1
|
Krasic J, Skara Abramovic L, Himelreich Peric M, Vanjorek V, Gangur M, Zovko D, Malnar M, Masic S, Demirovic A, Juric B, Ulamec M, Coric M, Jezek D, Kulis T, Sincic N. Testicular Germ Cell Tumor Tissue Biomarker Analysis: A Comparison of Human Protein Atlas and Individual Testicular Germ Cell Tumor Component Immunohistochemistry. Cells 2023; 12:1841. [PMID: 37508506 PMCID: PMC10378501 DOI: 10.3390/cells12141841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The accurate management of testicular germ cell tumors (TGCTs) depends on identifying the individual histological tumor components. Currently available data on protein expression in TGCTs are limited. The human protein atlas (HPA) is a comprehensive resource presenting the expression and localization of proteins across tissue types and diseases. In this study, we have compared the data from the HPA with our in-house immunohistochemistry on core TGCT diagnostic genes to test reliability and potential biomarker genes. We have compared the protein expression of 15 genes in TGCT patients and non-neoplastic testicles with the data from the HPA. Protein expression was converted into diagnostic positivity. Our study discovered discrepancies in three of the six core TGCT diagnostic genes, POU5F1, KIT and SOX17 in HPA. DPPA3, CALCA and TDGF1 were presented as potential novel TGCT biomarkers. MGMT was confirmed while RASSF1 and PRSS21 were identified as biomarkers of healthy testicular tissue. Finally, SALL4, SOX17, RASSF1 and PRSS21 dysregulation in the surrounding testicular tissue with complete preserved spermatogenesis of TGCT patients was detected, a potential early sign of neoplastic transformation. We highlight the importance of a multidisciplinary collaborative approach to fully understand the protein landscape of human testis and its pathologies.
Collapse
Affiliation(s)
- Jure Krasic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Lucija Skara Abramovic
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | - Marta Himelreich Peric
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Health Centre Zagreb-West, 10000 Zagreb, Croatia
| | - Vedran Vanjorek
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Gangur
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dragana Zovko
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marina Malnar
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Silvija Masic
- Ljudevit Jurak Clinical Department of Pathology and Cytology, University Clinical Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia
| | - Alma Demirovic
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Ljudevit Jurak Clinical Department of Pathology and Cytology, University Clinical Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Bernardica Juric
- Ljudevit Jurak Clinical Department of Pathology and Cytology, University Clinical Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia
| | - Monika Ulamec
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Ljudevit Jurak Clinical Department of Pathology and Cytology, University Clinical Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marijana Coric
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Pathology and Cytology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Davor Jezek
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tomislav Kulis
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Urology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
- Department of Urology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Nino Sincic
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
2
|
Low SYY, Cheng H, Zou R, Ng LP, Kuick CH, Syed Sulaiman NB, Chang KTE, Low DCY, Zhou L, Seow WT. Molecular exploration of paediatric intracranial germinomas from multi-ethnic Singapore. BMC Neurol 2020; 20:415. [PMID: 33187494 PMCID: PMC7666528 DOI: 10.1186/s12883-020-01981-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Germinomas (IG) account for up to 50% of all intracranial germ cell tumours. These tumours are reputed to be more prevalent in Oriental populations in comparison to Western cohorts. Biological characteristics of IG in other ethnic groups are unknown. Singapore is a multi-ethnic country with diverse cultures. Owing to inter-racial heterogeneity, the authors hypothesize there are molecular differences between paediatric IG patients in our local population. The aims of this study are exploratory: firstly, to identify molecular characteristics in this tumour type and circulating CSF unique to different racial cohorts; and next, to corroborate our findings with published literature. Methods This is a single-institution, retrospective study of prospectively collected data. Inclusion criteria encompass all paediatric patients with histologically confirmed IG. Excess CSF and brain tumour tissues are collected for molecular analysis. Tumour tissues are subjected to a next generation sequencing (NGS) targeted panel for KIT and PDGRA. All CSF samples are profiled via a high-throughput miRNA multiplexed workflow. Results are then corroborated with existing literature and public databases. Results In our cohort of 14 patients, there are KIT exon variants in the tumour tissues and CSF miRNAs corroborative with published studies. Separately, there are also KIT exon variants and miRNAs not previously highlighted in IG. A subgroup analysis demonstrates differential CSF miRNAs between Chinese and Malay IG patients. Conclusion This is the first in-depth molecular study of a mixed ethnic population of paediatric IGs from a Southeast Asian cohort. Validation studies are required to assess the relevance of novel findings in our study. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-020-01981-0.
Collapse
Affiliation(s)
- Sharon Yin Yee Low
- Neurosurgical Service, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore. .,Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore. .,SingHealth Duke-NUS Neuroscience Academic Clinical Program, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore. .,VIVA-KKH Paediatric Brain and Solid Tumours Laboratory, Singapore, Singapore.
| | - He Cheng
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore.,MiRXES Pte Ltd, 10 Biopolis Road, Chromos, Singapore, 138670, Singapore
| | - Ruiyang Zou
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore.,MiRXES Pte Ltd, 10 Biopolis Road, Chromos, Singapore, 138670, Singapore
| | - Lee Ping Ng
- Neurosurgical Service, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
| | - Chik Hong Kuick
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
| | - Nurfarhanah Bte Syed Sulaiman
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore.,VIVA-KKH Paediatric Brain and Solid Tumours Laboratory, Singapore, Singapore
| | - Kenneth Tou En Chang
- VIVA-KKH Paediatric Brain and Solid Tumours Laboratory, Singapore, Singapore.,Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
| | - David Chyi Yeu Low
- Neurosurgical Service, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore.,Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore.,SingHealth Duke-NUS Neuroscience Academic Clinical Program, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore
| | - Lihan Zhou
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore.,MiRXES Pte Ltd, 10 Biopolis Road, Chromos, Singapore, 138670, Singapore
| | - Wan Tew Seow
- Neurosurgical Service, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore.,Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore.,SingHealth Duke-NUS Neuroscience Academic Clinical Program, 11 Jalan Tan Tock Seng, Singapore, 30843, Singapore
| |
Collapse
|
3
|
Guerra F, Quintana S, Giustina S, Mendeluk G, Jufe L, Avagnina MA, Díaz LB, Palaoro LA. Investigation of EGFR/pi3k/Akt signaling pathway in seminomas. Biotech Histochem 2020; 96:125-137. [PMID: 32597316 DOI: 10.1080/10520295.2020.1776393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activation of the receptor for epidermal growth factor (EGFR) in some testicular tumors activates several signaling pathways. Some components of these pathways are phosphorylated or mutated in testicular germ tumors (TCGT), including EGFR, Kirstein ras oncogen (KRAS) and cell surface protein of the germ cell (KIT). The latter two activate RAF ⁄MEK⁄ERK and PI3 K⁄AKT, and interconnect with the EGFR/pI3 k/Akt pathway. We investigated the expression of EGFR/pI3 k/Akt pathway proteins in seminomas and in their precursor lesion, germinal cell neoplasia in situ (GCNIS) and related genetic mutations. We used immunohistochemistry for pEGFR, pI3 k and pAkt expression with a scoring system for 46 seminoma surgical specimens: 36 classical and 10 GCNIS. In 17 samples, the mutations of EGFR (exons 19 - 21), KIT (exons 11, 17) and KRAS (exons 2, 3) were investigated using qPCR and sequencing. Of the 36 seminomas studied, 22 (61%) expressed pEGFR. Ten samples exhibited high scores for pEGFR, pI3 k and pAkt. In 5 of 17 cases (33%) some mutation was exhibited in the exons studied: 21 of EGFR (2), 17 of EGFR (1), 3 of KRAS (1) and 11 of KIT (1). Six cases exhibited nuclear translocation of EGFR; of these, four exhibited mutations of EGFR, KRAS and KIT. Eight of ten of the GCNIS expressed a high pEGFR score (80%). In 2 of 6 cases (33%), mutation was detected in exon 21 of EGFR and one smear showed EGFR translocation to the nucleus. The translocation represents a subpopulation with worse prognosis for TCGT. The EGFR/pI3 k/Akt signaling pathway is linked to TDRG1, which regulates chemosensitivity to cisplatin; this is a mechanism of resistance to treatment. TDRG1 and the EGFR/pI3 k/pAkt pathway could be therapeutic targets for seminomas resistant to cisplatin.
Collapse
Affiliation(s)
- F Guerra
- Department of Clinical Biochemistry, Clinical Hospital (UBA), C.A.B.A., INFIBIOC , Córdoba, Argentina
| | - S Quintana
- Fares Taie Institute , Mar Del Plata, Buenos Aires, Argentina
| | - S Giustina
- Fares Taie Institute , Mar Del Plata, Buenos Aires, Argentina
| | - G Mendeluk
- Department of Clinical Biochemistry, Clinical Hospital (UBA), C.A.B.A., INFIBIOC , Córdoba, Argentina
| | - L Jufe
- Laboratory of Pathology, Ramos Mejía Hospital, C.A.B.A ., Argentina
| | - M A Avagnina
- Department of Pathology, Clinical Hospital (UBA), C.A.B.A ., Córdoba, Argentina
| | - L B Díaz
- Department of Pathology, Clinical Hospital (UBA), C.A.B.A ., Córdoba, Argentina
| | - L A Palaoro
- Department of Clinical Biochemistry, Clinical Hospital (UBA), C.A.B.A., INFIBIOC , Córdoba, Argentina
| |
Collapse
|
4
|
Galvez-Carvajal L, Sanchez-Muñoz A, Ribelles N, Saez M, Baena J, Ruiz S, Ithurbisquy C, Alba E. Targeted treatment approaches in refractory germ cell tumors. Crit Rev Oncol Hematol 2019; 143:130-138. [PMID: 31634730 DOI: 10.1016/j.critrevonc.2019.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/17/2019] [Indexed: 12/23/2022] Open
Abstract
Germ cell tumors (GCTs) are the most common type of solid tumor amongst patients between 15 and 35 years of age. They are also one of the types of tumor with the highest cure rate, due to their high sensitivity to cisplatin based chemotherapy. Nonetheless, around 15-20% of metastatic patients will not have curative options after a relapse on the first and second line. This proves that new therapeutic options for these refractory GCTs patients need to be developed. This article offers a bibliographic review of all studies using targeted treatment or immunotherapy for refractory GCTs patients.
Collapse
Affiliation(s)
- Laura Galvez-Carvajal
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Alfonso Sanchez-Muñoz
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Nuria Ribelles
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Maribel Saez
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Javier Baena
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Sofia Ruiz
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Catherine Ithurbisquy
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain
| | - Emilio Alba
- UGCI of Medical Oncology. Hospitales Regional and Universitario Virgen de la Victoria, IBIMA, UMA. Malaga, Spain.
| |
Collapse
|
5
|
Gaudichon J, Jakobczyk H, Debaize L, Cousin E, Galibert MD, Troadec MB, Gandemer V. Mechanisms of extramedullary relapse in acute lymphoblastic leukemia: Reconciling biological concepts and clinical issues. Blood Rev 2019; 36:40-56. [PMID: 31010660 DOI: 10.1016/j.blre.2019.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/03/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Long-term survival rates in childhood acute lymphoblastic leukemia (ALL) are currently above 85% due to huge improvements in treatment. However, 15-20% of children still experience relapses. Relapses can either occur in the bone marrow or at extramedullary sites, such as gonads or the central nervous system (CNS), formerly referred to as ALL-blast sanctuaries. The reason why ALL cells migrate to and stay in these sites is still unclear. In this review, we have attempted to assemble the evidence concerning the microenvironmental factors that could explain why ALL cells reside in such sites. We present criteria that make extramedullary leukemia niches and solid tumor metastatic niches comparable. Indeed, considering extramedullary leukemias as metastases could be a useful approach for proposing more effective treatments. In this context, we conclude with several examples of potential niche-based therapies which could be successfully added to current treatments of ALL.
Collapse
Affiliation(s)
- Jérémie Gaudichon
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology and Oncology Department, University Hospital, Caen, France.
| | - Hélène Jakobczyk
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Lydie Debaize
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Elie Cousin
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France
| | - Marie-Dominique Galibert
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France.
| | - Marie-Bérengère Troadec
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Virginie Gandemer
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France.
| |
Collapse
|
6
|
Frumento G, Zuo J, Verma K, Croft W, Ramagiri P, Chen FE, Moss P. CD117 (c-Kit) Is Expressed During CD8 + T Cell Priming and Stratifies Sensitivity to Apoptosis According to Strength of TCR Engagement. Front Immunol 2019; 10:468. [PMID: 30930902 PMCID: PMC6428734 DOI: 10.3389/fimmu.2019.00468] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 02/21/2019] [Indexed: 01/04/2023] Open
Abstract
CD117 (cKit) is the receptor for stem cell factor (SCF) and plays an important role in early haemopoiesis. We show that CD117 is also expressed following priming of mature human CD8+ T cells in vitro and is detectable following primary infection in vivo. CD117 expression is mediated through an intrinsic pathway and is suppressed by IL-12. Importantly, the extent of CD117 expression is inversely related to the strength of the activating stimulus and subsequent engagement with cell-bound SCF markedly increases susceptibility to apoptosis. CD117 is therefore likely to shape the pattern of CD8+ T cell immunodominance during a primary immune response by rendering cells with low avidity for antigen more prone to apoptosis. Furthermore, CD117+ T cells are highly sensitive to apoptosis mediated by galectin-1, a molecule commonly expressed within the tumor microenvironment, and CD117 expression may therefore represent a novel and potentially targetable mechanism of tumor immune evasion.
Collapse
Affiliation(s)
- Guido Frumento
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom.,NHS Blood and Transplant, Birmingham, United Kingdom
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom
| | - Wayne Croft
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom.,Centre for Computational Biology, University of Birmingham Birmingham, United Kingdom
| | - Pradeep Ramagiri
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom
| | - Frederick E Chen
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom.,NHS Blood and Transplant, Birmingham, United Kingdom.,Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust Birmingham, United Kingdom.,Royal London Hospital, Barts Health NHS Trust London, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham, United Kingdom.,Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust Birmingham, United Kingdom
| |
Collapse
|
7
|
Koli S, Mukherjee A, Reddy KVR. Retinoic acid triggers c-kit gene expression in spermatogonial stem cells through an enhanceosome constituted between transcription factor binding sites for retinoic acid response element (RARE), spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1) and E26 transformation-specific (ETS). Reprod Fertil Dev 2018; 29:521-543. [PMID: 28442062 DOI: 10.1071/rd15145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/14/2015] [Indexed: 12/19/2022] Open
Abstract
Restricted availability of retinoic acid (RA) in the testicular milieu regulates transcriptional activity of c-kit (KIT, CD117), which aids in the determination of spermatogonial stem-cell differentiation. The effect of RA on c-kit has been reported previously, but its mode of genomic action remains unresolved. We studied the molecular machinery guiding RA responsiveness to the c-kit gene using spermatogonial stem-cell line C18-4 and primary spermatogonial cells. A novel retinoic acid response element (RARE) positioned at -989 nucleotides upstream of the transcription start site (TSS) was identified, providing a binding site for a dimeric RA receptor (i.e. retinoic acid receptor gamma (RARγ) and retinoic X receptor). RA treatment influenced c-kit promoter activity, along with endogenous c-kit expression in C18-4 cells. A comprehensive promoter deletion assay using the pGL3B reporter system characterised the region spanning -271bp and -1011bp upstream of the TSS, which function as minimal promoter and maximal promoter, respectively. In silico analysis predicted that the region -1011 to +58bp comprised the distal enhancer RARE and activators such as spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1), specificity protein 1 (SP1) and four E26 transformation-specific (ETS) tandem binding sites at the proximal region. Gel retardation and chromatin immunoprecipitation (ChIP) assays showed binding for RARγ, PU.1 and SP1 to the predicted consensus binding sequences, whereas GABPα occupied only two out of four ETS binding sites within the c-kit promoter region. We propose that for RA response, an enhanceosome is orchestrated through scaffolding of a CREB-binding protein (CBP)/p300 molecule between RARE and elements in the proximal promoter region, controlling germ-line expression of the c-kit gene. This study outlines the fundamental role played by RARγ, along with other non-RAR transcription factors (PU.1, SP1 and GABPα), in the regulation of c-kit expression in spermatogonial stem cells in response to RA.
Collapse
Affiliation(s)
- Swanand Koli
- Division of Molecular Immunology and Microbiology, National Institute for Research in Reproductive Health, Indian Council of Medical Research, J.M Street, Parel, Mumbai-400 012, India
| | - Ayan Mukherjee
- Department of Biological Science, Kent State University, Kent, OH 44240, USA
| | - Kudumula Venkata Rami Reddy
- Division of Molecular Immunology and Microbiology, National Institute for Research in Reproductive Health, Indian Council of Medical Research, J.M Street, Parel, Mumbai-400 012, India
| |
Collapse
|
8
|
Elzinga-Tinke JE, Dohle GR, Looijenga LH. Etiology and early pathogenesis of malignant testicular germ cell tumors: towards possibilities for preinvasive diagnosis. Asian J Androl 2016; 17:381-93. [PMID: 25791729 PMCID: PMC4430936 DOI: 10.4103/1008-682x.148079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Malignant testicular germ cell tumors (TGCT) are the most frequent cancers in Caucasian males (20-40 years) with an 70% increasing incidence the last 20 years, probably due to combined action of (epi)genetic and (micro)environmental factors. It is expected that TGCT have carcinoma in situ(CIS) as their common precursor, originating from an embryonic germ cell blocked in its maturation process. The overall cure rate of TGCT is more than 90%, however, men surviving TGCT can present long-term side effects of systemic cancer treatment. In contrast, men diagnosed and treated for CIS only continue to live without these long-term side effects. Therefore, early detection of CIS has great health benefits, which will require an informative screening method. This review described the etiology and early pathogenesis of TGCT, as well as the possibilities of early detection and future potential of screening men at risk for TGCT. For screening, a well-defined risk profile based on both genetic and environmental risk factors is needed. Since 2009, several genome wide association studies (GWAS) have been published, reporting on single-nucleotide polymorphisms (SNPs) with significant associations in or near the genes KITLG, SPRY4, BAK1, DMRT1, TERT, ATF7IP, HPGDS, MAD1L1, RFWD3, TEX14, and PPM1E, likely to be related to TGCT development. Prenatal, perinatal, and postnatal environmental factors also influence the onset of CIS. A noninvasive early detection method for CIS would be highly beneficial in a clinical setting, for which specific miRNA detection in semen seems to be very promising. Further research is needed to develop a well-defined TGCT risk profile, based on gene-environment interactions, combined with noninvasive detection method for CIS.
Collapse
Affiliation(s)
| | | | - Leendert Hj Looijenga
- Department of Pathology, Laboratory of Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
9
|
|
10
|
Rijlaarsdam MA, Tax DMJ, Gillis AJM, Dorssers LCJ, Koestler DC, de Ridder J, Looijenga LHJ. Genome wide DNA methylation profiles provide clues to the origin and pathogenesis of germ cell tumors. PLoS One 2015; 10:e0122146. [PMID: 25859847 PMCID: PMC4479500 DOI: 10.1371/journal.pone.0122146] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/07/2015] [Indexed: 12/18/2022] Open
Abstract
The cell of origin of the five subtypes (I-V) of germ cell tumors (GCTs) are assumed to be germ cells from different maturation stages. This is (potentially) reflected in their methylation status as fetal maturing primordial germ cells are globally demethylated during migration from the yolk sac to the gonad. Imprinted regions are erased in the gonad and later become uniparentally imprinted according to fetal sex. Here, 91 GCTs (type I-IV) and four cell lines were profiled (Illumina’s HumanMethylation450BeadChip). Data was pre-processed controlling for cross hybridization, SNPs, detection rate, probe-type bias and batch effects. The annotation was extended, covering snRNAs/microRNAs, repeat elements and imprinted regions. A Hidden Markov Model-based genome segmentation was devised to identify differentially methylated genomic regions. Methylation profiles allowed for separation of clusters of non-seminomas (type II), seminomas/dysgerminomas (type II), spermatocytic seminomas (type III) and teratomas/dermoid cysts (type I/IV). The seminomas, dysgerminomas and spermatocytic seminomas were globally hypomethylated, in line with previous reports and their demethylated precursor. Differential methylation and imprinting status between subtypes reflected their presumed cell of origin. Ovarian type I teratomas and dermoid cysts showed (partial) sex specific uniparental maternal imprinting. The spermatocytic seminomas showed uniparental paternal imprinting while testicular teratomas exhibited partial imprinting erasure. Somatic imprinting in type II GCTs might indicate a cell of origin after global demethylation but before imprinting erasure. This is earlier than previously described, but agrees with the totipotent/embryonic stem cell like potential of type II GCTs and their rare extra-gonadal localization. The results support the common origin of the type I teratomas and show strong similarity between ovarian type I teratomas and dermoid cysts. In conclusion, we identified specific and global methylation differences between GCT subtypes, providing insight into their developmental timing and underlying developmental biology. Data and extended annotation are deposited at GEO (GSE58538 and GPL18809).
Collapse
Affiliation(s)
- Martin A. Rijlaarsdam
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - David M. J. Tax
- Faculty of Electrical Engineering, Mathematics and Computer Science Intelligent Systems—Delft Bioinformatics Lab, Technical University of Delft, Delft, The Netherlands
| | - Ad J. M. Gillis
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lambert C. J. Dorssers
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Devin C. Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jeroen de Ridder
- Faculty of Electrical Engineering, Mathematics and Computer Science Intelligent Systems—Delft Bioinformatics Lab, Technical University of Delft, Delft, The Netherlands
| | - Leendert H. J. Looijenga
- Department of Pathology, Erasmus MC Cancer Institute—University Medical Center Rotterdam, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
11
|
Cardoso HJ, Figueira MI, Correia S, Vaz CV, Socorro S. The SCF/c-KIT system in the male: Survival strategies in fertility and cancer. Mol Reprod Dev 2014; 81:1064-79. [PMID: 25359157 DOI: 10.1002/mrd.22430] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/25/2014] [Indexed: 12/18/2022]
Abstract
Maintaining the delicate balance between cell survival and death is of the utmost importance for the proper development of germ cells and subsequent fertility. On the other hand, the fine regulation of tissue homeostasis by mechanisms that control cell fate is a factor that can prevent carcinogenesis. c-KIT is a type III receptor tyrosine kinase activated by its ligand, stem cell factor (SCF). c-KIT signaling plays a crucial role in cell fate decisions, specifically controlling cell proliferation, differentiation, survival, and apoptosis. Indeed, deregulating the SCF/c-KIT system by attenuation or overactivation of its signaling strength is linked to male infertility and cancer, and rebalancing its activity via c-KIT inhibitors has proven beneficial in treating human tumors that contain gain-of-function mutations or overexpress c-KIT. This review addresses the roles of SCF and c-KIT in the male reproductive tract, and discusses the potential application of c-KIT target therapies in disorders of the reproductive system.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | |
Collapse
|
12
|
Rijlaarsdam MA, Looijenga LHJ. An oncofetal and developmental perspective on testicular germ cell cancer. Semin Cancer Biol 2014; 29:59-74. [PMID: 25066859 DOI: 10.1016/j.semcancer.2014.07.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 07/17/2014] [Indexed: 12/19/2022]
Abstract
Germ cell tumors (GCTs) represent a diverse group of tumors presumably originating from (early fetal) developing germ cells. Most frequent are the testicular germ cell cancers (TGCC). Overall, TGCC is the most frequent malignancy in Caucasian males (20-40 years) and remains an important cause of (treatment related) mortality in these young men. The strong association between the phenotype of TGCC stem cell components and their totipotent ancestor (fetal primordial germ cell or gonocyte) makes these tumors highly relevant from an onco-fetal point of view. This review subsequently discusses the evidence for the early embryonic origin of TGCCs, followed by an overview of the crucial association between TGCC pathogenesis, genetics, environmental exposure and the (fetal) testicular micro-environment (genvironment). This culminates in an evaluation of three genvironmentally modulated hallmarks of TGCC directly related to the oncofetal pathogenesis of TGCC: (1) maintenance of pluripotency, (2) cell cycle control/cisplatin sensitivity and (3) regulation of proliferation/migration/apoptosis by KIT-KITL mediated receptor tyrosine kinase signaling. Briefly, TGCC exhibit identifiable stem cell components (seminoma and embryonal carcinoma) and progenitors that show large and consistent similarities to primordial/embryonic germ cells, their presumed totipotent cells of origin. TGCC pathogenesis depends crucially on a complex interaction of genetic and (micro-)environmental, i.e. genvironmental risk factors that have only been partly elucidated despite significant effort. TGCC stem cell components also show a high degree of similarity with embryonic stem/germ cells (ES) in the regulation of pluripotency and cell cycle control, directly related to their exquisite sensitivity to DNA damaging agents (e.g. cisplatin). Of note, (ES specific) micro-RNAs play a pivotal role in the crossover between cell cycle control, pluripotency and chemosensitivity. Moreover, multiple consistent observations reported TGCC to be associated with KIT-KITL mediated receptor tyrosine kinase signaling, a pathway crucially implicated in proliferation, migration and survival during embryogenesis including germ cell development. In conclusion, TGCCs are a fascinating model for onco-fetal developmental processes especially with regard to studying cell cycle control, pluripotency maintenance and KIT-KITL signaling. The knowledge presented here contributes to better understanding of the molecular characteristics of TGCC pathogenesis, translating to identification of at risk individuals and enhanced quality of care for TGCC patients (diagnosis, treatment and follow-up).
Collapse
Affiliation(s)
- Martin A Rijlaarsdam
- Department of Pathology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Leendert H J Looijenga
- Department of Pathology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Correia S, Alves MR, Cavaco JE, Oliveira PF, Socorro S. Estrogenic regulation of testicular expression of stem cell factor and c-kit: implications in germ cell survival and male fertility. Fertil Steril 2014; 102:299-306. [DOI: 10.1016/j.fertnstert.2014.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 03/29/2014] [Accepted: 04/06/2014] [Indexed: 01/22/2023]
|
14
|
Zhao W, Ji X, Zhang F, Li L, Ma L. Embryonic stem cell markers. Molecules 2012; 17:6196-236. [PMID: 22634835 PMCID: PMC6268870 DOI: 10.3390/molecules17066196] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/31/2012] [Accepted: 05/04/2012] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cell (ESC) markers are molecules specifically expressed in ES cells. Understanding of the functions of these markers is critical for characterization and elucidation for the mechanism of ESC pluripotent maintenance and self-renewal, therefore helping to accelerate the clinical application of ES cells. Unfortunately, different cell types can share single or sometimes multiple markers; thus the main obstacle in the clinical application of ESC is to purify ES cells from other types of cells, especially tumor cells. Currently, the marker-based flow cytometry (FCM) technique and magnetic cell sorting (MACS) are the most effective cell isolating methods, and a detailed maker list will help to initially identify, as well as isolate ESCs using these methods. In the current review, we discuss a wide range of cell surface and generic molecular markers that are indicative of the undifferentiated ESCs. Other types of molecules, such as lectins and peptides, which bind to ESC via affinity and specificity, are also summarized. In addition, we review several markers that overlap with tumor stem cells (TSCs), which suggest that uncertainty still exists regarding the benefits of using these markers alone or in various combinations when identifying and isolating cells.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| | - Xiang Ji
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Fangfang Zhang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Liang Li
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| |
Collapse
|
15
|
Ferlin A, Pengo M, Pizzol D, Carraro U, Frigo AC, Foresta C. Variants in KITLG predispose to testicular germ cell cancer independently from spermatogenic function. Endocr Relat Cancer 2012; 19:101-8. [PMID: 22194441 DOI: 10.1530/erc-11-0340] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epidemiological data suggest an association and a common pathogenetic link between male infertility and testicular germ cell tumor (TGCT) development. Genome-wide studies identified that TGCT susceptibility is associated with KITLG (c-KIT ligand), which regulates the formation of primordial germ cells, from which TGCT is believed to arise and spermatogenesis develops. In this study, we analyzed the link between KITLG, TGCT, and spermatogenic disruption by performing an association study between the KITLG markers rs995030 and rs4471514 and 426 TGCT cases and 614 controls with normal and abnormal sperm count. We found that TGCT risk was increased more than twofold per copy of the major G allele and A allele in KITLG rs995030 and rs4471514 (odds ratio (OR)=2.38, 95% confidence interval (95% CI)=1.81-3.12; OR=2.43, 95% CI=1.86-3.17 respectively), and homozygotes for the risk allele had a sevenfold increased risk of TGCT. KITLG markers were strongly associated with seminoma subtype (per allele risk increased more than threefold, homozygote risk increased by 13- to 16-fold) and weakly with nonseminoma. KITLG markers were not associated with sperm production, as no difference was observed in men with normozoospermia and azoo-oligozoospermia, both in controls and in TGCT cases. In conclusion, this study provides evidence that KITLG variants are involved in TGCT development and they represent an independent and strong specific risk factor for TGCT independently from spermatogenic function. A shared genetic cause and a common pathogenetic link between TGCT development and impairment of spermatogenesis are not evident from this study.
Collapse
Affiliation(s)
- Alberto Ferlin
- Section of Clinical Pathology and Centre for Human Reproduction Pathology, Department of Histology, Microbiology and Medical Biotechnologies, University of Padova, Via Gabelli 63, 35121 Padova, Italy.
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Pediatric germ cell tumors represent a diverse group of tumors that present from in utero through adolescence at many nongonadal locations, from the neck to the sacrococcygeal region. Surgical resection remains the central element of management, and accurate surgical staging is essential to properly ascertain the correct risk-based treatment. The management for all benign tumors (mature and immature teratomas) and select completely resectable malignant tumors is surgery alone. Modern-day chemotherapy is extremely effective in infants and children with unresectable and metastatic disease and these children have a very high survival rate. The use of neoadjuvant chemotherapy allows vital organ preservation and there is no role for resection of vital structures at the time of initial presentation.
Collapse
Affiliation(s)
- Frederick J Rescorla
- Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
17
|
Cheng L, Roth LM, Zhang S, Wang M, Morton MJ, Zheng W, Abdul Karim FW, Montironi R, Lopez-Beltran A. KIT gene mutation and amplification in dysgerminoma of the ovary. Cancer 2010; 117:2096-103. [DOI: 10.1002/cncr.25794] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/09/2010] [Accepted: 10/15/2010] [Indexed: 01/04/2023]
|
18
|
Chui K, Trivedi A, Cheng CY, Cherbavaz DB, Dazin PF, Huynh ALT, Mitchell JB, Rabinovich GA, Noble-Haeusslein LJ, John CM. Characterization and functionality of proliferative human Sertoli cells. Cell Transplant 2010; 20:619-35. [PMID: 21054948 DOI: 10.3727/096368910x536563] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It has long been thought that mammalian Sertoli cells are terminally differentiated and nondividing postpuberty. For most previous in vitro studies immature rodent testes have been the source of Sertoli cells and these have shown little proliferative ability when cultured. We have isolated and characterized Sertoli cells from human cadaveric testes from seven donors ranging from 12 to 36 years of age. The cells proliferated readily in vitro under the optimized conditions used with a doubling time of approximately 4 days. Nuclear 5-ethynyl-2'-deoxyuridine (EdU) incorporation confirmed that dividing cells represented the majority of the population. Classical Sertoli cell ultrastructural features, lipid droplet accumulation, and immunoexpression of GATA-4, Sox9, and the FSH receptor (FSHr) were observed by electron and fluorescence microscopy, respectively. Flow cytometry revealed the expression of GATA-4 and Sox9 by more than 99% of the cells, and abundant expression of a number of markers indicative of multipotent mesenchymal cells. Low detection of endogenous alkaline phosphatase activity after passaging showed that few peritubular myoid cells were present. GATA-4 and SOX9 expression were confirmed by reverse transcription polymerase chain reaction (RT-PCR), along with expression of stem cell factor (SCF), glial cell line-derived neurotrophic factor (GDNF), and bone morphogenic protein 4 (BMP4). Tight junctions were formed by Sertoli cells plated on transwell inserts coated with fibronectin as revealed by increased transepithelial electrical resistance (TER) and polarized secretion of the immunoregulatory protein, galectin-1. These primary Sertoli cell populations could be expanded dramatically in vitro and could be cryopreserved. The results show that functional human Sertoli cells can be propagated in vitro from testicular cells isolated from adult testis. The proliferative human Sertoli cells should have important applications in studying infertility, reproductive toxicology, testicular cancer, and spermatogenesis, and due to their unique biological properties potentially could be useful in cell therapy.
Collapse
Affiliation(s)
- Kitty Chui
- MandalMed, Inc., San Francisco, CA 94107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
BACKGROUND Germ cell tumours (GCTs) represent an extraordinarily chemosensitive malignancy. However, 20-30% of patients with advanced disease cannot be cured by currently available treatments. The role of tyrosine kinase receptors has been widely studied in other malignancies. Yet, limited information is available on GCTs. METHODS One hundred and nine paraffin-embedded GCTs in 84 patients were assessed by immunohistochemistry for epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER-2)/neu and KIT (CD117) expression. Univariate and multivariate analyses were performed to evaluate their role as predictive and/or prognostic factors. RESULTS EGFR and HER-2/neu staining was detected in 28% and 13% of tumours, respectively, predominantly in nonseminomatous GCTs. KIT protein was almost universally expressed in seminomas (97%), being virtually absent in choriocarcinoma and teratocarcinoma subtypes. EGFR expression showed inverse association with tumour response of borderline significance. With a median follow-up of 10.6 years, no significant association was observed between the expression of any of these markers and relapse-free or overall survival. CONCLUSIONS EGFR, HER-2/neu and KIT have differential patterns of expression in GCTs according to histological subtypes. The expression of these markers in our series had no prognostic or predictive significance.
Collapse
|
20
|
Muciaccia B, Sette C, Paronetto MP, Barchi M, Pensini S, D'Agostino A, Gandini L, Geremia R, Stefanini M, Rossi P. Expression of a truncated form of KIT tyrosine kinase in human spermatozoa correlates with sperm DNA integrity. Hum Reprod 2010; 25:2188-202. [DOI: 10.1093/humrep/deq168] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
Rapley EA, Nathanson KL. Predisposition alleles for testicular germ cell tumour. Curr Opin Genet Dev 2010; 20:225-30. [DOI: 10.1016/j.gde.2010.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 02/18/2010] [Accepted: 02/19/2010] [Indexed: 01/21/2023]
|
22
|
Lau SK, Weiss LM, Chu PG. TCL1 protein expression in testicular germ cell tumors. Am J Clin Pathol 2010; 133:762-6. [PMID: 20395523 DOI: 10.1309/ajcpipu1mptbm2fq] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
We immunohistochemically studied TCL1 protein expression in different histologic types of 63 testicular germ cell tumors: 23 seminomas, 14 embryonal carcinomas, 4 teratomas, 2 yolk sac tumors, and 20 mixed germ cell tumors. The 20 mixed germ cell tumors contained components of seminoma (n = 10), embryonal carcinoma (n = 18), teratoma (n = 16), yolk sac tumor (n = 7), and choriocarcinoma (n = 3). We also examined 40 cases of intratubular germ cell neoplasia, unclassified type (IGCNU). Positive immunoreactivity for TCL1 was observed in 91% of the seminoma samples (30/33) and all IGCNU cases. In contrast, no TCL1 expression was detected among the nonseminomatous germ cell tumors. In the context of testicular germ cell neoplasia, the presence of TCL1 protein appears restricted to IGCNU and seminoma, suggesting association with an undifferentiated state and loss of protein expression with tumor differentiation. Immunohistochemical evaluation of TCL1 expression may have usefulness in substantiating a diagnosis of IGCNU or seminoma and in the separation of seminoma from nonseminomatous germ cell tumors.
Collapse
|
23
|
Bialas M, Borczynska A, Rozwadowska N, Fiszer D, Kosicki W, Jedrzejczak P, Kurpisz M. SCF and c-kit expression profiles in male individuals with normal and impaired spermatogenesis. Andrologia 2010; 42:83-91. [DOI: 10.1111/j.1439-0272.2009.00960.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
24
|
Bougherara H, Subra F, Crépin R, Tauc P, Auclair C, Poul MA. The Aberrant Localization of Oncogenic Kit Tyrosine Kinase Receptor Mutants Is Reversed on Specific Inhibitory Treatment. Mol Cancer Res 2009; 7:1525-33. [DOI: 10.1158/1541-7786.mcr-09-0138] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
25
|
Biermann K, Zhou H, Büttner R. [Molecular pathology of testicular germ cell tumors: an update]. DER PATHOLOGE 2009; 29:348-53. [PMID: 18633620 DOI: 10.1007/s00292-008-1014-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Germ cell tumors (GCT) comprise a heterogeneous group of benign and malignant tumors. Based on their different biological characteristics and their origin, five types of GCT are classified. Among these, malignant seminomatous and non-seminomatous GCT in males and females are designated as type II GCT. They occur most frequently as malignant testicular GCTs. Many characteristics of type II GCT can be linked to embryonic stem cells. Intratubular germ cell neoplasia, unclassified (IGCNU) is the precursor of type II GCT and derives from undifferentiated germ cells, gonocytes, which persist in the newborn testis and escape the normal differentiation process. It is suggested that Exon-17-activated mutations of the receptor tyrosine kinase, c-KIT, occur early in germ cell development and that gonocytes with an activated c-KIT receptor are restricted in their differentiation, thereby escaping normal development. New diagnostic markers for neoplastic germ cells, including OCT3/4 and AP-2gamma, are specifically detected in IGCNU, seminomas and embryonal carcinomas and are helpful in the differentiation of type II GCT from other malignant tumors.
Collapse
Affiliation(s)
- K Biermann
- Afdeling Pathologie, Josephine Nefkens Instituut, Erasmus University Medical Center, Dr. Molewaterplein 50, Postbus 2040, 3000 CA, 3015 GE, Rotterdam, Niederlande.
| | | | | |
Collapse
|
26
|
Unni SK, Modi DN, Pathak SG, Dhabalia JV, Bhartiya D. Stage-specific localization and expression of c-kit in the adult human testis. J Histochem Cytochem 2009; 57:861-9. [PMID: 19435714 DOI: 10.1369/jhc.2009.953737] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The c-kit receptor (KIT) and its ligand, stem cell factor (SCF), represent one of the key regulators of testicular formation, development, and function and have been extensively studied in various animal models. The present study was undertaken to characterize the pattern of localization and expression of c-kit in normal adult human testis. Immunohistochemical analysis showed that KIT is expressed in the cytoplasm of spermatogonia, acrosomal granules of spermatids, and Leydig cells. Interestingly, a rather heterogenous pattern of expression of the protein along the basement membrane was observed. Intense protein localization in spermatogonia was detected in stages I-III, whereas low expression was observed in stages IV-VI of the seminiferous epithelium, indicating that the expression of the molecule was stage specific. In situ hybridization studies revealed that the transcripts of the gene were also localized in a similar non-uniform pattern. To the best of our knowledge, such a stage-specific expression of KIT has not been reported previously in the human testis. The results of the present study may expand current knowledge about the c-kit/SCF system in human spermatogenesis.
Collapse
Affiliation(s)
- Sreepoorna K Unni
- Scientist D and Divisional Head, Stem Cell Biology Department, National Institute for Research in Reproductive Health (ICMR), J M Street, Parel, Mumbai 400012, India
| | | | | | | | | |
Collapse
|
27
|
Weinbauer GF, Wessels J. ‘Paracrine’ control of spermatogenesis. Andrologia 2009. [DOI: 10.1111/j.1439-0272.1999.tb01421.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
28
|
van de Geijn GJM, Hersmus R, Looijenga LHJ. Recent developments in testicular germ cell tumor research. ACTA ACUST UNITED AC 2009; 87:96-113. [DOI: 10.1002/bdrc.20140] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Hussain SA, Ma YT, Palmer DH, Hutton P, Cullen MH. Biology of testicular germ cell tumors. Expert Rev Anticancer Ther 2009; 8:1659-73. [PMID: 18925857 DOI: 10.1586/14737140.8.10.1659] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Germ cell tumors are derived from cells of the germ cell lineage and are the most common solid malignancies to affect young Caucasian men between the ages of 15 and 40 years. All testicular germ cell tumors develop from the same precursor lesion, intratubular germ cell neoplasia unclassified, which in turn is thought to arise from malignant transformation of a primordial germ cell or gonocyte. These tumors are characterized by extreme chemosensitivity and are considered a model for curative disease. In spite of this, a small subset of patients with metastatic disease fail to achieve a complete response with cisplatin-based chemotherapy or relapse from complete remission. Understanding the molecular biology may help the design of new therapies for those patients with a poor prognosis and could also improve the treatment of cancer in general. Current understanding of the role of genetic and epigenetic factors in the etiology of germ cell tumors and the biochemical mechanisms underlying chemotherapy sensitivity and resistance is discussed in detail in this review.
Collapse
Affiliation(s)
- Syed A Hussain
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, University Hospital Birmingham NHS Foundation Trust, Edgbaston, Birmingham , UK.
| | | | | | | | | |
Collapse
|
30
|
Ozer O, Zhao YD, Ostler KR, Akin C, Anastasi J, Vardiman JW, Godley LA. The identification and characterisation of novel KIT transcripts in aggressive mast cell malignancies and normal CD34+ cells. Leuk Lymphoma 2008; 49:1567-77. [PMID: 18766971 DOI: 10.1080/10428190802140865] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
KIT mutations have been identified in several malignancies, including acute myeloid leukemia (AML) and systemic mastocytosis (SM). Mast cell leukemia (MCL) is the most aggressive mast cell neoplasm, but has not been well studied due to its rarity. We identified novel KIT transcripts in two patients with MCL and two patients with SM with an associated hematological disorder, but not from two patients with SM. Similar novel KIT transcripts were also observed in normal CD34+ cells from bone marrow and umbilical cord blood, suggesting that altered KIT isoforms may be specific to the blast stage of hematopoietic precursors. The novel KIT proteins lack several domains including the ATP binding site, and one was inactive in a functional test for autophosphorylation. Our discovery of novel KIT transcripts underscores the importance of analysing entire protein encoding regions when studying genes of interest.
Collapse
Affiliation(s)
- Ozden Ozer
- Section of Hematopathology, Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Stoop H, Honecker F, van de Geijn GJM, Gillis AJM, Cools MC, de Boer M, Bokemeyer C, Wolffenbuttel KP, Drop SLS, de Krijger RR, Dennis N, Summersgill B, McIntyre A, Shipley J, Oosterhuis JW, Looijenga LHJ. Stem cell factor as a novel diagnostic marker for early malignant germ cells. J Pathol 2008; 216:43-54. [PMID: 18566970 DOI: 10.1002/path.2378] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Carcinoma in situ (CIS) of the testis is the pre-invasive stage of type II testicular germ cell tumours (TGCTs) of adolescents and adults. These tumours are the most frequently diagnosed cancer in Caucasian adolescents and young adults. In dysgenetic gonads, the precursor of type II GCTs can be either CIS or a lesion known as gonadoblastoma (GB). CIS/GB originates from a primordial germ cell (PGC)/gonocyte, ie an embryonic cell. CIS can be cured by local low-dose irradiation, with limited side effects on hormonal function. Therefore, strategies for early diagnosis of CIS are essential. Various markers are informative to diagnose CIS in adult testis by immunohistochemistry, including c-KIT, PLAP, AP-2gamma, NANOG, and POU5F1 (OCT3/4). OCT3/4 is the most informative and consistent in presence and expression level, resulting in intense nuclear staining. In the case of maturational delay of germ cells, frequently present in gonads of individuals at risk for type II (T)GCTs, use of these markers can result in overdiagnosis of malignant germ cells. This demonstrates the need for a more specific diagnostic marker to distinguish malignant germ cells from germ cells showing maturation delay. Here we report the novel finding that immunohistochemical detection of stem cell factor (SCF), the c-KIT ligand, is informative in this context. This was demonstrated in over 400 cases of normal (fetal, neonatal, infantile, and adult) and pathological gonads, as well as TGCT-derived cell lines, specifically in cases of CIS and GB. Both membrane-bound and soluble SCF were expressed, suggestive of an autocrine loop. SCF immunohistochemistry can be a valuable diagnostic tool, in addition to OCT3/4, to screen for precursor lesions of TGCTs, especially in patients with germ cell maturation delay.
Collapse
Affiliation(s)
- H Stoop
- Department of Pathology, Erasmus MC-Erasmus University Medical Center, Daniel den Hoed Cancer Center, Josephine Nefkens Institute, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Application of stem cell markers in search for neoplastic germ cells in dysgenetic gonads, extragonadal tumours, and in semen of infertile men. Cancer Treat Rev 2008; 34:348-67. [PMID: 18289797 DOI: 10.1016/j.ctrv.2007.12.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2007] [Revised: 12/17/2007] [Accepted: 12/18/2007] [Indexed: 01/08/2023]
Abstract
Germ cell tumours (GCTs) are a complex entity. Current areas of attention include early detection and avoidance of unnecessary over-treatment. Novel findings regarding diagnosis of GCTs located in various anatomical sites are described, particularly testicular GCTs and their common progenitor, carcinoma in situ (CIS). Recognition of CIS enables intervention before tumour development, but nevertheless, testicular GCTs are sporadically diagnosed at the pre-invasive stage where minimal treatment is necessary. As presence of CIS is asymptomatic, a simple screening method is needed when CIS is suspected (i.e. in males investigated for infertility). To develop approaches for early detection CIS gene expression studies have been performed showing many similarities with embryonic stem cells with confirmation of established markers (i.e. PLAP, OCT-3/4, KIT) and identification of novel markers (i.e. AP-2 gamma, NANOG). We have reported a very promising new approach of AP-2 gamma (or OCT3/4) based immunocytological semen analysis (specificity 93.6%, sensitivity 54.5%). Comparative studies of gonadal/extragonadal GCTs have revealed resemblance pointing towards similar, but not identical, origins. Moreover, infertility and testicular cancer are connected in the 'Testicular Dysgenesis Syndrome' and 25% of contralateral testes from testicular GCT patients harbour dysgenetic features, including impaired spermatogenesis. Thus, recent data have provided potential diagnostic tools including CIS detection in semen, microarray-based tumour classification, additional serological GCT markers, and novel stem cell markers for immunohistochemical diagnosis of gonadal and extragonadal GCTs. Many CIS candidate genes are yet uninvestigated, and information from these could increase knowledge about CIS tumour initiation/progression and be used for optimisation of a non-invasive detection method.
Collapse
|
33
|
Coffey J, Linger R, Pugh J, Dudakia D, Sokal M, Easton DF, Timothy Bishop D, Stratton M, Huddart R, Rapley EA. Somatic KIT mutations occur predominantly in seminoma germ cell tumors and are not predictive of bilateral disease: report of 220 tumors and review of literature. Genes Chromosomes Cancer 2008; 47:34-42. [PMID: 17943970 DOI: 10.1002/gcc.20503] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Mutations in the KIT gene occur in approximately 8% of all testicular germ cell tumors (TGCT) and KIT is the most frequently mutated known cancer gene. One report has shown that 93% of patients with bilateral disease have a mutation at codon 816 of the KIT gene. Importantly, this suggests that the identification of a mutation in KIT is predictive of the development of a contralateral TGCT. We investigated the frequency and type of mutations in KIT in a series of 220 tumors from 211 patients with TGCTs and extragonadal germ cell tumors. In 170 patients with unilateral TGCT and no additional germ cell tumour, we identified one exon 11 mutation in a patient with unilateral TGCT and eight activating KIT mutations in exon 17 (9/175, 5.1%). In 32 patients with bilateral TGCT, one patient had an activating KIT mutation in exon 17 (3.1%). The incidence of activating KIT mutations in sporadic TGCT vs. familial TGCT was not significantly different. All mutations were identified in seminomas. Three extragonadal primary germ cell tumors were examined and in one tumor an activating KIT mutation was demonstrated in the pineal germinoma. Interestingly, this mutation was also seen in the patient's testicular seminoma. We find no evidence for an increased frequency of KIT mutations in bilateral TGCT.
Collapse
Affiliation(s)
- Jerome Coffey
- Testicular Cancer Genetics Team, Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sakuma Y, Matsukuma S, Yoshihara M, Sakurai S, Nishii M, Kishida T, Kubota Y, Nagashima Y, Inayama Y, Sasaki T, Nakamura Y, Miura T, Kameda Y, Tsuchiya E, Miyagi Y. Mutations of c-kit gene in bilateral testicular germ cell tumours in Japan. Cancer Lett 2008; 259:119-26. [DOI: 10.1016/j.canlet.2007.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2007] [Revised: 09/29/2007] [Accepted: 10/03/2007] [Indexed: 11/24/2022]
|
35
|
Tu J, Fan L, Tao K, Zhu W, Li J, Lu G. Stem cell factor affects fate determination of human gonocytes in vitro. Reproduction 2007; 134:757-65. [DOI: 10.1530/rep-07-0161] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The stem cell factor (SCF), binding its tyrosine kinase receptor c-Kit, has been shown to play essential roles in the proliferation, differentiation, and survival of germline cells. However, few reports are available about the effect of SCF on the development of human gonocytes within the fetal testis. The objective of this study was to investigate whether SCF affects the biological behaviors of human gonocytes before or after they enter the mitotic arrest stage. Employing an organ culture system, we observed that addition of exogenous SCF could influence the morphology of human gonocytesin vitro. Moreover, SCF was able to trigger the colony formation of round gonocytes, which were characterized positive for alkaline phosphatase activity, Oct-4, SSEA-4, and c-Kit as well. We found that SCF exerted actions in a dose- and age-dependent manner, although the stimulatory effect lasted no more than 14 days. We also showed that SCF played a role in suppressing the apoptosis of human gonocytes. Blocking of SCF signaling with either phosphatidylinositol 3-kinase or mitogen-activated protein kinase inhibitor resulted in similar apoptotic features as well as the SCF-withdrawal cultures. Taken together, we report that SCF acts as a potent regulator in the fate determination of human gonocytes. Our studies should form the basis forin vitrostudies and facilitate investigation of the molecular mechanisms underlying this unique stage.
Collapse
|
36
|
Abstract
BACKGROUND An increasing incidence of testis tumors has been noted over the second half of the 20th century. Congenital malformation of the male genitalia, prenatal risk factors, nonspecific and specific exposures in adulthood, and male infertility have all been associated with the etiology of germ cell tumors. METHODS The histologic classification, pathology, and current concepts of testicular germ cell tumors are reviewed. RESULTS Germ cell tumors occur at all ages. The tumors are identified as pure form (those of one histologic type) and mixed form (more than one histologic type). Over half of germ cell tumors consist of more than one cell type, requiring appropriate sampling for the correct diagnosis and correlation with the serum tumor markers. Burned-out germ cell tumors may occur in patients with metastatic disease with no gross evidence of a testicular tumor. CONCLUSIONS Appropriate management of testis tumors relies on accurate pathology and classification of these tumors.
Collapse
Affiliation(s)
- Isabell A Sesterhenn
- Department of Genitourinary Pathology, Armed Forces Institute of Pathology, Washington, DC 20306, USA.
| | | |
Collapse
|
37
|
Olesen IA, Sonne SB, Hoei-Hansen CE, Rajpert-De Meyts E, Skakkebaek NE. Environment, testicular dysgenesis and carcinoma in situ testis. Best Pract Res Clin Endocrinol Metab 2007; 21:462-78. [PMID: 17875492 DOI: 10.1016/j.beem.2007.04.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The testicular dysgenesis syndrome (TDS) hypothesis proposes that a proportion of the male reproductive disorders-cryptorchidism, hypospadias, infertility and testicular cancer-may be symptoms of one underlying developmental disease, TDS, which is most likely a result of disturbed gonadal development in the embryo. TDS may be caused by genetic factors, environmental/life-style factors, or a combination of both. Some rare disorders of sex development of genetic origin are among the best-known examples of severe TDS. Among the environmental and life-style factors that are suspected to influence the hormonal milieu of the developing gonad are the endocrine disrupters. A prenatal exposure to commonly used chemicals, e.g. phthalates, may result in a TDS-like phenotype in rats. Currently, this animal model is the best model for TDS. In humans the situation is much more complex, and TDS exists in a wide range of phenotypes: from the mildest and most common form, in which impaired spermatogenesis is the only symptom, to the most severe cases, in which the patient may develop testicular cancer. It is of great importance that clinicians in different specialties treating patients with TDS are aware of the association between the different symptoms.
Collapse
Affiliation(s)
- Inge A Olesen
- University Department of Growth & Reproduction, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
38
|
Orfao A, Garcia-Montero AC, Sanchez L, Escribano L. Recent advances in the understanding of mastocytosis: the role of KIT mutations. Br J Haematol 2007; 138:12-30. [PMID: 17555444 DOI: 10.1111/j.1365-2141.2007.06619.x] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mastocytosis is a heterogeneous disorder characterised by the expansion and accumulation of mast cells in different organs and tissues. Mast cell physiology is closely dependent on activation of the stem cell factor/Kit signalling pathways and accumulating evidences confirm the physiopathological key role of activating KIT mutations (typically D816V) in mastocytosis and their relationship with the clinical manifestations of the disease. This paper reviews the most recent advances in the understanding of the molecular mechanisms associated with KIT mutations in mastocytosis, including recent data about the use of new therapies targeting the Kit molecule and its associated downstream signalling pathways.
Collapse
Affiliation(s)
- Alberto Orfao
- Centro de Investigación del Cáncer/IBMCC, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.
| | | | | | | |
Collapse
|
39
|
Yang B, Wu J, Maddodi N, Ma Y, Setaluri V, Longley BJ. Epigenetic control of MAGE gene expression by the KIT tyrosine kinase. J Invest Dermatol 2007; 127:2123-8. [PMID: 17495964 DOI: 10.1038/sj.jid.5700836] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Class I MAGE proteins include the MAGE-A, MAGE-B, and MAGE-C antigens, which are normally expressed only in male germ cells but may be aberrantly expressed in melanomas and other tumors. It is known that MAGE gene expression is epigenetically repressed by promoter region methylation in most cells but factors controlling MAGE gene promoter methylation have not been identified. Using transcript microarray analysis and immunoblotting we found that MAGE-A and MAGE-C mRNA and protein are selectively downregulated by pharmacologic inhibition of KIT in KIT-dependent mast cell lines. Methylation-specific polymerase chain reaction studies showed that the MAGE-A3 and MAGE-C2 gene promoter regions were de-methylated in the presence of activated KIT but became methylated on inhibition of KIT, consistent with the downregulation of mRNA and protein. This is early evidence of a tyrosine kinase affecting MAGE gene promoter region methylation and expression, and represents early evidence of a tyrosine kinase in the epigenetic control of gene expression. MAGE proteins suppress apoptosis and promote tumor survival, and are novel targets for functional manipulation and immunotherapy. Understanding the factors controlling MAGE gene expression may allow more effective therapeutic strategies targeting MAGE antigens.
Collapse
Affiliation(s)
- Bing Yang
- Department of Dermatology, University of Wisconsin Medical School, Madison, Wisconsin 53715, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Lau SK, Weiss LM, Chu PG. D2-40 immunohistochemistry in the differential diagnosis of seminoma and embryonal carcinoma: a comparative immunohistochemical study with KIT (CD117) and CD30. Mod Pathol 2007; 20:320-5. [PMID: 17277761 DOI: 10.1038/modpathol.3800749] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The distinction between seminoma and embryonal carcinoma based on morphology alone can sometimes be problematic, requiring the use of immunohistochemistry to facilitate diagnosis. D2-40 is a monoclonal antibody that reacts with an oncofetal antigen expressed by fetal germ cells and testicular germ cell tumors. The diagnostic value of D2-40 immunohistochemistry in distinguishing seminoma from embryonal carcinoma has not been determined. D2-40 immunoreactivity was evaluated in a series of testicular germ cell tumors and compared with that of KIT (CD117) and CD30, to assess the relative utility of this marker in discriminating between seminoma and embryonal carcinoma. Forty testicular germ cell neoplasms were examined, which included 19 seminomas, three embryonal carcinomas, three teratomas, one yolk sac tumor, and 14 mixed germ cell tumors. The 14 cases of mixed germ cell tumors contained components of seminoma (n=7), embryonal carcinoma (n=11), teratoma (n=10), yolk sac tumor (n=2), and choriocarcinoma (n=1). All cases of pure seminoma and the seminomatous components of mixed germ cell tumors exhibited positive immunoreactivity for D2-40. Focal positivity for D2-40 was also observed in 29% of the embryonal carcinoma samples. D2-40 immunoreactivity in seminomas was characterized by diffuse membrane staining, whereas for embryonal carcinomas, staining was focal and distributed along the apical surfaces of the neoplastic cells. Immunohistochemical staining for KIT was observed in 92% of the seminoma samples and in none of the embryonal carcinomas. Conversely, CD30 expression was identified in 93% of the embryonal carcinoma samples and in none of the seminomas. Other germ cell components showed no immunoreactivity for D2-40, KIT, or CD30. KIT and CD30 are effective immunohistochemical markers in separating seminoma from embryonal carcinoma. Although a highly sensitive marker for seminomas, D2-40 positivity was also observed in a subset of embryonal carcinomas, thus limiting the utility of this antibody for discriminating between these two malignancies.
Collapse
Affiliation(s)
- Sean K Lau
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | | | | |
Collapse
|
41
|
Biermann K, Klingmüller D, Koch A, Pietsch T, Schorle H, Büttner R, Zhou H. Diagnostic value of markers M2A, OCT3/4, AP-2γ, PLAP and c-KIT in the detection of extragonadal seminomas. Histopathology 2006; 49:290-7. [PMID: 16918976 DOI: 10.1111/j.1365-2559.2006.02496.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS To compare the suitability of new seminoma markers including transcription factors AP-2gamma, OCT3/4 and M2A for detection of metastatic and extragonadal seminomas with the two well-known markers c-KIT and PLAP. MATERIALS AND METHODS The immunohistochemical distribution of PLAP, c-KIT, M2A, AP-2gamma and OCT3/4 was examined in two pineal germinomas, 28 metastatic seminomas and 10 of their testicular primaries. Evaluation of specificity was achieved by additional tissue array studies on 75 malignancies other than germ cell tumours (GCT). Clinical data including serum PLAP were available in 18 patients. RESULTS Compared with other markers, significantly better staining results were observed with antibodies to M2A and AP-2gamma in all seminomatous GCT. In contrast, the staining pattern with antibodies to c-KIT, PLAP and OCT3/4 was variable or absent. The lowest specificity was obtained with c-KIT, which was expressed in a variety of non-GCT. The only M2A+ mesothelioma expressed no other seminoma markers. No correlation between serum PLAP level and tissue PLAP expression was found. CONCLUSIONS M2A and AP-2gamma are the most sensitive markers for seminoma metastases or primary extragonadal seminomas. Combination of these markers provides highly specific and clear results for detection of a seminomatous GCT.
Collapse
Affiliation(s)
- K Biermann
- Institute of Pathology, Department of Developmental Pathology, University of Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Cools M, Drop SLS, Wolffenbuttel KP, Oosterhuis JW, Looijenga LHJ. Germ cell tumors in the intersex gonad: old paths, new directions, moving frontiers. Endocr Rev 2006; 27:468-84. [PMID: 16735607 DOI: 10.1210/er.2006-0005] [Citation(s) in RCA: 274] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The risk for the development of germ cell tumors is an important factor to deal with in the management of patients with disorders of sex development (DSD). However, this risk is often hard to predict. Recently, major progress has been made in identifying gene-products related to germ cell tumor development (testis-specific protein-Y encoded and octamer binding transcription factor 3/4) and in recognizing early changes of germ cells (maturation delay, preneoplastic lesions, and in situ neoplasia). The newly recognized "undifferentiated gonadal tissue" has been identified as a gonadal differentiation pattern bearing a high risk for the development of gonadoblastoma. It is expected that the combination of these findings will allow for estimation of the risk for tumor development in the individual patient (high risk/intermediate risk/low risk). This article reviews the recent literature regarding the prevalence of germ cell tumors in patients with DSD. Some major limitations regarding this topic, including a confusing terminology referring to the different forms of intersex disorders and unclear criteria for the diagnosis of malignant germ cells at an early age (maturation delay vs. early steps in malignant transformation) are discussed. Thereafter, an overview of the recent advances that have been made in our knowledge of germ cell tumor development and the correct diagnosis of early neoplastic lesions in this patient population is provided. A new classification system for patients with DSD is proposed as a tool to refine our insight in the prevalence of germ cell tumors in specific diagnostic groups.
Collapse
Affiliation(s)
- Martine Cools
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Josephine Nefkens Institute, The Netherlands
| | | | | | | | | |
Collapse
|
43
|
Sung MT, MacLennan GT, Cheng L. Retroperitoneal seminoma in limited biopsies: morphologic criteria and immunohistochemical findings in 30 cases. Am J Surg Pathol 2006; 30:766-73. [PMID: 16723857 DOI: 10.1097/00000478-200606000-00015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The incidence of retroperitoneal seminoma is much less than that of its gonadal counterpart. Accurate diagnosis of retroperitoneal seminoma is critical, because it carries an excellent prognosis due to its favorable response to radiation therapy and/or cisplatin-based chemotherapy. However, correctly diagnosing a retroperitoneal seminoma may be challenging, especially when the biopsy material is limited. The present study was conducted to evaluate histologic findings and immunohistochemical staining patterns in biopsy specimens of retroperitoneal seminoma and to compare their utility as diagnostic tools. Thirty biopsy specimens of retroperitoneal seminoma were assessed for histologic characteristics and immunohistochemical expression of OCT4, c-kit, placental-like alkaline phosphatase, and cytokeratin AE1/AE3. The clinical information, morphologic features, and staining intensities and the percentages of positively staining tumor cells were analyzed. The mean age of patients was 38 years. Lymphocytic infiltration and nucleolar prominence in tumor cells were found in all 30 cases (100%). The incidence of other histologic characteristics were as follows: fibrous septa/stroma in 80% (24 cases), clear tumor cell cytoplasm in 70% (21 cases), tumor necrosis in 60% (18 cases), cellular pleomorphism in 53% (16 cases), granulomatous inflammation in 50% (15 cases), distinct cell borders in 46% (14 cases), intercellular edema in 23% (7 cases), and syncytiotrophoblasts in 3% (1 case). The mean mitotic count was 3 (range 0 to 15) per 10 high-power fields. All 30 cases (100%) of retroperitoneal seminoma revealed moderate to strong nuclear OCT4 staining in more than 50% of tumor cells. Twenty-one cases (70%) showed membranous expression of c-kit by tumor cells, with moderate to strong staining intensity in most cases. Variable degrees of staining for placental-like alkaline phosphatase were identified in 23 cases (77%) with occasional background staining artifact. Six cases (20%) displayed a positive cytokeratin AE1/AE3 staining pattern with weak to moderate intensity. In conclusion, the most common histologic findings in limited biopsy specimens of retroperitoneal seminoma were lymphocytic infiltration and nucleolar prominence in tumor cell nuclei. OCT4 immunostaining, with its superior sensitivity and easy interpretation compared with other markers, is a powerful tool for confirming the diagnosis of retroperitoneal seminoma.
Collapse
Affiliation(s)
- Ming-Tse Sung
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA, and Department of Pathology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, Taiwan
| | | | | |
Collapse
|
44
|
Camps C, Sirera R, Bremnes RM, Garde J, Safont MJ, Blasco A, Berrocal A, Sánchez JJ, Calabuig C, Martorell M. Analysis of c-kit expression in small cell lung cancer: prevalence and prognostic implications. Lung Cancer 2006; 52:343-7. [PMID: 16574270 DOI: 10.1016/j.lungcan.2006.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Revised: 02/13/2006] [Accepted: 02/13/2006] [Indexed: 02/07/2023]
Abstract
c-kit, a growth factor receptor with tyrosine kinase activity, plays an important role in the biology of cancer. Its expression has been documented in several malignancies. We performed a retrospective study in 85 patients diagnosed with small cell lung cancer (SCLC) to determine the prevalence and role of c-kit as a possible prognostic marker in this lung cancer malignancy. Demographic and clinical data were obtained from patient charts. c-kit, analyzed as immunohistochemical expression in paraffin-embedded tumour tissues, was observed in 60% of patients. All patients were former or present smokers. At diagnosis, 46% of the patients had limited disease (LD) and 54% extended disease (ED). c-kit expression was observed in 59% of LD and 61% of ED patients (p=0.4). Patients received a median of 4 cycles first-line combination chemotherapy (platinum and etoposide). In LD patients, time to progression (TTP) was 11.5 months in c-kit (+) versus 5.9 in c-kit (-) patients (p=0.14), and median survival 15.4 and 12.8 months, respectively (p=0.33). In the ED group, TTP was 5.5 months in c-kit (+) versus 3.8 in c-kit (-) patients (p=0.34), whereas median survival was 6.3 and 7.9 months, respectively (p=0.45). With the limited number of patients in mind, our findings tended towards an association between c-kit expression and survival in the LD group.
Collapse
Affiliation(s)
- Carlos Camps
- Servicio de Oncología Médica, Consorcio Hospital General Universitario de Valencia, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rajpert-De Meyts E. Developmental model for the pathogenesis of testicular carcinoma in situ: genetic and environmental aspects. Hum Reprod Update 2006; 12:303-23. [PMID: 16540528 DOI: 10.1093/humupd/dmk006] [Citation(s) in RCA: 304] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Carcinoma in situ testis (CIS), also known as intratubular germ cell neoplasia (ITGCN), is a pre-invasive precursor of testicular germ cell tumours, the commonest cancer type of male adolescents and young adults. In this review, evidence supporting the hypothesis of developmental origin of testicular germ cell cancer is summarized, and the current concepts regarding aetiology and pathogenesis of this disease are critically discussed. Comparative studies of cell surface proteins (e.g. PLAP and KIT), some of the germ cell-specific markers (e.g. MAGEA4, VASA, TSPY and NY-ESO-1), supported by studies of regulatory elements of the cell cycle (e.g. p53, CHK2 and p19-INK4d) demonstrated a close similarity of CIS to primordial germ cells and gonocytes, consistent with the pre-meiotic origin of CIS. Recent gene expression profiling studies showed that CIS cells closely resemble embryonic stem cells (ESCs). The abundance of factors associated with pluripotency (NANOG and OCT-3/4) and undifferentiated state (AP-2gamma) may explain the remarkable pluripotency of germ cell neoplasms, which are capable of differentiating to various somatic tissue components of teratomas. Impaired gonadal development resulting in the arrest of gonocyte differentiation and retention of its embryonic features, associated with an increasing genomic instability, is the most probable model for the pathogenesis of CIS. Genomic amplification of certain chromosomal regions, e.g. 12p, may facilitate survival of CIS and further invasive progression. Genetic studies, have so far not identified gene polymorphisms predisposing to the most common non-familial testicular cancer, but this research has only recently begun. Association of CIS with other disorders, such as congenital genital malformations and some forms of impaired spermatogenesis, all rising in incidence in a synchronous manner, led to the hypothesis that CIS might be a manifestation of testicular dysgenesis syndrome (TDS). The aetiology of TDS including testicular cancer remains to be elucidated, but epidemiological trends suggest a primary role for environmental factors, probably combined with genetic susceptibility.
Collapse
Affiliation(s)
- Ewa Rajpert-De Meyts
- University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark.
| |
Collapse
|
46
|
Bakhshi S, Singh D, Karak AK, Thulkar S. Childhood primary mesenteric seminoma. Indian J Pediatr 2006; 73:241-3. [PMID: 16567922 DOI: 10.1007/bf02825492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We report an 11-year-old child who presented with an abdominal lump and was diagnosed as having an extragonal primary mesenteric seminoma. Patient was treated with 4 cycles of combination chemotherapy cisplatin, etoposide and bleomycin; he is now disease free for 2 years. We discuss and review extragonadal germ cell tumors arising from the mesentery and their management.
Collapse
Affiliation(s)
- Sameer Bakhshi
- Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, New Delhi, India.
| | | | | | | |
Collapse
|
47
|
Miettinen M, Lasota J. KIT (CD117): a review on expression in normal and neoplastic tissues, and mutations and their clinicopathologic correlation. Appl Immunohistochem Mol Morphol 2006; 13:205-20. [PMID: 16082245 DOI: 10.1097/01.pai.0000173054.83414.22] [Citation(s) in RCA: 359] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD117 (KIT) is a type III receptor tyrosine kinase operating in cell signal transduction in several cell types. Normally KIT is activated (phosphorylated) by binding of its ligand, the stem cell factor. This leads to a phosphorylation cascade ultimately activating various transcription factors in different cell types. Such activation regulates apoptosis, cell differentiation, proliferation, chemotaxis, and cell adhesion. KIT-dependent cell types include mast cells, some hematopoietic stem cells, germ cells, melanocytes, and Cajal cells of the gastrointestinal tract, and neoplasms of these cells are examples of KIT-positive tumors. Other KIT-positive normal cells include epithelial cells in skin adnexa, breast, and subsets of cerebellar neurons. KIT positivity has been variably reported in sarcomas such as angiosarcoma, Ewing sarcoma, synovial sarcoma, leiomyosarcoma, and MFH; results of the last three are controversial. The variations in published data may result from incomplete specificity of some polyclonal antibodies, possibly contributed by too high dilutions. Also, KIT is expressed in pulmonary and other small cell carcinomas, adenoid cystic carcinoma, renal chromophobe carcinoma, thymic, and some ovarian and few breast carcinomas. A good KIT antibody reacts with known KIT positive cells, and smooth muscle cells and fibroblasts are negative. KIT deficiency due to hereditary nonsense/missense mutations leads to disruption of KIT-dependent functions such as erythropoiesis, skin pigmentation, fertility, and gastrointestinal motility. Conversely, pathologic activation of KIT through gain-of-function mutations leads to neoplasia of KIT-dependent and KIT-positive cell types at least in three different systems: mast cells/myeloid cells--mastocytosis/acute myeloid leukemia, germ cells--seminoma, and Cajal cells--gastrointestinal stromal tumors (GISTs). KIT tyrosine kinase inhibitors such as imatinib mesylate are the generally accepted treatment of metastatic GISTs, and their availability has prompted an active search for other treatment targets among KIT-positive tumors such as myeloid leukemias and small cell carcinoma of the lung, with variable and often nonconvincing results.
Collapse
Affiliation(s)
- Markku Miettinen
- Department of Soft Tissue Pathology, Armed Forces Institute of Pathology, Washington, DC 20306-6000, USA.
| | | |
Collapse
|
48
|
Cools M, Honecker F, Stoop H, Veltman JD, de Krijger RR, Steyerberg E, Wolffenbuttel KP, Bokemeyer C, Lau YFC, Drop SLS, Looijenga LHJ. Maturation delay of germ cells in fetuses with trisomy 21 results in increased risk for the development of testicular germ cell tumors. Hum Pathol 2006; 37:101-11. [PMID: 16360422 DOI: 10.1016/j.humpath.2005.09.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Accepted: 09/24/2005] [Indexed: 11/23/2022]
Abstract
Trisomy 21 is associated with an increased risk for the occurrence of germ cell tumors in males. The development of these tumors is thought to be related to events in fetal life. A delay in the maturation of germ cells is one of the mechanisms that have been proposed for the development of these tumors in high-risk groups such as intersex patients. To investigate whether a delay in germ cell development also occurs in trisomy 21, we examined the gonads of 30 fetuses, neonates, and infants with trisomy 21 (19 males and 11 females) for the expression of several immunohistochemical germ cell markers throughout pregnancy and compared them with a series of 46 age-matched controls. The results of our study reveal a significant delay in germ cell development in fetuses with trisomy 21, especially in males. Prolonged expression of octamer binding transcription factor 3/4, in combination with an increased expression of testis-specific protein, Y-encoded, might have pathogenetic relevance for the development of testicular germ cell tumors in this population.
Collapse
Affiliation(s)
- Martine Cools
- Department of Pathology, Josephine Nefkens Institute, Erasmus MC-University Medical Center, Daniel den Hoed, 3000 DR Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hoei-Hansen CE, Sehested A, Juhler M, Lau YFC, Skakkebaek NE, Laursen H, Rajpert-de Meyts E. New evidence for the origin of intracranial germ cell tumours from primordial germ cells: expression of pluripotency and cell differentiation markers. J Pathol 2006; 209:25-33. [PMID: 16456896 DOI: 10.1002/path.1948] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Primary intracranial germ cell tumours are rare neoplasms that occur in children and adolescents. This study examined both the biology and the origin of these tumours, as it has been hypothesized that they originate from a totipotent primordial germ cell. We applied recent knowledge from gonadal germ cell tumours and analysed expression of a wide panel of stem cell-related proteins (C-KIT, OCT-3/4 (POU5F1), AP-2gamma (TFAP2C), and NANOG) and developmentally regulated germ cell-specific proteins (including MAGE-A4, NY-ESO-1, and TSPY). Expression at the protein level was analysed in 21 children and young adults with intracranial germinomas and non-germinomas, contributing to a careful description of these unusual tumours and adding to the understanding of pathogenesis. Stem cell related proteins were highly expressed in intracranial germ cell tumours, and many similarities were detected with their gonadal equivalents, including a close similarity with primordial germ cells. A notable difference was the sex-specific expression of TSPY, a gene previously implicated in the origin of gonadoblastoma. TSPY was only detected in germ cell tumours in the central nervous system (CNS) from males, suggesting that it is not required for the initiation of malignant germ cell transformation. The expression of genes associated with embryonic stem cell pluripotency in CNS germ cell tumours strongly suggests that these tumours are derived from cells that retain, at least partially, an embryonic stem cell-like phenotype, which is a hallmark of primordial germ cells.
Collapse
Affiliation(s)
- C E Hoei-Hansen
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
50
|
Coffey J, Birtle AJ, Cogill G, Christmas TJ, Rapley EA, Huddart RA. A discussion of the biology of testicular cancer and current concepts in the management of stage I and bilateral disease. Clin Oncol (R Coll Radiol) 2005; 17:441-7. [PMID: 16149288 DOI: 10.1016/j.clon.2005.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This case was the subject of a Grand Round Presentation at the Royal Marsden Hospital, Sutton, UK on 8 June 2004. A case of metachronous, bilateral testicular germ-cell tumours (TGCTs) arising in a patient with a family history of this disease was presented. The second primary was managed conservatively. The rationale and outcome of this approach was presented, along with a discussion of the management of early stage TGCTs and the genetics of familial and bilateral disease.
Collapse
Affiliation(s)
- J Coffey
- Academic Urology Unit, Royal Marsden Hospital, Sutton, UK
| | | | | | | | | | | |
Collapse
|