1
|
Donohoe ME, Morey R, Li Y, Pizzo D, Kallol S, Cho HY, Soncin F, Parast MM. Identification of HTRA4 as a Transcriptional Target of p63 in Trophoblast. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1162-1170. [PMID: 38880601 PMCID: PMC11220921 DOI: 10.1016/j.ajpath.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 06/18/2024]
Abstract
The placenta plays a crucial role in pregnancy success. ΔNp63α (p63), a transcription factor from the TP53 family, is highly expressed in villous cytotrophoblasts (CTBs), the epithelial stem cells of the human placenta, and is involved in CTB maintenance and differentiation. We examined the mechanisms of action of p63 by identifying its downstream targets. Gene expression changes were evaluated following overexpression and knockdown of p63 in the JEG3 choriocarcinoma cell line, using microarray-based RNA profiling. High-temperature requirement A4 (HTRA4), a placenta-specific serine protease involved in trophoblast differentiation and altered in preeclampsia, was identified as a gene reciprocally regulated by p63, and its expression was characterized in primary human placental tissues by RNA-sequencing and in situ hybridization. Potential p63 DNA-binding motifs were identified in the HTRA4 promoter, and p63 occupancy at some of these sites was confirmed using chromatin immunoprecipitation, followed by quantitative PCR in both JEG3 and trophoblast stem cells. These data begin to identify members of the transcriptional network downstream of p63, thus laying the groundwork for probing mechanisms by which this important transcription factor regulates trophoblast stemness and differentiation.
Collapse
Affiliation(s)
- Mary E Donohoe
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Robert Morey
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Yingchun Li
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Sampada Kallol
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Hee-Young Cho
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
2
|
Bogani G, Ray-Coquard I, Mutch D, Vergote I, Ramirez PT, Prat J, Concin N, Ngoi NYL, Coleman RL, Enomoto T, Takehara K, Denys H, Lorusso D, Takano M, Sagae S, Wimberger P, Segev Y, Kim SI, Kim JW, Herrera F, Mariani A, Brooks RA, Tan D, Paolini B, Chiappa V, Longo M, Raspagliesi F, Benedetti Panici P, Di Donato V, Caruso G, Colombo N, Pignata S, Zannoni G, Scambia G, Monk BJ. Gestational choriocarcinoma. Int J Gynecol Cancer 2023; 33:1504-1514. [PMID: 37758451 DOI: 10.1136/ijgc-2023-004704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Gestational choriocarcinoma accounts for 5% of gestational trophoblastic neoplasms. Approximately 50%, 25%, and 25% of gestational choriocarcinoma occur after molar pregnancies, term pregnancies, and other gestational events, respectively. The FIGO scoring system categorizes patients into low (score 0 to 6) and high risk (score 7 or more) choriocarcinoma. Single-agent and multi-agent chemotherapy are used in low- and high-risk patients, respectively. Chemotherapy for localized disease has a goal of eradication of disease without surgery and is associated with favorable prognosis and fertility preservation. Most patients with gestational choriocarcinoma are cured with chemotherapy; however, some (<5.0%) will die as a result of multi-drug resistance, underscoring the need for novel approaches in this group of patients. Although there are limited data due to its rarity, the treatment response with immunotherapy is high, ranging between 50-70%. Novel combinations of immune checkpoint inhibitors with targeted therapies (including VEGFR-2 inhibitors) are under evaluation. PD-L1 inhibitors are considered a potential important opportunity for chemo-resistant patients, and to replace or de-escalate chemotherapy to avoid or minimize chemotherapy toxicity. In this review, the Rare Tumor Working Group and the European Organization for Research and Treatment of Cancer evaluated the current landscape and further perspective in the management of patients diagnosed with gestational choriocarcinoma.
Collapse
Affiliation(s)
- Giorgio Bogani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Isabelle Ray-Coquard
- Centre Leon Berard, LYON CEDEX 08, France
- Hesper lab, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - David Mutch
- Washington University in Saint Louis, St Louis, Missouri, USA
| | - Ignace Vergote
- Department of Gynecology and Obstetrics, Gynecologic Oncology, Leuven Cancer Institute, Catholic University Leuven, Leuven, Belgium
| | - Pedro T Ramirez
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, Texas, USA
| | - Jaime Prat
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Nicole Concin
- Department of Gynecology and Obstetrics; Innsbruck Medical Univeristy, Innsbruck, Austria
| | | | | | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Belgium
| | - Kazuhiro Takehara
- Department of Gynecologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | | | | | - Masashi Takano
- Department of Obstetrics and Gynecology, National Defense Medical College, Tokorozawa, Japan
| | - Satoru Sagae
- Gynecologic Oncology, Tokeidai Kinen Byoin, Sapporo, Japan
| | - Pauline Wimberger
- Gyncology and Obstetrics, Technische Universitat Dresden Medizinische Fakultat Carl Gustav Carus, Dresden, Germany
| | - Yakir Segev
- Obstetrics and Gynecology, Carmel Hospital, Haifa, Israel
| | - Se Ik Kim
- Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Korea (the Republic of)
| | - Jae-Weon Kim
- Obstetrics and gynecology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Fernanda Herrera
- Centre Hospitalier Universitaire Vaudois Departement doncologie CHUV-UNIL, Lausanne, Switzerland
| | - Andrea Mariani
- Gynecologic Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Rebecca A Brooks
- Section of Gynecologic Oncology, University of Chicago Medicine, Chicago, Illinois, USA
| | - David Tan
- National University Cancer Institute, Singapore
| | - Biagio Paolini
- Istituto Nazionale per lo Studio e la Cura dei Tumori, Milano, Italy
| | - Valentina Chiappa
- Department of Gynecologic Oncology, IRCCS National Cancer Institute, Milan, Italy
| | | | | | | | | | | | - Nicoletta Colombo
- Medical Gynecologic Oncology Unit; University of Milan Bicocca; Milan; Italy, European Institute of Oncology, Milano, Italy
| | - Sandro Pignata
- Gynaecological Oncology, National Cancer Institute Napels, Naples, Italy
| | - Gianfranco Zannoni
- Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna e del Bambino, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Bradley J Monk
- Virginia G Piper Cancer Center - Biltmore Cancer Center, Phoenix, Arizona, USA
| |
Collapse
|
3
|
HOPX: A Unique Homeodomain Protein in Development and Tumor Suppression. Cancers (Basel) 2022; 14:cancers14112764. [PMID: 35681746 PMCID: PMC9179269 DOI: 10.3390/cancers14112764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Homeobox (HOX) genes encode homeodomain proteins that regulate a wide range of molecular pathways. The homeodomain is highly conserved and binds to DNA. One exception is homeodomain-only protein (HOPX) that lacks DNA-binding capacity. HOPX plays a crucial role in development and its functional impairment is associated with a variety of diseases, including cancer. Loss of HOPX function occurs in a wide range of cancer types, where it functions as a tumor suppressor gene. Understanding the molecular mechanisms by which HOPX regulates carcinogenesis will likely lead to the development of new therapeutic approaches. Abstract Homeobox genes are master regulators of morphogenesis and differentiation by acting at the top of genetic hierarchies and their deregulation is associated with a variety of human diseases. They usually contain a highly conserved sequence that codes for the homeodomain of the protein, a specialized motif with three α helices and an N-terminal arm that aids in DNA binding. However, one homeodomain protein, HOPX, is unique among its family members in that it lacks the capacity to bind DNA and instead functions by interacting with transcriptional regulators. HOPX plays crucial roles in organogenesis and is expressed in both embryonic and adult stem cells. Loss of HOPX expression is common in cancer, where it functions primarily as a tumor suppressor gene. In this review, we describe the function of HOPX in development and discuss its role in carcinogenesis.
Collapse
|
4
|
The role of HOPX in normal tissues and tumor progression. Biosci Rep 2020; 40:221873. [PMID: 31934721 PMCID: PMC6997107 DOI: 10.1042/bsr20191953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
The homeodomain-only protein homeobox (HOPX) as the smallest homeodomain protein, lacks certain conserved residues required for DNA binding. Through our literature search, we reviewed the current understandings of HOPX in normal tissues and tumor progression. HOPX was initially identified as a critical transcription factor in various normal tissues, which interacted with serum response factor (SRF) or other substance to regulate normal physiological function. However, HOPX is at a low expression or methylation level in tumors. These data indicated that HOPX may play a very important role in regulating differentiation phenotype and tumor suppressive function. We predicted the prognosis of HOPX in tumors from TCGA database and discussed the downstream genes of HOPX. To understand how HOPX is involved in the mechanisms between physical and pathological conditions could lead to novel therapeutic strategies for treatment.
Collapse
|
5
|
Eder N, Roncaroli F, Domart MC, Horswell S, Andreiuolo F, Flynn HR, Lopes AT, Claxton S, Kilday JP, Collinson L, Mao JH, Pietsch T, Thompson B, Snijders AP, Ultanir SK. YAP1/TAZ drives ependymoma-like tumour formation in mice. Nat Commun 2020; 11:2380. [PMID: 32404936 PMCID: PMC7220953 DOI: 10.1038/s41467-020-16167-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/17/2020] [Indexed: 11/09/2022] Open
Abstract
YAP1 gene fusions have been observed in a subset of paediatric ependymomas. Here we show that, ectopic expression of active nuclear YAP1 (nlsYAP5SA) in ventricular zone neural progenitor cells using conditionally-induced NEX/NeuroD6-Cre is sufficient to drive brain tumour formation in mice. Neuronal differentiation is inhibited in the hippocampus. Deletion of YAP1's negative regulators LATS1 and LATS2 kinases in NEX-Cre lineage in double conditional knockout mice also generates similar tumours, which are rescued by deletion of YAP1 and its paralog TAZ. YAP1/TAZ-induced mouse tumours display molecular and ultrastructural characteristics of human ependymoma. RNA sequencing and quantitative proteomics of mouse tumours demonstrate similarities to YAP1-fusion induced supratentorial ependymoma. Finally, we find that transcriptional cofactor HOPX is upregulated in mouse models and in human YAP1-fusion induced ependymoma, supporting their similarity. Our results show that uncontrolled YAP1/TAZ activity in neuronal precursor cells leads to ependymoma-like tumours in mice.
Collapse
Affiliation(s)
- Noreen Eder
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Federico Roncaroli
- Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Salford and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biology, University of Manchester, Manchester, M13 9PT, UK
| | | | - Stuart Horswell
- Bioinformatics and Biostatistics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Felipe Andreiuolo
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Helen R Flynn
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Andre T Lopes
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Suzanne Claxton
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - John-Paul Kilday
- Centre for Paediatric, Teenage and Young Adult Cancer, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Lucy Collinson
- Electron Microscopy Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jun-Hao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Barry Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Ambrosius P Snijders
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| |
Collapse
|
6
|
Ooizumi Y, Katoh H, Yokota M, Watanabe M, Yamashita K. Epigenetic silencing of HOPX is critically involved in aggressive phenotypes and patient prognosis in papillary thyroid cancer. Oncotarget 2019; 10:5906-5918. [PMID: 31666923 PMCID: PMC6800262 DOI: 10.18632/oncotarget.27187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 08/12/2019] [Indexed: 01/14/2023] Open
Abstract
HOPX is involved in multiple organ development and acts as a tumor suppressor in various cancers. Epigenetic silencing of HOPX via its promoter methylation has been shown frequent and cancer-specific in human cancers. The proliferation of thyroid cancer cells and cancer progression are strongly influenced by epigenetic alterations as well as genetic changes. Papillary thyroid cancer (PTC) comprises the vast majority of thyroid cancers and exhibits slow progression. However, ~10% of patients still show disease recurrence and refractoriness to treatment. Accordingly, it is important approach to research epigenetic mechanisms in PTC progression to find useful biomarkers. Here, we aimed to seek into the roles and clinical impact of epigenetic silencing of HOPX in PTC. The promoter methylation of HOPX was observed in five of six human thyroid cancer cell lines. Down-regulation of HOPX was seen in three cell lines including PTC line K1, and demethylating agents restored HOPX expression. The promoter methylation was observed with high sensitivity and specificity in human PTC tissues. HOPX promoter methylation independently predicted disease recurrence in PTC patients. Epigenetic silencing of HOPX was associated with Ki-67 expression. Of note, HOPX promoter methylation was dramatically associated with worse prognosis especially in patients with stage I PTC. Forced HOPX expression suppressed cell proliferation, invasive activities, and anchorage-independent growth in vitro. HOPX promoter methylation is frequent and cancer-specific event, leading to aggressive phenotype in PTC. Epigenetic silencing of HOPX may be a clue to tackle cancer progression and have clinical impact as a novel biomarker in PTC.
Collapse
Affiliation(s)
- Yosuke Ooizumi
- Department of Surgery, Kitasato University Hospital, Kanagawa, Japan
| | - Hiroshi Katoh
- Breast and Endocrine Surgery, Kitasato University Hospital, Kanagawa, Japan
| | - Mitsuo Yokota
- Department of Surgery, Kitasato University Hospital, Kanagawa, Japan.,Breast and Endocrine Surgery, Kitasato University Hospital, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Surgery, Kitasato University Hospital, Kanagawa, Japan
| | - Keishi Yamashita
- Department of Surgery, Kitasato University Hospital, Kanagawa, Japan.,Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University Hospital, Kanagawa, Japan
| |
Collapse
|
7
|
Kikuchi M, Katoh H, Waraya M, Tanaka Y, Ishii S, Tanaka T, Nishizawa N, Yokoi K, Minatani N, Ema A, Kosaka Y, Tanino H, Yamashita K, Watanabe M. Epigenetic silencing of HOPX contributes to cancer aggressiveness in breast cancer. Cancer Lett 2016; 384:70-78. [PMID: 27756570 DOI: 10.1016/j.canlet.2016.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 01/18/2023]
Abstract
Epigenetic silencing of HOPX has been shown to be frequent and specific in human cancers. HOPX is thought as a tumor suppressor gene and its promoter methylation is the main mechanism of down-regulation. In non-hereditary breast cancer, since roles of epigenetic modifications are more critical than in other cancers, the aim of this study is to seek into the roles and clinical relevance of epigenetic silencing of HOPX. Down-regulation of HOPX was observed in all human breast cancer cell lines tested. The promoter methylation was found in six of seven cell lines, and demethylating agents restored HOPX expression. The promoter methylation was cancer-specific in human breast tissues. Forced expression of HOPX attenuated anchorage-independent growth in vitro. HOPX promoter methylation independently predicted worse prognosis of breast cancer patients. Of note, HOPX promoter methylation was significantly associated with HER2 positivity as well as advanced lymph node metastasis. HOPX promoter methylation is not only frequent and cancer-specific but also associated with aggressive phenotype in breast cancer. Epigenetic silencing of HOPX may have clinical potential as a biomarker in the treatment strategy of breast cancer patients.
Collapse
Affiliation(s)
- Mariko Kikuchi
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiroshi Katoh
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mina Waraya
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoko Tanaka
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Satoru Ishii
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Toshimichi Tanaka
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Nobuyuki Nishizawa
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Keigo Yokoi
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Naoko Minatani
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Akira Ema
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshimasa Kosaka
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hirokazu Tanino
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Keishi Yamashita
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Surgery, Kitasato University School of Medicine, Kanagawa, Japan.
| |
Collapse
|
8
|
Mariotto A, Pavlova O, Park HS, Huber M, Hohl D. HOPX: The Unusual Homeodomain-Containing Protein. J Invest Dermatol 2016; 136:905-911. [PMID: 27017330 DOI: 10.1016/j.jid.2016.01.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 12/23/2015] [Accepted: 01/04/2016] [Indexed: 01/15/2023]
Abstract
The homeodomain-only protein homeobox (HOPX) is the smallest known member of the homeodomain-containing protein family, atypically unable to bind DNA. HOPX is widely expressed in diverse tissues, where it is critically involved in the regulation of proliferation and differentiation. In human skin, HOPX controls epidermal formation through the regulation of late differentiation markers, and HOPX expression correlates with the level of differentiation in cutaneous pathologies. In mouse skin, Hopx was additionally identified as a lineage tracing marker of quiescent hair follicle stem cells. This review discusses current knowledge of HOPX structure and function in normal and pathological conditions.
Collapse
Affiliation(s)
- Anita Mariotto
- Laboratory of Cutaneous Biology, Service of Dermatology and Venereology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Olesya Pavlova
- Laboratory of Cutaneous Biology, Service of Dermatology and Venereology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Hyun-Sook Park
- Laboratory of Cutaneous Biology, Service of Dermatology and Venereology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Marcel Huber
- Laboratory of Cutaneous Biology, Service of Dermatology and Venereology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Daniel Hohl
- Laboratory of Cutaneous Biology, Service of Dermatology and Venereology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.
| |
Collapse
|
9
|
Chen Y, Yang L, Cui T, Pacyna-Gengelbach M, Petersen I. HOPX is methylated and exerts tumour-suppressive function through Ras-induced senescence in human lung cancer. J Pathol 2014; 235:397-407. [DOI: 10.1002/path.4469] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 09/24/2014] [Accepted: 10/10/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Yuan Chen
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | - Linlin Yang
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | - Tiantian Cui
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| | | | - Iver Petersen
- Institute of Pathology, University Hospital Jena; Friedrich-Schiller-University Jena; Ziegelmühlenweg 1 07743 Jena Germany
| |
Collapse
|
10
|
The homeobox only protein homeobox (HOPX) and colorectal cancer. Int J Mol Sci 2013; 14:23231-43. [PMID: 24287901 PMCID: PMC3876040 DOI: 10.3390/ijms141223231] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/31/2013] [Accepted: 11/01/2013] [Indexed: 12/13/2022] Open
Abstract
The HOP (homeobox only protein) homeobox (HOPX) is most closely related to the homeobox protein that contains a homeobox-like domain but lacks certain conserved residues required for DNA binding. Here, we review the current understanding of HOPX in the progression of colorectal cancer (CRC). HOPX was initially reported as a differentiation marker and is expressed in various normal tissues. In the colon, HOPX is expressed uniquely in the quiescent stem cell, +4, and in differentiated mucosal cells of the colon. HOPX expression is markedly suppressed in a subset of cancers, mainly in an epigenetic manner. CRC may include separate entities which are differentially characterized by HOPX expression from a prognostic point of view. HOPX itself can regulate epigenetics, and defective expression of HOPX can result in loss of tumor suppressive function and differentiation phenotype. These findings indicate that HOPX may be both a central regulator of epigenetic dynamics and a critical determinant for differentiation in human cells. HOPX downstream targets were identified in CRC cell lines and hold promise as candidates for therapeutic targets of CRC, such as EphA2 or AP-1. Further analysis will elucidate and confirm the precise role of such proteins in CRC progression.
Collapse
|
11
|
Mak VCY, Lee L, Siu MKY, Wong OGW, Lu X, Ngan HYS, Wong ESY, Cheung ANY. Downregulation of ASPP2 in choriocarcinoma contributes to increased migratory potential through Src signaling pathway activation. Carcinogenesis 2013; 34:2170-7. [PMID: 23671128 DOI: 10.1093/carcin/bgt161] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gestational choriocarcinoma is a malignant tumor derived from placental trophoblast and the most aggressive member of gestational trophoblastic disease (GTD). Apoptosis-stimulating protein of p53-2 (ASPP2) is a member of ASPP family that transactivates p53 and thereby functions as a tumor suppressor. In this study, the expression profile of ASPP2 in choriocarcinoma was examined in comparison with normal placentas and hydatidiform moles, the latter being a type of GTD that carries malignant potential. Downregulation of ASPP2 messenger RNA and protein was demonstrated in choriocarcinoma by quantitative PCR and immunohistochemistry. ASPP2-transfected choriocarcinoma cells (JEG-3 and JAR) showed an increase in apoptosis and a decrease in cell migration as detected by TdT-mediated dUTP nick end labeling and wound healing assays, respectively, illustrating the complex action of ASPP2 on cell functions other than programmed cell death. Activated Src is known to be important in tumor progression. Transfection of ASPP2 but not ASPP1, another tumor-suppressive ASPP, was found to be related to subsequent decreased Src-pY416 phosphorylation, suggesting an inactivating effect of ASPP2 on Src. Moreover, this ASPP2-mediated inactivation of Src could be abolished by RNA interference with C-terminal Src kinase (Csk), a kinase that can inhibit Src activation. Our findings suggested that the ability of ASPP2 to attenuate Src activation was specific to ASPP2 in a Csk-dependent manner. Taken together, we demonstrated a loss of tumor-suppressive ASPP2 in choriocarcinoma with effects on cell migration and apoptosis. We also unveiled a possible mechanistic link between ASPP2 and Csk/Src signaling pathway, implicating the multiple cellular functions of ASPP2.
Collapse
|
12
|
Waraya M, Yamashita K, Katoh H, Ooki A, Kawamata H, Nishimiya H, Nakamura K, Ema A, Watanabe M. Cancer specific promoter CpG Islands hypermethylation of HOP homeobox (HOPX) gene and its potential tumor suppressive role in pancreatic carcinogenesis. BMC Cancer 2012; 12:397. [PMID: 22958219 PMCID: PMC3488580 DOI: 10.1186/1471-2407-12-397] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Accepted: 08/31/2012] [Indexed: 12/25/2022] Open
Abstract
Background We have recently identified HOP hoemobox (HOPX) as a tumor suppressor gene candidate, characterized by tumor-specific promoter DNA hypermethylation in human cancers, and it can remarkably inhibit tumors’ aggressive phenotypes. In this current study, we for the first time examined methylation level of HOPX and tested the functional relevance in pancreatic cancer (PC). Methods Clinical features of HOPX promoter hypermethylation was investigated in 89 PC tissues, and immunohistochemistry was added. We also examined its functional relevance in phenotype assays such as soft agar, proliferation, invasion, and cell cycle analysis. Results PC tissues had HOPX gene hypermethylation as compared to the corresponding normal pancreas tissues, and its uniqueness was robust to discriminate tumor from normal tissues (AUC = 0.85, P < 0.0001). Unexpectedly, HOPX was increased in expression in tumor tissues, and immunohistochemistry revealed its predominant expression in the Langerhans islet cells, where HOPX was reduced in expression for PC cells with promoter hypermethylation. HOPX transfectants exhibited G1 arrest with subG1 accumulation, and inhibited tumor forming and invasive ability. Conclusion Defective expression of HOPX which is consistent with promoter DNA hypermethylation may explain aggressive phenotype of pancreatic cancer, and intense expression of HOPX in the Langerhans cells may in turn uniquely contribute to pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Mina Waraya
- Department of Surgery, Kitasato University Hospital, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0375, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Sudheer S, Bhushan R, Fauler B, Lehrach H, Adjaye J. FGF inhibition directs BMP4-mediated differentiation of human embryonic stem cells to syncytiotrophoblast. Stem Cells Dev 2012; 21:2987-3000. [PMID: 22724507 DOI: 10.1089/scd.2012.0099] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling is known to support differentiation of human embryonic stem cells (hESCs) into mesoderm and extraembryonic lineages, whereas other signaling pathways can largely influence this lineage specification. Here, we set out to reinvestigate the influence of ACTIVIN/NODAL and fibroblast growth factor (FGF) pathways on the lineage choices made by hESCs during BMP4-driven differentiation. We show that BMP activation, coupled with inhibition of both ACTIVIN/NODAL and FGF signaling, induces differentiation of hESCs, specifically to βhCG hormone-secreting multinucleated syncytiotrophoblast and does not support induction of embryonic and extraembryonic lineages, extravillous trophoblast, and primitive endoderm. It has been previously reported that FGF2 can switch BMP4-induced hESC differentiation outcome to mesendoderm. Here, we show that FGF inhibition alone, or in combination with either ACTIVIN/NODAL inhibition or BMP activation, supports hESC differentiation to hCG-secreting syncytiotrophoblast. We show that the inhibition of the FGF pathway acts as a key in directing BMP4-mediated hESC differentiation to syncytiotrophoblast.
Collapse
Affiliation(s)
- Smita Sudheer
- Department of Vertebrate Genomics, Molecular Embryology and Aging Group, Max Planck Institute for Molecular Genetics, Berlin, Germany.
| | | | | | | | | |
Collapse
|
14
|
Gene expression signatures differentiate adenocarcinoma of lung and breast origin in effusions. Hum Pathol 2011; 43:684-94. [PMID: 21937081 DOI: 10.1016/j.humpath.2011.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/11/2011] [Accepted: 06/15/2011] [Indexed: 02/02/2023]
Abstract
Lung and breast adenocarcinoma at advanced stages commonly involve the serosal cavities, giving rise to malignant effusions. The aim of the present study was to compare the global gene expression patterns of metastases from these 2 malignancies, to expand and improve the diagnostic panel of biomarkers currently available for their differential diagnosis, as well as to define type-specific biological targets. Gene expression profiles of 7 breast and 4 lung adenocarcinoma effusions were analyzed using the HumanRef-8 BeadChip from Illumina. Differentially expressed candidate genes were validated using quantitative real-time polymerase chain reaction and immunohistochemistry. Unsupervised hierarchical clustering using all 54,675 genes in the array separated lung from breast adenocarcinoma samples. We identified 289 unique probes that were significantly differentially expressed in the 2 cancers by greater than 2-fold using moderated t statistics, of which 65 and 224 were overexpressed in breast and lung adenocarcinoma, respectively. Genes overexpressed in breast adenocarcinoma included TFF1, TFF3, FOXA1, CA12, PITX1, RARRES1, CITED4, MYC, TFAP2A, EFHD1, TOB1, SPDEF, FASN, and TH. Genes overexpressed in lung adenocarcinoma included TITF1, SFTPG, MMP7, EVA1, GPR116, HOP, SCGB3A2, and MET. The differential expression of 15 genes was validated by quantitative real-time PCR, and differences in 8 gene products were confirmed by immunohistochemistry. Expression profiling distinguishes breast adenocarcinoma from lung adenocarcinoma and identifies genes that are differentially expressed in these 2 tumor types. The molecular signatures unique to these cancers may facilitate their differential diagnosis and may provide a molecular basis for therapeutic target discovery.
Collapse
|
15
|
Alifrangis C, Seckl MJ. Genetics of gestational trophoblastic neoplasia: an update for the clinician. Future Oncol 2011; 6:1915-23. [PMID: 21142864 DOI: 10.2217/fon.10.153] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Gestational trophoblastic disease is a spectrum of disorders ranging from premalignant hydatidiform moles through to malignant invasive moles, choriocarcinoma and rare placental site trophoblastic tumor. The latter are often collectively referred to as gestational trophoblastic tumors or neoplasia (GTN). Although most women can expect to be cured of their disease, many interesting questions arise in the management of gestational trophoblastic disease. Current issues pertain to diagnosis of GTN, predicting progression from hydatidiform moles to GTN and the emergence of drug resistance in GTN. Our understanding of the genetics of GTN has helped us answer some of these questions but many remain unresolved. This article seeks to address recent advances in the genetics of GTN in relation to diagnosis, etiology, prognosis and treatment.
Collapse
Affiliation(s)
- Constantine Alifrangis
- Charing Cross Gestational Trophoblastic Disease Screening & Management Centre, Dept of Medical Oncology, Charing Cross Hospital Campus of Imperial College NHS Healthcare Trust, Fulham Palace Road, London, UK
| | | |
Collapse
|
16
|
Obarzanek-Fojt M, Favre B, Kypriotou M, Ryser S, Huber M, Hohl D. Homeodomain-only protein HOP is a novel modulator of late differentiation in keratinocytes. Eur J Cell Biol 2011; 90:279-90. [PMID: 21256618 DOI: 10.1016/j.ejcb.2010.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 11/03/2010] [Accepted: 11/03/2010] [Indexed: 02/03/2023] Open
Abstract
The homeodomain-only protein (HOP) contains an atypical homeodomain which is unable to bind to DNA due to mutations in residues important for DNA binding. Recently, HOP was reported to regulate proliferation/differentiation homeostasis in different cell types. In the present study, we performed transcriptional profiling of cultured primary human keratinocytes and noted a robust induction of HOP upon calcium-induced cell differentiation. Immunohistochemistry of human skin localized HOP to the granular layer in the epidermis. Overexpression of HOP using a lentiviral vector up-regulated FLG and LOR expression during keratinocyte differentiation. Conversely, decreasing HOP expression using small interfering RNA markedly reduced the calcium-induced expression of late markers of differentiation in vitro, with the most prominent effect on profilaggrin (FLG) mRNA. Moreover, mRNA levels of profilaggrin and loricrin were downregulated in the epidermis of HOP knockout mice. Analysis of skin disorders revealed altered HOP expression in lichen planus, psoriasis and squamous cell carcinoma (SCC). Our data indicate that HOP is a novel modulator of late terminal differentiation in keratinocytes.
Collapse
Affiliation(s)
- Magdalena Obarzanek-Fojt
- Department of Dermatology, University Hospital of Lausanne (CHUV), CH-1011 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
17
|
Genomic and epigenetic profiles of gastric cancer: Potential diagnostic and therapeutic applications. Surg Today 2010; 41:24-38. [DOI: 10.1007/s00595-010-4370-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 04/22/2010] [Indexed: 02/07/2023]
|
18
|
Expression of the homeobox gene, HOPX, is modulated by cell differentiation in human keratinocytes and is involved in the expression of differentiation markers. Eur J Cell Biol 2010; 89:537-46. [PMID: 20226564 DOI: 10.1016/j.ejcb.2010.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 12/30/2009] [Accepted: 01/15/2010] [Indexed: 11/20/2022] Open
Abstract
Homeodomain only protein X (HOPX), an unusual homeodomain protein, was originally identified as a key regulator of cardiac development. We first demonstrated that the expression of HOPX was dependent on the differentiation of human keratinocytes and has an effect on the expression of differentiation markers. HOPX was suppressed in proliferating human keratinocytes and was gradually induced by calcium-triggered differentiation of human keratinocytes. In the epidermis, HOPX is highly expressed in the terminally differentiated suprabasal layers. Among the transcript variants of HOPX, the variant 3 driven by promoter A was the main transcript and it was regulated by cell differentiation in human keratinocytes. The expression of HOPX was induced through the phorbol-12-myristate-13-acetate (PMA)-dependent protein kinase C (PKC) signaling pathway, and not by the demethylating agent, 5-aza-dC (5-aza-2'-deoxycitidine) suggesting the suppression of HOPX is not associated with DNA methylation in human keratinocytes. The RNA interference (RNAi) silencing experiment showed that the knockdown of HOPX expression resulted in the increase of such differentiation markers as involucrin and loricrin. Exogenous expression of HOPX down-regulated the expression of differentiation marker genes in immortalized human keratinocytes (HaCaT). Collectively, HOPX is modulated by cell differentiation in human keratinocytes and this might contribute to homeostasis of keratinocytes by controlling differentiation-dependent genes.
Collapse
|
19
|
Kyritsis AP. Management of primary intracranial germ cell tumors. J Neurooncol 2009; 96:143-9. [PMID: 19588227 DOI: 10.1007/s11060-009-9951-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 06/19/2009] [Indexed: 10/20/2022]
Abstract
Primary intracranial germ cell tumors are rare and usually localized in the pineal and the suprasellar regions. They are divided into the following histologic types: germinoma, teratoma (mature, immature, malignant), choriocarcinoma, embryonal carcinoma, endodermal sinus tumor (yolk sac tumor), and mixed tumors. Clinically, they are manifested with ocular signs or signs of obstructive hydrocephalus. Localized germinomas are treated with radiation therapy and exhibit a relatively good prognosis. Chemotherapy is reserved for disseminated germinomas. Mature teratomas are treated with surgery. The rest of germ cell tumors are managed with various combinations of surgery, chemotherapy, and radiotherapy depending on the tumor type. If the tumors secrete beta-human chorionic gonadotrophin (hCG) or alpha-fetoprotein (FP), these tumor markers can be used to accurately monitor response to treatment. Prognosis is best for germinomas and mature teratomas and worst for choriocarcinomas and embryonal carcinomas.
Collapse
Affiliation(s)
- Athanassios P Kyritsis
- Department of Neurology, University of Ioannina School of Medicine, University Campus, Ioannina 45110, Greece.
| |
Collapse
|
20
|
Cheung ANY, Zhang HJ, Xue WC, Siu MKY. Pathogenesis of choriocarcinoma: clinical, genetic and stem cell perspectives. Future Oncol 2009; 5:217-31. [PMID: 19284380 DOI: 10.2217/14796694.5.2.217] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Choriocarcinoma is a unique malignant neoplasm composed of mononuclear cytotrophoblasts and multinucleated syncytiotrophoblasts that produce human chorionic gonadotrophin. Choriocarcinoma can occur after a pregnancy, as a component of germ cell tumors, or in association with a poorly differentiated somatic carcinoma, each with distinct clinical features. Cytogenetic and molecular studies, predominantly on gestational choriocarcinoma, revealed the impact of oncogenes, tumor suppressor genes and imprinting genes on its pathogenesis. The role of stem cells in various types of choriocarcinoma has been studied recently. This review will discuss how such knowledge can enhance our understanding of the pathogenesis of choriocarcinoma, enable exploration of novel anti-choriocarcinoma targeted therapy and possibly improve our insight on embryological and placental development.
Collapse
Affiliation(s)
- Annie N Y Cheung
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| | | | | | | |
Collapse
|
21
|
Yamaguchi S, Asanoma K, Takao T, Kato K, Wake N. Homeobox gene HOPX is epigenetically silenced in human uterine endometrial cancer and suppresses estrogen-stimulated proliferation of cancer cells by inhibiting serum response factor. Int J Cancer 2009; 124:2577-88. [PMID: 19173292 DOI: 10.1002/ijc.24217] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
HOPX (homeodomain only protein X) is a newly identified homeobox gene whose loss of expression has been reported for several types of neoplasm. Although we found most human uterine endometrial cancers (HEC) defective in HOPX expression, genetic mutations in the HOPX gene were undetectable. As is the case with several tumor suppressor genes, the promoter region of HOPX is densely methylated in HEC tissue samples obtained by laser capture microdissection. HOPX mRNA and protein levels were reduced in the majority of samples, and this correlated with hypermethylation of the HOPX promoter. Forced expression of HOPX resulted in a partial block in cell proliferation, in vivo tumorigenicity and c-fos gene expression in HEC and MCF7 cells in response to 17beta-estradiol (E(2)) stimulation. Analysis of the serum response element (SRE) of c-fos gene promoter showed that the effect of HOPX expression is associated with inhibition of E(2)-induced c-fos activation through the serum response factor (SRF) motif. Knockdown of HOPX in immortalized human endometrial cells resulted in accelerated proliferation. Our study indicates that transcriptional silencing of HOPX results from hypermethylation of the HOPpromoter, which leads to HEC development.
Collapse
Affiliation(s)
- Shinichiro Yamaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
22
|
Strath J, Georgopoulos LJ, Kellam P, Blair GE. Identification of genes differentially expressed as result of adenovirus type 5- and adenovirus type 12-transformation. BMC Genomics 2009; 10:67. [PMID: 19200380 PMCID: PMC2651901 DOI: 10.1186/1471-2164-10-67] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Accepted: 02/06/2009] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cells transformed by human adenoviruses (Ad) exhibit differential capacities to induce tumours in immunocompetent rodents; for example, Ad12-transformed rodent cells are oncogenic whereas Ad5-transformed cells are not. The E1A gene determines oncogenic phenotype, is a transcriptional regulator and dysregulates host cell gene expression, a key factor in both cellular transformation and oncogenesis. To reveal differences in gene expression between cells transformed with oncogenic and non-oncogenic adenoviruses we have performed comparative analysis of transcript profiles with the aim of identifying candidate genes involved in the process of neoplastic transformation. RESULTS Analysis of microarray data revealed that a total of 232 genes were differentially expressed in Ad12 E1- or Ad5 E1-transformed BRK cells compared to untransformed baby rat kidney (BRK) cells. Gene information was available for 193 transcripts and using gene ontology (GO) classifications and literature searches it was possible to assign known or suggested functions to 166 of these identified genes. A subset of differentially-expressed genes from the microarray was further examined by real-time PCR and Western blotting using BRK cells immortalised by Ad12 E1A or Ad5 E1A in addition to Ad12 E1- or Ad5 E1-transformed BRK cells. Up-regulation of RelA and significant dysregulation of collagen type I mRNA transcripts and proteins were found in Ad-transformed cells. CONCLUSION These results suggest that a complex web of cellular pathways become altered in Ad-transformed cells and that Ad E1A is sufficient for the observed dysregulation. Further work will focus on investigating which splice variant of Ad E1A is responsible for the observed dysregulation at the pathway level, and the mechanisms of E1A-mediated transcriptional regulation.
Collapse
Affiliation(s)
- Janet Strath
- Institute of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | | | | | | |
Collapse
|
23
|
Kondoh N, Ishikawa T, Ohkura S, Arai M, Hada A, Yamazaki Y, Kitagawa Y, Shindoh M, Takahashi M, Ando T, Sato Y, Izumo T, Hitomi K, Yamamoto M. Gene expression signatures that classify the mode of invasion of primary oral squamous cell carcinomas. Mol Carcinog 2008; 47:744-56. [PMID: 18449855 DOI: 10.1002/mc.20431] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
To identify molecular signatures and establish a new diagnostic model for progressive oral squamous cell carcinoma (OSCC). Total RNAs were isolated from primary OSCCs from both node-positive and -negative patients and used in cDNA microarray analysis. To identify marker genes representing a malignant phenotype, their expression was further examined by quantitative reverse transcription-PCR (QRT-PCR) in 64 OSCC tissues. Using Fisher's linear discriminant analysis (LDA) fitted with a stepwise increment method, we created discriminatory predictor models. The stability of these models was examined using leave-one-out cross validation. Immunohistochemical analysis was performed. Among the 16,600 possible target cDNAs in the array analysis, 83 genes demonstrated significantly differential signals (>2-fold). We further identified 53 marker genes that can be implicated in the Yamamoto-Kohama's (YKs) mode of invasion for OSCCs (P < 0.06). Using LDA fitted with a stepwise increment method, we created four discriminatory predictor models based on 16- to 25-gene signatures which could best distinguish the five established grades of YKs mode of invasion. Leave-one out validation demonstrated that the stability of these models was 92-95%. For validation, we also examined an independent set of 13 primary OSCCs; the predictor models determined the invasion status from 77% to 100% (on average, 85%) fidelity with the pathological observations. TGM3 protein expression was markedly suppressed in highly invasive OSCCs. We reveal novel gene expression alterations during the progression of OSCC, and have constructed prediction models for the evaluation of the invasion status of these cancers.
Collapse
Affiliation(s)
- Nobuo Kondoh
- Department of Biochemistry, Asahi University, School of Dentistry, Mizuho-shi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Shih IM, Kuo KT. Power of the eternal youth: Nanog expression in the gestational choriocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:911-4. [PMID: 18755845 DOI: 10.2353/ajpath.2008.080624] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Ie-Ming Shih
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| | | |
Collapse
|
25
|
De Toni A, Zbinden M, Epstein JA, Ruiz i Altaba A, Prochiantz A, Caillé I. Regulation of survival in adult hippocampal and glioblastoma stem cell lineages by the homeodomain-only protein HOP. Neural Dev 2008; 3:13. [PMID: 18507846 PMCID: PMC2416439 DOI: 10.1186/1749-8104-3-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 05/28/2008] [Indexed: 01/01/2023] Open
Abstract
Background Homeodomain proteins play critical roles in shaping the development of the embryonic central nervous system in mammals. After birth, neurogenic activities are relegated to stem cell niches, which include the subgranular layer of the dentate gyrus of the hippocampus. Here, we have analyzed the function of HOP (Homeodomain only protein) in this stem cell niche and in human glioblastomas. Results We find that HOP is strongly expressed by radial astrocytes of the dentate gyrus in mice, which are stem cells that give rise to hippocampal granular neurons throughout adulthood. Deletion or down-regulation of HOP results in a decrease of apoptosis of these stem cells without changes in proliferation, and in an increase in the number of newly formed granule neurons. We also find that human glioblastomas largely lack HOP expression and that reintroduction of HOP function in glioma cells cultured as gliomaspheres leads to enhanced apoptosis in a subset of cases. In these cells, HOP function decreases clonogenicity. Conclusion These data suggest that HOP participates in the regulation of the adult mouse hippocampal stem cell niche by negatively affecting cell survival. In addition, HOP may work as a tumor suppressor in a subset of glioblastomas. HOP function thus appears to be critical in the adult brain in a region of continued plasticity, and its deregulation may contribute to disease.
Collapse
|
26
|
Yamashita K, Kim MS, Park HL, Tokumaru Y, Osada M, Inoue H, Mori M, Sidransky D. HOP/OB1/NECC1 promoter DNA is frequently hypermethylated and involved in tumorigenic ability in esophageal squamous cell carcinoma. Mol Cancer Res 2008; 6:31-41. [PMID: 18234960 DOI: 10.1158/1541-7786.mcr-07-0213] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Promoter DNA hypermethylation with gene silencing is a common feature of human cancer, and cancer-prone methylation is believed to be a landmark of tumor suppressor genes (TSG). Identification of novel methylated genes would not only aid in the development of tumor markers but also elucidate the biological behavior of human cancers. We identified several epigenetically silenced candidate TSGs by pharmacologic unmasking of esophageal squamous cell carcinoma (ESCC) cell lines by demethylating agents (5-aza-2'-deoxycitidine and trichostatin A) combined with ESCC expression profiles using expression microarray. HOP/OB1/NECC1 was identified as an epigenetically silenced candidate TSG and further examined for (a) expression status, (b) methylation status, and (c) functional involvement in cancer cell lines. (a) The HOP gene encodes two putative promoters (promoters A and B) associated with two open reading frames (HOPalpha and HOPbeta, respectively), and HOPalpha and HOPbeta were both down-regulated in ESCC independently. (b) Promoter B harbors dense CpG islands, in which we found dense methylation in a cancer-prone manner (55% in tumor tissues by TaqMan methylation-specific PCR), whereas promoter A does not harbor CpG islands. HOPbeta silencing was associated with DNA methylation of promoter B in nine ESCC cell lines tested, and reactivated by optimal conditions of demethylating agents, whereas HOPalpha silencing was not reactivated by such treatments. Forced expression of HOP suppressed tumorigenesis in soft agar in four different squamous cell carcinoma cell lines. More convincingly, RNA interference knockdown of HOP in TE2 cells showed drastic restoration of the oncogenic phenotype. In conclusion, HOP is a putative TSG that harbors tumor inhibitory activity, and we for the first time showed that the final shutdown process of HOP expression is linked to promoter DNA hypermethylation under the double control of the discrete promoter regions in cancer.
Collapse
Affiliation(s)
- Keishi Yamashita
- Department of Otolaryngology, Head and Neck Cancer Research Division, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen Y, Pacyna-Gengelbach M, Deutschmann N, Niesporek S, Petersen I. Homeobox gene HOP has a potential tumor suppressive activity in human lung cancer. Int J Cancer 2007; 121:1021-7. [PMID: 17417779 DOI: 10.1002/ijc.22753] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The homeobox containing gene HOP (Homeodomain Only Protein) was identified in the developing heart and lung where it functions downstream of Nkx2.5 and Nkx2.1 to modulate cardiac and lung gene expression. Previously, we found that HOP was downregulated in lung cancer. In this study, we constructed an expression vector containing the full-length cDNA of HOP and transfected it into a lung cancer cell line H2170. Stable transfection led to an increased expression of HOP confirmed by Northern blot analysis. HOP positive transfectants remarkably reduced the growth rate and the ability of anchorage-independent growth in soft agar, and moreover suppressed the tumor formation in nude mice compared to controls. Transient transfection of Nkx2.1 into H2170 resulted in the overexpression of HOP, and correspondingly, siRNA silencing of Nkx2.1 reduced the expression of HOP in lung cancer cells. Treatment with a differentiation modulating agent 5-bromodeoxyuridine (BrdU) led to restoration of HOP expression in a small cell lung cancer cell line H526. In 29 paired primary lung tumor samples, loss of heterozygosity (LOH) analysis was performed by using the 3 microsatellite markers D4S189, D4S231 and D4S392 around the region of chromosome 4q12 where HOP locates. LOH was only found in 4 out 23 cases (17.4%) indicating that allelic loss is a rare genetic event not responsible for the downregulation of HOP in lung cancer. Taken together, our data suggest that HOP is a potential tumor suppressor possibly involved in lung cancer differentiation, and functions downstream of Nkx2.1.
Collapse
Affiliation(s)
- Yuan Chen
- Institute of Pathology, University Hospital Charité, Schumannstr 20-21, D-10098 Berlin, Germany
| | | | | | | | | |
Collapse
|
28
|
Asanoma K, Kato H, Yamaguchi S, Shin CH, Liu ZP, Kato K, Inoue T, Miyanari Y, Yoshikawa K, Sonoda K, Fukushima K, Wake N. HOP/NECC1, A Novel Regulator of Mouse Trophoblast Differentiation. J Biol Chem 2007; 282:24065-74. [PMID: 17576768 DOI: 10.1074/jbc.m701380200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Homeodomain-only protein/not expressed in choriocarcinoma clone 1 (HOP/NECC1) is a newly identified gene that modifies the expression of cardiac-specific genes and thereby regulates heart development. More recently, HOP/NECC1 was reported to be a suppressor of choriocarcinogenesis. Here, we examined the temporal expression profile of HOP/NECC1 in wild-type mouse placenta. We found that E8.5-E9.5 wild-type placenta expressed HOP/NECC1 in the giant cell and spongiotrophoblast layers. HOP/NECC1 (-/-) placenta exhibited marked propagation of giant cell layers and, in turn reduction of spongiotrophoblast formation. We demonstrated SRF transcriptional activity increased in the differentiating trophoblasts and forced expression of SRF in a trophoblast stem (TS) cell line induces the differentiation into giant cells. Negative regulation of SRF (serum response factor) by the binding of HOP/NECC1 protein contributed at least in part to the generation of these placental defects. Gradual induction of HOP/NECC1 in response to differentiation stimuli may result in the decision to differentiate into a particular type of trophoblastic cell lineage and result in non-lethal defects shown by the HOP/NECC1 (-/-) placentas.
Collapse
Affiliation(s)
- Kazuo Asanoma
- Division of Molecular and Cell Therapeutics, Kyushu University, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Gestational trophoblastic neoplasia comprises a unique group of human neoplastic diseases that derive from fetal trophoblastic tissues and represent semiallografts in patients. This group is composed of choriocarcinoma, placental-site trophoblastic tumour, and epithelioid trophoblastic tumour, and many forms are derived from the precursor lesions, hydatidiform moles. Although most patients with gestational trophoblastic neoplasia are cured by chemotherapy and tumour resection, some patients suffer from metastatic diseases that are refractory to conventional chemotherapy. Therefore, new therapeutic regimens are needed to reduce the toxic effects associated with current chemotherapy and to salvage the occasional non-operable patients with recurrent and chemoresistant disease. Until the fundamental biology of gestational trophoblastic neoplasia becomes more clearly understood, development of a new treatment will remain empirical. This review will briefly summarise the recent advances in understanding the molecular aetiology of this group of diseases and highlight the molecules that can be potentially used for therapeutic targets to treat metastatic gestational trophoblastic neoplasia.
Collapse
Affiliation(s)
- Ie-Ming Shih
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
30
|
Lemaire F, Millon R, Muller D, Rabouel Y, Bracco L, Abecassis J, Wasylyk B. Loss of HOP tumour suppressor expression in head and neck squamous cell carcinoma. Br J Cancer 2004; 91:258-61. [PMID: 15213722 PMCID: PMC2409811 DOI: 10.1038/sj.bjc.6601952] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We report that homeodomain-only protein (HOP) is expressed in the suprabasal layer of normal upper aerodigestive tract epithelium and expression strongly decreases in hypopharyngeal carcinoma. Interestingly, HOP has very recently been shown to be a tumour suppressor involved in differentiation, suggesting that HOP may have a similar role in head and neck squamous cell carcinoma (HNSSC).
Collapse
Affiliation(s)
- F Lemaire
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, 1 Rue Laurent Fries, BP 10142, 67404 Illkirch cedex, France
| | - R Millon
- UPRES EA 34-30, Centre Paul Strauss, 3 rue de la Porte de l'Hôpital, 67085 Strasbourg, France
| | - D Muller
- UPRES EA 34-30, Centre Paul Strauss, 3 rue de la Porte de l'Hôpital, 67085 Strasbourg, France
| | - Y Rabouel
- UPRES EA 34-30, Centre Paul Strauss, 3 rue de la Porte de l'Hôpital, 67085 Strasbourg, France
| | - L Bracco
- Exonhit Therapeutics S.A., 65 Boulevard Masséna, Paris F-75013, France
| | - J Abecassis
- UPRES EA 34-30, Centre Paul Strauss, 3 rue de la Porte de l'Hôpital, 67085 Strasbourg, France
| | - B Wasylyk
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, 1 Rue Laurent Fries, BP 10142, 67404 Illkirch cedex, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, 1 Rue Laurent Fries, BP 10142, 67404 Illkirch cedex, France. E-mail:
| |
Collapse
|
31
|
Abstract
Serial analysis of gene expression (SAGE) was applied to compare expression profiles of normal thyroid tissue and papillary thyroid carcinoma (PTC). A SAGE tag corresponding to the partial cDNA for the small protein 31 (SMAP31) is upregulated approximately 13-fold in papillary thyroid cancer (PTC) and was selected for further research. BLAST-searching the human genome database reveals that the SMAP31 gene is located on chromosome 4q11-12 and contains 6 exons. Alternative splicing results in seven transcripts encoding 2 possible open reading frames (ORF) of 73 and 95 amino acids. Database searching in GenBank's dbEST shows that SMAP31 transcripts are expressed mainly in brain, heart, gingiva, and lung tissue. Thyroid tissue contains three transcripts caused by alternatively splicing in the 5' untranslated region (UTR), which all encode an identical ORF of 73 amino acids. Homology search shows that this protein contains a homeobox domain. Thyroid and/or thyroid carcinoma-specific expression of SMAP31 is studied using Northern blot and reverse transcriptase-polymerase chain reaction (RT-PCR) on a multiple tissue panel. RT-PCR experiments on a cDNA panel containing samples from different normal and tumor tissues shows expression of SMAP31 mRNA in brain, placenta, lung, heart, thyroid and thyroid carcinoma. SMAP31 expression is elevated in 4 of 6 PTC tumor samples compared to 4 normal thyroid controls.
Collapse
Affiliation(s)
- Erwin Pauws
- Laboratory of Pediatric Endocrinology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
32
|
Matsuda T, Wake N. Genetics and molecular markers in gestational trophoblastic disease with special reference to their clinical application. Best Pract Res Clin Obstet Gynaecol 2004; 17:827-36. [PMID: 14614883 DOI: 10.1016/s1521-6934(03)00096-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gestational trophoblastic disease (GTD) encompasses a diverse group of lesions with specific cytogenetic and molecular pathogenesis. Although cytogenetic studies have been extensively reported, the molecular pathogenesis is poorly understood. We will summarize some of the recent molecular observations and correlate them with the pathology of GTD. Complete mole is androgenetic in origin. Thus, if a monoallelic contribution can be shown in complete mole, this would render the gene susceptible to functional inactivation by 'one-hit' kinetics. Alternatively, uniparental transmission of genes that are subject to parental imprinting in humans would impair their regulation. Loss of NECC1 expression, biallelic deletions at the critical (7p12-7q11.23) region and enhanced H19 expression in choriocarcinoma would reflect the genetic features exhibited by the putative forerunner, complete mole. In addition to the unique genetic features shown in GTD, alterations in gene expression profiles accompanied by malignant conversion of trophoblasts would facilitate the development of choriocarcinogenesis from complete mole. With recent advances in molecular techniques, further work is still necessary to provide a better understanding and useful markers for persistent trophoblastic disease. These may provide useful prognostic indications that may guide the different diagnosis of GTD.
Collapse
Affiliation(s)
- Takao Matsuda
- Department of Reproductive Physiology and Gynecology, Medical Institute of Bioregulation, Kyushu University, Beppu, Oita, Japan.
| | | |
Collapse
|