1
|
Stock A, Calaminus G, Weisthoff M, Serfling J, Pietsch T, Bison B, Pham M, Warmuth-Metz M. Imaging in malignant germ cell tumors involving the hypothalamo-neurohypophyseal axis: the evaluation of the posterior pituitary bright spot is essential. Neuroradiology 2024; 66:1405-1416. [PMID: 38844695 PMCID: PMC11246258 DOI: 10.1007/s00234-024-03384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/29/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE Malignant intracranial germ cell tumors (GCTs) are rare diseases in Western countries. They arise in midline structures and diagnosis is often delayed. We evaluated imaging characteristics and early tumor signs of suprasellar and bifocal GCT on MRI. METHODS Patients with the diagnosis of a germinoma or non-germinomatous GCT (NGGCT) who received non-contrast sagittal T1WI on MRI pre-therapy were included. Loss of the posterior pituitary bright spot (PPBS), the expansion and size of the tumor, and the expansion and infiltration of surrounding structures were evaluated. Group comparison for histologies and localizations was performed. RESULTS A total of 102 GCT patients (median age at diagnosis 12.3 years, range 4.4-33.8; 57 males; 67 in suprasellar localization) were enrolled in the study. In the suprasellar cohort, NGGCTs (n = 20) were noticeably larger than germinomas (n = 47; p < .001). Each tumor showed involvement of the posterior lobe or pituitary stalk. A PPBS loss (total n = 98) was observed for each localization and entity in more than 90% and was related to diabetes insipidus. Osseous infiltration was observed exclusively in suprasellar GCT (significantly more frequent in NGGCT; p = .004). Time between the first MRI and therapy start was significantly longer in the suprasellar cohort (p = .005), with an even greater delay in germinoma compared to NGGCT (p = .002). The longest interval to treatment had circumscribed suprasellar germinomas (median 312 days). CONCLUSION A loss of the PPBS is a hint of tumor origin revealing small tumors in the neurohypophysis. Using this sign in children with diabetes insipidus avoids a delay in diagnosis.
Collapse
Affiliation(s)
- Annika Stock
- Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, D-97080, Wuerzburg, Germany.
| | - Gabriele Calaminus
- Department of Pediatric Hematology/Oncology, University Children's Hospital Bonn, Bonn, Germany
| | - Mathilda Weisthoff
- Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, D-97080, Wuerzburg, Germany
- Department of Radiology, University Hospital Cologne, Colonge, Germany
| | - Julia Serfling
- Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, D-97080, Wuerzburg, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Brigitte Bison
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Neuroradiological Reference Center for the pediatric brain tumor (HIT) studies of the German Society of Pediatric Oncology and Hematology, University Hospital Wuerzburg until 2020; Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg Since 2021, Augsburg, Germany
| | - Mirko Pham
- Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, D-97080, Wuerzburg, Germany
| | - Monika Warmuth-Metz
- Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, D-97080, Wuerzburg, Germany
| |
Collapse
|
2
|
Zhou J, Wu C, Li S. CNS Germ Cell Tumors: Molecular Advances, Significance in Risk Stratification and Future Directions. Brain Sci 2024; 14:445. [PMID: 38790424 PMCID: PMC11119131 DOI: 10.3390/brainsci14050445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Central Nervous System Germ Cell Tumors (CNS GCTs) represent a subtype of intracranial malignant tumors characterized by highly heterogeneous histology. Current diagnostic methods in clinical practice have notable limitations, and treatment strategies struggle to achieve personalized therapy based on patient risk stratification. Advances in molecular genetics, biology, epigenetics, and understanding of the tumor microenvironment suggest the diagnostic potential of associated molecular alterations, aiding risk subgroup identification at diagnosis. Furthermore, they suggest the existence of novel therapeutic approaches targeting chromosomal alterations, mutated genes and altered signaling pathways, methylation changes, microRNAs, and immune checkpoints. Moving forward, further research is imperative to explore the pathogenesis of CNS GCTs and unravel the intricate interactions among various molecular alterations. Additionally, these findings require validation in clinical cohorts to assess their role in the diagnosis, risk stratification, and treatment of patients.
Collapse
Affiliation(s)
| | | | - Shouwei Li
- Department of Neuro-Oncology (No.6 Neurosurgery Department), Sanbo Brain Hospital, Capital Medical University, No.50. Yi-Ke-Song, Xiangshan, Haidian District, Beijing 100093, China; (J.Z.); (C.W.)
| |
Collapse
|
3
|
Toll SA, Flore LA, Gorsi HS, Marupudi NI, Mody S, Kupsky W, Wang ZJ. Intracranial Germinoma in Two Caucasian American Siblings With Autism Spectrum Disorder. J Pediatr Hematol Oncol 2024; 46:106-111. [PMID: 38277627 DOI: 10.1097/mph.0000000000002821] [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] [Received: 03/06/2023] [Accepted: 12/17/2023] [Indexed: 01/28/2024]
Abstract
Intracranial germ cell tumors (IGCTs) comprise 3% to 5% of all pediatric brain tumors in the West, with a significantly higher prevalence in Asia. Although these tumors are histologically diverse, repeated somatic variants have been demonstrated. Chromosomal aneuploidies, such as Klinefelter and Down syndromes, are associated with IGCTs, but no familial germline tumor syndromes are currently known. Here, we report the novel case of 2 American siblings with underlying autism spectrum disorder who developed intracranial germinoma within months of each other, in the absence of external risk factors. Extensive genetic testing was performed, including karyotyping, chromosomal microarray, and whole exome and whole genome sequencing, and did not identify any variants accounting for the phenotypes. Despite the absence of overlapping variants, a recent retrospective review demonstrated a threefold greater prevalence of autism spectrum disorder in patients with intracranial germinoma compared with national prevalence. This report highlights the complexity of tumor development, as well as the need for further research regarding IGCTs in a neurodivergent population.
Collapse
Affiliation(s)
- Stephanie A Toll
- Department of Pediatrics, Division of Hematology/Oncology
- Central Michigan University School of Medicine, Mt Pleasant
| | - Leigh Anne Flore
- Department of Pediatrics, Division of Genetics, Children's Hospital of Michigan
- Central Michigan University School of Medicine, Mt Pleasant
| | - Hamza S Gorsi
- Department of Pediatrics, Division of Hematology/Oncology
- Central Michigan University School of Medicine, Mt Pleasant
| | | | - Swati Mody
- Central Michigan University School of Medicine, Mt Pleasant
- Department of Radiology, C.S. Mott Children's Hospital, Ann Arbor, MI
| | - William Kupsky
- Department of Pathology, Detroit Medical Center, Detroit
| | - Zhihong Joanne Wang
- Department of Pediatrics, Division of Hematology/Oncology, Children's Hospital of Richmond, Richmond, VA
| |
Collapse
|
4
|
Abbas M, Enani MZ, Alsabban Z, Meliti A, Homoud M. Primary anterior visual pathway germinoma in a 13-year-old boy: A case report. Surg Neurol Int 2024; 15:48. [PMID: 38468649 PMCID: PMC10927171 DOI: 10.25259/sni_929_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/21/2024] [Indexed: 03/13/2024] Open
Abstract
Background Primary optic nerve and chiasmal germinomas are very rare. These lesions can commonly be mistaken for optic pathway gliomas based on imaging alone. It is radiosensitive and cured in most of the cases. Case Description We report a rare case of a 13-year-old boy with primary bilateral optic nerves and chiasmal germinoma who underwent partial surgical resection followed by radiotherapy. Follow-up brain imaging after two months post-radiotherapy showed interval regression of the tumor. Our literature review identified that 12 reported cases of primary anterior visual pathway germinoma had been reported to regress significantly post-radiotherapy alone or with chemotherapy. Conclusion Histologic correlation is essential for appropriate treatment, alleviating symptoms, and avoiding irreversible vision loss.
Collapse
Affiliation(s)
- Mosab Abbas
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mariam Zuhair Enani
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Zehour Alsabban
- Department of Radiology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Abdelrazak Meliti
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mohammed Homoud
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
Takami H, Satomi K, Fukuoka K, Nakamura T, Tanaka S, Mukasa A, Saito N, Suzuki T, Yanagisawa T, Sugiyama K, Kanamori M, Kumabe T, Tominaga T, Tamura K, Maehara T, Nonaka M, Asai A, Yokogami K, Takeshima H, Iuchi T, Kobayashi K, Yoshimoto K, Sakai K, Nakazato Y, Matsutani M, Nagane M, Nishikawa R, Ichimura K. Distinct patterns of copy number alterations may predict poor outcome in central nervous system germ cell tumors. Sci Rep 2023; 13:15760. [PMID: 37735187 PMCID: PMC10514291 DOI: 10.1038/s41598-023-42842-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
We have previously reported that 12p gain may predict the presence of malignant components and poor prognosis for CNS germ cell tumor (GCT). Recently, 3p25.3 gain was identified as an independent predictor of poor prognosis for testicular GCT. Eighty-one CNS GCTs were analyzed. Copy number was calculated using methylation arrays. Five cases (6.2%) showed 3p25.3 gain, but only among the 40 non-germinomatous GCTs (NGGCTs) (5/40, 12.5%; p = 0.03). Among NGGCTs, those with a yolk sac tumor component showed a significantly higher frequency of 3p25.3 gain (18.2%) than those without (1.5%; p = 0.048). NGGCTs with gain showed significantly shorter progression-free survival (PFS) than those without (p = 0.047). The 3p25.3 gain and 12p gain were independent from each other. The combination of 3p25.3 gain and/or 12p gain was more frequent among NGGCTs with malignant components (69%) than among those without (29%; p = 0.02). Germinomas containing a higher number of copy number alterations showed shorter PFS than those with fewer (p = 0.03). Taken together, a finding of 3p25.3 gain may be a copy number alteration specific to NGGCTs and in combination with 12p gain could serve as a marker of negative prognosis or treatment resistance. Germinoma with frequent chromosomal instability may constitute an unfavorable subgroup.
Collapse
Affiliation(s)
- Hirokazu Takami
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Kaishi Satomi
- Department of Pathology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka City, Tokyo, 181-8611, Japan
| | - Kohei Fukuoka
- Departments of Hematology/Oncology, Saitama Children's Medical Center, 1-2, Shintoshin, Chuo-ku, Saitama City, Saitama, 330-8777, Japan
| | - Taishi Nakamura
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama City, Kanagawa, 236-0004, Japan
| | - Shota Tanaka
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Nobuhito Saito
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomonari Suzuki
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama, 350-1298, Japan
| | - Takaaki Yanagisawa
- Department of Neurosurgery, Jikei University, 3-25-8, Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neuro-Oncology Program, Faculty of Medicine, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi, 980-8574, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi, 980-8574, Japan
- Department of Neurosurgery, Kitasato University, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai City, Miyagi, 980-8574, Japan
| | - Kaoru Tamura
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Taketoshi Maehara
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-0034, Japan
| | - Masahiro Nonaka
- Department of Neurosurgery, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Akio Asai
- Department of Neurosurgery, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Kiyotaka Yokogami
- Department of Neurosurgery, University of Miyazaki Faculty of Medicine, 5200, Kihara, Kiyotakecho, Miyazaki, 889-1692, Japan
| | - Hideo Takeshima
- Department of Neurosurgery, University of Miyazaki Faculty of Medicine, 5200, Kihara, Kiyotakecho, Miyazaki, 889-1692, Japan
| | - Toshihiko Iuchi
- Department of Neurosurgery, Chiba Cancer Center, 666-2, Nitona-cho, Chuo-ku, Chiba, 260-0801, Japan
| | - Keiichi Kobayashi
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka City, Tokyo, 181-8611, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Kyusyu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keiichi Sakai
- Shinshu Ueda Medical Center, 1-27-21, Midorigaoka, Ueda City, Nagano, 386-8610, Japan
| | - Yoichi Nakazato
- Department of Pathology, Hidaka Hospital, 886, Nakaomachi, Takasaki City, Gunma, 370-0001, Japan
| | - Masao Matsutani
- Gotanda Rehabilitation Hospital, 8-20, Nishi-gotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka City, Tokyo, 181-8611, Japan
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama, 350-1298, Japan
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
6
|
Maia R, Miranda A, Geraldo AF, Sampaio L, Ramaglia A, Tortora D, Severino M, Rossi A. Neuroimaging of pediatric tumors of the sellar region-A review in light of the 2021 WHO classification of tumors of the central nervous system. Front Pediatr 2023; 11:1162654. [PMID: 37416813 PMCID: PMC10320298 DOI: 10.3389/fped.2023.1162654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Sellar/suprasellar tumors comprise about 10% of all pediatric Central Nervous System (CNS) tumors and include a wide variety of entities, with different cellular origins and distinctive histological and radiological findings, demanding customized neuroimaging protocols for appropriate diagnosis and management. The 5th edition of the World Health Organization (WHO) classification of CNS tumors unprecedently incorporated both histologic and molecular alterations into a common diagnostic framework, with a great impact in tumor classification and grading. Based on the current understanding of the clinical, molecular, and morphological features of CNS neoplasms, there have been additions of new tumor types and modifications of existing ones in the latest WHO tumor classification. In the specific case of sellar/suprasellar tumors, changes include for example separation of adamantinomatous and papillary craniopharyngiomas, now classified as distinct tumor types. Nevertheless, although the current molecular landscape is the fundamental driving force to the new WHO CNS tumor classification, the imaging profile of sellar/suprasellar tumors remains largely unexplored, particularly in the pediatric population. In this review, we aim to provide an essential pathological update to better understand the way sellar/suprasellar tumors are currently classified, with a focus on the pediatric population. Furthermore, we intend to present the neuroimaging features that may assist in the differential diagnosis, surgical planning, adjuvant/neoadjuvant therapy, and follow-up of this group of tumors in children.
Collapse
Affiliation(s)
- Rúben Maia
- Department of Neuroradiology, Centro Hospitalar Universitário São João, Porto, Portugal
| | - André Miranda
- Diagnostic Neuroradiology Unit, Imaging Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Ana Filipa Geraldo
- Diagnostic Neuroradiology Unit, Imaging Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Luísa Sampaio
- Department of Neuroradiology, Centro Hospitalar Universitário São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Antonia Ramaglia
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Domenico Tortora
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| |
Collapse
|
7
|
Han YP, Lin HW, Li H. Cancer Stem Cells in Tumours of the Central Nervous System in Children: A Comprehensive Review. Cancers (Basel) 2023; 15:3154. [PMID: 37370764 DOI: 10.3390/cancers15123154] [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: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer stem cells (CSCs) are a subgroup of cells found in various kinds of tumours with stem cell characteristics, such as self-renewal, induced differentiation, and tumourigenicity. The existence of CSCs is regarded as a major source of tumour recurrence, metastasis, and resistance to conventional chemotherapy and radiation treatment. Tumours of the central nervous system (CNS) are the most common solid tumours in children, which have many different types including highly malignant embryonal tumours and midline gliomas, and low-grade gliomas with favourable prognoses. Stem cells from the CNS tumours have been largely found and reported by researchers in the last decade and their roles in tumour biology have been deeply studied. However, the cross-talk of CSCs among different CNS tumour types and their clinical impacts have been rarely discussed. This article comprehensively reviews the achievements in research on CSCs in paediatric CNS tumours. Biological functions, diagnostic values, and therapeutic perspectives are reviewed in detail. Further investigations into CSCs are warranted to improve the clinical practice in treating children with CNS tumours.
Collapse
Affiliation(s)
- Yi-Peng Han
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Hou-Wei Lin
- Department of Paediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Department of Paediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, Jiaxing 314001, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| |
Collapse
|
8
|
Yang M, Wang J, Zhang L, Liu J. Update on MRI in pediatric intracranial germ cell tumors-The clinical and radiological features. Front Pediatr 2023; 11:1141397. [PMID: 37215600 PMCID: PMC10192609 DOI: 10.3389/fped.2023.1141397] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Intracranial germ cell tumors (iGCTs) are uncommon brain tumors that mainly occur in children. Differing in histology, location, and gender of the patients, iGCTs are often divided into germinomas and non-germinomatous germ cell tumors (NGGCTs). Early diagnosis and timely treatment are crucial to iGCTs, the subtypes of which have substantial variations. This review summarized the clinical and radiological features of iGCTs at different sites, and reviewed the recent advances in neuroimaging of iGCTs, which can help predict tumor subtypes early and guide clinical decision-making.
Collapse
Affiliation(s)
| | | | - Lin Zhang
- Correspondence: Lin Zhang Jungang Liu
| | | |
Collapse
|
9
|
Primary midbrain germinomas: Report of a rare case with an updated review of the literature. Clin Neurol Neurosurg 2023; 227:107643. [PMID: 36863221 DOI: 10.1016/j.clineuro.2023.107643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/04/2023]
Abstract
Intracranial germinomas are most commonly extra-axial germ cell tumors that are predominantly found in the pineal and suprasellar regions. Primary intra-axial midbrain germinomas are extremely rare, with only eight reported cases. Here we present a 30-year-old man who presented with severe neurological deficits, with an MRI that showed a heterogeneously enhancing mass with ill-defined margins in the midbrain, and with surrounding vasogenic edema extending to the thalamus. The presumptive preoperative differential diagnosis included glial tumors and lymphoma. The patient underwent a right paramedian suboccipital craniotomy and biopsy obtained through the supracerebellar infratentorial transcollicular approach. The histopathological diagnosis was reported as pure germinoma. After patient discharge, he received chemotherapy with carboplatin and etoposide, followed by radiotherapy. Follow-up MRI at up to 26 months showed no contrast-enhancing lesions but a mild T2 FLAIR hyperintensity adjacent to the resection cavity. Differential diagnosis of midbrain lesions can be challenging and should include glial tumors, primary central nervous system lymphoma, germ cell tumors, and metastasis. Accurate diagnosis requires adequate tissue sampling. In this report, we present a very rare case of a primary intra-axial germinoma of the midbrain which is biopsied via a transcollicular approach. This report is also unique as it provides the first surgical video of an open biopsy and the microscopic appearance of an intra-axial primary midbrain germinoma via a transcollicular approach.
Collapse
|
10
|
Cheng H, Shang D, Zhou R. Germline stem cells in human. Signal Transduct Target Ther 2022; 7:345. [PMID: 36184610 PMCID: PMC9527259 DOI: 10.1038/s41392-022-01197-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
The germline cells are essential for the propagation of human beings, thus essential for the survival of mankind. The germline stem cells, as a unique cell type, generate various states of germ stem cells and then differentiate into specialized cells, spermatozoa and ova, for producing offspring, while self-renew to generate more stem cells. Abnormal development of germline stem cells often causes severe diseases in humans, including infertility and cancer. Primordial germ cells (PGCs) first emerge during early embryonic development, migrate into the gentile ridge, and then join in the formation of gonads. In males, they differentiate into spermatogonial stem cells, which give rise to spermatozoa via meiosis from the onset of puberty, while in females, the female germline stem cells (FGSCs) retain stemness in the ovary and initiate meiosis to generate oocytes. Primordial germ cell-like cells (PGCLCs) can be induced in vitro from embryonic stem cells or induced pluripotent stem cells. In this review, we focus on current advances in these embryonic and adult germline stem cells, and the induced PGCLCs in humans, provide an overview of molecular mechanisms underlying the development and differentiation of the germline stem cells and outline their physiological functions, pathological implications, and clinical applications.
Collapse
Affiliation(s)
- Hanhua Cheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| | - Dantong Shang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China
| | - Rongjia Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| |
Collapse
|
11
|
Takami H, Ichimura K. Biomarkers for risk-based treatment modifications for CNS germ cell tumors: Updates on biological underpinnings, clinical trials, and future directions. Front Oncol 2022; 12:982608. [PMID: 36132131 PMCID: PMC9483213 DOI: 10.3389/fonc.2022.982608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 12/05/2022] Open
Abstract
CNS germ cell tumors (GCTs) preferentially occur in pediatric and adolescent patients. GCTs are located predominantly in the neurohypophysis and the pineal gland. Histopathologically, GCTs are broadly classified into germinomas and non-germinomatous GCTs (NGGCTs). In general, germinoma responds well to chemotherapy and radiation therapy, with a 10-year overall survival (OS) rate of approximately 90%. In contrast, NGGCTs have a less favorable prognosis, with a five-year OS of approximately 70%. Germinomas are typically treated with platinum-based chemotherapy and whole-ventricular radiation therapy, while mature teratomas can be surgically cured. Other NGGCTs require intensive chemotherapy with radiation therapy, including whole brain or craniospinal irradiation, depending on the dissemination status and protocols. Long-term treatment-related sequelae, including secondary neoplasms and cerebrovascular events, have been well recognized. These late effects have a tremendous impact in later life, especially since patients are mostly affected in childhood or young adults. Intending to minimize the treatment burden on patients, the identification of biomarkers for treatment stratification and evaluation of treatment response is of critical importance. Recently, tumor cell content in germinomas has been shown to be closely related to prognosis, suggesting that cases with low tumor cell content may be safely treated with a less intensive regimen. Among the copy number alterations, the 12p gain is the most prominent and has been shown to be a negative prognostic factor in NGGCTs. MicroRNA clusters (mir-371-373) were also revealed to be a hallmark of GCTs, demonstrating the potential for the application of liquid biopsy in the diagnosis and detection of recurrence. Recurrent mutations have been detected in the MAPK or PI3K pathways, most typically in KIT and MTOR and low genome-wide methylation has been demonstrated in germinoma; this most likely reflects the cell-of-origin primordial germ cells for this tumor type. These alterations can also be leveraged for liquid biopsies of cell-free DNA and may potentially be targeted for treatment in the future. Advancements in basic research will be translated into clinical practice and can directly impact patient management. Additional understanding of the biology and pathogenesis of GCTs will lead to the development of better-stratified clinical trials, ultimately resulting in improved treatment outcomes and a reduction in long-term treatment-related adverse effects.
Collapse
Affiliation(s)
- Hirokazu Takami
- Department of Neurosurgery, The University of Tokyo Hospital, Tokyo, Japan
- *Correspondence: Hirokazu Takami,
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Satomi K, Takami H, Fukushima S, Yamashita S, Matsushita Y, Nakazato Y, Suzuki T, Tanaka S, Mukasa A, Saito N, Kanamori M, Kumabe T, Tominaga T, Kobayashi K, Nagane M, Iuchi T, Yoshimoto K, Tamura K, Maehara T, Sakai K, Sugiyama K, Yokogami K, Takeshima H, Nonaka M, Asai A, Ushijima T, Matsutani M, Nishikawa R, Ichimura K. 12p gain is predominantly observed in non-germinomatous germ cell tumors and identifies an unfavorable subgroup of central nervous system germ cell tumors. Neuro Oncol 2021; 24:834-846. [PMID: 34698864 DOI: 10.1093/neuonc/noab246] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) germ cell tumors (GCTs) are neoplasms predominantly arising in pediatric and young adult populations. While germinomas generally respond to chemotherapy and radiation, non-germinomatous GCTs (NGGCTs) require more intensive treatment. This study aimed to determine whether 12p gain could predict the prognosis of CNS GCTs. METHODS Eighty-two CNS GCTs were included in this study. The 12p gain was defined by an additional 12p in the background of potential polyploidy or polysomy. Cases were analyzed using an Illumina methylation 450K array for copy number investigations and validated by fluorescence in situ hybridization (FISH). RESULTS A 12p gain was found in 25-out-of-82 cases (30%) and was more frequent in NGGCTs (12% of germinoma cases and 50% of NGGCT cases), particularly in cases with malignant components, such as immature teratoma, yolk sac tumor, choriocarcinoma, and embryonal carcinoma. 12p gain and KIT mutation were mutually exclusive events. The presence of 12p gain correlated with shorter progression-free (PFS) and overall survival (OS) (10-year OS: 59% vs 94%, with and without 12p gain, respectively, P = 0.0002), even with histology and tumor markers incorporated in the multivariate analysis. Among NGGCTs, 12p gain still had prognostic significance for PFS and OS (10-year OS: 47% vs. 90%, respectively, P = 0.02). The 12p copy number status was shared among histological components in mixed GCTs. CONCLUSIONS 12p gain may predict the presence of malignant components of NGGCTs, and poor prognosis of the patients. It may be associated with early tumorigenesis of CNS GCT.
Collapse
Affiliation(s)
- Kaishi Satomi
- Department of Diagnostic Pathology, National Cancer Center Hospital.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | - Hirokazu Takami
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute.,Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Shintaro Fukushima
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | | | - Yuko Matsushita
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | | | - Tomonari Suzuki
- Department of NeuroOncology/Neurosurgery, Saitama Medical University International Medical Center
| | - Shota Tanaka
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Akitake Mukasa
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo.,Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University
| | - Nobuhito Saito
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Toshihiro Kumabe
- Department of Neurosurgery, Tohoku University Graduate School of Medicine.,Department of Neurosurgery, Kitasato University
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | | | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine
| | | | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University.,Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Kaoru Tamura
- Department of Functional Neurosurgery, Tokyo Medical and Dental University
| | - Taketoshi Maehara
- Department of Functional Neurosurgery, Tokyo Medical and Dental University
| | - Keiichi Sakai
- Department of Neurosurgery, Shinshu Ueda Medical Center
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neurooncology Program, Cancer Treatment Center, Hiroshima University Hospital
| | - Kiyotaka Yokogami
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki
| | - Hideo Takeshima
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki
| | - Masahiro Nonaka
- Department of Neurosurgery, Kansai Medical University Hospital
| | - Akio Asai
- Department of Neurosurgery, Kansai Medical University Hospital
| | | | | | - Ryo Nishikawa
- Department of NeuroOncology/Neurosurgery, Saitama Medical University International Medical Center
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute.,Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine
| |
Collapse
|
13
|
Nakamura H, Takami H, Yanagisawa T, Kumabe T, Fujimaki T, Arakawa Y, Karasawa K, Terashima K, Yokoo H, Fukuoka K, Sonoda Y, Sakurada K, Mineharu Y, Soejima T, Fujii M, Shinojima N, Hara J, Yamasaki K, Fujimura J, Yamasaki F, Takahashi M, Suzuki T, Sato I, Nishikawa R, Sugiyama K. The Japan Society for Neuro-Oncology Guideline on the Diagnosis and Treatment of Central Nervous System Germ Cell Tumors. Neuro Oncol 2021; 24:503-515. [PMID: 34671804 DOI: 10.1093/neuonc/noab242] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Primary CNS germ cell tumors (GCTs) are rare neoplasms predominantly observed in the pediatric and young adult populations. In line with the hypothesis that the primordial germ cell is the cell-of-origin, histopathological examinations for this pathology involve a diverse range of components mirroring the embryogenic developmental dimensions. Chemotherapy and radiotherapy are the mainstays of treatment, with surgery having a limited role for diagnosis and debulking of residual tissue after treatment. While better management has been achieved over recent decades by modifying radiation coverage and selecting appropriate chemotherapy, standardization of treatment remains challenging, partly due to the low volume of cases encountered in each institution. As the incidence is higher in East Asia, including Japan, the Japan Society for Neuro-Oncology established a multidisciplinary task force to create an evidence-based guideline for CNS GCTs. This guideline provides recommendations for multiple dimensions of clinical management for CNS GCTs, with particular focus on diagnostic measures including serum markers, treatment algorithms including surgery, radiotherapy and chemotherapy, and under-investigated but important areas such as treatment for recurrent cases, long-term follow-up protocols and long-term sequelae. This guideline serves the purpose of helping healthcare professionals keep up to date with current knowledge and standards of management for patients with this rare disease in daily clinical practice, as well as driving future translational and clinical research by recognizing unmet needs concerning this tumor.
Collapse
Affiliation(s)
- Hideo Nakamura
- Department of Neurosurgery, Kurume University School of Medicine
| | - Hirokazu Takami
- Department of Neurosurgery, The University of Tokyo Hospital
| | | | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine
| | | | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine
| | - Katsuyuki Karasawa
- Division of Radiation Oncology/Department of Radiology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital
| | - Keita Terashima
- Division of Neuro-Oncology, National Center for Child Health and Development
| | - Hideaki Yokoo
- Department of Human Pathology, Gunma University Graduate School of Medicine
| | - Kohei Fukuoka
- Department of Hematology and Oncology, Saitama Children's Medical Center
| | | | - Kaori Sakurada
- Department of Neurosurgery, Yamagata University Hospital
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine
| | | | - Motoaki Fujii
- Department of Radiation Therapy, Mitsui Memorial Hospital
| | - Naoki Shinojima
- Department of Neurosurgery, Kumamoto University School of Medicine
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital
| | - Kai Yamasaki
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital
| | - Junya Fujimura
- Department of Pediatrics, Juntendo University Faculty of Medicine
| | | | - Mayu Takahashi
- Department of Neurosurgery, University of Occupational and Environmental Health
| | - Tomonari Suzuki
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center
| | - Iori Sato
- Department of Family Nursing, School of Health Sciences and Nursing, Graduate School of Medicine, The University of Tokyo
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neuro-Oncology Program, Hiroshima University Hospital
| | | |
Collapse
|
14
|
Au HK, Peng SW, Guo CL, Lin CC, Wang YL, Kuo YC, Law TY, Ho HN, Ling TY, Huang YH. Niche Laminin and IGF-1 Additively Coordinate the Maintenance of Oct-4 Through CD49f/IGF-1R-Hif-2α Feedforward Loop in Mouse Germline Stem Cells. Front Cell Dev Biol 2021; 9:646644. [PMID: 34381769 PMCID: PMC8351907 DOI: 10.3389/fcell.2021.646644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/03/2021] [Indexed: 01/16/2023] Open
Abstract
The mechanism on how extracellular matrix (ECM) cooperates with niche growth factors and oxygen tension to regulate the self-renewal of embryonic germline stem cells (GSCs) still remains unclear. Lacking of an appropriate in vitro cell model dramatically hinders the progress. Herein, using a serum-free culture system, we demonstrated that ECM laminin cooperated with hypoxia and insulin-like growth factor 1 receptor (IGF-1R) to additively maintain AP activity and Oct-4 expression of AP+GSCs. We found the laminin receptor CD49f expression in d2 testicular GSCs that were surrounded by laminin. Laminin and hypoxia significantly increased the GSC stemness-related genes, including Hif-2α, Oct-4, IGF-1R, and CD49f. Cotreatment of IGF-1 and laminin additively increased the expression of IGF-IR, CD49f, Hif-2α, and Oct-4. Conversely, silencing IGF-1R and/or CD49f decreased the expression of Hif-2α and Oct-4. The underlying mechanism involved CD49f/IGF1R-(PI3K/AKT)-Hif-2α signaling loop, which in turn maintains Oct-4 expression, symmetric self-renewal, and cell migration. These findings reveal the additive niche laminin/IGF-IR network during early GSC development.
Collapse
Affiliation(s)
- Heng-Kien Au
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan.,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Syue-Wei Peng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| | - Chien-Chia Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Lin Wang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Che Kuo
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsz-Yau Law
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Nerng Ho
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Taipei Municipal Wanfang Hospital, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yen-Hua Huang
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Comprehensive Cancer Center of Taipei Medical University, Taipei, Taiwan.,The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
To Be or Not to Be a Germ Cell: The Extragonadal Germ Cell Tumor Paradigm. Int J Mol Sci 2021; 22:ijms22115982. [PMID: 34205983 PMCID: PMC8199495 DOI: 10.3390/ijms22115982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023] Open
Abstract
In the human embryo, the genetic program that orchestrates germ cell specification involves the activation of epigenetic and transcriptional mechanisms that make the germline a unique cell population continuously poised between germness and pluripotency. Germ cell tumors, neoplasias originating from fetal or neonatal germ cells, maintain such dichotomy and can adopt either pluripotent features (embryonal carcinomas) or germness features (seminomas) with a wide range of phenotypes in between these histotypes. Here, we review the basic concepts of cell specification, migration and gonadal colonization of human primordial germ cells (hPGCs) highlighting the analogies of transcriptional/epigenetic programs between these two cell types.
Collapse
|
16
|
Takami H, Perry A, Graffeo CS, Giannini C, Narita Y, Nakazato Y, Saito N, Nishikawa R, Matsutani M, Ichimura K, Daniels DJ. Comparison on epidemiology, tumor location, histology, and prognosis of intracranial germ cell tumors between Mayo Clinic and Japanese consortium cohorts. J Neurosurg 2021; 134:446-456. [PMID: 32005022 DOI: 10.3171/2019.11.jns191576] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/27/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Central nervous system (CNS) germ cell tumors (GCTs) are rare malignant neoplasms that arise predominantly in adolescents and young adults. CNS GCTs demonstrate characteristic trends in national associations, with implications for both tumor incidence and genetics. Although the incidence of CNS GCTs is markedly higher in East Asia than Western countries, direct comparative analyses between these CNS GCT populations are limited. METHODS In Japan, to facilitate the genomic analyses of CNS GCTs, the Intracranial Germ Cell Tumor Genome Analysis Consortium was established in 2011, and more than 200 cases of GCTs are available for both tumor tissue and clinical data, which is organized by the National Cancer Center (NCC) Japan. At the Mayo Clinic, there have been 98 cases of intracranial GCTs treated by the Department of Neurologic Surgery since 1988. In this paper, the authors compared the epidemiology, clinical presentation including location and histology, and prognosis between cases treated in the US and Japan. RESULTS There was no significant difference in age and sex distributions between the databases. However, there was a significant difference in the tumor locations; specifically, the frequency of basal ganglia was higher in the NCC database compared with the Mayo Clinic (8.4% vs 0%, p = 0.008), and bifocal location (neurohypophysis and pineal gland) was higher at the Mayo Clinic than at the NCC (18.8% vs 5.8%, p = 0.002). There was no difference in histological subdivisions between the databases. There was no difference in progression-free survival (PFS) and overall survival (OS) of germinoma cases and OS of nongerminomatous GCT (NGGCT) cases treated with chemotherapy and radiation therapy covering whole ventricles. However, PFS of NGGCTs differed significantly, and was better in the NCC cohorts (p = 0.04). CONCLUSIONS There appears to be a differential distribution of GCTs by neuroanatomical location between major geographic and national groups. Further study is warranted to better characterize any underlying genomic, epigenetic, or environmental factors that may be driving the phenotypic differences.
Collapse
Affiliation(s)
- Hirokazu Takami
- Departments of1Neurologic Surgery and
- 3Division of Brain Tumor Translational Research, National Cancer Center, Tokyo, Japan
- 4Department of Neurosurgery, Faculty of Medicine, University of Tokyo, Japan
| | | | | | - Caterina Giannini
- 2Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Narita
- 5Department of Neurosurgery and Neuro-oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoichi Nakazato
- 6Department of Pathology, Hidaka Hospital, Gunma, Japan; and
| | - Nobuhito Saito
- 4Department of Neurosurgery, Faculty of Medicine, University of Tokyo, Japan
| | - Ryo Nishikawa
- 7Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Masao Matsutani
- 7Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Koichi Ichimura
- 3Division of Brain Tumor Translational Research, National Cancer Center, Tokyo, Japan
| | | |
Collapse
|
17
|
Pediatric Suprasellar Germ Cell Tumors: A Clinical and Radiographic Review of Solitary vs. Bifocal Tumors and Its Therapeutic Implications. Cancers (Basel) 2020; 12:cancers12092621. [PMID: 32937871 PMCID: PMC7565935 DOI: 10.3390/cancers12092621] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Suprasellar germ cell tumors (S-GCTs) are rare, presenting in either solitary or multifocal fashion. In this study, we retrospectively examine 22 solitary S-GCTs and 20 bifocal germ cell tumors (GCTs) over a 30-year period and demonstrate clinical, radiographic, and prognostic differences between the two groups with therapeutic implications. Compared to S-GCTs, bifocal tumors were almost exclusively male, exhibited higher rate of metastasis, and had worse rates of progression free and overall survival trending toward significance. We also introduce a novel magnetic resonance (MR) imaging classification of suprasellar GCT into five types: a IIIrd ventricle floor tumor extending dorsally with or without an identifiable pituitary stalk (Type Ia, Ib), ventrally (Type III), in both directions (Type II), small lesions at the IIIrd ventricle floor extending to the stalk (Type IV), and tumor localized in the stalk (Type V). S-GCTs almost uniformly presented as Type I-III, while most bifocal GCTs were Type IV with a larger pineal mass. These differences are significant as bifocal GCTs representing concurrent primaries or subependymal extension may be treated with whole ventricle radiation, while cerebrospinal fluid (CSF)-borne metastases warrant craniospinal irradiation (CSI). Although further study is necessary, we recommend CSI for bifocal GCTs exhibiting high-risk features such as metastasis or non-germinomatous germ cell tumor histology.
Collapse
|
18
|
Chen H, Yin G, Cui N, Lin H. Rare Primary Embryonal Carcinoma in the Brachial Plexus: A Case Report and Literature Review. Neurosurgery 2020; 87:E152-E155. [PMID: 31555806 DOI: 10.1093/neuros/nyz361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/01/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND AND IMPORTANCE Primary tumors of the brachial plexus are rare. Most are benign and characterized as Schwannoma and neurofibroma, whereas malignant peripheral nerve sheath tumors are less common. Here, we report a rare case of primary embryonal carcinoma in the brachial plexus. CLINICAL PRESENTATION A 17-yr-old male presented with a 3-mo history of a mass growing in the left supraclavicular region over the middle part of the clavicle. Magnetic resonance imaging revealed a well-defined mass (diameter 2.5 cm) straddling the brachial plexus. After surgical resection, and the mass was histologically confirmed to be an embryonal carcinoma. CONCLUSION Primary embryonal carcinoma in the brachial plexus has not been reported previously. This case highlights the importance of considering the possibility that some primary brachial plexus tumors may be malignant and should be treated promptly.
Collapse
Affiliation(s)
- Huihao Chen
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Gang Yin
- Department of Orthopedic Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Na Cui
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Haodong Lin
- Department of Orthopedic Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Babakhanzadeh E, Nazari M, Ghasemifar S, Khodadadian A. Some of the Factors Involved in Male Infertility: A Prospective Review. Int J Gen Med 2020; 13:29-41. [PMID: 32104049 PMCID: PMC7008178 DOI: 10.2147/ijgm.s241099] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/23/2020] [Indexed: 01/04/2023] Open
Abstract
Infertility is defined as the inability of couples to have a baby after one year of regular unprotected intercourse, affecting 10 to 15% of couples. According to the latest WHO statistics, approximately 50-80 million people worldwide sufer from infertility, and male factors are responsible for approximately 20-30% of all infertility cases. The diagnosis of infertility in men is mainly based on semen analysis. The main parameters of semen include: concentration, appearance and motility of sperm. Causes of infertility in men include a variety of things including hormonal disorders, physical problems, lifestyle problems, psychological issues, sex problems, chromosomal abnormalities and single-gene defects. Despite numerous efforts by researchers to identify the underlying causes of male infertility, about 70% of cases remain unknown. These statistics show a lack of understanding of the mechanisms involved in male infertility. This article focuses on the histology of testicular tissue samples, the male reproductive structure, factors affecting male infertility, strategies available to find genes involved in infertility, existing therapeutic methods for male infertility, and sperm recovery in infertile men.
Collapse
Affiliation(s)
- Emad Babakhanzadeh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Nazari
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sina Ghasemifar
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Khodadadian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
20
|
Abstract
Human germ cell tumours (GCTs) are derived from stem cells of the early embryo and the germ line. They occur in the gonads (ovaries and testes) and also in extragonadal sites, where migrating primordial germ cells are located during embryogenesis. This group of heterogeneous neoplasms is unique in that their developmental potential is in effect determined by the latent potency state of their cells of origin, which are reprogrammed to omnipotent, totipotent or pluripotent stem cells. Seven GCT types, defined according to their developmental potential, have been identified, each with distinct epidemiological and (epi)genomic features. Heritable predisposition factors affecting the cells of origin and their niches likely explain bilateral, multiple and familial occurrences of the different types of GCTs. Unlike most other tumour types, GCTs are rarely caused by somatic driver mutations, but arise through failure to control the latent developmental potential of their cells of origin, resulting in their reprogramming. Consistent with their non-mutational origin, even the malignant tumours of the group are characterized by wild-type TP53 and high sensitivity for DNA damage. However, tumour progression and the rare occurrence of treatment resistance are driven by embryonic epigenetic state, specific (sub)chromosomal imbalances and somatic mutations. Thus, recent progress in understanding GCT biology supports a comprehensive developmental pathogenetic model for the origin of all GCTs, and provides new biomarkers, as well as potential targets for treatment of resistant disease.
Collapse
Affiliation(s)
- J Wolter Oosterhuis
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands.
| | - Leendert H J Looijenga
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
21
|
Zapka P, Dörner E, Dreschmann V, Sakamato N, Kristiansen G, Calaminus G, Vokuhl C, Leuschner I, Pietsch T. Type, Frequency, and Spatial Distribution of Immune Cell Infiltrates in CNS Germinomas: Evidence for Inflammatory and Immunosuppressive Mechanisms. J Neuropathol Exp Neurol 2019; 77:119-127. [PMID: 29237087 DOI: 10.1093/jnen/nlx106] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Central nervous system germinomas are characterized by a massive immune cell infiltrate. We systematically characterized these immune cells in 28 germinomas by immunophenotyping and image analysis. mRNA expression was analyzed by Nanostring technology and in situ RNA hybridization. Tumor infiltrating lymphocytes (TILs) were composed of 61.8% ± 3.1% (mean ± SE) CD3-positive T cells, including 45.2% ± 3.5% of CD4-positive T-helper cells, 23.4% ± 1.5% of CD8-positive cytotoxic T cells, 5.5% ± 0.9% of FoxP3-positive regulatory T cells, and 11.9% ±1.3% PD-1-positive TILs. B cells accounted for 35.8% ± 2.9% of TILs and plasma cells for 9.3% ± 1.6%. Tumor-associated macrophages consisted of clusters of activated PD-L1-positive macrophages and interspersed anti-inflammatory macrophages expressing CD163. Germinoma cells did not express PD-L1. Expression of genes encoding immune cell markers and cytokines was high and comparable to mRNA levels in lymph node tissue. IFNG and IL10 mRNA was detected in subfractions of TILs and in PD-L1-positive macrophages. Taken together, the strong immune reaction observed in germinomas involves inflammatory as well as various suppressive mechanisms. Expression of PD-1 and PD-L1 and infiltration of cytotoxic T cells are biomarkers predictive of response to anti-PD-1/PD-L1 therapies, constituting a rationale for possible novel treatment approaches.
Collapse
Affiliation(s)
- Pia Zapka
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Evelyn Dörner
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Verena Dreschmann
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Noriaki Sakamato
- Department of Diagnostic Pathology/Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Glen Kristiansen
- Department of Pathology, University of Bonn Medical Center, Bonn, Germany
| | - Gabriele Calaminus
- Department of Pediatric Haematology and Oncology, University of Bonn Medical Center, Bonn, Germany
| | - Christian Vokuhl
- Pediatric Tumor Registry, Pediatric Pathology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Ivo Leuschner
- Pediatric Tumor Registry, Pediatric Pathology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Torsten Pietsch
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
22
|
Lau YFC, Li Y, Kido T. Battle of the sexes: contrasting roles of testis-specific protein Y-encoded (TSPY) and TSPX in human oncogenesis. Asian J Androl 2019; 21:260-269. [PMID: 29974883 PMCID: PMC6498724 DOI: 10.4103/aja.aja_43_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022] Open
Abstract
The Y-located testis-specific protein Y-encoded (TSPY) and its X-homologue TSPX originated from the same ancestral gene, but act as a proto-oncogene and a tumor suppressor gene, respectively. TSPY has specialized in male-specific functions, while TSPX has assumed the functions of the ancestral gene. Both TSPY and TSPX harbor a conserved SET/NAP domain, but are divergent at flanking structures. Specifically, TSPX contains a C-terminal acidic domain, absent in TSPY. They possess contrasting properties, in which TSPY and TSPX, respectively, accelerate and arrest cell proliferation, stimulate and inhibit cyclin B-CDK1 phosphorylation activities, have no effect and promote proteosomal degradation of the viral HBx oncoprotein, and exacerbate and repress androgen receptor (AR) and constitutively active AR variant, such as AR-V7, gene transactivation. The inhibitory domain has been mapped to the carboxyl acidic domain in TSPX, truncation of which results in an abbreviated TSPX exerting positive actions as TSPY. Transposition of the acidic domain to the C-terminus of TSPY results in an inhibitory protein as intact TSPX. Hence, genomic mutations/aberrant splicing events could generate TSPX proteins with truncated acidic domain and oncogenic properties as those for TSPY. Further, TSPY is upregulated by AR and AR-V7 in ligand-dependent and ligand-independent manners, respectively, suggesting the existence of a positive feedback loop between a Y-located proto-oncogene and male sex hormone/receptors, thereby amplifying the respective male oncogenic actions in human cancers and diseases. TSPX counteracts such positive feedback loop. Hence, TSPY and TSPX are homologues on the sex chromosomes that function at the two extremes of the human oncogenic spectrum.
Collapse
Affiliation(s)
- Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Yunmin Li
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| |
Collapse
|
23
|
Weissferdt A, Kalhor N, Rodriguez Canales J, Fujimoto J, Wistuba II, Moran CA. Primary Mediastinal Yolk Sac Tumors: An Immunohistochemical Analysis of 14 Cases. Appl Immunohistochem Mol Morphol 2019; 27:125-133. [DOI: 10.1097/pai.0000000000000442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Bowzyk Al-Naeeb A, Murray M, Horan G, Harris F, Kortmann RD, Nicholson J, Ajithkumar T. Current Management of Intracranial Germ Cell Tumours. Clin Oncol (R Coll Radiol) 2018; 30:204-214. [DOI: 10.1016/j.clon.2018.01.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/25/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022]
|
25
|
Schulte SL, Waha A, Steiger B, Denkhaus D, Dörner E, Calaminus G, Leuschner I, Pietsch T. CNS germinomas are characterized by global demethylation, chromosomal instability and mutational activation of the Kit-, Ras/Raf/Erk- and Akt-pathways. Oncotarget 2018; 7:55026-55042. [PMID: 27391150 PMCID: PMC5342399 DOI: 10.18632/oncotarget.10392] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/13/2016] [Indexed: 11/25/2022] Open
Abstract
CNS germinomas represent a unique germ cell tumor entity characterized by undifferentiated tumor cells and a high response rate to current treatment protocols. Limited information is available on their underlying genomic, epigenetic and biological alterations. We performed a genome-wide analysis of genomic copy number alterations in 49 CNS germinomas by molecular inversion profiling. In addition, CpG dinucleotide methylation was studied by immunohistochemistry for methylated cytosine residues. Mutational analysis was performed by resequencing of candidate genes including KIT and RAS family members. Ras/Erk and Akt pathway activation was analyzed by immunostaining with antibodies against phospho-Erk, phosho-Akt, phospho-mTOR and phospho-S6. All germinomas coexpressed Oct4 and Kit but showed an extensive global DNA demethylation compared to other tumors and normal tissues. Molecular inversion profiling showed predominant genomic instability in all tumors with a high frequency of regional gains and losses including high level gene amplifications. Activating mutations of KIT exons 11, 13, and 17 as well as a case with genomic KIT amplification and activating mutations or amplifications of RAS gene family members including KRAS, NRAS and RRAS2 indicated mutational activation of crucial signaling pathways. Co-activation of Ras/Erk and Akt pathways was present in 83% of germinomas. These data suggest that CNS germinoma cells display a demethylated nuclear DNA similar to primordial germ cells in early development. This finding has a striking coincidence with extensive genomic instability. In addition, mutational activation of Kit-, Ras/Raf/Erk- and Akt- pathways indicate the biological importance of these pathways and their components as potential targets for therapy.
Collapse
Affiliation(s)
| | - Andreas Waha
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Barbara Steiger
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Dorota Denkhaus
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Evelyn Dörner
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Gabriele Calaminus
- Department of Pediatric Hematology/Oncology, University of Bonn Medical Center, Bonn, Germany
| | - Ivo Leuschner
- Kiel Paediatric Tumor Registry, Department of Paediatric Pathology, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
26
|
Kuo YC, Au HK, Hsu JL, Wang HF, Lee CJ, Peng SW, Lai SC, Wu YC, Ho HN, Huang YH. IGF-1R Promotes Symmetric Self-Renewal and Migration of Alkaline Phosphatase + Germ Stem Cells through HIF-2α-OCT4/CXCR4 Loop under Hypoxia. Stem Cell Reports 2018; 10:524-537. [PMID: 29307582 PMCID: PMC5830933 DOI: 10.1016/j.stemcr.2017.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 12/22/2022] Open
Abstract
Hypoxia cooperates with endocrine signaling to maintain the symmetric self-renewal proliferation and migration of embryonic germline stem cells (GSCs). However, the lack of an appropriate in vitro cell model has dramatically hindered the understanding of the mechanism underlying this cooperation. Here, using a serum-free system, we demonstrated that hypoxia significantly induced the GSC mesenchymal transition, increased the expression levels of the pluripotent transcription factor OCT4 and migration-associated proteins (SDF-1, CXCR4, IGF-1, and IGF-1R), and activated the cellular expression and translocalization of the CXCR4-downstream proteins ARP3/pFAK. The underlying mechanism involved significant IGF-1/IGF-1R activation of OCT4/CXCR4 expression through HIF-2α regulation. Picropodophyllin-induced inhibition of IGF-1R phosphorylation significantly suppressed hypoxia-induced SDF-1/CXCR4 expression and cell migration. Furthermore, transactivation between IGF-1R and CXCR4 was involved. In summary, we demonstrated that niche hypoxia synergistically cooperates with its associated IGF-1R signaling to regulate the symmetric division (self-renewal proliferation) and cell migration of alkaline phosphatase-positive GSCs through HIF-2α-OCT4/CXCR4 during embryogenesis. Hypoxia regulated AP+GSC self-renewal and cell migration via IGF-1R and CXCR4 Hypoxia increased IGF1/IGF-1R and SDF-1/CXCR4 to promote AP+GSC migration Crosstalk of IGF-1/IGF-1R and SDF-1/CXCR4 signaling in AP+GSCs under hypoxia Inhibition of IGF-1R phosphorylation suppressed hypoxia-induced cell migration
Collapse
Affiliation(s)
- Yung-Che Kuo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Heng-Kien Au
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, 11031 Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Medical University Hospital, 11031 Taipei, Taiwan
| | - Jue-Liang Hsu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, 91201 Pingtung, Taiwan
| | - Hsiao-Feng Wang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Chiung-Ju Lee
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Syue-Wei Peng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Ssu-Chuan Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Yu-Chih Wu
- Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 11031 Taipei, Taiwan
| | - Hong-Nerng Ho
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, 10002 Taipei, Taiwan; Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, National Taiwan University and Hospital, 10041 Taipei, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, 11031 Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 11031 Taipei, Taiwan; Comprehensive Cancer Center of Taipei Medical University, 10031 Taipei, Taiwan; The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 10031 Taipei, Taiwan.
| |
Collapse
|
27
|
Nagasawa DT, Lagman C, Sun M, Yew A, Chung LK, Lee SJ, Bui TT, Ooi YC, Robison RA, Zada G, Yang I. Pineal germ cell tumors: Two cases with review of histopathologies and biomarkers. J Clin Neurosci 2017; 38:23-31. [PMID: 28189312 DOI: 10.1016/j.jocn.2016.12.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/27/2016] [Indexed: 02/08/2023]
Abstract
Pineal germ cell tumors (GCTs) are primarily seen in pediatric and Asian populations. These tumors are divided into germinomatous and non-germinomatous GCTs (NGGCTs). GCTs are thought to arise by misplacement of totipotent stem cells en route to gonads during embryogenesis. Intracranial GCTs display an affinity to develop along the pineal-suprasellar axis and have variable manifestations dependent upon the location of the tumor. Management and outcomes are driven by histopathologies. In this study, we highlight two cases of pineal GCTs and present a review of the literature with an emphasis on histopathologies and biomarkers.
Collapse
Affiliation(s)
- Daniel T Nagasawa
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Carlito Lagman
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Michael Sun
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Andrew Yew
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lawrance K Chung
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Seung J Lee
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Timothy T Bui
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yinn Cher Ooi
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - R Aaron Robison
- Department of Neurosurgery, Keck School of Medicine of the University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Gabriel Zada
- Department of Neurosurgery, Keck School of Medicine of the University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Isaac Yang
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States; Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States; Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
28
|
Mandal GK, Das I, Paul R, Mustafi SM. Primary central nervous system teratoma with sarcomatous transformation in a young girl: Report of a rare case. Asian J Neurosurg 2016; 11:458. [PMID: 27695574 PMCID: PMC4974995 DOI: 10.4103/1793-5482.181117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 13-year-old girl presented with chief complaints of severe headache and vomiting followed by hemiparesis. Radiological examination suggested a space occupying lesion in the right parietal lobe. Craniotomy and debulking of the tumor mass were done. Histopathological and subsequent immunohistochemical examination showed a tumor composed of fascicle of atypical spindle cells which revealed reactivity to vimentin with interspersed areas of well-differentiated cartilage tissue. Hence, the diagnosis of teratoma with sarcomatous transformation was given. Detailed discussion including review of literature has been made regarding different aspect of the tumor.
Collapse
Affiliation(s)
- Gautam Kumar Mandal
- Department of Pathology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Indranil Das
- Department of Pathology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Rahul Paul
- Department of Pathology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Saunak Mitra Mustafi
- Department of Pathology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
29
|
Li B, Jiang Q, Zhang S, Zhou Y, Zhang QF, OuYang L. Primary yolk sac tumor of the gluteus: a case report and literature review. Onco Targets Ther 2016; 9:4715-9. [PMID: 27536133 PMCID: PMC4976902 DOI: 10.2147/ott.s106924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Yolk sac tumor (YST) is a common malignant primitive germ cell tumor that often exhibits differentiation into endodermal structures. They most commonly occur in childhood and adolescence and are rare after the age of 40 years. Derived from the yolk sac during the embryonic period, YSTs can occur in the gonads and germ cells because the tumor cells migrate from the yolk sac toward the gonads. Here, we present a rare case of primary gluteus YST in a 3-year-old girl. She received BEP chemotherapy (bleomycin + etoposide + cisplatin) after surgical resection. There was no evidence of recurrence 7 months after primary treatment.
Collapse
Affiliation(s)
- Bo Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang
| | - Qianqian Jiang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang
| | - Shitai Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang
| | - Qing-Fu Zhang
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Ling OuYang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang
| |
Collapse
|
30
|
Abstract
CNS germ cell tumors (GCT) are rare tumors that arise in midline brain regions (mostly pineal or suprasellar). They are of two types, germinoma and nongerminomatous GCT (NGGCT) which include teratoma, choriocarcinoma, yolk sac, embyronal carcinoma and mixed GCT. Tissue is needed for diagnosis unless serum or cerebrospinal fluid markers, b-HCG or AFP, are elevated. Germinomas can be cured with radiation therapy (RT) alone (whole ventricle fields, if localized), but chemotherapy may permit RT dose-reduction. Best outcomes for NGGCT are with RT and chemotherapy. Craniospinal RT is needed for all disseminated tumors and best survival for localized NGGCT has included craniospinal RT. Recent genetic findings in CNS GCT may lead to therapies targeting their oncogenic pathways.
Collapse
Affiliation(s)
- Patricia L Robertson
- Departments of Pediatrics & Neurology, University of Michigan Medical School, Pediatric Neuro-Oncology, 12-718 C.S. Mott Children’s Hospital, 1540 E. Hospital Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
ZHAO XUANZHONG, WANG FENG, HOU MINGXING. Expression of stem cell markers nanog and PSCA in gastric cancer and its significance. Oncol Lett 2016; 11:442-448. [PMID: 26870231 PMCID: PMC4727111 DOI: 10.3892/ol.2015.3884] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 07/16/2015] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to determine the expression of stem cell markers Nanog compared with PSCA in gastric cancer tissues and adjacent normal tissues, and to investigate the association between tumor stem cells and initiation, progression, metastasis, and prognosis of gastric cancer. One hundred chemotherapy- and radiotherapy-naive patients with pathologically confirmed gastric cancer were enrolled from the General Surgery Department and Surgical Oncology Department of the Affiliated Hospital of Inner Mongolia Medical University (Hohhot, P.R. China), between October 2011 and June 2013. Surgically resected specimens of cancer tissues and adjacent normal tissues (>5 cm from the boundary of cancerous component) were collected. The mRNA expression levels of Nanog and PSCA in those tissues was determined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The correlation between the expression of stem cell markers Nanog and PSCA in gastric cancer tissues and clinicopathological factors was analyzed. The qPCR results demonstrated that the relative expression of Nanog was increased in gastric cancer tissues compared with in the adjacent tissues (P<0.05); and relative expression of PSCA was reduced in gastric cancer tissues compared with adjacent tissues (P<0.05). The expression of Nanog and PSCA in gastric cancer tissues was associated with tumor differentiation. The expression of Nanog was increased in poorly-differentiated and undifferentiated tumors compared with moderately- and well-differentiated tumors (P<0.05). The expression of PSCA was reduced in poorly differentiated and undifferentiated tumors compared with moderately- and well-differentiated tumors (P<0.05). However, the expression of Nanog and PSCA was not associated with age, gender, tumor size, TNM stage, depth of invasion, or lymph node metastasis. Therefore, Nanog and PSCA may have potential as molecular markers to reflect the differentiation status of gastric cancer.
Collapse
Affiliation(s)
- XUANZHONG ZHAO
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| | - FENG WANG
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| | - MINGXING HOU
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| |
Collapse
|
32
|
Yu B, Cai H, Xu Z, Xu T, Zou Q, Gu M. Expressions of stem cell transcription factors Nanog and Oct4 in renal cell carcinoma tissues and clinical significance. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1818-1823. [PMID: 26631537 DOI: 10.3109/21691401.2015.1105238] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We aimed to detect the expressions of stem cell transcription factors Nanog and Oct4 in renal cell carcinoma (RCC) tissues. Nanog and Oct4 mRNA expressions in RCC tissues significantly exceeded those in paracancerous tissues (p < 0.01 and p < 0.05), being positively correlated with histological grade (p < 0.01 and p < 0.05) and TNM stage (p < 0.05). With increasing TNM stage (p < 0.01) and lymphatic metastasis (p < 0.05), the positive expression rate of Nanog protein increased. RCC patients with low Nanog and Oct4 expressions in tumor tissues had significantly higher survival rates (p < 0.05). High Nanog and Oct4 expressions may be potential therapeutic targets.
Collapse
Affiliation(s)
- Bin Yu
- a Department of Urology , Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University , Nanjing , China
| | - Hongzhou Cai
- a Department of Urology , Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University , Nanjing , China
| | - Zicheng Xu
- a Department of Urology , Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University , Nanjing , China
| | - Ting Xu
- a Department of Urology , Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University , Nanjing , China
| | - Qing Zou
- a Department of Urology , Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University , Nanjing , China
| | - Min Gu
- b Department of Urology , First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| |
Collapse
|
33
|
Abstract
The transcription factor OCT4 is an established diagnostic marker for central nervous system (CNS) germinoma. However, no data are available to date concerning the expression of its downstream target undifferentiated embryonic cell transcription factor 1 (UTF1) in CNS germ cell tumours. We examined 21 CNS germinomas and two mixed CNS germ cell tumours for UTF1 and the post-transcriptional regulator LIN28 immunohistochemical expression. We compared the profile to established diagnostic germinoma markers and to the expression in six testicular and four metastatic germ cell tumours as well as 150 CNS tumours of various backgrounds. We found UTF1 expression in 23 of 23 and LIN28 in 20 of 23 CNS germ cell tumours. The established germinoma markers cKIT (23/23), OCT4 (21/23) and placental alkaline phosphatase (PLAP) (19/21) were also frequently expressed in our cohort. In terms of signal intensity and frequency, UTF1 showed similar results as cKIT but staining was superior to OCT4, PLAP and LIN28. OCT4 was absent in all CNS metastases and haemangioblastomas, while UTF1 was weakly observed in two metastases.With a sensitivity of 100% and a specificity of 97% in the detection of CNS germinomas, UTF1 serves as a new reliable alternative in the diagnostic setting of CNS germ cell tumours.
Collapse
|
34
|
Weissferdt A, Rodriguez-Canales J, Liu H, Fujimoto J, Wistuba II, Moran CA. Primary mediastinal seminomas: a comprehensive immunohistochemical study with a focus on novel markers. Hum Pathol 2015; 46:376-83. [PMID: 25576290 DOI: 10.1016/j.humpath.2014.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/05/2014] [Accepted: 11/12/2014] [Indexed: 02/03/2023]
Abstract
Primary mediastinal seminomas are unusual tumors that can present in a pure form or as part of a mixed germ cell tumor. Contrary to testicular seminomas, little is known about the expression of novel immunohistochemical markers in mediastinal seminomas. This study investigates the immunohistochemical features of these tumors with a focus on novel markers. Thirty-two cases of primary mediastinal seminomas were reviewed; and representative whole-tissue sections were selected for immunohistochemical studies using antibodies directed against high molecular weight cytokeratin 5/6 (CK5/6), low molecular weight cytokeratin (CAM5.2), octamer-binding transcription factor 3/4 (OCT3/4), spalt-like transcription factor 4 (SALL4), GATA binding protein 3 (GATA-3), sry-related HMG box 2 (SOX2), SOX17, human T cell leukemia/lymphoma 1 (TCL1), glypican 3, melanoma associated antigen C2 (MAGEC2), and paired box gene 8 (Pax8). The percentage of positive tumor cells as well as the intensity of staining was evaluated and scored. Thirty-one cases (97%) expressed SOX17, whereas 29 cases (91%) were positive for OCT3/4 and SALL4, respectively. Twenty-eight cases (88%) expressed MAGEC2 and CAM5.2, respectively. Two cases (6%) were positive for Pax8, and a single case (3%) was positive for TCL1. None of the cases stained with CK5/6, GATA-3, SOX2, or glypican 3. Similar to testicular seminomas, mediastinal seminomas show consistent expression of OCT3/4, SALL4, SOX17, and MAGEC2 and are negative for SOX2, glypican 3, GATA-3, and CK5/6. Pax8 positivity is only inconsistently identified in mediastinal seminomas. Contrary to their testicular counterparts, mediastinal tumors show diffuse expression of low-molecular-weight cytokeratin in up to 90% of cases and are commonly negative for TCL1. Although there is some immunohistochemical overlap between testicular and mediastinal seminomas, considerable differences also exist and should be acknowledged when dealing with these tumors.
Collapse
Affiliation(s)
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030
| | - Hui Liu
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030
| | - Cesar A Moran
- Department of Pathology, MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
35
|
Bartkova J, Hoei-Hansen CE, Krizova K, Hamerlik P, Skakkebæk NE, Rajpert-De Meyts E, Bartek J. Patterns of DNA damage response in intracranial germ cell tumors versus glioblastomas reflect cell of origin rather than brain environment: implications for the anti-tumor barrier concept and treatment. Mol Oncol 2014; 8:1667-78. [PMID: 25066726 DOI: 10.1016/j.molonc.2014.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 06/24/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022] Open
Abstract
The DNA damage response (DDR) machinery becomes commonly activated in response to oncogenes and during early stages of development of solid malignancies, with an exception of testicular germ cell tumors (TGCTs). The active DDR signaling evokes cell death or senescence but this anti-tumor barrier can be breached by defects in DDR factors, such as the ATM-Chk2-p53 pathway, thereby allowing tumor progression. The DDR barrier is strongly activated in brain tumors, particularly gliomas, due to oxidative damage and replication stress. Here, we took advantage of rare human primary intracranial germ cell tumors (PIGCTs), to address the roles of cell-intrinsic factors including cell of origin, versus local tissue environment, in the constitutive DDR activation in vivo. Immunohistochemical analysis of 7 biomarkers on a series of 21 PIGCTs (germinomas and other subtypes), 20 normal brain specimens and 20 glioblastomas, revealed the following: i) The overall DDR signaling (γH2AX) and activation of the ATM-Chk2-p53 pathway were very low among the PIGCTs, reminiscent of TGCTs, and contrasting sharply with strong DDR activation in glioblastomas; ii) Except for one case of embryonal carcinoma, there were no clear aberrations in the ATM-Chk2-p53 pathway components among the PIGCT cohort; iii) Subsets of PIGCTs showed unusual cytosolic localization of Chk2 and/or ATM. Collectively, these results show that PIGCTs mimic the DDR activation patterns of their gonadal germ cell tumor counterparts, rather than the brain tumors with which they share the tissue environment. Hence cell-intrinsic factors and cell of origin dictate the extent of DDR barrier activation and also the ensuing pressure to select for DDR defects. Our data provide conceptually important insights into the role of DNA damage checkpoints in intracranial tumorigenesis, with implications for the differential biological responses of diverse tumor types to endogenous stress as well as to genotoxic treatments such as ionizing radiation or chemotherapy.
Collapse
Affiliation(s)
| | - Christina E Hoei-Hansen
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics, Rigshospitalet, Copenhagen, Denmark
| | - Katerina Krizova
- Danish Cancer Society Research Center, Copenhagen, Denmark; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petra Hamerlik
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Niels E Skakkebæk
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- University Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark.
| | - Jiri Bartek
- Danish Cancer Society Research Center, Copenhagen, Denmark; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
36
|
Huang YH, Lin MH, Wang PC, Wu YC, Chiang HL, Wang YL, Chang JH, Huang YK, Gu SY, Ho HN, Ling TY. Hypoxia inducible factor 2α/insulin-like growth factor receptor signal loop supports the proliferation and Oct-4 maintenance of mouse germline stem cells. Mol Hum Reprod 2014; 20:526-37. [PMID: 24598112 DOI: 10.1093/molehr/gau016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hypoxia inducible factor 2α (HIF-2α) is critical for primordial germ cell (PGC) survival as knockout of HIF-2α (HIF-2α(-/-)) decreases both expression of Oct-4 and PGC number in genital ridge. Hypoxia is known to stabilize HIF-2α protein from proteasomal degradation. However, little is known about the hypoxia-associated endocrinal signaling in HIF-2α expression. The current work demonstrates a role for an endocrine insulin-like growth factor-I receptor (IGF-IR)-PI3K/Akt-mTOR-HIF-2α regulatory loop in the proliferation and Oct-4 maintenance of PGC-like alkaline phosphatase positive mouse germline stem cells (AP(+)GSCs). We found that hypoxia greatly increased the cell proliferation and the levels of nuclear Oct-4/HIF-2α protein of AP(+)GSCs. The hypoxic-AP(+)GSCs presented stronger stemness ability for germ cell differentiation than normoxic, with expressions of c-KIT (differentiation germ cell marker), VASA (differentiation germ cell marker) and SCP3 (meiotic marker) using a renal capsule transplantation assay. Meanwhile, hypoxia significantly increased the expression levels of secreted-IGF-I and IGF-IR. The IGF-I dose dependently increased the HIF-2α expression levels in AP(+)GSCs; and, the inhibition of IGF-IR by RNA interference (shIGF-IR) or LY294002 (PI3K inhibitor)/Rapamycin (mTOR inhibitor) effectively suppressed the IGF-I- and/or hypoxia-induced HIF-2α and Oct-4 expression, suggesting that the IGF-IR and its downstream Akt/mTOR signaling are involved in the IGF-I/hypoxia effects. Additionally, knockdown of HIF-2α dramatically suppressed Oct-4 and IGF-IR protein levels in AP(+)GSC cells. In conclusion, the present study demonstrates a regulatory loop of IGF-IR-PI3K/Akt-mTOR-HIF-2α in proliferation and Oct-4 maintenance of PGC-like AP(+)GSCs under hypoxia. This finding provides insights into the niche endocrinology underlying early germ cell development.
Collapse
Affiliation(s)
- Y-H Huang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan Center for Reproductive Medicine and Sciences, Taipei Medical University Hospital, Taipei, Taiwan
| | - M-H Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - P-C Wang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-C Wu
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - H-L Chiang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-L Wang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - J-H Chang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-K Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| | - S-Y Gu
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| | - H-N Ho
- Graduate Institute of Clinical Genomics, College of Medicine, National Taiwan University, Taipei, Taiwan Division of Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei Medical University, Taipei, Taiwan
| | - T-Y Ling
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| |
Collapse
|
37
|
Mutually exclusive mutations of KIT and RAS are associated with KIT mRNA expression and chromosomal instability in primary intracranial pure germinomas. Acta Neuropathol 2014; 127:911-25. [PMID: 24452629 DOI: 10.1007/s00401-014-1247-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/14/2014] [Indexed: 01/13/2023]
Abstract
Intracranial germ cell tumors (iGCTs) are the second most common brain tumors among children under 15 in Japan. The pathogenesis of iGCTs is largely unexplored. Although a subset of iGCTs is known to have KIT mutation, its impact on the biology and patients' survival has not been established. In this study, we investigated genes involved in the KIT signaling pathway. 65 iGCTs (30 pure germinomas, 14 teratomas, 18 mixed GCTs, 2 yolk sac tumors, 1 choriocarcinoma) were screened for mutation of KIT, KRAS, NRAS, HRAS, BRAF, PDGFRA, and IDH1 by direct sequencing. KIT expression was examined by immunohistochemistry and quantitative PCR. Chromosomal status was analyzed by array-comparative genomic hybridization (aCGH). Somatic mutations were detected only in KIT and RAS, which were frequently observed in pure germinomas (60.0 %), but rare in non-germinomatous GCTs (NGGCTs) (8.6 %). All KIT/RAS mutations were mutually exclusive. Regardless of the mutation status or mRNA expression, the KIT protein was expressed in all germinomas, while only in 54.3 % of NGGCTs. Amplification of KIT was found in one pure germinoma by aCGH. In pure germinomas, high expression of KIT mRNA was associated with the presence of KIT/RAS alterations and severe chromosomal instability. Our results indicate that alterations of the KIT signaling pathway play an important role in the development of germinomas. Pure germinomas may develop through two distinct pathogeneses: one with KIT/RAS alterations, elevated KIT mRNA expression and severe chromosomal instability, and the other through yet an unidentified mechanism without any of the above abnormalities.
Collapse
|
38
|
Cao J, Li L, Chen C, Lv C, Meng F, Zeng L, Li Z, Wu Q, Zhao K, Pan B, Cheng H, Chen W, Xu K. RNA interference-mediated silencing of NANOG leads to reduced proliferation and self-renewal, cell cycle arrest and apoptosis in T-cell acute lymphoblastic leukemia cells via the p53 signaling pathway. Leuk Res 2013; 37:1170-7. [PMID: 23683786 DOI: 10.1016/j.leukres.2013.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 04/13/2013] [Accepted: 04/18/2013] [Indexed: 01/23/2023]
Abstract
NANOG is critical for maintaining the self-renewal and proliferative properties of embryonic stem cells. Here we found that cultured T-cell acute lymphoblastic leukemia (T-ALL) cells, as well as human primary T-ALL cells, express a functional variant of NANOG. NANOG mRNA is derived predominantly from a retrogene locus termed NANOGP8. Furthermore, we showed that RNA interference-mediated NANOG knockdown inhibited cell proliferation, reduced self-renewal, promoted apoptosis and arrested the cell cycle through a p53-mediated pathway in leukemic cells. These findings demonstrate the oncogenic potential of this pluripotent gene in human T-ALL cells.
Collapse
Affiliation(s)
- Jiang Cao
- Department of Hematology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Taniguchi M, Nishihara M, Sasayama T, Takahashi Y, Kohmura E. A rapidly expanding immature teratoma originating from a neurohypophyseal germinoma. Neuropathol Appl Neurobiol 2013; 39:445-8. [DOI: 10.1111/nan.12000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 10/22/2012] [Indexed: 11/30/2022]
Affiliation(s)
- M. Taniguchi
- Department of Neurosurgery; Kobe University Graduate School of Medicine; Chuo-ku; Japan
| | - M. Nishihara
- Department of Neurosurgery; Nishi-Kobe Medical Center; Nishi-ku; Kobe; Hyogo; Japan
| | - T. Sasayama
- Department of Neurosurgery; Kobe University Graduate School of Medicine; Chuo-ku; Japan
| | - Y. Takahashi
- Division of Diabetes, Metabolism and Endocrinology; Department of Internal Medicine; Kobe University Graduate School of Medicine; Chuo-ku; Japan
| | - E. Kohmura
- Department of Neurosurgery; Kobe University Graduate School of Medicine; Chuo-ku; Japan
| |
Collapse
|
40
|
Abstract
Background: Primary intracranial germ cell tumors are rare (ICGCTs) and usually localized in the pineal and suprasellar regions of the brain. They are divided into histologic types: Germinoma, teratoma choriocarcinoma, embryonal carcinoma, yolk sac tumor, and malignant mixed germ cell tumors (MMGCTs). Neuroimaging evaluation is useful to distinguish between the types of ICGCTs. Germinoma is highly sensitive to radiotherapy or/and chemotherapy, and is potentially curable without surgery. MMGCTs are treated with the combination of chemotherapy and radiation, with a poorer prognosis. ICGCTs warrant correct pre-operative diagnosis in order to decide on appropriate management. Aim: To report the clinicopathological and immunohistochemical findings in three cases of primary intracranial germ-cell tumor. Materials and Methods: Three cases of intracranial germ cell tumors inclusive of both genders and all brain regions were retrieved from the archives of the Anatomical Pathology Department at King Abdul Aziz University Hospital, Jeddah between the years, 1995-2011, through a computerized search. Results: Based on histological examination, they were respectively diagnosed as MMGCTs (two cases) and germinoma. Clinical, radiological, pathological characteristics and immunohistochemical profile of the three ICGCTs are presented along with the review of recent literature. Conclusion: ICGCTs are rare tumors affecting males more than females, and at the end of three years follow-up in the present study, treatment morbidity appears to be low with no recurrence observed among surviving patients, indicating that suprasellar and basal ganglia ICGCTs may have a favorable prognosis regardless of histological type. Pineal MMGCTs may have an aggressive course.
Collapse
Affiliation(s)
- Shagufta Tahir Mufti
- Department of Anatomic Pathology, Faculty of Medicine, King Abdulaziz University and Hospital, Jeddah, Kingdom of Saudi Arabia
| | | |
Collapse
|
41
|
Tan C, Scotting PJ. Stem cell research points the way to the cell of origin for intracranial germ cell tumours. J Pathol 2012; 229:4-11. [DOI: 10.1002/path.4098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 08/15/2012] [Accepted: 08/22/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Chris Tan
- Children's Brain Tumour Research Centre, School of Biology; University of Nottingham, Queen's Medical Centre; Nottingham UK
| | - Paul J Scotting
- Children's Brain Tumour Research Centre, School of Biology; University of Nottingham, Queen's Medical Centre; Nottingham UK
| |
Collapse
|
42
|
Haskins WE, Eedala S, Jadhav YA, Labhan MS, Pericherla VC, Perlman EJ. Insights on neoplastic stem cells from gel-based proteomics of childhood germ cell tumors. Pediatr Blood Cancer 2012; 58:722-8. [PMID: 21793190 PMCID: PMC3204330 DOI: 10.1002/pbc.23282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 06/22/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Childhood germ cell tumors (cGCTs), believed to arise from transformed primordial germ cells by an unknown mechanism, provide a unique model system for investigating cell signaling, pluripotency, and the microenvironment of neoplastic stem cells (NSCs) in vivo. This is the first report of proteomics of cGCTs. PROCEDURE Four dysgerminomas (DYSs) and four childhood endodermal sinus tumors (cESTs), resembling self-renewing and differentiating NSCs, respectively, were selected. Proteomic studies were performed by 2-DE, SDS-PAGE, and cLC/MS/MS with protein database searching. RESULTS 2-DE: 9 of 941 spots were differentially regulated with greater than a twofold change in spot volume for at least three of four gels in each group. Two of nine spots had P values for the t-test analysis of comparisons less than 0.001, while the remaining spots had P values from 0.013 to 0.191. Top-ranked proteins were identified in nine of nine spots with 4.0-38% sequence coverage. APOA1, CRK, and PDIA3 were up-regulated in cESTs. TFG, TYMP, VCP, RBBP, FKBP4, and BiP were up-regulated in DYSs. SDS-PAGE: Up-regulation of NF45 and FKBP4 was observed in four of four cESTs and DYSs, respectively. The fold-changes observed correspond with characteristic genetic changes. CONCLUSION Differential regulation of FKBP4 and NF45, combined with previous research on immunosuppressant binding, suggests that glucocorticoid receptor signaling merits further investigation in cGCTs and NSCs.
Collapse
Affiliation(s)
- William E. Haskins
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Medicine, Division of Hematology & Medical Oncology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229,Correspondence: William E. Haskins, Ph.D., Dept. of Biology-BSE 3.108A, The University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249-0662, , Phone: (210)563-4492, Fax: (210)458-5658
| | - Sruthi Eedala
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Y.L. Avinash Jadhav
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Manbir S. Labhan
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Vidya C. Pericherla
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Elizabeth J. Perlman
- Department of Pathology, Northwestern University’s Feinberg School of Medicine and Robert H. Lurie Cancer Center, Chicago, IL, 60614
| |
Collapse
|
43
|
Implication of expression of Nanog in prostate cancer cells and their stem cells. ACTA ACUST UNITED AC 2012; 32:242-246. [DOI: 10.1007/s11596-012-0043-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Indexed: 12/29/2022]
|
44
|
Jorsal T, Rørth M. Intracranial germ cell tumours. A review with special reference to endocrine manifestations. Acta Oncol 2012; 51:3-9. [PMID: 22150165 DOI: 10.3109/0284186x.2011.586000] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
EPIDEMIOLOGY Intracranial germ cell tumours (icGCTs) represent 3-15% of primary paediatric intracranial neoplasms with a considerable geographical variation in incidence. Ninety percent of patients diagnosed with icGCTs are under 20 years of age. PATHOLOGY Histologic characteristics and investigation of the tumour markers β-human chorionic gonadotropin (β-hCG) and alpha-fetoprotein (AFP) help define the different categories of icGCTs. The tumours are divided into two major groups called germinomas and non-germinomatous GCTs (NGGCTs). CLINICAL PRESENTATION The clinical symptoms depend on the size and location of tumour in the brain, which is most commonly in the pineal or suprasellar region. Pineal GCTs often present with neurological symptoms because of their tendency to cause increased intracranial pressure. Suprasellar GCTs are often accompanied by endocrine abnormalities such as diabetes insipidus (DI), growth retardation and precocious or delayed puberty. DIAGNOSIS A combination of clinical findings, endocrine and tumour marker evaluation, spinal fluid cytology, magnetic resonance imaging (MRI) and biopsy helps verifying the diagnosis of an icGCT. A summary of published data (n = 97) revealed that >90% of patients at diagnosis had at least one endocrine abnormality, DI being the most common (>80%). TREATMENT Classification of tumour is important for choice of treatment and for prognosis. A combination of chemotherapy and radiotherapy is often used, since most icGCTs have a great sensitivity to these treatment modalities. CONCLUSION Endocrine symptoms are very frequently appearing in patients with icGCTs and they can present long before neuroimaging verification of tumour is possible. It is of the outmost importance to have the diagnosis of icGCTs in mind when children, adolescents and young adults are presenting with endocrine irregularities, because most icGCTs are very sensitive to radiotherapy and chemotherapy, and early onset of treatment is important in order to minimize morbidity and mortality.
Collapse
Affiliation(s)
- Tina Jorsal
- Department of Oncology, Rigshospitalet, Denmark.
| | | |
Collapse
|
45
|
Schneider DT, Terenziani M, Cecchetto G, Olson TA. Gonadal and Extragonadal Germ Cell Tumors, Sex Cord Stromal and Rare Gonadal Tumors. RARE TUMORS IN CHILDREN AND ADOLESCENTS 2012. [DOI: 10.1007/978-3-642-04197-6_39] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
46
|
Hypoxia impairs primordial germ cell migration in zebrafish (Danio rerio) embryos. PLoS One 2011; 6:e24540. [PMID: 21931746 PMCID: PMC3169607 DOI: 10.1371/journal.pone.0024540] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 08/12/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND As a global environmental concern, hypoxia is known to be associated with many biological and physiological impairments in aquatic ecosystems. Previous studies have mainly focused on the effect of hypoxia in adult animals. However, the effect of hypoxia and the underlying mechanism of how hypoxia affects embryonic development of aquatic animals remain unclear. METHODOLOGY/PRINCIPAL FINDINGS In the current study, the effect of hypoxia on primordial germ cell (PGC) migration in zebrafish embryos was investigated. Hypoxic embryos showed PGC migration defect as indicated by the presence of mis-migrated ectopic PGCs. Insulin-like growth factor (IGF) signaling is required for embryonic germ line development. Using real-time PCR, we found that the mRNA expression levels of insulin-like growth factor binding protein (IGFBP-1), an inhibitor of IGF bioactivity, were significantly increased in hypoxic embryos. Morpholino knockdown of IGFBP-1 rescued the PGC migration defect phenotype in hypoxic embryos, suggesting the role of IGFBP-1 in inducing PGC mis-migration. CONCLUSIONS/SIGNIFICANCE This study provides novel evidence that hypoxia disrupts PGC migration during embryonic development in fish. IGF signaling is shown to be one of the possible mechanisms for the causal link between hypoxia and PGC migration. We propose that hypoxia causes PGC migration defect by inhibiting IGF signaling through the induction of IGFBP-1.
Collapse
|
47
|
HESRG: a novel biomarker for intracranial germinoma and embryonal carcinoma. J Neurooncol 2011; 106:251-9. [PMID: 21861197 DOI: 10.1007/s11060-011-0673-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/29/2011] [Indexed: 12/16/2022]
Abstract
The novel stem cell-related gene, HESRG, was first identified by our group, and its expression pattern in human tumors remains unknown. In this study, we used RT-PCR to systematically investigate the expression of HESRG in various types of intracranial tumors and found that HESRG was expressed only in germinoma and embryonal carcinoma, but hardly at all in other types of brain tumors. Real-time PCR results further confirmed this expression pattern. Subsequently, we tested 134 intracranial non-germ cell tumors and 64 intracranial germ cell tumors by immunohistochemistry. Our results showed that HESRG was expressed strongly and diffusively in the nuclei of tumor cells in intracranial germinoma and embryonal carcinoma as well as in human embryonic stem cells. No positive staining signal was observed in any other type of intracranial tumors. In germinomas, 25 of 31 showed intensive (3+) expression, four cases showed moderate (2+) immunostaining and the remaining 2 cases showed weak (1+) immunostaining. In embryonal carcinoma, 6 of 9 showed intensive (3+) immunostaining and 3 of 9 showed moderate (2+) immunostaining. These results suggest that HESRG is a novel, sensitive and specific biomarker for intracranial germinoma and embryonal carcinoma.
Collapse
|
48
|
Kido T, Schubert S, Schmidtke J, Chris Lau YF. Expression of the human TSPY gene in the brains of transgenic mice suggests a potential role of this Y chromosome gene in neural functions. J Genet Genomics 2011; 38:181-91. [PMID: 21621739 DOI: 10.1016/j.jgg.2011.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 03/09/2011] [Accepted: 03/28/2011] [Indexed: 01/13/2023]
Abstract
The testis specific protein Y-encoded (TSPY) is a member of TSPY/SET/NAP1 superfamily, encoded within the gonadoblastoma locus on the Y chromosome. TSPY shares a highly conserved SET/NAP-domain responsible for protein--protein interaction among TSPY/SET/NAP1 proteins. Accumulating data, so far, support the role of TSPY as the gonadoblastoma gene, involved in germ cell tumorigenesis. The X-chromosome homolog of TSPY, TSPX is expressed in various tissues at both fetal and adult stages, including the brain, and is capable of interacting with the multi-domain adapter protein CASK, thereby influencing the synaptic and transcriptional functions and developmental regulation of CASK in the brain and other neural tissues. Similar to TSPX, we demonstrated that TSPY could interact with CASK at its SET/NAP-domain in cultured cells. Transgenic mice harboring a human TSPY gene and flanking sequences showed specific expression of the human TSPY transgene in both testis and brain. The neural expression pattern of the human TSPY gene overlapped with those of the endogenous mouse Cask and Tspx gene. Similarly with TSPX, TSPY was co-localized with CASK in neuronal axon fibers in the brain, suggesting a potential role(s) of TSPY in development and/or physiology of the nervous system.
Collapse
Affiliation(s)
- Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center, and Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | | | | | | |
Collapse
|
49
|
Abstract
The gonadoblastoma locus on the human Y chromosome (GBY) is postulated to serve normal functions in spermatogenesis, but could exert oncogenic properties in predisposing susceptible germ cells to tumorigenesis in incompatible niches such as streaked gonads in XY sex reversed patients or dysfunctional testis in males. The testis-specific protein Y-linked (TSPY) repeat gene has recently been demonstrated to be the putative gene for GBY, based on its location on the GBY critical region, expression patterns in early and late stages of gonadoblastoma and ability to induce gonadoblastoma-like structures in the ovaries of transgenic female mice. Over-expression of TSPY accelerates G(2)/M progression in the cell cycle by enhancing the mitotic cyclin B-CDK1 kinase activities. Currently the normal functions of TSPY in spermatogenesis are uncertain. Expression studies of TSPY, and its X-homologue, TSPX, in normal human testis suggest that TSPY is co-expressed with cyclin B1 in spermatogonia and various stages of spermatocytes while TSPX is principally expressed in Sertoli cells in the human testis. The co-expression pattern of TSPY and cyclin B1 in spermatogonia and spermatocytes suggest respectively that 1) TSPY is important for male spermatogonial cell replication and renewal in the testis; and 2) TSPY could be a catalyst/meiotic factor essential for augmenting the activities of cyclin B-cyclin dependent kinases, important for the differentiation of the spermatocytes in prophase I and in preparation for consecutive rounds of meiotic divisions without an intermediate interphase during spermatogenesis.
Collapse
Affiliation(s)
- Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, Veterans Affairs Medical Center, University of California, San Francisco, CA 94121, USA.
| | | | | |
Collapse
|
50
|
Blum R, Gupta R, Burger PE, Ontiveros CS, Salm SN, Xiong X, Kamb A, Wesche H, Marshall L, Cutler G, Wang X, Zavadil J, Moscatelli D, Wilson EL. Molecular signatures of the primitive prostate stem cell niche reveal novel mesenchymal-epithelial signaling pathways. PLoS One 2010; 5:e13024. [PMID: 20941365 PMCID: PMC2948007 DOI: 10.1371/journal.pone.0013024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 08/05/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Signals between stem cells and stroma are important in establishing the stem cell niche. However, very little is known about the regulation of any mammalian stem cell niche as pure isolates of stem cells and their adjacent mesenchyme are not readily available. The prostate offers a unique model to study signals between stem cells and their adjacent stroma as in the embryonic prostate stem cell niche, the urogenital sinus mesenchyme is easily separated from the epithelial stem cells. Here we investigate the distinctive molecular signals of these two stem cell compartments in a mammalian system. METHODOLOGY/PRINCIPAL FINDINGS We isolated fetal murine urogenital sinus epithelium and urogenital sinus mesenchyme and determined their differentially expressed genes. To distinguish transcripts that are shared by other developing epithelial/mesenchymal compartments from those that pertain to the prostate stem cell niche, we also determined the global gene expression of epidermis and dermis of the same embryos. Our analysis indicates that several of the key transcriptional components that are predicted to be active in the embryonic prostate stem cell niche regulate processes such as self-renewal (e.g., E2f and Ap2), lipid metabolism (e.g., Srebp1) and cell migration (e.g., Areb6 and Rreb1). Several of the enriched promoter binding motifs are shared between the prostate epithelial/mesenchymal compartments and their epidermis/dermis counterparts, indicating their likely relevance in epithelial/mesenchymal signaling in primitive cellular compartments. Based on differential gene expression we also defined ligand-receptor interactions that may be part of the molecular interplay of the embryonic prostate stem cell niche. CONCLUSIONS/SIGNIFICANCE We provide a comprehensive description of the transcriptional program of the major regulators that are likely to control the cellular interactions in the embryonic prostatic stem cell niche, many of which may be common to mammalian niches in general. This study provides a comprehensive source for further studies of mesenchymal/epithelial interactions in the prostate stem cell niche. The elucidation of pathways in the normal primitive niche may provide greater insight into mechanisms subverted during abnormal proliferative and oncogenic processes. Understanding these events may result in the development of specific targeted therapies for prostatic diseases such as benign prostatic hypertrophy and carcinomas.
Collapse
Affiliation(s)
- Roy Blum
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Rashmi Gupta
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| | - Patricia E. Burger
- Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Christopher S. Ontiveros
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| | - Sarah N. Salm
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
- Department of Science, Borough of Manhattan Community College/City University of New York, New York, New York, United States of America
| | - Xiaozhong Xiong
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| | - Alexander Kamb
- Amgen Inc, South San Francisco, California, United States of America
| | - Holger Wesche
- Amgen Inc, South San Francisco, California, United States of America
| | - Lisa Marshall
- Amgen Inc, South San Francisco, California, United States of America
| | - Gene Cutler
- Amgen Inc, South San Francisco, California, United States of America
| | - Xiangyun Wang
- Pfizer Inc, Groton, Connecticut, United States of America
| | - Jiri Zavadil
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- NYU Cancer Institute, New York University School of Medicine, New York, New York, United States of America
- Center for Health Informatics and Bioinformatics, New York University Medical Center, New York, New York, United States of America
| | - David Moscatelli
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
- NYU Cancer Institute, New York University School of Medicine, New York, New York, United States of America
| | - E. Lynette Wilson
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
- Division of Immunology, University of Cape Town, Cape Town, South Africa
- Department of Urology, New York University School of Medicine, New York, New York, United States of America
- NYU Cancer Institute, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|