments are required to confirm it. In Brief Viral transduction was used to over express Nanos2 in mouse mesenchymal stromal/stem cells (mMSCs). Induced expression of Nanos2 downregulated the expression of immune-related genes and proteins. These modified mMSCs switched to an immunosuppressive state, even in the presence of pro-inflammatory cytokines; and could also contribute to tumor progression in a mouse model.
Collapse
Affiliation(s)
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Moein Farshchian
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
3
|
RANKL regulates testicular cancer growth and Denosumab treatment has suppressive effects on GCNIS and advanced seminoma. Br J Cancer 2022; 127:408-421. [PMID: 35418213 PMCID: PMC9345904 DOI: 10.1038/s41416-022-01810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Testicular germ cell tumours (TGCTs) have a high sensitivity to chemotherapy and a high cure rate, although with serious adverse effects. In the search for tumour suppressive drugs, the RANKL inhibitor Denosumab, used to treat osteoporosis, came up as a candidate since RANKL signalling was recently identified in the testis. METHODS Expression of RANKL, RANK and OPG, and the effects of RANKL inhibition were investigated in human TGCTs, TGCT-derived cell-lines, and TGCT-xenograft models. Serum RANKL was measured in TGCT-patients. RESULTS RANKL, RANK, and OPG were expressed in germ cell neoplasia in situ (GCNIS), TGCTs, and TGCT-derived cell lines. RANKL-inhibition reduced proliferation of seminoma-derived TCam-2 cells, but had no effect on embryonal carcinoma-derived NTera2 cells. Pretreatment with Denosumab did not augment the effect of cisplatin in vitro. However, inhibition of RANKL in vivo reduced tumour growth exclusively in the TCam-2-xenograft model and Denosumab-treatment decreased proliferation in human GCNIS cultures. In TGCT-patients serum RANKL had no prognostic value. CONCLUSIONS This study shows that the RANKL signalling system is expressed in GCNIS and seminoma where RANKL inhibition suppresses tumour growth in vitro and in vivo. Future studies are needed to determine whether RANKL is important for the malignant transformation or transition from GCNIS to invasive tumours.
Collapse
|
4
|
To Be or Not to Be a Germ Cell: The Extragonadal Germ Cell Tumor Paradigm. Int J Mol Sci 2021; 22:ijms22115982. [PMID: 34205983 PMCID: PMC8199495 DOI: 10.3390/ijms22115982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023] Open
Abstract
In the human embryo, the genetic program that orchestrates germ cell specification involves the activation of epigenetic and transcriptional mechanisms that make the germline a unique cell population continuously poised between germness and pluripotency. Germ cell tumors, neoplasias originating from fetal or neonatal germ cells, maintain such dichotomy and can adopt either pluripotent features (embryonal carcinomas) or germness features (seminomas) with a wide range of phenotypes in between these histotypes. Here, we review the basic concepts of cell specification, migration and gonadal colonization of human primordial germ cells (hPGCs) highlighting the analogies of transcriptional/epigenetic programs between these two cell types.
Collapse
|
5
|
Lobo J, Constâncio V, Guimarães-Teixeira C, Leite-Silva P, Miranda-Gonçalves V, Sequeira JP, Pistoni L, Guimarães R, Cantante M, Braga I, Maurício J, Looijenga LHJ, Henrique R, Jerónimo C. Promoter methylation of DNA homologous recombination genes is predictive of the responsiveness to PARP inhibitor treatment in testicular germ cell tumors. Mol Oncol 2021; 15:846-865. [PMID: 33513287 PMCID: PMC8024740 DOI: 10.1002/1878-0261.12909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most common cancers in men aged 15-39 years and are divided into two major groups, seminomas and nonseminomas. Novel treatment options are required for these patients, to limit side effects of chemotherapy. We hypothesized that promoter methylation of relevant homologous recombination (HR) genes might be predictive of response to poly-ADP ribose polymerase inhibitors (PARPis) in TGCTs. We report a study pipeline combining in silico, in vitro, and clinical steps. By using several databases and in silico tools, we identified BRCA1, RAD51C, PALB2, RAD54B, and SYCP3 as the most relevant genes for further investigation and pinpointed specific CpG sites with pronounced negative correlation to gene expression. Nonseminomas displayed significantly higher methylation levels for all target genes, where increased methylation was observed in patients with more differentiated subtypes and higher disease burden. We independently performed second-line targeted validation in tissue series from TGCT patients. A moderate and/or strong anti-correlation between gene expression (assessed by RNA-sequencing) and promoter methylation (assessed by 450k array) was found, for all of the targets. As a proof of concept, we demonstrated the sensitivity of TGCT cell lines to Olaparib, which associated with differential methylation levels of a subset of targets, namely BRCA1 and RAD51C. Our findings support the use of HR genes promoter methylation as a predictor of the therapeutic response to PARPis in patients with TGCT.
Collapse
Affiliation(s)
- João Lobo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Vera Constâncio
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Catarina Guimarães-Teixeira
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Pedro Leite-Silva
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - José Pedro Sequeira
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Laura Pistoni
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Biology, University of Pisa, Italy
| | - Rita Guimarães
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Mariana Cantante
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Isaac Braga
- Department of Urology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Joaquina Maurício
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | | | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal
| |
Collapse
|
6
|
Xu Y, Xie J. Etomoxir regulates the differentiation of male germ cells by specifically reducing H3K27ac level. BMC DEVELOPMENTAL BIOLOGY 2021; 21:5. [PMID: 33517883 PMCID: PMC7849134 DOI: 10.1186/s12861-020-00237-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Background Fatty acid oxidation plays an important role in a variety of developing and mature organ systems. However, the role of this metabolic pathway in different stages of testis development remains unknown. Here, we elucidate the mechanisms by which fatty acid oxidation regulates the maintenance and differentiation of gonocytes and spermatogonial stem cells. Results During E13.5-E15.5, male germ cells gradually enter the mitotic arrest phase, while the expression of CPT1A, a rate-limiting enzyme for fatty acid oxidation, gradually increases. Therefore, we treated pregnant mice (E13.5 to E15.5) with etomoxir, which is an inhibitor of CPT1A. Etomoxir-treated mice showed no difference in embryonic morphology; however, etomoxir-treated male gonocytes exited mitotic arrest, and cells of the gonad underwent apoptosis. In addition, etomoxir-treated mice at P7 displayed impaired homing of spermatogonia and increased cell apoptosis. We further demonstrated that inhibition of fatty acid oxidation in gonads was associated with gonocyte differentiation events and the histone modification H3K27ac. Conclusions Inhibiting fatty acid oxidation can specifically reduce the level of H3K27ac in the reproductive crest, which may be the cause of the down-regulation of male differentiation-specific gene expression, which ultimately leads to the male primordial germ cells exited from mitotic arrest. Our work uncovers metabolic reprogramming during male gonadal development, revealing that it plays an important role in the maintenance of gonocytes in a differentiated and quiescent state during foetal testis development.
Collapse
Affiliation(s)
- Yushan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
7
|
Zhang F, Liu R, Liu C, Zhang H, Lu Y. Nanos3, a cancer-germline gene, promotes cell proliferation, migration, chemoresistance, and invasion of human glioblastoma. Cancer Cell Int 2020; 20:197. [PMID: 32508533 PMCID: PMC7249350 DOI: 10.1186/s12935-020-01272-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background Radiotherapy, chemotherapy, and surgery have made crucial strides in glioblastoma treatment, yet they often fail; thus, new treatment and new detection methods are needed. Aberrant expression of Nanos3 has been functionally associated with various cancers. Here, we sought to identify the clinical significance and potential mechanisms of Nanos3 in human glioblastoma. Methods Nanos3 expression was studied in nude mouse glioblastoma tissues and glioblastoma cell lines by immunohistochemistry, Western blot, and RT-PCR. Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing assay was performed to generate the Nanos3 knockdown glioblastoma cell lines. The effects of Nanos3 on glioblastoma cells proliferation, migration, invasion, chemoresistance, germ cell characteristics, and tumor formation were analyzed by CCK8, transwell, cell survival experiments and alkaline phosphatase staining in vitro and in nude mouse models in vivo. Correlation between the expression of stemness proteins and the expression of Nanos3 was evaluated by Western blot. Results We found that Nanos3 was strongly expressed in both glioblastoma cell lines and tissues. Western blot and sequencing assays showed that the Nanos3 knockdown glioblastoma cell lines were established successfully, and we discovered that Nanos3 deletion reduced the proliferation, migration, and invasion of glioblastoma cells in vitro (P < 0.05). Nanos3 knockdown enhanced the sensitivity of glioblastoma cells to doxorubicin (DOX) and temozolomide (TMZ) (P < 0.05), and Nanos3+/- glioblastoma cell lines did not show the characteristics of the germline cells. In addition, Nanos3 deletion inhibited subcutaneous xenograft tumor growth in vivo (P < 0.001). Moreover, the oncogenesis germline protein levels of CD133, Oct4, Ki67, and Dazl decreased significantly in glioblastoma cells following Nanos3 knockdown. Conclusions Both in vitro and in vivo assays suggest that Nanos3, which is a cancer-germline gene, initiates the tumorigenesis of glioblastoma via acquiring the oncogenesis germline traits. These data demonstrate that ectopic germline traits are necessary for glioblastoma growth.
Collapse
Affiliation(s)
- Fengyu Zhang
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Jing-an District, Shanghai, 200040 China
| | - Ruilai Liu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Jing-an District, Shanghai, 200040 China
| | - Cheng Liu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Jing-an District, Shanghai, 200040 China
| | - Haishi Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Jing-an District, Shanghai, 200040 China
| | - Yuan Lu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Jing-an District, Shanghai, 200040 China
| |
Collapse
|
8
|
Guioli S, Zhao D, Nandi S, Clinton M, Lovell-Badge R. Oestrogen in the chick embryo can induce chromosomally male ZZ left gonad epithelial cells to form an ovarian cortex that can support oogenesis. Development 2020; 147:dev181693. [PMID: 32001442 PMCID: PMC7055392 DOI: 10.1242/dev.181693] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/16/2020] [Indexed: 12/25/2022]
Abstract
In chickens, the embryonic ovary differentiates into two distinct domains before meiosis: a steroidogenic core (the female medulla), overlain by the germ cell niche (the cortex). The differentiation of the medulla is a cell-autonomous process based on chromosomal sex identity (CASI). In order to address the extent to which cortex differentiation depends on intrinsic or extrinsic factors, we generated models of gonadal intersex by mixing ZW (female) and ZZ (male) cells in gonadal chimeras, or by altering oestrogen levels of ZW and ZZ embryos. We found that CASI does not apply to the embryonic cortex. Both ZW and ZZ cells can form the cortex and this can happen independently of the phenotypic sex of the medulla as long as oestrogen is provided. We also show that the cortex-promoting activity of oestrogen signalling is mediated via estrogen receptor alpha within the left gonad epithelium. However, the presence of a medulla with an 'intersex' or male phenotype may compromise germ cell progression into meiosis, causing cortical germ cells to remain in an immature state in the embryo.
Collapse
Affiliation(s)
| | - Debiao Zhao
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Sunil Nandi
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Michael Clinton
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | | |
Collapse
|
9
|
Rajpert-De Meyts E. Testicular germ cell cancer: recent developments in biology and clinical management. Andrology 2019; 7:391-393. [DOI: 10.1111/andr.12675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- E. Rajpert-De Meyts
- Department of Growth and Reproduction; Copenhagen University Hospital (Rigshospitalet); Copenhagen Denmark
| |
Collapse
|
10
|
Abstract
Testicular germ cell tumors are a diverse group of neoplasms, consisting of the prepubertal type 1 tumors, pure teratoma, and pure yolk sac tumor, the type 2 tumors, which are biologically malignant, preceded by germ cell neoplasia in situ, and harbor chromosome 12p abnormalities, and the type 3 tumor, spermatocytic tumor, which features chromosome 9p amplification. These arise in distinct clinical settings, and harbor distinct genetic abnormalities, immunohistochemical properties, and morphologic features. Here we have attempted to unify embryology, morphology, immunohistochemistry, and genetics in order to distill this fascinating group of neoplasms into what we hope is a useful framework for understanding their classification.
Collapse
|
11
|
Andries V, De Keuckelaere E, Staes K, Hochepied T, Taminau J, Lemeire K, Birembaut P, Berx G, van Roy F. A new mouse model to study the role of ectopic Nanos3 expression in cancer. BMC Cancer 2019; 19:598. [PMID: 31208373 PMCID: PMC6580527 DOI: 10.1186/s12885-019-5807-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND NANOS3 is a gene conserved throughout evolution. Despite the quite low conservation of Nanos sequences between different organisms and even between Nanos paralogs, their role in germ cell development is remarkably universal. Human Nanos3 expression is normally restricted to the gonads and the brain. However, ectopic activation of this gene has been detected in various human cancers. Until now, Nanos3 and other Nanos proteins have been studied almost exclusively in germ cell development. METHODS Transgenic mice were generated by targeted insertion of a human Nanos3 cDNA into the ROSA26 locus. The transgene could be spatiotemporally induced by Cre recombinase activity removing an upstream floxed STOP cassette. A lung tumor model with ectopic Nanos3 expression was based on the lung-specific activation of the reverse tetracycline transactivator gene, in combination with a tetO-CMV promoter controlling Cre expression. When doxycycline was provided to the mice, Cre was activated leading to deletion of TP53 alleles and activation of both oncogenic KRasG12D and Nanos3. Appropriate controls were foreseen. Tumors and tumor-derived cell cultures were analyzed in various ways. RESULTS We describe the successful generation of Nanos3LSL/- and Nanos3LSL/LSL mice in which an exogenous human NANOS3 gene can be activated in vivo upon Cre expression. These mice, in combination with different conditional and doxycycline-inducible Cre lines, allow the study of the role of ectopic Nanos3 expression in several cancer types. The Nanos3LSL mice were crossed with a non-small cell lung cancer (NSCLC) mouse model based on conditional expression of oncogenic KRas and homozygous loss of p53. This experiment demonstrated that ectopic expression of Nanos3 in the lungs has a significant negative effect on survival. Enhanced bronchiolar dysplasia was observed when Nanos3-expressing NSCLC mice were compared with control NSCLC mice. An allograft experiment, performed with cell cultures derived from primary lung tumors of control and Nanos3-expressing NSCLC mice, revealed lymph node metastasis in mice injected with Nanos3-expressing NSCLC cells. CONCLUSIONS A new mouse model was generated allowing examination of Nanos3-associated pathways and investigation of the influence of ectopic Nanos3 expression in various cancer types. This model might identify Nanos3 as an interesting target in cancer therapeutics.
Collapse
Affiliation(s)
- Vanessa Andries
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Evi De Keuckelaere
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Katrien Staes
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Tino Hochepied
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Joachim Taminau
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kelly Lemeire
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Philippe Birembaut
- INSERM UMRS 1250, Department of Biopathology, CHU Maison-Blanche, University Hospital of Reims & University of Reims Champagne-Ardenne, rue Cognacq-Jay 45, 51092, Reims, France
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Frans van Roy
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
| |
Collapse
|
12
|
Macdonald J, Kilcoyne KR, Sharpe RM, Kavanagh Á, Anderson RA, Brown P, Smith LB, Jørgensen A, Mitchell RT. DMRT1 repression using a novel approach to genetic manipulation induces testicular dysgenesis in human fetal gonads. Hum Reprod 2019; 33:2107-2121. [PMID: 30272154 PMCID: PMC6195803 DOI: 10.1093/humrep/dey289] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/04/2018] [Indexed: 01/16/2023] Open
Abstract
STUDY QUESTION Does loss of DMRT1 in human fetal testis alter testicular development and result in testicular dysgenesis? SUMMARY ANSWER DMRT1 repression in human fetal testis alters the expression of key testicular and ovarian determining genes, and leads to focal testicular dysgenesis. WHAT IS KNOWN ALREADY Testicular dysgenesis syndrome (TDS) is associated with common testicular disorders in young men, but its etiology is unknown. DMRT1 has been shown to play a role in the regulation of sex differentiation in the vertebrate gonad. Downregulation of DMRT1 in male mice results in trans-differentiation of Sertoli cells into granulosa (FOXL2+) cells resulting in an ovarian gonadal phenotype. STUDY DESIGN, SIZE, DURATION To determine the effect of DMRT1 repression on human fetal testes, we developed a novel system for genetic manipulation, which utilizes a Lentivral delivered miRNA during short-term in vitro culture (2 weeks). A long-term (4–6 weeks) ex vivo xenograft model was used to determine the subsequent effects of DMRT1 repression on testicular development and maintenance. We included first and second-trimester testis tissue (8–20 weeks gestation; n = 12) in the study. PARTICIPANTS/MATERIALS, SETTING, METHODS Human fetal testes were cultured in vitro and exposed to either of two DMRT1 miRNAs (miR536, miR641), or to scrambled control miRNA, for 24 h. This was followed by a further 14 days of culture (n = 3–4), or xenografting (n = 5) into immunocompromised mice for 4–6 weeks. Tissues were analyzed by histology, immunohistochemistry, immunofluorescence and quantitative RT-PCR. Endpoints included histological evaluation of seminiferous cord integrity, mRNA expression of testicular, ovarian and germ cell genes, and assessment of cell number and protein expression for proliferation, apoptosis and pluripotency factors. Statistical analysis was performed using a linear mixed effect model. MAIN RESULTS AND THE ROLE OF CHANCE DMRT1 repression (miR536/miR641) resulted in a loss of DMRT1 protein expression in a sub-population of Sertoli cells of first trimester (8–11 weeks gestation) human fetal testis; however, this did not affect the completion of seminiferous cord formation or morphological appearance. In second-trimester testis (12–20 weeks gestation), DMRT1 repression (miR536/miR641) resulted in disruption of seminiferous cords with absence of DMRT1 protein expression in Sertoli (SOX9+) cells. No differences in proliferation (Ki67+) were observed and apoptotic cells (CC3+) were rare. Expression of the Sertoli cell associated gene, SOX8, was significantly reduced (miR536, 34% reduction, P = 0.031; miR641 36% reduction, P = 0.026), whilst SOX9 expression was unaffected. Changes in expression of AMH (miR536, 100% increase, P = 0.033), CYP26B1 (miR641, 38% reduction, P = 0.05) and PTGDS (miR642, 30% reduction, P = 0.0076) were also observed. Amongst granulosa cell associated genes, there was a significant downregulation in R-spondin 1 expression (miR536, 76% reduction, P < 0.0001; miR641, 49% reduction, P = 0.046); however, there were no changes in expression of the granulosa cell marker, FOXL2. Analysis of germ cell associated genes demonstrated a significant increase in the expression of the pluripotency gene OCT4 (miR536, 233%, P < 0.001). We used the xenograft system to investigate the longer-term effects of seminiferous cord disruption via DMRT1 repression. As was evident in vitro for second-trimester samples, DMRT1 repression resulted in focal testicular dysgenesis similar to that described in adults with TDS. These dysgenetic areas were devoid of germ cells, whilst expression of FOXL2 within the dysgenetic areas, indicated trans-differentiation from a male (Sertoli cell) to female (granulosa cell) phenotype. LIMITATIONS, REASONS FOR CAUTION Human fetal testis tissue is a limited resource; however, we were able to demonstrate significant effects of DMRT1 repression on the expression of germ and somatic cell genes, in addition to the induction of focal testicular dysgenesis, using these limited samples. In vitro culture may not reflect all aspects of human fetal testis development and function; however, the concurrent use of the xenograft model which represents a more physiological system supports the validity of the in vitro findings. WIDER IMPLICATIONS OF THE FINDINGS Our findings have important implications for understanding the role of DMRT1 in human testis development and in the origin of testicular dysgenesis. In addition, we provide validation of a novel system that can be used to determine the effects of repression of genes that have been implicated in gonadal development and associated human reproductive disorders. STUDY FUNDING/COMPETING INTEREST(S) This project was funded by a Wellcome Trust Intermediate Clinical Fellowship (Grant No. 098522) awarded to RTM. LBS was supported by MRC Programme Grant MR/N002970/1. RAA was supported by MRC Programme Grant G1100357/1. RMS was supported by MRC Programme Grant G33253. This work was undertaken in the MRC Centre for Reproductive Health which is funded by the MRC Centre grant MR/N022556/1. The funding bodies had no input into the conduct of the research or the production of this manuscript. The authors have declared no conflicts of interest.
Collapse
Affiliation(s)
- Joni Macdonald
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Karen R Kilcoyne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Áine Kavanagh
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Pamela Brown
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK.,School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, NSW, Australia
| | - Anne Jørgensen
- University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK.,Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, Scotland, UK
| |
Collapse
|
13
|
Feichtinger J, McFarlane RJ. Meiotic gene activation in somatic and germ cell tumours. Andrology 2019; 7:415-427. [PMID: 31102330 PMCID: PMC6766858 DOI: 10.1111/andr.12628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022]
Abstract
Background Germ cell tumours are uniquely associated with the gametogenic tissues of males and females. A feature of these cancers is that they can express genes that are normally tightly restricted to meiotic cells. This aberrant gene expression has been used as an indicator that these cancer cells are attempting a programmed germ line event, meiotic entry. However, work in non‐germ cell cancers has also indicated that meiotic genes can become aberrantly activated in a wide range of cancer types and indeed provide functions that serve as oncogenic drivers. Here, we review the activation of meiotic factors in cancers and explore commonalities between meiotic gene activation in germ cell and non‐germ cell cancers. Objectives The objectives of this review are to highlight key questions relating to meiotic gene activation in germ cell tumours and to offer possible interpretations as to the biological relevance in this unique cancer type. Materials and Methods PubMed and the GEPIA database were searched for papers in English and for cancer gene expression data, respectively. Results We provide a brief overview of meiotic progression, with a focus on the unique mechanisms of reductional chromosome segregation in meiosis I. We then offer detailed insight into the role of meiotic chromosome regulators in non‐germ cell cancers and extend this to provide an overview of how this might relate to germ cell tumours. Conclusions We propose that meiotic gene activation in germ cell tumours might not indicate an unscheduled attempt to enter a full meiotic programme. Rather, it might simply reflect either aberrant activation of a subset of meiotic genes, with little or no biological relevance, or aberrant activation of a subset of meiotic genes as positive tumour evolutionary/oncogenic drivers. These postulates provide the provocation for further studies in this emerging field.
Collapse
Affiliation(s)
- J Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria.,OMICS Center Graz, BioTechMed Graz, Graz, Austria
| | - R J McFarlane
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Bangor, Gwynedd, UK
| |
Collapse
|
14
|
Hilbold E, Bergmann M, Fietz D, Kliesch S, Weidner W, Langeheine M, Rode K, Brehm R. Immunolocalization of DMRTB1 in human testis with normal and impaired spermatogenesis. Andrology 2019; 7:428-440. [PMID: 30920770 DOI: 10.1111/andr.12617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The transcription factor DMRTB1 plays a pivotal role in coordinating the transition between mitosis and meiosis in murine germ cells. No reliable data are available for human testis. OBJECTIVES The present study aims to examine the testicular expression pattern of DMRTB1 in men showing normal and impaired spermatogenesis. MATERIALS AND METHODS Immunohistochemistry was performed using 54 human testicular biopsy specimens and a commercial rabbit polyclonal anti-DMRTB1 primary antibody. RT-PCR complemented immunohistochemistry. To further characterize immunopositive cells and possible co-localization, the proliferation marker Ki-67, the tumor marker PLAP, and an anti-DMRT1 antibody were used. RESULTS In men with normal spermatogenesis, a strong immunoreactivity was detectable in a subset of spermatogonia (38.34 ± 2.14%). Some spermatocytes showed a weak immunostaining. Adjacent Sertoli cells were immunonegative. Compared with a hematoxylin and eosin overview staining, these immunopositive cells were almost exclusively identified as Apale and B spermatogonia and primary spermatocytes in (pre-)leptotene, zygotene, and pachytene stages. In patients with spermatogenic arrest at spermatogonial level, an altered staining pattern was found. No immunoreactivity was detected in Sertoli cells in Sertoli cell-only syndrome. In germ cell neoplasia in situ (GCNIS) tubules, except for a few (0.4 ± 0.03%), pre-invasive tumor cells were immunonegative. Seminoma cells showed no immunostaining. DISCUSSION According to previous findings in mice, it seems reasonable that DMRTB1 is expressed in these normal germ cell populations. Moreover, altered staining pattern in spermatogenic arrest at spermatogonial stage suggests a correlation with mitosis and transformation into B spermatogonia. The absence of DMRTB1 in GCNIS cells and tumor cells might be associated with uncontrolled neoplastic cell proliferation and progression into invasive germ cell tumors. Further research is required to elucidate, for example, the role of DMRTB1 in the malignant transformation of human germ cells. CONCLUSION Our data indicate a relevant role for DMRTB1 regarding the entry of spermatogonia into meiosis in men.
Collapse
Affiliation(s)
- E Hilbold
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - M Bergmann
- Institute for Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - D Fietz
- Institute for Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - S Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - W Weidner
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - M Langeheine
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - K Rode
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - R Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
15
|
Mérida-García A, Díaz-Serrano A, Bernard B, Del Mar Galera M, de Velasco G, Sepúlveda JM, Fernández L, Medina J, Castellano D. Update on the management of patients with intermediate and poor-risk testicular germ cell tumors and new biological insights. Cancer Treat Res Commun 2019; 19:100117. [PMID: 30684681 DOI: 10.1016/j.ctarc.2018.100117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/17/2018] [Accepted: 12/28/2018] [Indexed: 11/26/2022]
Affiliation(s)
- Antonio Mérida-García
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain.
| | - Asunción Díaz-Serrano
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain
| | - Brandon Bernard
- Division of Medical Oncology, University of Colorado Denver, 12801 E. 17th Avenue, Aurora, CO 80045, USA
| | - María Del Mar Galera
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain
| | - Guillermo de Velasco
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain
| | - Juan Manuel Sepúlveda
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain
| | - Lourdes Fernández
- Division of Medical Oncology, Virgen de la Salud Hospital, Av. de Barber, 30, 45004 Toledo, Spain
| | - Javier Medina
- Division of Medical Oncology, Virgen de la Salud Hospital, Av. de Barber, 30, 45004 Toledo, Spain
| | - Daniel Castellano
- Division of Medical Oncology, 12 de Octubre University Hospital, Avenida de Córdoba s/n 28041 Madrid, Spain
| |
Collapse
|
16
|
Tsang M, Gantchev J, Netchiporouk E, Moreau L, Ghazawi FM, Glassman S, Sasseville D, Litvinov IV. A study of meiomitosis and novel pathways of genomic instability in cutaneous T-cell lymphomas (CTCL). Oncotarget 2018; 9:37647-37661. [PMID: 30701021 PMCID: PMC6340880 DOI: 10.18632/oncotarget.26479] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
Genomic instability is a hallmark of cancer and an enabling factor for genetic alterations that drive cancer development and progression. The clashing of mitosis and aberrantly expressed meiosis machineries, which may contribute to genomic instability, has been coined cancer “meiomitosis”. LINE-1 retrotransposition, a process active in germ cells, acts outside of the meiotic machinery to create DNA double strand breaks (DNA DSBs) and has played an important role in the evolution of the human genome. We have previously demonstrated that in CTCL several cancer testis/meiotic genes are expressed. Furthermore, this cancer exhibits extensive and ongoing chromosomal/microsatellite instability. In this study we analyzed immortalized patient-derived cells and primary CTCL patient samples using RT-PCR, western blotting and confocal microscopy and found that proteins critically involved in meiosis and LINE-1 retrotransposition are expressed and are associated with chromosomal instability and DNA DSB formation. Using cell cycle synchronization, we show G1/S phase-transition-specific expression of meiosis proteins. Using the Alu retrotransposition assay, we demonstrate the functional activity of LINE-1 retrotransposon in CTCL. Histone acetyltransferase inhibition results in downregulation of the ectopic germ cell programs and concomitant decrease in DNA DSBs foci formation. Notably, LINE-1 and meiosis genes were expressed across a panel of other solid tumor cell lines. Taken together, our results indicate that malignant cells in culture undergo “cancer meiomitosis” rather than the classic mitosis division. The ectopic expression of meiosis genes and reactivation of LINE-1 may be contributing to genomic instability and represent novel targets for immunotherapy in this and other cancers.
Collapse
Affiliation(s)
- Matthew Tsang
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Jennifer Gantchev
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Elena Netchiporouk
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Linda Moreau
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Feras M Ghazawi
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Steven Glassman
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Denis Sasseville
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Ivan V Litvinov
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada.,Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| |
Collapse
|
17
|
Jørgensen A, Macdonald J, Nielsen JE, Kilcoyne KR, Perlman S, Lundvall L, Langhoff Thuesen L, Juul Hare K, Frederiksen H, Andersson AM, Skakkebæk NE, Juul A, Sharpe RM, Rajpert-De Meyts E, Mitchell RT. Nodal Signaling Regulates Germ Cell Development and Establishment of Seminiferous Cords in the Human Fetal Testis. Cell Rep 2018; 25:1924-1937.e4. [PMID: 30428358 DOI: 10.1016/j.celrep.2018.10.064] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 01/26/2023] Open
Abstract
Disruption of human fetal testis development is widely accepted to underlie testicular germ cell cancer (TGCC) origin and additional disorders within testicular dysgenesis syndrome (TDS). However, the mechanisms for the development of testicular dysgenesis in humans are unclear. We used ex vivo culture and xenograft approaches to investigate the importance of Nodal and Activin signaling in human fetal testis development. Inhibition of Nodal, and to some extent Activin, signaling disrupted seminiferous cord formation, abolished AMH expression, reduced androgen secretion, and decreased gonocyte numbers. Subsequent xenografting of testicular tissue rescued the disruptive effects on seminiferous cords and somatic cells but not germ cell effects. Stimulation of Nodal signaling increased the number of germ cells expressing pluripotency factors, and these persisted after xenografting. Our findings suggest a key role for Nodal signaling in the regulation of gonocyte differentiation and early human testis development with implications for the understanding of TGCC and TDS origin.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Joni Macdonald
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Karen R Kilcoyne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Signe Perlman
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Lene Lundvall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Lea Langhoff Thuesen
- Department of Obstetrics and Gynaecology, Hvidovre University Hospital, Kettegård Alle 30, Hvidovre, Denmark
| | - Kristine Juul Hare
- Department of Obstetrics and Gynaecology, Hvidovre University Hospital, Kettegård Alle 30, Hvidovre, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Niels E Skakkebæk
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - 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
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
18
|
Yang SY, Lee HJ, Lee HC, Hwang YS, Park YH, Ono T, Han JY. The dynamic development of germ cells during chicken embryogenesis. Poult Sci 2018; 97:650-657. [PMID: 29126291 DOI: 10.3382/ps/pex316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/25/2017] [Indexed: 01/05/2023] Open
Abstract
Appropriate regulation of cell proliferation during embryogenesis is crucial for the maintenance of germness. An in-depth understanding of germ cell developmental processes may yield valuable information on germ cell biology and applied sciences. However, direct evidences about germ cell proliferation and cell cycling during avian embryonic development has not been well-studied. Thus, we explored chicken germ cell dynamics during embryonic development via flow cytometry employing a germ cell-specific anti-cVASA antibody (the chicken VASA homolog is termed CVH) and propidium iodide staining. The numbers of male germ cells increased significantly during early embryonic development, but proliferation was decreased significantly with accumulation at the G0/G1 phase after embryonic d 14 (E.14), indicating initiation of mitotic arrest in the testis. On the other hand, the number of female germ cells increased significantly throughout embryogenesis, and proliferating cells were continuously evident in the ovary to the time of hatching, although gradual accumulation of cells at the G2/M phase was also evident. 5-ethynyl-2΄-deoxyuridine (EdU) incorporation analysis revealed that populations of mitotically active germ cells existed in both sexes during late embryogenesis, indicating either the maintenance of stem cell populations, or asynchronous meiosis. Collectively, these results indicate that chicken germ cells exhibited conserved developmental processes that were clearly sexually dimorphic.
Collapse
Affiliation(s)
- Seo Yeong Yang
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Hong Jo Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Hyung Chul Lee
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Young Sun Hwang
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Young Hyun Park
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Tamao Ono
- Division of Animal Science Faculty of Agriculture, Shinshu University, 8304 Minamiminowa, Nagano 399-4598, Japan
| | - Jae Yong Han
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,Institute for Biomedical Sciences, Shinshu University, Minamiminowa, Nagano 399-4598, Japan
| |
Collapse
|
19
|
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
|
20
|
Juel Mortensen L, Blomberg Jensen M, Christiansen P, Rønholt AM, Jørgensen A, Frederiksen H, Nielsen JE, Loya AC, Grønkær Toft B, Skakkebæk NE, Rajpert-De Meyts E, Juul A. Germ Cell Neoplasia in Situ and Preserved Fertility Despite Suppressed Gonadotropins in a Patient With Testotoxicosis. J Clin Endocrinol Metab 2017; 102:4411-4416. [PMID: 29029242 DOI: 10.1210/jc.2017-01761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/29/2017] [Indexed: 01/01/2023]
Abstract
CONTEXT Testotoxicosis is an autosomal-dominant, male-limited disorder. Activating mutations in the luteinizing hormone receptor gene (LHCGR) cause high autonomous testosterone secretion, resulting in early-onset peripheral precocious puberty. Little is known about long-term consequences of testotoxicosis. CASE DESCRIPTION We present a rare case of a patient followed for 25 years with two remarkable outcomes: preserved fertility and germ cell neoplasia in situ (GCNIS). He presented with precocious puberty at 10 months of age and was diagnosed with testotoxicosis due to a de novo heterozygous Asp578Tyr mutation in LHCGR. Testicular biopsy in childhood showed Leydig cell hyperplasia with altered cell maturation. From infancy throughout adulthood, elevated testosterone and estradiol, low inhibin B and anti-Müllerian hormone, and completely suppressed follicle-stimulating hormone and luteinizing hormone were noted. Height acceleration and advanced bone age resulted in a reduced final height. Semen analysis revealed ongoing spermatogenesis, and the patient fathered a child by natural conception. Ketoconazole treatment decreased circulating testosterone in childhood, supported by experimental suppression of testosterone production in his adult testis tissue cultured ex vivo. At 25 years of age, ultrasound revealed a testicular tumor, identified as a Leydig cell adenoma, but unexpectedly with GCNIS present in adjacent seminiferous tubules. CONCLUSION The case illustrates that absence of gonadotropins but high intratesticular testosterone concentration is sufficient for spermatogenesis and to allow fatherhood. Our study is also the first description, to our knowledge, of GCNIS in a patient with testotoxicosis. We recommend regular clinical examination and ultrasonic evaluation of the testes in these patients due to potential increased risk of malignancy.
Collapse
Affiliation(s)
- Li Juel Mortensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard University
| | - Martin Blomberg Jensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard University
| | - Peter Christiansen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | - Anand C Loya
- Department of Pathology, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Niels E Skakkebæk
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Denmark
| |
Collapse
|
21
|
Rudolph C, Melau C, Nielsen JE, Vile Jensen K, Liu D, Pena-Diaz J, Rajpert-De Meyts E, Rasmussen LJ, Jørgensen A. Involvement of the DNA mismatch repair system in cisplatin sensitivity of testicular germ cell tumours. Cell Oncol (Dordr) 2017; 40:341-355. [PMID: 28536927 DOI: 10.1007/s13402-017-0326-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Testicular germ cell tumours (TGCT) are highly sensitive to cisplatin-based chemotherapy, but patients with tumours containing differentiated teratoma components are less responsive to this treatment. The cisplatin sensitivity in TGCT has previously been linked to the embryonic phenotype in the majority of tumours, although the underlying mechanism largely remains to be elucidated. The aim of this study was to investigate the role of the DNA mismatch repair (MMR) system in the cisplatin sensitivity of TGCT. METHODS The expression pattern of key MMR proteins, including MSH2, MSH6, MLH1 and PMS2, were investigated during testis development and in the pathogenesis of TGCT, including germ cell neoplasia in situ (GCNIS). The TGCT-derived cell line NTera2 was differentiated using retinoic acid (10 μM, 6 days) after which MMR protein expression and activity, as well as cisplatin sensitivity, were investigated in both undifferentiated and differentiated cells. Finally, the expression of MSH2 was knocked down by siRNA in NTera2 cells after which the effect on cisplatin sensitivity was examined. RESULTS MMR proteins were expressed in proliferating cells in the testes, while in malignant germ cells MMR protein expression was found to coincide with the expression of the pluripotency factor OCT4, with no or low expression in the more differentiated yolk sac tumours, choriocarcinomas and teratomas. In differentiated NTera2 cells we found a significantly (p < 0.05) lower expression of the MMR and pluripotency factors, as well as a reduced MMR activity and cisplatin sensitivity, compared to undifferentiated NTera2 cells. Also, we found that partial knockdown of MSH2 expression in undifferentiated NTera2 cells resulted in a significantly (p < 0.001) reduced cisplatin sensitivity. CONCLUSION This study reports, for the first time, expression of the MMR system in fetal gonocytes, from which GCNIS cells are derived. Our findings in primary TGCT specimens and TGCT-derived cells suggest that a reduced sensitivity to cisplatin in differentiated TGCT components could result from a reduced expression of MMR proteins, in particular MSH2 and MLH1, which are involved in the recognition of cisplatin adducts and in activation of the DNA damage response pathway to initiate apoptosis.
Collapse
Affiliation(s)
- Christiane Rudolph
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Cecilie Melau
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - John E Nielsen
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Kristina Vile Jensen
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Dekang Liu
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Javier Pena-Diaz
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Lene Juel Rasmussen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Anne Jørgensen
- University Department of Growth and Reproduction (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
22
|
Giannoulatou E, Maher GJ, Ding Z, Gillis AJM, Dorssers LCJ, Hoischen A, Rajpert-De Meyts E, McVean G, Wilkie AOM, Looijenga LHJ, Goriely A. Whole-genome sequencing of spermatocytic tumors provides insights into the mutational processes operating in the male germline. PLoS One 2017; 12:e0178169. [PMID: 28542371 PMCID: PMC5439955 DOI: 10.1371/journal.pone.0178169] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022] Open
Abstract
Adult male germline stem cells (spermatogonia) proliferate by mitosis and, after puberty, generate spermatocytes that undertake meiosis to produce haploid spermatozoa. Germ cells are under evolutionary constraint to curtail mutations and maintain genome integrity. Despite constant turnover, spermatogonia very rarely form tumors, so-called spermatocytic tumors (SpT). In line with the previous identification of FGFR3 and HRAS selfish mutations in a subset of cases, candidate gene screening of 29 SpTs identified an oncogenic NRAS mutation in two cases. To gain insights in the etiology of SpT and into properties of the male germline, we performed whole-genome sequencing of five tumors (4/5 with matched normal tissue). The acquired single nucleotide variant load was extremely low (~0.2 per Mb), with an average of 6 (2-9) non-synonymous variants per tumor, none of which is likely to be oncogenic. The observed mutational signature of SpTs is strikingly similar to that of germline de novo mutations, mostly involving C>T transitions with a significant enrichment in the ACG trinucleotide context. The tumors exhibited extensive aneuploidy (50-99 autosomes/tumor) involving whole-chromosomes, with recurrent gains of chr9 and chr20 and loss of chr7, suggesting that aneuploidy itself represents the initiating oncogenic event. We propose that SpT etiology recapitulates the unique properties of male germ cells; because of evolutionary constraints to maintain low point mutation rate, rare tumorigenic driver events are caused by a combination of gene imbalance mediated via whole-chromosome aneuploidy. Finally, we propose a general framework of male germ cell tumor pathology that accounts for their mutational landscape, timing and cellular origin.
Collapse
Affiliation(s)
- Eleni Giannoulatou
- Clinical Genetics Group, MRC-Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Geoffrey J. Maher
- Clinical Genetics Group, MRC-Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Zhihao Ding
- Clinical Genetics Group, MRC-Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ad J. M. Gillis
- Department of Pathology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lambert C. J. Dorssers
- Department of Pathology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | | | - Gilean McVean
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Andrew O. M. Wilkie
- Clinical Genetics Group, MRC-Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leendert H. J. Looijenga
- Department of Pathology, Erasmus MC—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anne Goriely
- Clinical Genetics Group, MRC-Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Rosario R, Childs AJ, Anderson RA. RNA-binding proteins in human oogenesis: Balancing differentiation and self-renewal in the female fetal germline. Stem Cell Res 2017; 21:193-201. [PMID: 28434825 PMCID: PMC5446320 DOI: 10.1016/j.scr.2017.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/29/2017] [Accepted: 04/13/2017] [Indexed: 12/11/2022] Open
Abstract
Primordial germ cells undergo three significant processes on their path to becoming primary oocytes: the initiation of meiosis, the formation and breakdown of germ cell nests, and the assembly of single oocytes into primordial follicles. However at the onset of meiosis, the germ cell becomes transcriptionally silenced. Consequently translational control of pre-stored mRNAs plays a central role in coordinating gene expression throughout the remainder of oogenesis; RNA binding proteins are key to this regulation. In this review we examine the role of exemplars of such proteins, namely LIN28, DAZL, BOLL and FMRP, and highlight how their roles during germ cell development are critical to oogenesis and the establishment of the primordial follicle pool. RNA-binding proteins (RBPs) are key regulators of gene expression during oogenesis. RBPs LIN28, DAZL, BOLL and FMRP display stage-specific expression in fetal oocytes. LIN28 and DAZL may regulate self-renewal and progression into meiosis respectively. BOLL and FMRP may be involved in the later stages of prophase I and oocyte growth. RBPs may have critical roles in establishing the ovarian reserve during fetal life.
Collapse
Affiliation(s)
- Roseanne Rosario
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew J Childs
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
24
|
Abstract
Fetal germ cell development is tightly regulated by the somatic cell environment, and is characterised by cell cycle states that differ between XY and XX gonads. In the testis, gonocytes enter G1/G0 arrest from 12.5 days post coitum (dpc) in mice and maintain cell cycle arrest until after birth. Failure to correctly maintain G1/G0 arrest can result in loss of germ cells or, conversely, germ cell tumours. High mobility group box containing transcription factor 1 (HBP1) is a transcription factor that was previously identified in fetal male germ cells at the time of embryonic cell cycle arrest. In somatic cells, HBP1 is classified as a tumour suppressor protein, known to regulate proliferation and senescence. We therefore investigated the possible role of HBP1 in the initiation and maintenance of fetal germ cell G1/G0 arrest using the mouse model. We identified two splice variants of Hbp1, both of which are expressed in XY and XX fetal gonads, but only one of which is localised to the nucleus in in vitro assays. To investigate Hbp1 loss of function, we used embryonic stem (ES) cells carrying a Genetrap mutation for Hbp1 to generate mice lacking Hbp1 function. We found that Hbp1-genetrap mouse mutant germ cells proliferated correctly throughout development, and adult males were viable and fertile. Multiple Hbp1-LacZ reporter mouse lines were generated, unexpectedly revealing Hbp1 embryonic expression in hair follicles, eye and limbs. Lastly, in a model of defective germ cell G1/G0 arrest, the Rb1-knockout model, we found no evidence for Hbp1 mis-regulation, suggesting that the reported RB1-HBP1 interaction is not critical in the germline, despite co-expression.
Collapse
|
25
|
Taylor-Weiner A, Zack T, O'Donnell E, Guerriero JL, Bernard B, Reddy A, Han GC, AlDubayan S, Amin-Mansour A, Schumacher SE, Litchfield K, Turnbull C, Gabriel S, Beroukhim R, Getz G, Carter SL, Hirsch MS, Letai A, Sweeney C, Van Allen EM. Genomic evolution and chemoresistance in germ-cell tumours. Nature 2016; 540:114-118. [PMID: 27905446 PMCID: PMC5553306 DOI: 10.1038/nature20596] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/02/2016] [Indexed: 01/04/2023]
Abstract
Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.
Collapse
Affiliation(s)
- Amaro Taylor-Weiner
- Division of Medical Sciences, Harvard University, Boston, Massachusetts 02115, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Travis Zack
- Division of Medical Sciences, Harvard University, Boston, Massachusetts 02115, USA
- Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elizabeth O'Donnell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Jennifer L Guerriero
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Brandon Bernard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Anita Reddy
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - G Celine Han
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Saud AlDubayan
- Division of Genetics and Genomics, Department of Medicine, Boston Children's Hospital, Massachusetts 02115, USA
- Department of Medicine, King Saud bin Abdulaziz University for Health Sciences, Saudi Arabia
| | - Ali Amin-Mansour
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Steven E Schumacher
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Kevin Litchfield
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
- William Harvey Research Institute, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK
| | - Clare Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
- William Harvey Research Institute, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK
| | - Stacey Gabriel
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Rameen Beroukhim
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Gad Getz
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Scott L Carter
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 , USA
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Eliezer M Van Allen
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
26
|
Savage P. Chemotherapy curable malignancies and cancer stem cells: a biological review and hypothesis. BMC Cancer 2016; 16:906. [PMID: 27871274 PMCID: PMC5117562 DOI: 10.1186/s12885-016-2956-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/15/2016] [Indexed: 12/14/2022] Open
Abstract
Background Cytotoxic chemotherapy brings routine cures to only a small select group of metastatic malignancies comprising gestational trophoblast tumours, germ cell tumours, acute leukemia, Hodgkin’s disease, high grade lymphomas and some of the rare childhood malignancies. We have previously postulated that the extreme sensitivity to chemotherapy for these malignancies is linked to the on-going high levels of apoptotic sensitivity that is naturally linked with the unique genetic events of nuclear fusion, meiosis, VDJ recombination, somatic hypermutation, and gastrulation that have occurred within the cells of origin of these malignancies. In this review we will examine the cancer stem cell/cancer cell relationship of each of the chemotherapy curable malignancies and how this relationship impacts on the resultant biology and pro-apoptotic sensitivity of the varying cancer cell types. Discussion In contrast to the common epithelial cancers, in each of the chemotherapy curable malignancies there are no conventional hierarchical cancer stem cells. However cells with cancer stem like qualities can arise stochastically from within the general tumour cell population. These stochastic stem cells acquire a degree of resistance to DNA damaging agents but also retain much of the key characteristics of the cancer cells from which they develop. We would argue that the balance between the acquired resistance of the stochastic cancer stem cell and the inherent chemotherapy sensitivity of parent tumour cell determines the overall chemotherapy curability of each diagnosis. Summary The cancer stem cells in the chemotherapy curable malignancies appear to have two key biological differences from those of the more common chemotherapy incurable malignancies. The first difference is that the conventional hierarchical pattern of cancer stem cells is absent in each of the chemotherapy curable malignancies. The other key difference, we suggest, is that the stochastic stem cells in the chemotherapy curable malignancies take on a significant aspect of the biological characteristics of their parent cancer cells. This action includes for the chemotherapy curable malignancies the heightened pro-apoptotic sensitivity linked to their respective associated unique genetic events. For the chemotherapy curable malignancies the combination of the relationship of their cancer stem cells combined with the extreme inherent sensitivity to induction of apoptosis from DNA damaging agents plays a key role in determining their overall curability with chemotherapy.
Collapse
|
27
|
Abstract
Testicular germ cell tumours are at the crossroads of developmental and neoplastic processes. Their cause has not been fully elucidated but differences in incidences suggest that a combination of genetic and environment factors are involved, with environmental factors predominating early in life. Substantial progress has been made in understanding genetic susceptibility in the past 5 years on the basis of the results of large genome-wide association studies. Testicular germ cell tumours are highly sensitive to radiotherapy and chemotherapy and hence have among the best outcomes of all tumours. Because the tumours occur mainly in young men, preservation of reproductive function, quality of life after treatment, and late effects are crucial concerns. In this Seminar, we provide an overview of advances in the understanding of the epidemiology, genetics, and biology of testicular germ cell tumours. We also summarise the consensus on how to treat testicular germ cell tumours and focus on a few controversies and improvements in the understanding of late effects of treatment and quality of life for survivors.
Collapse
Affiliation(s)
- Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark; International Center for Research and Research Training in Endocrine Disrupting Effects on Male Reproduction and Child Health, Copenhagen, Denmark
| | - Katherine A McGlynn
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Keisei Okamoto
- Department of Urology, Shiga University of Medical Science, Tsukinowa, Seta, Shiga, Japan.
| | - Michael A S Jewett
- Departments of Surgery (Urology) and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada
| | - Carsten Bokemeyer
- Department of Oncology, Haematology, Bone Marrow Transplantation with section Pneumology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
28
|
Lanza DG, Dawson EP, Rao P, Heaney JD. Misexpression of cyclin D1 in embryonic germ cells promotes testicular teratoma initiation. Cell Cycle 2016; 15:919-30. [PMID: 26901436 DOI: 10.1080/15384101.2016.1149272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Testicular teratomas result from anomalies in embryonic germ cell development. In the 129 family of inbred mouse strains, teratomas arise during the same developmental period that male germ cells normally enter G1/G0 mitotic arrest and female germ cells initiate meiosis (the mitotic:meiotic switch). Dysregulation of this switch associates with teratoma susceptibility and involves three germ cell developmental abnormalities seemingly critical for tumor initiation: delayed G1/G0 mitotic arrest, retention of pluripotency, and misexpression of genes normally restricted to embryonic female and adult male germ cells. One misexpressed gene, cyclin D1 (Ccnd1), is a known regulator of cell cycle progression and an oncogene in many tissues. Here, we investigated whether Ccnd1 misexpression in embryonic germ cells is a determinant of teratoma susceptibility in mice. We found that CCND1 localizes to teratoma-susceptible germ cells that fail to enter G1/G0 arrest during the mitotic:meiotic switch and is the only D-type cyclin misexpressed during this critical developmental time frame. We discovered that Ccnd1 deficiency in teratoma-susceptible mice significantly reduced teratoma incidence and suppressed the germ cell proliferation and pluripotency abnormalities associated with tumor initiation. Importantly, Ccnd1 expression was dispensable for somatic cell development and male germ cell specification and maturation in tumor-susceptible mice, implying that the mechanisms by which Ccnd1 deficiency reduced teratoma incidence were germ cell autonomous and specific to tumorigenesis. We conclude that misexpression of Ccnd1 in male germ cells is a key component of a larger pro-proliferative program that disrupts the mitotic:meiotic switch and predisposes 129 inbred mice to testicular teratocarcinogenesis.
Collapse
Affiliation(s)
- Denise G Lanza
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA
| | - Emily P Dawson
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA
| | - Priya Rao
- b Department of Pathology , MD Anderson Cancer Center, The University of Texas , Houston , TX , USA
| | - Jason D Heaney
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,c Dan L Duncan Cancer Center, Baylor College of Medicine , Houston , TX , USA.,d Center For Reproductive Medicine, Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
29
|
Manku G, Hueso A, Brimo F, Chan P, Gonzalez-Peramato P, Jabado N, Gayden T, Bourgey M, Riazalhosseini Y, Culty M. Changes in the expression profiles of claudins during gonocyte differentiation and in seminomas. Andrology 2015; 4:95-110. [PMID: 26588606 DOI: 10.1111/andr.12122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 12/15/2022]
Abstract
Testicular germ cell tumors (TGCTs) are the most common type of cancer in young men and their incidence has been steadily increasing for the past decades. TGCTs and their precursor carcinoma in situ (CIS) are thought to arise from the deficient differentiation of gonocytes, precursors of spermatogonial stem cells. However, the mechanisms relating failed gonocyte differentiation to CIS formation remain unknown. The goal of this study was to uncover genes regulated during gonocyte development that would show abnormal patterns of expression in testicular tumors, as prospective links between failed gonocyte development and TGCT. To identify common gene and protein signatures between gonocytes and seminomas, we first performed gene expression analyses of transitional rat gonocytes, spermatogonia, human normal testicular, and TGCT specimens. Gene expression arrays, pathway analysis, and quantitative real-time PCR analysis identified cell adhesion molecules as a functional gene category including genes downregulated during gonocyte differentiation and highly expressed in seminomas. In particular, the mRNA and protein expressions of claudins 6 and 7 were found to decrease during gonocyte transition to spermatogonia, and to be abnormally elevated in seminomas. The dynamic changes in these genes suggest that they may play important physiological roles during gonocyte development. Moreover, our findings support the idea that TGCTs arise from a disruption of gonocyte differentiation, and position claudins as interesting genes to further study in relation to testicular cancer.
Collapse
Affiliation(s)
- G Manku
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada.,Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - A Hueso
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - F Brimo
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada.,Pathology, McGill University, Montreal, QC, Canada
| | - P Chan
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada.,Surgery, McGill University, Montreal, QC, Canada
| | - P Gonzalez-Peramato
- Department of Pathology, La Paz University Hospital, Universidad Autonoma de Madrid, Madrid, Spain
| | - N Jabado
- Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - T Gayden
- Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - M Bourgey
- Department of Genome Quebec Innovation Centre, McGill University, Montreal, QC, Canada
| | - Y Riazalhosseini
- Department of Genome Quebec Innovation Centre, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - M Culty
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada.,Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Molecular biology of testicular germ cell tumors. Clin Transl Oncol 2015; 18:550-6. [PMID: 26482724 DOI: 10.1007/s12094-015-1423-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/29/2015] [Indexed: 10/22/2022]
Abstract
Testicular germ cell tumors (TGCTs) are the most common solid tumors in young adult men. They constitute a unique pathology because of their embryonic and germ origin and their special behavior. Genetic predisposition, environmental factors involved in their development and genetic aberrations have been under study in many works throughout the last years trying to explain the susceptibility and the transformation mechanism of TGCTs. Despite the high rate of cure in this type of tumors because its particular sensitivity to cisplatin, there are tumors resistant to chemotherapy for which it is needed to find new therapies. In the present work, it has been carried out a literature review on the most important molecular aspects involved in the onset and development of such tumors, as well as a review of the major developments regarding prognostic factors, new prognostic biomarkers and the possibility of new targeted therapies.
Collapse
|
31
|
Jørgensen A, Lindhardt Johansen M, Juul A, Skakkebaek NE, Main KM, Rajpert-De Meyts E. Pathogenesis of germ cell neoplasia in testicular dysgenesis and disorders of sex development. Semin Cell Dev Biol 2015; 45:124-37. [PMID: 26410164 DOI: 10.1016/j.semcdb.2015.09.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/21/2015] [Indexed: 12/29/2022]
Abstract
Development of human gonads is a sex-dimorphic process which evolved to produce sex-specific types of germ cells. The process of gonadal sex differentiation is directed by the action of the somatic cells and ultimately results in germ cells differentiating to become functional gametes through spermatogenesis or oogenesis. This tightly controlled process depends on the proper sequential expression of many genes and signalling pathways. Disturbances of this process can be manifested as a large spectrum of disorders, ranging from severe disorders of sex development (DSD) to - in the genetic male - mild reproductive problems within the testicular dysgenesis syndrome (TDS), with large overlap between the syndromes. These disorders carry an increased but variable risk of germ cell neoplasia. In this review, we discuss the pathogenesis of germ cell neoplasia associated with gonadal dysgenesis, especially in individuals with 46,XY DSD. We summarise knowledge concerning development and sex differentiation of human gonads, with focus on sex-dimorphic steps of germ cell maturation, including meiosis. We also briefly outline the histopathology of germ cell neoplasia in situ (GCNIS) and gonadoblastoma (GDB), which are essentially the same precursor lesion but with different morphological structure dependent upon the masculinisation of the somatic niche. To assess the risk of germ cell neoplasia in different types of DSD, we have performed a PubMed search and provide here a synthesis of the evidence from studies published since 2006. We present a model for pathogenesis of GCNIS/GDB in TDS/DSD, with the risk of malignancy determined by the presence of the testis-inducing Y chromosome and the degree of masculinisation. The associations between phenotype and the risk of neoplasia are likely further modulated in each individual by the constellation of the gene polymorphisms and environmental factors.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Marie Lindhardt Johansen
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Anders Juul
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Niels E Skakkebaek
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Katharina M Main
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
32
|
Jørgensen A, Nielsen JE, Perlman S, Lundvall L, Mitchell RT, Juul A, Rajpert-De Meyts E. Ex vivo culture of human fetal gonads: manipulation of meiosis signalling by retinoic acid treatment disrupts testis development. Hum Reprod 2015; 30:2351-63. [PMID: 26251460 DOI: 10.1093/humrep/dev194] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/13/2015] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION What are the effects of experimentally manipulating meiosis signalling by addition of retinoic acid (RA) in cultured human fetal gonads? SUMMARY ANSWER RA-treatment accelerated meiotic entry in cultured fetal ovary samples, while addition of RA resulted in a dysgenetic gonadal phenotype in fetal testis cultures. WHAT IS KNOWN ALREADY One of the first manifestations of sex differentiation is the initiation of meiosis in fetal ovaries. In contrast, meiotic entry is actively prevented in the fetal testis at this developmental time-point. It has previously been shown that RA-treatment mediates initiation of meiosis in human fetal ovary ex vivo. STUDY DESIGN, SIZE, DURATION This was a controlled ex vivo study of human fetal gonads treated with RA in 'hanging-drop' tissue cultures. The applied experimental set-up preserves germ cell-somatic niche interactions and the investigated outcomes included tissue integrity and morphology, cell proliferation and survival and the expression of markers of meiosis and sex differentiation. PARTICIPANTS/MATERIALS, SETTING, METHODS Tissue from 24 first trimester human fetuses was included in this study, all from elective terminations at gestational week (GW) 7-12. Gonads were cultured for 2 weeks with and without addition of 1 µM RA. Samples were subsequently formalin-fixed and investigated by immunohistochemistry and cell counting. Proteins investigated and quantified included; octamer-binding transcription factor 4 (OCT4), transcription factor AP-2 gamma (AP2γ) (embryonic germ cell markers), SRY (sex determining region Y)-box 9 (SOX9), anti-Müllerian hormone (AMH) (immature Sertoli cell markers), COUP transcription factor 2 (COUP-TFII) (marker of interstitial cells), forkhead box L2 (FOXL2) (granulosa cell marker), H2A histone family, member X (γH2AX) (meiosis marker), doublesex and mab-3 related transcription factor 1 (DMRT1) (meiosis regulator), cleaved poly ADP ribose polymerase (PARP), cleaved Caspase 3 (apoptosis markers) and Ki-67 antigen (Ki-67) (proliferation marker). Also, proliferation was determined using a 5'-bromo-2'-deoxyuridine (BrdU) incorporation assay. MAIN RESULTS AND THE ROLE OF CHANCE A novel ex vivo 'hanging-drop' culture model for human fetal gonads was successfully established. Continued proliferation of cells without signs of increased apoptosis was observed after 2 weeks of culture. In cultured fetal ovaries treated with RA, an increased number of meiotic germ cells (P < 0.05) and DMRT1-positive oogonia initiating meiosis (P < 0.05) was observed, which is in agreement with a previous study. In fetal testes, RA-treatment resulted in a decreased number of gonocytes (P < 0.05), a reduced percentage of proliferating gonocytes (P < 0.05), altered expression pattern of the somatic cell markers AMH and COUP-TFII, as well as disrupted seminiferous cord structure and testis morphology. LIMITATIONS, REASONS FOR CAUTION The number of samples included in this study was relatively small due to the limited availability of human fetal tissue. WIDER IMPLICATIONS OF THE FINDINGS The hanging-drop culture, similarly to other organ culture approaches, allows studies of germ cell-somatic niche interactions and determination of effects after manipulating specific signalling pathways. Our novel finding of disrupted fetal testis development after treatment with RA indicates that abnormal meiosis regulation can potentially cause gonadal dysgenesis. Further studies will elucidate the exact mechanisms and timing of observed effects. STUDY FUNDING/COMPETING INTERESTS This work was supported in part by an ESPE Research Fellowship, sponsored by Novo Nordisk A/S to A.Jø. Additional funding for this project was obtained from The Research Council of the Capital Region of Denmark (E.R.-D.M.), The Research Fund at Rigshospitalet (A.Ju. and J.E.N.), Familien Erichssens Fund (A.Jø.), Dagmar Marshalls Fund (A.Jø.) and Aase & Ejnar Danielsens Fund (A.Jø.). The authors have no conflicts of interest.
Collapse
Affiliation(s)
- A Jørgensen
- University Department of Growth and Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - J E Nielsen
- University Department of Growth and Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - S Perlman
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - L Lundvall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | - R T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh EH9 1LF, UK
| | - A Juul
- University Department of Growth and Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - E Rajpert-De Meyts
- University Department of Growth and Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Lawaetz AC, Almstrup K. Involvement of epigenetic modifiers in the pathogenesis of testicular dysgenesis and germ cell cancer. Biomol Concepts 2015; 6:219-27. [DOI: 10.1515/bmc-2015-0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/10/2015] [Indexed: 12/27/2022] Open
Abstract
AbstractTesticular germ cell cancer manifests mainly in young adults as a seminoma or non-seminoma. The solid tumors are preceded by the presence of a non-invasive precursor cell, the carcinoma in situ cell (CIS), which shows great similarity to fetal germ cells. It is therefore hypothesized that the CIS cell is a fetal germ cell that has been arrested during development due to testicular dysgenesis. CIS cells retain a fetal and open chromatin structure, and recently several epigenetic modifiers have been suggested to be involved in testicular dysgenesis in mice. We here review the possible involvement of epigenetic modifiers with a focus on jumonji C enzymes in the development of testicular dysgenesis and germ cell cancer in men.
Collapse
Affiliation(s)
- Andreas C. Lawaetz
- 1University Department of Growth and Reproduction, Section GR-5064, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Kristian Almstrup
- 1University Department of Growth and Reproduction, Section GR-5064, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
34
|
Grelet S, Andries V, Polette M, Gilles C, Staes K, Martin AP, Kileztky C, Terryn C, Dalstein V, Cheng CW, Shen CY, Birembaut P, Van Roy F, Nawrocki-Raby B. The human NANOS3 gene contributes to lung tumour invasion by inducing epithelial-mesenchymal transition. J Pathol 2015; 237:25-37. [PMID: 25904364 DOI: 10.1002/path.4549] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/30/2015] [Accepted: 04/17/2015] [Indexed: 01/01/2023]
Abstract
We have explored the role of the human NANOS3 gene in lung tumour progression. We show that NANOS3 is over-expressed by invasive lung cancer cells and is a prognostic marker for non-small cell lung carcinomas (NSCLCs). NANOS3 gene expression is restricted in testis and brain and is regulated by epigenetic events. It is up-regulated in cultured cells undergoing epithelial - mesenchymal transition (EMT). NANOS3 over-expression in human NSCLC cell lines enhances their invasiveness by up-regulating EMT, whereas its silencing induces mesenchymal - epithelial transition. NANOS3 represses E-cadherin at the transcriptional level and up-regulates vimentin post-transcriptionally. Also, we show that NANOS3 binds mRNAs encoding vimentin and regulates the length of their poly(A) tail. Finally, NANOS3 can also protect vimentin mRNA from microRNA-mediated repression. We thus demonstrate a role for NANOS3 in the acquisition of invasiveness by human lung tumour cells and propose a new mechanism of post-transcriptional regulation of EMT.
Collapse
Affiliation(s)
- Simon Grelet
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France
| | - Vanessa Andries
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | - Myriam Polette
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Christine Gilles
- Laboratory of Developmental and Tumour Biology, GIGA-Cancer, University of Liège, Belgium
| | - Katrien Staes
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | | | - Claire Kileztky
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France
| | - Christine Terryn
- Plateforme Imagerie Cellulaire et Tissulaire, University of Reims Champagne-Ardenne, France
| | - Véronique Dalstein
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Chun-Wen Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Philippe Birembaut
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Frans Van Roy
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | | |
Collapse
|
35
|
Abstract
Spermatocytic seminomas affect 0.3-0.8 per one million men. This tumor is not, as the name might suggest, a variant of classical seminoma but a distinct nosological entity, which differs markedly from all other germ cell tumors in its epidemiology, peculiar morphology, oncogenesis and clinical outcome. There are no racial differences in the incidence and risk factors are completely unknown. Patients are significantly older than is the case for other germ cell tumors with an average of 53.5 years; nevertheless, more than 25 % are younger than 40 years. Spermatocytic seminoma arises from differentiated spermatogonia, not from intratubular germ cell neoplasms. The tumor-specific gain of chromosome 9 seems to be the most important event in the oncogenesis. Conventional spermatocytic seminoma consists of three different cell types which give the tumor a highly aggressive appearance, although in actual fact, the tumor has a very favorable outcome, with few reported cases of general metastatic spread. Anaplastic spermatocytic seminoma, a recently described variant, also takes mostly a benign course; however, spermatocytic seminomas combined with sarcomas are extremely malignant.
Collapse
Affiliation(s)
- G Mikuz
- Institut für Pathologie, Medizinische Universität Innsbruck, Müllerstr. 44, 6020, Innsbruck, Österreich,
| |
Collapse
|
36
|
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
|
37
|
Intratubular germ cell neoplasia of the human testis: heterogeneous protein expression and relation to invasive potential. Mod Pathol 2014; 27:1255-1266. [PMID: 24457464 PMCID: PMC4012991 DOI: 10.1038/modpathol.2013.246] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/16/2013] [Accepted: 11/26/2013] [Indexed: 01/11/2023]
Abstract
Testicular germ cell cancer develops from premalignant intratubular germ cell neoplasia, unclassified cells that are believed to arise from failure of normal maturation of fetal germ cells from gonocytes (OCT4(+)/MAGEA4(-)) into pre-spermatogonia (OCT4(-)/MAGEA4(+)). Intratubular germ cell neoplasia cell subpopulations based on stage of germ cell differentiation have been described, however the importance of these subpopulations in terms of invasive potential has not been reported. We hypothesized that cells expressing an immature (OCT4(+)/MAGEA4(-)) germ cell profile would exhibit an increased proliferation rate compared with those with a mature profile (OCT4(+)/MAGEA4(+)). Therefore, we performed triple immunofluorescence and stereology to quantify the different intratubular germ cell neoplasia cell subpopulations, based on expression of germ cell (OCT4, PLAP, AP2γ, MAGEA4, VASA) and proliferation (Ki67) markers, in testis sections from patients with preinvasive disease, seminoma, and non-seminoma. We compared these subpopulations with normal human fetal testis and with seminoma cells. Heterogeneity of protein expression was demonstrated in intratubular germ cell neoplasia cells with respect to gonocyte and spermatogonial markers. It included an embryonic/fetal germ cell subpopulation lacking expression of the definitive intratubular germ cell neoplasia marker OCT4, that did not correspond to a physiological (fetal) germ cell subpopulation. OCT4(+)/MAGEA4(-) cells showed a significantly increased rate of proliferation compared with the OCT4(+)/MAGEA4(+) population (12.8 versus 3.4%, P<0.0001) irrespective of histological tumor type, reflected in the predominance of OCT4(+)/MAGEA4(-) cells in the invasive tumor component. Surprisingly, OCT4(+)/MAGEA4(-) cells in patients with preinvasive disease showed significantly higher proliferation compared to those with seminoma or non-seminoma (18.1 versus 10.2 versus 7.2%, P<0.05, respectively). In conclusion, this study has demonstrated that OCT4(+)/MAGEA4(-) cells are the most frequent and most proliferative cell population in tubules containing intratubular germ cell neoplasia, which appears to be an important factor in determining invasive potential of intratubular germ cell neoplasia to seminomas.
Collapse
|
38
|
Yu M, Mu H, Niu Z, Chu Z, Zhu H, Hua J. miR-34c enhances mouse spermatogonial stem cells differentiation by targeting Nanos2. J Cell Biochem 2014; 115:232-42. [PMID: 24038201 DOI: 10.1002/jcb.24655] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 08/14/2013] [Indexed: 12/19/2022]
Abstract
miRNAs are expressed in many mammalian cells, acting specific roles in regulating gene expression or mediating special mRNAs cleavage by targeting their 3'-untranslated region (3'UTR). Some miRNAs are essential and important for animal development. However, it is still unclear what the relationship is between miR-34c and mammalian spermatogonial stem cells (SSCs). We found that a conserved microRNA-34c through its target-Nanos2, regulating SSCs' differentiation in mouse. Immunohistochemistry analysis of Nanos2 and miR-34c FISH results revealed the opposite expression trends between them. Seven bioinformatics websites and programs predicted that miR-34c has interaction sites in Nanos2's 3'UTR. Dual-luciferase reporter vector and mutated dual-luciferase reporter vector analysis validated that they are interacted. After transfection miR-34c mimics into mouse SSCs, or miR-34c lentiviral vector in vitro co-cultivation with seminiferous tubules, and Western blot analysis demonstrated that miR-34c over-expression could suppress Nanos2 expression in post-transcription level. Our experiments identified that miR-34c may promote meiosis process by interacting with Nanos2 leading up-regulation of Stra8 in mouse spermatogonial stem cells.
Collapse
Affiliation(s)
- Meng Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | | | | | | | | | | |
Collapse
|
39
|
Bartkova J, Hoei-Hansen CE, Krizova K, Hamerlik P, Skakkebæk NE, Rajpert-De Meyts E, Bartek J. Patterns of DNA damage response in intracranial germ cell tumors versus glioblastomas reflect cell of origin rather than brain environment: implications for the anti-tumor barrier concept and treatment. Mol Oncol 2014; 8:1667-78. [PMID: 25066726 DOI: 10.1016/j.molonc.2014.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 06/24/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022] Open
Abstract
The DNA damage response (DDR) machinery becomes commonly activated in response to oncogenes and during early stages of development of solid malignancies, with an exception of testicular germ cell tumors (TGCTs). The active DDR signaling evokes cell death or senescence but this anti-tumor barrier can be breached by defects in DDR factors, such as the ATM-Chk2-p53 pathway, thereby allowing tumor progression. The DDR barrier is strongly activated in brain tumors, particularly gliomas, due to oxidative damage and replication stress. Here, we took advantage of rare human primary intracranial germ cell tumors (PIGCTs), to address the roles of cell-intrinsic factors including cell of origin, versus local tissue environment, in the constitutive DDR activation in vivo. Immunohistochemical analysis of 7 biomarkers on a series of 21 PIGCTs (germinomas and other subtypes), 20 normal brain specimens and 20 glioblastomas, revealed the following: i) The overall DDR signaling (γH2AX) and activation of the ATM-Chk2-p53 pathway were very low among the PIGCTs, reminiscent of TGCTs, and contrasting sharply with strong DDR activation in glioblastomas; ii) Except for one case of embryonal carcinoma, there were no clear aberrations in the ATM-Chk2-p53 pathway components among the PIGCT cohort; iii) Subsets of PIGCTs showed unusual cytosolic localization of Chk2 and/or ATM. Collectively, these results show that PIGCTs mimic the DDR activation patterns of their gonadal germ cell tumor counterparts, rather than the brain tumors with which they share the tissue environment. Hence cell-intrinsic factors and cell of origin dictate the extent of DDR barrier activation and also the ensuing pressure to select for DDR defects. Our data provide conceptually important insights into the role of DNA damage checkpoints in intracranial tumorigenesis, with implications for the differential biological responses of diverse tumor types to endogenous stress as well as to genotoxic treatments such as ionizing radiation or chemotherapy.
Collapse
Affiliation(s)
| | - Christina E Hoei-Hansen
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics, Rigshospitalet, Copenhagen, Denmark
| | - Katerina Krizova
- Danish Cancer Society Research Center, Copenhagen, Denmark; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petra Hamerlik
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Niels E Skakkebæk
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark.
| | - Jiri Bartek
- Danish Cancer Society Research Center, Copenhagen, Denmark; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
40
|
Schroeder JA, Siegmund HI, Roesch W, Hadziselimovic F, Hofstaedter F. Male infertility: assessment of juvenile testicular dysfunction and risk for malignancy in cryptorchid boys based on resin section evaluation. Ultrastruct Pathol 2014; 37:373-7. [PMID: 24047353 DOI: 10.3109/01913123.2013.818603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Infertility is sometimes more a man's problem than a woman's, failure of one or both of the testes to descend (cryptorchidism) being the most frequent genital malformation in boys. Untreated, the undescended testis impairs germ cell development and significantly reduces adult fertility. A-dark spermatogonia are the stem cells for all future spermatozoa, and their depletion can be reliably estimated in resin semithin sections. Additionally, there is an increased risk of testicular preneoplasia in the form of carcinoma in situ (CIS) cells. The authors report how the pathologic biopsy examination of juvenile cryptorchid testes can assess infertility and malignancy risk.
Collapse
|
41
|
van den Driesche S, Sharpe RM, Saunders PT, Mitchell RT. Regulation of the germ stem cell niche as the foundation for adult spermatogenesis: A role for miRNAs? Semin Cell Dev Biol 2014; 29:76-83. [DOI: 10.1016/j.semcdb.2014.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 12/19/2022]
|
42
|
Jørgensen A, Young J, Nielsen JE, Joensen UN, Toft BG, Rajpert-De Meyts E, Loveland KL. Hanging drop cultures of human testis and testis cancer samples: a model used to investigate activin treatment effects in a preserved niche. Br J Cancer 2014; 110:2604-14. [PMID: 24781282 PMCID: PMC4021512 DOI: 10.1038/bjc.2014.160] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/10/2014] [Accepted: 03/04/2014] [Indexed: 12/15/2022] Open
Abstract
Background: Testicular germ cell tumours of young adults, seminoma or non-seminomas, are preceded by a pre-invasive precursor, carcinoma in situ (CIS), understood to arise through differentiation arrest of embryonic germ cells. Knowledge about the malignant transformation of germ cells is currently limited by the lack of experimental models. The aim of this study was to establish an experimental tissue culture model to maintain normal and malignant germ cells within their niche and allow investigation of treatment effects. Methods: Human testis and testis cancer specimens from orchidectomies were cultured in ‘hanging drops' and effects of activin A and follistatin treatment were investigated in seminoma cultures. Results: Testis fragments with normal spermatogenesis or CIS cells were cultured for 14 days with sustained proliferation of germ cells and CIS cells and without increased apoptosis. Seminoma cultures survived 7 days, with proliferating cells detectable during the first 5 days. Activin A treatment significantly reduced KIT transcript and protein levels in seminoma cultures, thereby demonstrating a specific treatment response. Conclusions: Hanging drop cultures of human testis and testis cancer samples can be employed to delineate mechanisms governing growth of normal, CIS and tumorigenic germ cells retained within their niche.
Collapse
Affiliation(s)
- A Jørgensen
- University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - J Young
- 1] Department of Anatomy and Developmental Biology, Monash University, Melbourne, Clayton, Victoria 3800, Australia [2] Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Clayton, Victoria 3800, Australia
| | - J E Nielsen
- University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - U N Joensen
- Department of Urology, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - B G Toft
- Department of Pathology, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - E Rajpert-De Meyts
- University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - K L Loveland
- 1] Department of Anatomy and Developmental Biology, Monash University, Melbourne, Clayton, Victoria 3800, Australia [2] Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Clayton, Victoria 3800, Australia
| |
Collapse
|
43
|
The chemosensitivity of testicular germ cell tumors. Cell Oncol (Dordr) 2014; 37:79-94. [PMID: 24692098 DOI: 10.1007/s13402-014-0168-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2014] [Indexed: 12/13/2022] Open
Abstract
Although rare cancers overall, testicular germ cell tumors (TGCTs) are the most common type of cancer in young males below 40 years of age. Both subtypes of TGCTs, i.e., seminomas and non-seminomas, are highly curable and the majority of even metastatic patients may expect to be cured. These high cure rates are not due to the indolent nature of these cancers, but rather to their sensitivity to chemotherapy (and for seminomas to radiotherapy). The delineation of the cause of chemosensitivity at the molecular level is of paramount importance, because it may provide insights into the minority of TGCTs that are chemo-resistant and, thereby, provide opportunities for specific therapeutic interventions aimed at reverting them to chemosensitivity. In addition, delineation of the molecular basis of TGCT chemo-sensitivity may be informative for the cause of chemo-resistance of other more common types of cancer and, thus, may create new therapeutic leads. p53, a frequently mutated tumor suppressor in cancers in general, is not mutated in TGCTs, a fact that has implications for their chemo-sensitivity. Oct4, an embryonic transcription factor, is uniformly expressed in the seminoma and embryonic carcinoma components of non-seminomas, and its interplay with p53 may be important in the chemotherapy response of these tumors. This interplay, together with other features of TGCTs such as the gain of genetic material from the short arm of chromosome 12 and the association with disorders of testicular development, will be discussed in this paper and integrated in a unifying hypothesis that may explain their chemo-sensitivity.
Collapse
|
44
|
Boublikova L, Buchler T, Stary J, Abrahamova J, Trka J. Molecular biology of testicular germ cell tumors: Unique features awaiting clinical application. Crit Rev Oncol Hematol 2014; 89:366-85. [DOI: 10.1016/j.critrevonc.2013.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/30/2013] [Accepted: 10/01/2013] [Indexed: 01/29/2023] Open
|
45
|
Cost NG, Lubahn JD, Adibi M, Romman A, Wickiser JE, Raj GV, Sagalowsky AI, Margulis V. A comparison of pediatric, adolescent, and adult testicular germ cell malignancy. Pediatr Blood Cancer 2014; 61:446-51. [PMID: 24106160 DOI: 10.1002/pbc.24773] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/21/2013] [Indexed: 01/24/2023]
Abstract
BACKGROUND Testicular germ cell tumors (T-GCTs) occur from infancy to adulthood, and are the most common solid tumor in adolescent and young adult males. Traditionally, pediatric T-GCTs were perceived as more indolent than adult T-GCTs. However, there are few studies comparing these groups and none that specifically evaluate adolescents. METHODS An institutional database of T-GCT patients was reviewed and patients were categorized into Pediatric, aged 0-12 years, Adolescent, aged 13-19 years, and Adult, older than 20 years, cohorts. Demographics, tumor characteristics, disease stage, treatment, event-free survival (EFS), and overall survival (OS) were compared between groups. RESULTS Overall, 413 patients (20 pediatric, 39 adolescent, 354 adult) met study criteria and were followed for a median of 2.0 years (0.1-23.6). Adolescents presented with more advanced stage than children (P = 0.018) or adults (P = 0.008). There was a higher rate of events in Adolescents (13, 33.3%) than in Adults (61, 17.2%) or Children (2, 10.0%). Three-year EFS was 87.2% in the Pediatric group, 59.9% in Adolescents and 80.0% in Adults (P = 0.011). In a multivariate analysis, controlling for stage, IGCCCG risk, and histology, the hazard ratio (HR) for an event was: 1 (Reference) for Adults, HR = 0.82 (95% CI 0.19-3.46; P = 0.33) for the Pediatric group, and HR = 2.22 (95% CI 1.21-4.07; P = 0.01) for Adolescents. Five-year OS was 100% in the Pediatric group, 84.8% in Adolescents, and 92.8% in Adults (P = 0.388). CONCLUSION Lower EFS in adolescent T-GCT patients was observed than in either children or adults. Elucidating factors associated with inferior outcomes in adolescents is an important focus of future research.
Collapse
Affiliation(s)
- Nicholas G Cost
- Division of Urologic Oncology, University of Texas Southwestern Medical Center, Dallas, Texas; Division of Urology, University of Colorado School of Medicine, Aurora, Colorado
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Mu H, Wu J, Zhu H, Li N, Tang F, Yao X, Yang C, Peng S, Li G, Hua J. The function of Msx1 gene in promoting meiosis of dairy goat male germline stem cells (mGSCs). Cell Biochem Funct 2013; 31:629-35. [PMID: 24123057 DOI: 10.1002/cbf.3010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/21/2013] [Accepted: 09/23/2013] [Indexed: 02/02/2023]
Abstract
During sequential stages of meiosis, numerous cytoplasmic and nuclear events take place in which many germline and non-germline genes involved. It is demonstrated that the germline gene Stra8 and synaptonemal complex protein 3 (Scp3) play an important role in the meiosis. Recently, studies showed Msx1, a DNA-binding protein taking part in the skeletal development, also having a functional attractive factor to Stra8 and Scp3 in the meiosis. In this study, we cloned the gene Msx1 then transfected the Msx1 constructed recombination plasmid, pMsx1-Ires2-AcGFP, into the dairy goat germline stem cells (male germline stem cells) and analysed the effects of Msx1 on the expression of Stra8 and Scp3. The results showed that Msx1 could enhance the expression of Stra8 and Scp3 and promote the meiosis in goat testicular cells. Bmp4 activated the expression of Msx1 and Stra8. This study suggests that Msx1 plays an important role in spermatogenesis and meiosis.
Collapse
Affiliation(s)
- Hailong Mu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Blomberg Jensen M, Lieben L, Nielsen JE, Willems A, Jørgensen A, Juul A, Toppari J, Carmeliet G, Rajpert-De Meyts E. Characterization of the testicular, epididymal and endocrine phenotypes in the Leuven Vdr-deficient mouse model: targeting estrogen signalling. Mol Cell Endocrinol 2013; 377:93-102. [PMID: 23850520 DOI: 10.1016/j.mce.2013.06.036] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 06/12/2013] [Accepted: 06/30/2013] [Indexed: 12/21/2022]
Abstract
Vitamin D is a key factor for calcium and bone homeostasis, but signalling through the vitamin D receptor (VDR) seems also to be important for testicular function. To test the functional role of vitamin D signalling we examined the male reproductive system of the Leuven Vdr-ablated (Vdr(-/-)) mice, previously established as a model for hereditary vitamin D-resistant rickets. We investigated reproductive hormones, changes in gene expression and histological phenotype of eleven Vdr(-/-), eight Vdr(+/-) and nine Vdr(+/+) mice. Testicular and epididymal histology were grossly normal in Vdr(-/-) mice. Accordingly, no differences were found in serum concentrations of testosterone, estradiol, LH, and FSH or testicular expression of Cyp19a1, Ersα, Cyp17a1, Star, Insl3, Inhbb, and Amh. However, a significantly lower ERβ expression was found in testis of Vdr(+/-) and Vdr(-/-) mice, conversely epididymal expressions of ERα and the estrogen-target gene Aqp9 were higher. In conclusion, vitamin D seems dispensable for murine spermatogenesis and sex hormone production, but aberrant estrogen-signalling may elicit some of the VDR-mediated effects on male reproduction.
Collapse
|
48
|
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
|