1
|
Conese M, Napolitano O, Laselva O, Di Gioia S, Nappi L, Trabace L, Matteo M. The Oncogenic Theory of Preeclampsia: Is Amniotic Mesenchymal Stem Cells-Derived PLAC1 Involved? Int J Mol Sci 2023; 24:ijms24043612. [PMID: 36835024 PMCID: PMC9962629 DOI: 10.3390/ijms24043612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
The pathomechanisms of preeclampsia (PE), a complication of late pregnancy characterized by hypertension and proteinuria, and due to improper placentation, are not well known. Mesenchymal stem cells derived from the amniotic membrane (AMSCs) may play a role in PE pathogenesis as placental homeostasis regulators. PLACenta-specific protein 1 (PLAC1) is a transmembrane antigen involved in trophoblast proliferation that is found to be associated with cancer progression. We studied PLAC1 in human AMSCs obtained from control subjects (n = 4) and PE patients (n = 7), measuring the levels of mRNA expression (RT-PCR) and secreted protein (ELISA on conditioned medium). Lower levels of PLAC1 mRNA expression were observed in PE AMSCs as compared with Caco2 cells (positive controls), but not in non-PE AMSCs. PLAC1 antigen was detectable in conditioned medium obtained from PE AMSCs, whereas it was undetectable in that obtained from non-PE AMSCs. Our data suggest that abnormal shedding of PLAC1 from AMSC plasma membranes, likely by metalloproteinases, may contribute to trophoblast proliferation, supporting its role in the oncogenic theory of PE.
Collapse
Affiliation(s)
- Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
- Correspondence:
| | - Ottavio Napolitano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| | - Onofrio Laselva
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| | - Luigi Nappi
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| | - Maria Matteo
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli 121, 71122 Foggia, Italy
| |
Collapse
|
2
|
Chen Y, Stagg C, Schlessinger D, Nagaraja R. PLAC1 affects cell to cell communication by interacting with the desmosome complex. Placenta 2021; 110:39-45. [PMID: 34118612 DOI: 10.1016/j.placenta.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION X-linked PLAC1 is highly expressed in placenta during embryogenesis, and when ablated in mice, causes aberrant placental cell layer organization. It is also highly expressed in many types of cancer cell-lines. Although it has been shown that it promotes AKT phosphorylation in cancer cells, the exact mechanism by which it influences placental layer differentiation is unclear. METHODS To investigate the mechanism of action of PLAC1 we did cell fractionation and immunoprecipitation of the protein and Mass Spectrometry analysis to identify its interaction partners. The associated proteins were directly tested for interactions by co-transfection with PLAC1 and immunoprecipitation. Mutations in the ZP-N domain of PLAC1 were introduced to assess its involvement in the interactions. RESULTS We provide evidence that Desmoglein-2 (DSG2), a component of the membrane-associated desmosomal complex, directly interacts with PLAC1. Mutations of cysteines in ZP-N domain disrupt the interaction between PLAC1 and DSG-2. DISCUSSION Because desmosomes are responsible for establishing lateral cell-cell junctions, we suggest that direct interaction with the lateral junction protein complex may be implicated in the PLAC1 effects on cell-cell interactions, and thereby on the layer structure of the placenta.
Collapse
Affiliation(s)
- Yaohui Chen
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Carole Stagg
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Ramaiah Nagaraja
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Oliveira MDMS, Salgado CDM, Viana LR, Gomes-Marcondes MCC. Pregnancy and Cancer: Cellular Biology and Mechanisms Affecting the Placenta. Cancers (Basel) 2021; 13:1667. [PMID: 33916290 PMCID: PMC8037654 DOI: 10.3390/cancers13071667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer during pregnancy is rarely studied due to its low incidence (1:1000). However, as a result of different sociocultural and economic changes, women are postponing pregnancy, so the number of pregnant women with cancer has been increasing in recent years. The importance of studying cancer during pregnancy is not only based on maternal and foetal prognosis, but also on the evolutionary mechanisms of the cell biology of trophoblasts and neoplastic cells, which point out similarities between and suggest new fields for the study of cancer. Moreover, the magnitude of how cancer factors can affect trophoblastic cells, and vice versa, in altering the foetus's nutrition and health is still a subject to be understood. In this context, the objective of this narrative review was to show that some researchers point out the importance of supplementing branched-chain amino acids, especially leucine, in experimental models of pregnancy associated with women with cancer. A leucine-rich diet may be an interesting strategy to preserve physiological placenta metabolism for protecting the mother and foetus from the harmful effects of cancer during pregnancy.
Collapse
Affiliation(s)
| | | | - Lais Rosa Viana
- Nutrition and Cancer Laboratory, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Sao Paulo 13083-862, Brazil; (M.d.M.S.O.); (C.d.M.S.)
| | - Maria Cristina Cintra Gomes-Marcondes
- Nutrition and Cancer Laboratory, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Sao Paulo 13083-862, Brazil; (M.d.M.S.O.); (C.d.M.S.)
| |
Collapse
|
4
|
Mahmoudi AR, Ghods R, Rakhshan A, Madjd Z, Bolouri MR, Mahmoudian J, Rahdan S, Shokri MR, Dorafshan S, Shekarabi M, Zarnani AH. Discovery of a potential biomarker for immunotherapy of melanoma: PLAC1 as an emerging target. Immunopharmacol Immunotoxicol 2020; 42:604-613. [PMID: 33106058 DOI: 10.1080/08923973.2020.1837865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Melanoma has increased in incidence worldwide prompting investigators to search for new biomarkers for targeted immunotherapy of this disease. Placenta specific 1 (PLAC1) is a new member of cancer-testis antigens with widespread expression in many types of cancer. Here, we aimed to study for the first time the expression pattern of PLAC1 in skin cancer samples including cutaneous melanoma, basal cell carcinoma (BCC), squamous cell carcinoma (SCC) in comparison to normal skin and nevus tissues and potential therapeutic effect of anti-PLAC1 antibody in melanoma cancer cell lines in vitro. MATERIALS AND METHODS Polyclonal and monoclonal antibodies were applied for immunohistochemical profiling of PLAC1 expression using tissue microarray. The cytotoxic action of anti-PLAC1 antibody alone or as an antibody drug conjugate (with anti-neoplastic agent SN38) was investigated in melanoma cell lines. RESULTS We observed that 100% (39 of 39) of melanoma tissues highly expressed PLAC1 with both cytoplasmic and surface expression pattern. Investigation of PLAC1 expression in BCC (n = 110) samples showed negative results. Cancer cells in SCC samples (n = 66) showed very weak staining. Normal skin tissues and nevus samples including congenital melanocytic nevus failed to express PLAC1. Anti-PLAC1-SN38 exerted a specific pattern of cytotoxicity in a dose- and time-dependent manner in melanoma cells expressing surface PLAC1. CONCLUSIONS Our findings re-inforce the concept of re-expression of embryonic/placental tissue antigens in cancer and highlight the possibility of melanoma targeted therapy by employing anti-PLAC1 antibodies. The data presented here should lead to the future research on targeted immunotherapy of patients with melanoma.
Collapse
Affiliation(s)
- Ahmad-Reza Mahmoudi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Rakhshan
- Department of Pathology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Bolouri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Mahmoudian
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Shaghayegh Rahdan
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Dorafshan
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shekarabi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Roldán DB, Grimmler M, Hartmann C, Hubich-Rau S, Beißert T, Paret C, Cagna G, Rohde C, Wöll S, Koslowski M, Türeci Ö, Sahin U. PLAC1 is essential for FGF7/FGFRIIIb-induced Akt-mediated cancer cell proliferation. Oncotarget 2020; 11:1862-1875. [PMID: 32499871 PMCID: PMC7244013 DOI: 10.18632/oncotarget.27582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/14/2020] [Indexed: 12/20/2022] Open
Abstract
PLAC1 (placenta enriched 1) is a mammalian trophoblast-specific protein. Aberrant expression of PLAC1 is observed in various human cancers, where it is involved in the motility, migration, and invasion of tumor cells, which are associated with the phosphoinositide 3-kinase (PI3K)/AKT pathway. We previously demonstrated that AKT activation mediates the downstream effects of PLAC1; however, the molecular mechanisms of PLAC1-induced AKT-mediated tumor-related processes are unclear. We studied human choriocarcinoma and breast cancer cell lines to explore the localization and receptor-ligand interactions, as well as the downstream effects of PLAC1. We show secretion and adherence of PLAC1 to the extracellular matrix, where it forms a trimeric complex with fibroblast growth factor 7 (FGF7) and its receptor, FGF receptor 2 IIIb (FGFR2IIIb). We further show that PLAC1 signaling via FGFR2IIIb activates AKT phosphorylation in cancer cell lines. As the FGF pathway is of major interest in anticancer therapeutic strategies, these data further promote PLAC1 as a promising anticancer drug target.
Collapse
Affiliation(s)
- Diana Barea Roldán
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Matthias Grimmler
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: DiaSys Diagnostic Systems GmbH, Holzheim, Germany
- These authors contributed equally to this work
| | - Christoph Hartmann
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
- These authors contributed equally to this work
| | - Stefanie Hubich-Rau
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Tim Beißert
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Paret
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Giuseppe Cagna
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Lonza Pharma & Biotech, Cologne, Germany
| | - Christoph Rohde
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
| | - Stefan Wöll
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Michael Koslowski
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Formerly of University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: GammaDelta Therapeutics, London, United Kingdom
| | - Özlem Türeci
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- Ci3 Cluster for Individualized Immune Intervention, Mainz, Germany
| | - Ugur Sahin
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
6
|
Yilmaz N, Timur H, Ugurlu EN, Yilmaz S, Ozgu-Erdinc AS, Erkilinc S, Inal HA. Placenta specific protein-1 in recurrent pregnancy loss and in In Vitro Fertilisation failure: a prospective observational case-control study. J OBSTET GYNAECOL 2019; 40:843-848. [PMID: 31791163 DOI: 10.1080/01443615.2019.1674263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Observations from studies have provided evidence that Placenta-specific protein1 (PLAC1) is important for the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. The aim of this study is to investigate whether maternal serum PLAC1 levels have any impact on etiopathogenesis of recurrent pregnancy loss (RPL) and repeated implantation failure after In Vitro Fertilisation (RIF). We conducted a prospective observational case-control study in a Research Hospital. Twenty-eight patients with RPL (group 1), 30 patients with unexplained infertility and RIF (group 2), 29 fertile patients (group 3) were included. The demographic features and serum PLAC1 levels were compared. There was a significant difference in PLAC1 levels between the groups (group 1 = 19.71 + 16.55 ng/ml; group 2 = 4.82 + 1.44 ng/ml; group 3 = 0.89 + 0.62 ng/ml, respectively) (p=.001). Positive correlation was found between serum PLAC1 levels and abortion rates (r = 0.64; p=.001), a negative correlation was found between serum PLAC1 levels and live birth rates (r = -0.69; p=.001). PLAC1 might have a negative effect on implantation in RPL and RIF. There may be a subgroup of PLAC with different bioactivity. There are no relevant studies conducted among these populations, further large-scale studies are needed to assess the molecular role of PLAC1 on implantation.IMPACT STATEMENTWhat is already known about this subject? PLAC1 (placenta-specific protein-1) gene is located on the X chromosome which encodes for a protein that is thought to be important for placental development although its role has not been clearly defined. Studies in the literature have provided evidence that PLAC1 has an important role in the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. Several reports over the past few years have demonstrated PLAC1 expression in a variety of human tumours including lung cancers, breast cancer, hepatocellular and colorectal cancers, gastric cancers and uterine cancers.What do the results of this study add? There have been no previous studies conducted among patients with recurrent pregnancy loss (RPL) or repeated implantation failure after In Vitro Fertilisation (RIF) that have searched for any association between PLAC1 levels and implantation failure. This study has demonstrated higher PLAC1 levels in infertile women with RIF and RPL for the first time; suggesting that it could have a negative effect on implantation in these populations. PLAC1 could be detected in the serum as a biomarker that is associated with RIF and RPL. What are the implications of these findings for clinical practice and/or further research? Defining the precise role of PLAC1 during implantation will provide new insight into understanding of poor reproductive outcomes such as RIF and RPL and help in developing treatment strategies. Further large-scale studies with more patients are needed to uncover the clinical value of PLAC1 as a biomarker to predict repeated implantation failure and RPL.
Collapse
Affiliation(s)
- Nafiye Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hakan Timur
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Evin Nil Ugurlu
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Saynur Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - A Seval Ozgu-Erdinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Selcuk Erkilinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hasan Ali Inal
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| |
Collapse
|
7
|
Mahmoudian J, Ghods R, Nazari M, Jeddi-Tehrani M, Ghahremani MH, Ghaffari-Tabrizi-Wizsy N, Ostad SN, Zarnani AH. PLAC1: biology and potential application in cancer immunotherapy. Cancer Immunol Immunother 2019; 68:1039-1058. [PMID: 31165204 PMCID: PMC11028298 DOI: 10.1007/s00262-019-02350-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/24/2019] [Indexed: 12/29/2022]
Abstract
The emergence of immunotherapy has revolutionized medical oncology with unprecedented advances in cancer treatment over the past two decades. However, a major obstacle in cancer immunotherapy is identifying appropriate tumor-specific antigens to make targeted therapy achievable with fewer normal cells being impaired. The similarity between placentation and tumor development and growth has inspired many investigators to discover antigens for effective immunotherapy of cancers. Placenta-specific 1 (PLAC1) is one of the recently discovered placental antigens with limited normal tissue expression and fundamental roles in placental function and development. There is a growing body of evidence showing that PLAC1 is frequently activated in a wide variety of cancer types and promotes cancer progression. Based on the restricted expression of PLAC1 in testis, placenta and a wide variety of cancers, we have designated this molecule with new terminology, cancer-testis-placenta (CTP) antigen, a feature that PLAC1 shares with many other cancer testis antigens. Recent reports from our lab provide compelling evidence on the preferential expression of PLAC1 in prostate cancer and its potential utility in prostate cancer immunotherapy. PLAC1 may be regarded as a potential CTP antigen for targeted cancer immunotherapy based on the available data on its promoting function in cancer development and also its expression in cancers of different histological origin. In this review, we will summarize current data on PLAC1 with emphasis on its association with cancer development and immunotherapy.
Collapse
Affiliation(s)
- Jafar Mahmoudian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran
| | | | - Seyed Nasser Ostad
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran.
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Nafisi Building, Enghelab St., Tehran, 1417613151, Iran.
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Yang L, Zha TQ, He X, Chen L, Zhu Q, Wu WB, Nie FQ, Wang Q, Zang CS, Zhang ML, He J, Li W, Jiang W, Lu KH. Placenta-specific protein 1 promotes cell proliferation and invasion in non-small cell lung cancer. Oncol Rep 2017; 39:53-60. [PMID: 29138842 PMCID: PMC5783604 DOI: 10.3892/or.2017.6086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/26/2017] [Indexed: 01/08/2023] Open
Abstract
Pulmonary carcinoma-associated proteins have emerged as crucial players in governing fundamental biological processes such as cell proliferation, apoptosis and metastasis in human cancers. Placenta-specific protein 1 (PLAC1) is a cancer-related protein, which is activated and upregulated in a variety of malignant tissues, including prostate cancer, gastric adenocarcinoma, colorectal, epithelial ovarian and breast cancer. However, its biological role and clinical significance in non-small cell lung cancer (NSCLC) development and progression are still unknown. In the present study, we found that PLAC1 was significantly upregulated in NSCLC tissues, and its expression level was associated with advanced pathological stage and it was also correlated with shorter progression-free survival of lung cancer patients. Furthermore, knockdown of PLAC1 expression by siRNA inhibited cell proliferation, induced apoptosis and impaired invasive ability in NSCLC cells partly via regulation of epithelial-mesenchymal transition (EMT)-related protein expression. Our findings present that increased PLAC1 could be identified as a negative prognostic biomarker in NSCLC and regulate cell proliferation and invasion. Thus, we conclusively demonstrated that PLAC1 plays a key role in NSCLC development and progression, which may provide novel insights on the function of tumor-related gene-driven tumorigenesis.
Collapse
Affiliation(s)
- Li Yang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Tian-Qi Zha
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang He
- Department of Digestive, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Liang Chen
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Quan Zhu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wei-Bing Wu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Feng-Qi Nie
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Qian Wang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Chong-Shuang Zang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Mei-Ling Zhang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Jing He
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Li
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Wen Jiang
- Department of Biochemistry, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| | - Kai-Hua Lu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Gulou, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
9
|
Reinholt BM, Bradley JS, Jacobs RD, Ealy AD, Johnson SE. Tissue organization alters gene expression in equine induced trophectoderm cells. Gen Comp Endocrinol 2017; 247:174-182. [PMID: 28161437 DOI: 10.1016/j.ygcen.2017.01.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/26/2017] [Accepted: 01/29/2017] [Indexed: 12/29/2022]
Abstract
Rapid morphological and gene expression changes occur during the early formation of a mammalian blastocyst. Critical to successful retention of the blastocyst and pregnancy is a functional trophectoderm (TE) that supplies the developing embryo with paracrine factors and hormones. The contribution of TE conformational changes to gene expression was examined in equine induced trophoblast (iTr) cells. Equine iTr cells were cultured as monolayers or in suspension to form spheres. The spheres are hollow and structurally reminiscent of native equine blastocysts. Total RNA was isolated from iTr monolayers and spheres and analyzed by RNA sequencing. An average of 32.2 and 31million aligned reads were analyzed for the spheres and monolayers, respectively. Forty-four genes were unique to monolayers and 45 genes were expressed only in spheres. Conformation did not affect expression of CDX2, POU5F1, TEAD4, ETS2, ELF3, GATA2 or TFAP2A, the core gene network of native TE. Bioinformatic analysis was used to identify classes of genes differentially expressed in response to changes in tissue shape. In both iTr spheres and monolayers, the majority of the differentially expressed genes were associated with binding activity in cellular, developmental and metabolic processes. Inherent to protein:protein interactions, several receptor-ligand families were identified in iTr cells with enrichment of genes coding for PI3-kinase and MAPK signaling intermediates. Our results provide evidence for ligand initiated kinase signaling pathways that underlie early trophectoderm structural changes.
Collapse
Affiliation(s)
- Brad M Reinholt
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jennifer S Bradley
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Robert D Jacobs
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Alan D Ealy
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.
| |
Collapse
|
10
|
Liso A, Massenzio F, Stracci F. PLAC1 immunization does not induce infertility in mice. Immunotherapy 2017; 9:481-486. [DOI: 10.2217/imt-2017-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Placenta specific 1 (PLAC1) is a protein rarely expressed in normal cells, except it is important for placental development, with a possible role in the establishment of the mother–fetus interface. The gene is also highly active in a wide variety of cancers and therefore, immunization with PLAC1 peptides could possibly be part of future immunotherapeutic strategies. We investigated whether vaccination against PLAC1 could induce infertility. Materials & methods: We inoculated female mice with PLAC1 peptides, put them in mating, measured antibody response (ELISA assay) and checked, in immunohistochemistry, binding of the induced antibodies to the native antigen. Results: We demonstrated that mice consistently develop antibody responses. We also demonstrated that female mice, after being inoculated with the PLAC1 peptide mix, do became pregnant and can give birth to normal infants. Conclusion: PLAC1 antigens as a specific anti-cancer vaccine could induce anti-PLAC1 antibodies which do not necessarily cause infertility.
Collapse
Affiliation(s)
- Arcangelo Liso
- Department of Medicine & Surgery, University of Foggia, viale L. Pinto,1 71122 – Foggia, Italy
| | - Francesca Massenzio
- Department of Medicine & Surgery, University of Foggia, viale L. Pinto,1 71122 – Foggia, Italy
| | - Fabrizio Stracci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
11
|
Devor EJ, Gonzalez-Bosquet J, Warrier A, Reyes HD, Ibik NV, Schickling BM, Newtson A, Goodheart MJ, Leslie KK. p53 mutation status is a primary determinant of placenta-specific protein 1 expression in serous ovarian cancers. Int J Oncol 2017; 50:1721-1728. [PMID: 28339050 PMCID: PMC5403493 DOI: 10.3892/ijo.2017.3931] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/16/2017] [Indexed: 12/04/2022] Open
Abstract
Placenta-specific protein 1 (PLAC1) expression is co-opted in numerous human cancers. As a consequence of PLAC1 expression, tumor cells exhibit enhanced proliferation and invasiveness. This characteristic is associated with increased aggressiveness and worse patient outcomes. Recently, the presence of the tumor suppressor p53 was shown in vitro to inhibit PLAC1 transcription by compromising the P1, or distal/cancer, promoter. We sought to determine if this phenomenon occurs in primary patient tumors as well. Furthermore, we wanted to know if p53 mutation influenced PLAC1 expression as compared with wild-type. We chose to study serous ovarian tumors as they are well known to have a high rate of p53 mutation. We report herein that the phenomenon of PLAC1 transcription repression does occur in serous ovarian carcinomas but only when TP53 is wild-type. We find that mutant or absent p53 protein de-represses PLAC1 transcription. We further propose that the inability of mutant p53 to repress PLAC1 transcription is due to the fact that the altered TP53 protein is unable to occupy a putative p53 binding site in the PLAC1 P1 promoter thus allowing transcription to occur. Finally, we show that PLAC1 transcript number is significantly negatively correlated with patient survival in our samples. Thus, we suggest that characterizing tumors for TP53 mutation status, p53 protein status and PLAC1 transcription could be used to predict likely prognosis and inform treatment options in patients diagnosed with serous ovarian cancer.
Collapse
Affiliation(s)
- Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Jesus Gonzalez-Bosquet
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Akshaya Warrier
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Henry D Reyes
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Nonye V Ibik
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Brandon M Schickling
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andreea Newtson
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Michael J Goodheart
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine and The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
12
|
Chang WL, Wang H, Cui L, Peng NN, Fan X, Xue LQ, Yang Q. PLAC1 is involved in human trophoblast syncytialization. Reprod Biol 2016; 16:218-224. [PMID: 27692364 DOI: 10.1016/j.repbio.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022]
Abstract
Placenta specific protein 1 (PLAC1) is thought to be important for murine and human placentation because of its abundant expression in placenta; however, the trophoblast subtypes that express PLAC1 at the fetomaternal interface and the major role of PLAC1 in placentation are still unclear. This study investigated the expression pattern of PLAC1 at the human fetomaternal interface and its involvement in trophoblast syncytialization. Localization of PLAC1 at the fetomaternal interface was studied using in situ hybridization (ISH) and immunohistochemistry (IHC) assays. Real time RT-PCR and Western Blot were employed to exhibit the expression pattern of PLAC1 during human spontaneous syncytialization of term primary cytotrophoblast cells (CTBs). Spontaneous syncytialization of a primary term CTBs model transfected with siRNA specific to PLAC1 was used to investigate the role of PLAC1 during human trophoblast syncytialization. The results showed that PLAC1 was mainly expressed in the human villous syncytiotrophoblast (STB) layer throughout gestation, and the expression level of PLAC1 was significantly elevated during human trophoblast syncytialization. Down-regulation of PLAC1 via specific PLAC1 siRNA transfection attenuated spontaneous syncytialization of primary term CTBs (p<0.05) as indicated by cell fusion index and the expression patterns of the corresponding markers. These data demonstrate the facilitative role of PLAC1 in normal human trophoblast syncytialization.
Collapse
Affiliation(s)
- Wen-Lin Chang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China; State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKUHKUST Medical Center, Shenzhen, China
| | - Huiying Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lina Cui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Graduate School of Chinese Academy of Sciences, Beijing, China
| | - Nan-Ni Peng
- Reproductive Medical Center of Luohu Hospital Shenzhen, Shenzhen, Guangdong, China
| | - Xiujun Fan
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Li-Qun Xue
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| | - Qing Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
13
|
Muto M, Fujihara Y, Tobita T, Kiyozumi D, Ikawa M. Lentiviral Vector-Mediated Complementation Restored Fetal Viability but Not Placental Hyperplasia in Plac1-Deficient Mice. Biol Reprod 2015; 94:6. [PMID: 26586843 DOI: 10.1095/biolreprod.115.133454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/13/2015] [Indexed: 12/25/2022] Open
Abstract
The X-linked Plac1 gene is maternally expressed in trophoblast cells during placentation, and its disruption causes placental hyperplasia and intrauterine growth restriction. In contrast, Plac1 is also reported to be one of the upregulated genes in the hyperplastic placenta generated by nuclear transfer. However, the effect of overexpressed Plac1 on placental formation and function remained unaddressed. We complemented the Plac1 knockout placental dysfunction by lentiviral vector-mediated, placenta-specific Plac1 transgene expression. Whereas fetal development and the morphology of maternal blood sinuses in the labyrinth zone improved, placental hyperplasia remained, with an expanded the junctional zone that migrated and encroached into the labyrinth zone. Further experiments revealed that wild-type placenta with transgenically expressed Plac1 resulted in placental hyperplasia without the encroaching of the junctional zone. Our findings suggest that Plac1 is involved in trophoblast cell proliferation, differentiation, and migration. Its proper expression is required for normal placentation and fetal development.
Collapse
Affiliation(s)
- Masanaga Muto
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tomohiro Tobita
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
14
|
Gundling WE, Wildman DE. A review of inter- and intraspecific variation in the eutherian placenta. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140072. [PMID: 25602076 PMCID: PMC4305173 DOI: 10.1098/rstb.2014.0072] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The placenta is one of the most morphologically variable mammalian organs. Four major characteristics are typically discussed when comparing the placentas of different eutherian species: placental shape, maternal-fetal interdigitation, intimacy of the maternal-fetal interface and the pattern of maternal-fetal blood flow. Here, we describe the evolution of three of these features as well as other key aspects of eutherian placentation. In addition to interspecific anatomical variation, there is also variation in placental anatomy and function within a single species. Much of this intraspecific variation occurs in response to different environmental conditions such as altitude and poor maternal nutrition. Examinations of variation in the placenta from both intra- and interspecies perspectives elucidate different aspects of placental function and dysfunction at the maternal-fetal interface. Comparisons within species identify candidate mechanisms that are activated in response to environmental stressors ultimately contributing to the aetiology of obstetric syndromes such as pre-eclampsia. Comparisons above the species level identify the evolutionary lineages on which the potential for the development of obstetric syndromes emerged.
Collapse
Affiliation(s)
- William E Gundling
- Institute for Genomic Biology, University of Illinois, Urbana, IL, USA Department of Molecular and Integrative Physiology, University of Illinois, Urbana, IL, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Derek E Wildman
- Institute for Genomic Biology, University of Illinois, Urbana, IL, USA Department of Molecular and Integrative Physiology, University of Illinois, Urbana, IL, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
15
|
Ghods R, Ghahremani MH, Madjd Z, Asgari M, Abolhasani M, Tavasoli S, Mahmoudi AR, Darzi M, Pasalar P, Jeddi-Tehrani M, Zarnani AH. High placenta-specific 1/low prostate-specific antigen expression pattern in high-grade prostate adenocarcinoma. Cancer Immunol Immunother 2014; 63:1319-27. [PMID: 25186610 PMCID: PMC11029513 DOI: 10.1007/s00262-014-1594-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/05/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND The scarcity of effective therapeutic approaches for prostate cancer (PCa) has encouraged steadily growing interest for the identification of novel antigenic targets. Placenta-specific 1 (PLAC1) is a novel cancer-testis antigen with reported ectopic expression in a variety of tumors and cancer cell lines. The purpose of the present study was to investigate for the first time the differential expression of PLAC1 in PCa tissues. METHODS We investigated the differential expression of PLAC1 in PCa, high-grade prostatic intraepithelial neoplasia (HPIN), benign prostatic hyperplasia (BPH), and nonneoplastic/nonhyperplastic prostate tissues using microarray-based immunohistochemistry (n = 227). The correlation of PLAC1 expression with certain clinicopathological parameters and expression of prostate-specific antigen (PSA), as a prostate epithelial cell differentiation marker, were investigated. RESULTS Placenta-specific 1 (PLAC1) expression was increased in a stepwise manner from BPH to PCa, which expressed highest levels of this molecule, while in a majority of normal tissues, PLAC1 expression was not detected. Moreover, PLAC1 expression was positively associated with Gleason score (p ≤ 0.001). Interestingly, there was a negative correlation between PLAC1 and PSA expression in patients with PCa and HPIN (p ≤ 0.01). Increment of PLAC1 expression increased the odds of PCa and HPIN diagnosis (OR 49.45, 95 % CI for OR 16.17-151.25). CONCLUSION Our findings on differential expression of PLAC1 in PCa plus its positive association with Gleason score and negative correlation with PSA expression highlight the potential usefulness of PLAC1 for targeted PC therapy especially for patients with advanced disease.
Collapse
Affiliation(s)
- Roya Ghods
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- Department of Pharmacology-Toxicology, Faculty of Medicine, Tehran University of Medical Sciences, TUMS, Tehran, Iran
- School of Advanced Technologies in Medicine, Eastern side of Tehran University, 88, Italia St, P.O. box: 1417755469, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Sanaz Tavasoli
- Department of Nutrition, Science and Research Branch, Azad University, Tehran, Iran
| | - Ahmad-Reza Mahmoudi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Maryam Darzi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Parvin Pasalar
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, TUMS, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Immunology Research Center, Iran University of Medical Sciences, IUMS, Hemmat Highway, P.O. box: 1449614535, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
16
|
Ghafouri-Fard S, Shamsi R, Seifi-Alan M, Javaheri M, Tabarestani S. Cancer-testis genes as candidates for immunotherapy in breast cancer. Immunotherapy 2014; 6:165-79. [PMID: 24491090 DOI: 10.2217/imt.13.165] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer-testis (CT) antigens are tumor-associated antigens attracting immunologists for their possible application in the immunotherapy of cancer. Several clinical trials have assessed their therapeutic potentials in cancer patients. Breast cancers, especially triple-negative cancers are among those with significant expression of CT genes. Identification of CT genes with high expression in cancer patients is the prerequisite for any immunotherapeutic approach. CT genes have gained attention not only for immunotherapy of cancer patients, but also for immunoprevention in high-risk individuals. Many CT genes have proved to be immunogenic in breast cancer patients suggesting the basis for the development of polyvalent vaccines.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | | | | | | | | |
Collapse
|
17
|
Chang WL, Yang Q, Zhang H, Lin HY, Zhou Z, Lu X, Zhu C, Xue LQ, Wang H. Role of placenta-specific protein 1 in trophoblast invasion and migration. Reproduction 2014; 148:343-52. [PMID: 24989904 DOI: 10.1530/rep-14-0052] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Placenta-specific protein 1 (PLAC1), a placenta-specific gene, is known to be involved in the development of placenta in both humans and mice. However, the precise role of PLAC1 in placental trophoblast function remains unclear. In this study, the localization of PLAC1 in human placental tissues and its physiological significance in trophoblast invasion and migration are investigated by technical studies including real-time RT-PCR, in situ hybridization, immunohistochemistry, and functional studies by utilizing cell invasion and migration assays in the trophoblast cell line HTR8/SVneo as well as the primary inducing extravillous trophoblasts (EVTs). The results show that PLAC1 is mainly detected in the trophoblast columns and syncytiotrophoblast of the first-trimester human placental villi, as well as in the EVTs that invade into the maternal decidua. Knockdown of PLAC1 by RNA interference significantly suppresses the invasion and migration of HTR8/SVneo cells and shortens the distance of the outgrowth of the induced EVTs from the cytotrophoblast column of the explants. All the above data suggests that PLAC1 plays an important role in human placental trophoblast invasion and migration.
Collapse
Affiliation(s)
- Wen-Lin Chang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Qing Yang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hui Zhang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hai-Yan Lin
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Zhi Zhou
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Xiaoyin Lu
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Cheng Zhu
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Li-Qun Xue
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hongmei Wang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| |
Collapse
|
18
|
Wang X, Baddoo MC, Yin Q. The placental specific gene, PLAC1, is induced by the Epstein-Barr virus and is expressed in human tumor cells. Virol J 2014; 11:107. [PMID: 24912876 PMCID: PMC4072619 DOI: 10.1186/1743-422x-11-107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/30/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Epstein-Barr virus (EBV) is a causal agent in a number of malignancies in humans including hematopoietic tumors and non-hematopoietic tumors. Burkitt's lymphoma cell lines containing the Epstein-Barr virus have been shown to form tumors in nude mice while clonal derivatives of such cell lines in which the viral genome has been lost do not (JID 177: 1194-1201, 1998; JV 72: 9150-9156, 1998; JV 68: 6069-6073, 1994). The re-introduction of EBV into these EBV negative BLs reconstitutes the tumor phenotype. Thus, EBV-induced cellular genes play critical role in EBV-related tumors. METHODS AND RESULTS In an attempt to identify cellular genes regulated by EBV that may contribute to its tumorigenic properties, we have enforced genome loss in the Burkitt's lymphoma (BL) line, MutuI, by introducing a dominant negative form of the episomal replication factor, EBNA1 and carried out gene array analysis. One of the genes identified by this analysis is PLAC1, a gene originally identified as being expressed exclusively in placental tissue. Real time RT-PCR analysis verified higher expression in EBV positive vs. EBV negative Mutu clones. Analysis of a panel of RNAs from 20 normal tissues demonstrated the highest level of expression in placenta but significant expression was also observed in testis and brain cerebellum. PLAC1 expression was also observed in non-BL tumor cell lines derived from breast, ovary, and prostate. Lastly, expression of PLAC1 was found to be higher in some primary breast tumors compared to normal adjacent tissues. CONCLUSION This data suggests that the EBV-induced PLAC1 is a member of the cancer/testis group of tumor antigens.
Collapse
Affiliation(s)
| | | | - Qinyan Yin
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, SL9, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| |
Collapse
|
19
|
Ghods R, Ghahremani MH, Darzi M, Mahmoudi AR, Yeganeh O, Bayat AA, Pasalar P, Jeddi-Tehrani M, Zarnani AH. Immunohistochemical characterization of novel murine monoclonal antibodies against human placenta-specific 1. Biotechnol Appl Biochem 2014; 61:363-9. [PMID: 24237073 DOI: 10.1002/bab.1177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 11/08/2013] [Indexed: 11/08/2022]
Abstract
Human PLAC1 (placenta-specific 1) is a new member of cancer-testis antigens with 212 amino acids, and its expression is restricted to placenta and at much lower levels to testis. Recently, ectopic expression of the PLAC1 transcript has been demonstrated in a wide range of human tumors and cancer cell lines with a proposed function in tumor cell growth. No monoclonal anti-PLAC1 antibody applicable to immunohis-tochemical staining is available so far. To better understand the PLAC1 expression and localization, we aimed to produce monoclonal antibodies (mAbs) against the extracellular region of PLAC1. Mice were immunized with a synthetic peptide corresponding to the C-terminal 11 amino acids of PLAC1 conjugated with a carrier protein. Hybridomas were produced by standard protocol and screened for positive reactivity by enzyme-linked immunosorbent assay. Reactivity of final two clones was then assessed by Western blotting (WB), immunohistochemistry (IHC), and immunocytochemistry (ICC). Both clones showed a specific immunostaining pattern in human term placenta as the positive control. Reactivity was mostly localized to the cytoplasm of syncytiotrophoblasts. One of the clones showed an excellent staining signal in breast, ovary, and prostate cancer cell lines. Importantly, no reactivity was observed with human lymph node cells or prostate. None of the mAbs were able to detect PLAC1 in Western blot. Based on the present results, these mAbs can be used for detection of PLAC1 in IHC and ICC techniques.
Collapse
Affiliation(s)
- Roya Ghods
- Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran; Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Role of IGF2BP3 in trophoblast cell invasion and migration. Cell Death Dis 2014; 5:e1025. [PMID: 24457969 PMCID: PMC4040666 DOI: 10.1038/cddis.2013.545] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/27/2013] [Accepted: 11/28/2013] [Indexed: 01/06/2023]
Abstract
The insulin-like growth factor-2 mRNA-binding protein 3 (IGF2BP3) is a member of a highly conserved protein family that is expressed specifically in placenta, testis and various cancers, but is hardly detectable in normal adult tissues. IGF2BP3 has important roles in RNA stabilization and translation, especially during early stages of both human and mouse embryogenesis. Placenta is an indispensable organ in mammalian reproduction that connects developing fetus to the uterine wall, and is responsible for nutrient uptake, waste elimination and gas exchange. Fetus development in the maternal uterine cavity depends on the specialized functional trophoblast. Whether IGF2BP3 plays a role in trophoblast differentiation during placental development has never been examined. The data obtained in this study revealed that IGF2BP3 was highly expressed in human placental villi during early pregnancy, especially in cytotrophoblast cells (CTBs) and trophoblast column, but a much lower level of IGF2BP3 was detected in the third trimester placental villi. Furthermore, the expression level of IGF2BP3 in pre-eclamptic (PE) placentas was significantly lower than the gestational age-matched normal placentas. The role of IGF2BP3 in human trophoblast differentiation was shown by in vitro cell invasion and migration assays and an ex vivo explant culture model. Our data support a role of IGF2BP3 in promoting trophoblast invasion and suggest that abnormal expression of IGF2BP3 might be associated with the etiology of PE.
Collapse
|
21
|
Fant ME, Fuentes J, Kong X, Jackman S. The nexus of prematurity, birth defects, and intrauterine growth restriction: a role for plac1-regulated pathways. Front Pediatr 2014; 2:8. [PMID: 24600606 PMCID: PMC3930911 DOI: 10.3389/fped.2014.00008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/24/2014] [Indexed: 12/28/2022] Open
Abstract
Epidemiological studies have demonstrated an increased prevalence of birth defects and intrauterine growth restriction (IUGR) among infants born prematurely suggesting they share common biological determinants. The identification of key regulatory pathways contributing to this nexus is essential to ongoing efforts to develop effective intervention strategies. Plac1 is a paternally imprinted and X-linked gene that conforms to this paradigm. Examination of a mutant mouse model has confirmed that Plac1 is essential for normal placental development and function. Moreover, it is expressed throughout the developing embryo indicating that it also has broad relevance to embryogenesis. Most notably, its absence in the developing embryo is associated with abnormal brain development and an increased risk of lethal, postnatal hydrocephalus identifying it as a novel, X-linked determinant of brain development. The essential and non-redundant roles of Plac1 in placental and neurological development represent a novel regulatory paradigm for embryonic growth and pregnancy maintenance. Regulatory pathways influenced, in part, by Plac1 are likely to contribute to the observed nexus of IUGR, prematurity, and birth defects.
Collapse
Affiliation(s)
- Michael E Fant
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA ; Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida , Tampa, FL , USA ; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Juan Fuentes
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Xiaoyuan Kong
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Suzanne Jackman
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| |
Collapse
|
22
|
Jackman SM, Kong X, Fant ME. Plac1 (placenta-specific 1) is essential for normal placental and embryonic development. Mol Reprod Dev 2012; 79:564-72. [PMID: 22729990 DOI: 10.1002/mrd.22062] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 06/05/2012] [Indexed: 12/28/2022]
Abstract
Plac1 is a recently identified, X-linked gene whose expression is restricted primarily to cells of the trophoblast lineage. It localizes to a chromosomal locus previously implicated in placental growth. We therefore sought to determine if Plac1 is necessary for placental and embryonic development by examining a mutant mouse model. Plac1 ablation resulted in placentomegaly and mild intrauterine growth retardation (IUGR). At E16.5, knockout (KO) and heterozygous (Het) placentae of the Plac1-null allele inherited from the mother (X(m-) X) weighed approximately 100% more than wildtype (WT) placentae, whereas the corresponding embryos weighed 7-12% less. Histologically, Plac1 mutants exhibited an expanded spongiotrophoblast layer that invaded the labyrinth. By contrast, Het placentae that inherited the null allele from the father (XX(p-) ) exhibited normal growth and were histologically indistinguishable from WT placentae, consistent with paternal imprinting of Plac1. When examined across gestation, WT and X(m-) X placental weights peaked at E16.5 and decreased slightly thereafter. KO placentae (X(m-) X(p-) and X(m-) Y), however, continued to increase in weight after E16.5, consistent with a functional role for the paternal Plac1 allele. Subsequent analysis confirmed that the paternal allele partially escapes complete X-inactivation and thus contributes to placental growth regulation. Additionally, although male Plac1 KO mice can survive, they exhibit decreased viability as a consequence of events occurring late in gestation or shortly after birth. Thus, Plac1 is a paternally imprinted, X-linked gene essential for normal placental and embryonic development.
Collapse
Affiliation(s)
- Suzanne M Jackman
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | |
Collapse
|
23
|
Differential microRNA expression analysis in blastocysts by whole mount in situ hybridization and reverse transcription quantitative polymerase chain reaction on laser capture microdissection samples. Anal Biochem 2012; 423:93-101. [DOI: 10.1016/j.ab.2012.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 12/22/2022]
|
24
|
Chen Y, Moradin A, Schlessinger D, Nagaraja R. RXRα and LXR activate two promoters in placenta- and tumor-specific expression of PLAC1. Placenta 2011; 32:877-84. [PMID: 21937108 DOI: 10.1016/j.placenta.2011.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/19/2011] [Accepted: 08/24/2011] [Indexed: 01/05/2023]
Abstract
PLAC1 expression, first characterized as restricted to developing placenta among normal tissues, is also found in a wide range of tumors and transformed cell lines. To understand the basis for its unusual expression profile, we have analyzed the gene structure and its mode of transcription. We find that the gene has a hitherto unique feature, with two promoters, P1 and P2, separated by 105 kb. P2 has been described before. Here we define P1 and show that it and P2 are activated by RXRα in conjunction with LXRα or LXRβ. In placenta, P2 is the preferred promoter, whereas various tumor cell lines tend to express predominantly either one or the other promoter. Furthermore, when each promoter is fused to a luciferase reporter gene and transfected into cancer cell lines, the promoter corresponding to the more active endogenous promoter is preferentially transcribed. Joint expression of activating nuclear receptors can partially account for the restricted expression of PLAC1 in placenta, and may be co-opted for preferential P1 or P2 PLAC1 expression in various tumor cells.
Collapse
Affiliation(s)
- Y Chen
- Laboratory of Genetics, National Institute on Aging, Bayview Research Center, 251 Bayview Blvd, RM 10B117, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
25
|
Dioxin exposure of human CD34+ hemopoietic cells induces gene expression modulation that recapitulates its in vivo clinical and biological effects. Toxicology 2011; 283:18-23. [DOI: 10.1016/j.tox.2011.01.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 01/25/2011] [Accepted: 01/28/2011] [Indexed: 11/22/2022]
|
26
|
Fant M, Farina A, Nagaraja R, Schlessinger D. PLAC1 (Placenta-specific 1): a novel, X-linked gene with roles in reproductive and cancer biology. Prenat Diagn 2010; 30:497-502. [PMID: 20509147 PMCID: PMC4627609 DOI: 10.1002/pd.2506] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Placenta-specific 1 (PLAC1) is a recently described X-linked gene with expression restricted primarily to cells derived from trophoblast lineage during embryonic development. PLAC1 localizes to a region of the X chromosome thought to be important in placental development although its role in this process has not been defined. This review summarizes our current understanding of its expression, regulation, and function. PLAC1 is expressed throughout human pregnancy by the differentiated trophoblast and localizes to membranous structures in the syncytiotrophoblast, including the microvillous plasma membrane surface. Recent studies have demonstrated that PLAC1 is also expressed by a wide variety of human cancers. Studies of the PLAC1 promoter regions indicate that its expression in both normal placenta and cancer cells is driven by specific interactions involving a combination of transcription factors. Although functional insight into PLAC1 in the normal trophoblast is lacking, preliminary studies suggest that cancer-derived PLAC1 has the potential to promote tumor growth and function. In addition, it also appears to elicit a specific immunologic response that may influence survival in some cancer patients, suggesting that it may provide a therapeutic target for the treatment of some cancers. We also discuss a potential role for PLAC1 as a biomarker predictive of specific pregnancy complications, such as preeclampsia.
Collapse
Affiliation(s)
- Michael Fant
- Department of Pediatrics, University of South Florida College of Medicine, Tampa, FL 33606-350, USA.
| | | | | | | |
Collapse
|
27
|
Mouillet JF, Chu T, Nelson DM, Mishima T, Sadovsky Y. MiR-205 silences MED1 in hypoxic primary human trophoblasts. FASEB J 2010; 24:2030-9. [PMID: 20065103 DOI: 10.1096/fj.09-149724] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Acting through degradation of target mRNA or inhibition of translation, microRNAs (miRNAs) regulate development, differentiation, and cellular response to diverse cues. We analyzed changes in miRNA expression in human placental trophoblasts exposed to hypoxia, which may result from hypoperfusion and placental injury. Using an miRNA microarray screen, confirmed by Northern blot analysis, we defined a set of seven miRNAs (miR-93, miR-205, miR-224, miR-335, miR-424, miR-451, and miR-491) that are differentially regulated in primary trophoblasts exposed to hypoxia. We combined in silico prediction of miRNA targets with gene expression profiling data to identify a series of potential targets for the miRNAs, which were further analyzed using luciferase reporter assays. Among experimentally confirmed targets, we found that the transcriptional coactivator MED1, which plays an important role in placental development, is a target for miR-205. Using gain- and loss-of-function assays, we confirmed that miR-205 interacts with a specific target in the 3'-UTR sequence of MED1 and silences MED1 expression in human trophoblasts exposed to hypoxia, suggesting that miR-205 plays a role in trophoblast injury.
Collapse
Affiliation(s)
- Jean-Francois Mouillet
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
28
|
Koslowski M, Türeci O, Biesterfeld S, Seitz G, Huber C, Sahin U. Selective activation of trophoblast-specific PLAC1 in breast cancer by CCAAT/enhancer-binding protein beta (C/EBPbeta) isoform 2. J Biol Chem 2009; 284:28607-15. [PMID: 19652226 DOI: 10.1074/jbc.m109.031120] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The trophoblast-specific gene PLAC1 (placenta-specific 1) is ectopically expressed in a wide range of human malignancies, most frequently in breast cancer, and is essentially involved in cancer cell proliferation, migration, and invasion. Here we show that basal activity of the PLAC1 promoter is selectively controlled by ubiquitous transcription factor SP1 and isoform 2 of CCAAT/enhancer-binding protein beta that we found to be selectively expressed in placental tissue and cancer cells. Binding of both factors to their respective elements within the PLAC1 promoter was essential to attain full promoter activity. Estrogen receptor alpha (ERalpha) signaling further augmented transcription and translation of PLAC1 and most likely accounts for the positive correlation between PLAC1 expression levels and the ERalpha status we observed in primary breast cancer specimens. DNA affinity precipitation and chromatin immunoprecipitation assays revealed that transactivation of the PLAC1 promoter by ligand-activated ERalpha is based on a nonclassical pathway independent of estrogen-response elements, by tethering of ERalpha to DNA-bound CCAAT/enhancer-binding protein beta-2, and SP1. Our findings provide first insight into a novel and hitherto unknown regulatory mechanism governing selective activation of trophoblast-specific gene expression in breast cancer.
Collapse
Affiliation(s)
- Michael Koslowski
- Department of Internal Medicine III, Experimental and Translational Oncology, Johannes Gutenberg University, 55131 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Affiliation(s)
- Saara M. Rawn
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, and the Graduate Program in Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; ,
| | - James C. Cross
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, and the Graduate Program in Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; ,
| |
Collapse
|
30
|
Dong XY, Peng JR, Ye YJ, Chen HS, Zhang LJ, Pang XW, Li Y, Zhang Y, Wang S, Fant ME, Yin YH, Chen WF. Plac1 is a tumor-specific antigen capable of eliciting spontaneous antibody responses in human cancer patients. Int J Cancer 2008; 122:2038-43. [PMID: 18183594 DOI: 10.1002/ijc.23341] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunoselection and tumor evasion constitutes one of the major obstacles in cancer immunotherapy. A potential solution to this problem is the development of polyvalent vaccines, and the identification of more tumor-specific antigens is a prerequisite for the development of cancer vaccines. To identify novel tumor-specific antigens, suppression subtractive hybridization (SSH) was performed to isolate genes differentially expressed in human hepatocellular cancer (HCC) tissues. PLAC1 (PLACenta-specific 1) was one of the genes identified highly expressed in HCC tissues but not in paired noncancerous tissues. Further analyses revealed its expression in several other types of cancer tissues as well as tumor cell lines, but not in normal tissues except for placenta. Among HCC samples tested, 32% (22/69) showed PLAC1 mRNA expression while the protein was detected in 23.3% (7/30). A serological survey revealed that 3.8% (4/101) of HCC patients had anti-PLAC1 antibody response, suggesting the immunogenicity of PLAC1 in HCC patients. PLAC1 represents a new class of tumor associated antigen with restricted expression in placenta and cancer tissues, that may serve as a target for cancer vaccination.
Collapse
Affiliation(s)
- Xue-Yuan Dong
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Torry DS, Leavenworth J, Chang M, Maheshwari V, Groesch K, Ball ER, Torry RJ. Angiogenesis in implantation. J Assist Reprod Genet 2007; 24:303-15. [PMID: 17616801 PMCID: PMC3455012 DOI: 10.1007/s10815-007-9152-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Implantation failure and early pregnancy loss are common following natural conceptions and they are particularly important clinical hurdles to overcome following assisted reproduction attempts. The importance of adequate vascular development and maintenance during implantation has recently become a major focus of investigation. MATERIALS AND METHODS Review of current published literature was undertaken to summerize the cells and cell products that regulate tissue vascularity during implantation. RESULTS Vascular development at the maternal fetal interface can be regulated by a number of different cell types; two principal candidates are trophoblast and natural killer cells. A wide range of soluble factors, some with well established angiogenic functions as well as other more novel factors, can contribute to vascular development and maintenance at the maternal-fetal interface. CONCLUSIONS Robust vascular development occurs during implantation and early placentation of normal pregnancies. Studies to define the extent and mechanisms by which defects in vascularity contribute to human implantation failure and early miscarriage need to be undertaken.
Collapse
Affiliation(s)
- Donald S Torry
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, P.O. Box 19626, Springfield, IL 62794-9626, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Koslowski M, Sahin U, Mitnacht-Kraus R, Seitz G, Huber C, Türeci O. A placenta-specific gene ectopically activated in many human cancers is essentially involved in malignant cell processes. Cancer Res 2007; 67:9528-34. [PMID: 17909063 DOI: 10.1158/0008-5472.can-07-1350] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification and functional characterization of tumor-specific genes is a prerequisite for the development of targeted cancer therapies. Using an integrated data mining and experimental validation approach for the discovery of new targets for antibody therapy of cancer, we identified PLAC1. PLAC1 is a placenta-specific gene with no detectable expression in any other normal human tissue. However, it is frequently aberrantly activated and highly expressed in a variety of tumor types, in particular breast cancer. RNAi-mediated silencing of PLAC1 in MCF-7 and BT-549 breast cancer cells profoundly impairs motility, migration, and invasion and induces a G1-S cell cycle block with nearly complete abrogation of proliferation. Knockdown of PLAC1 is associated with decreased expression of cyclin D1 and reduced phosphorylation of AKT kinase. Moreover, PLAC1 is localized on the surface of cancer cells and is accessible for antibodies which antagonize biological functions of this molecule. These features, in summary, make PLAC1 an attractive candidate for targeted immunotherapeutic approaches.
Collapse
Affiliation(s)
- Michael Koslowski
- Department of Internal Medicine III, Division of Experimental and Translational Oncology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Bhat P, Anderson DA. Hepatitis B virus translocates across a trophoblastic barrier. J Virol 2007; 81:7200-7. [PMID: 17442714 PMCID: PMC1933314 DOI: 10.1128/jvi.02371-06] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Accepted: 04/08/2007] [Indexed: 12/20/2022] Open
Abstract
Mother-infant transmission of hepatitis B virus (HBV) accounts for up to 30% of worldwide chronic infections. The mechanism and high-risk period of HBV transmission from mother to infant are unknown. Although largely prevented by neonatal vaccination, significant transmission continues to occur in high-risk populations. It is unclear whether HBV can traverse an intact epithelial barrier to infect a new host. Transplacental transmission of a number of viruses relies on transcytotic pathways across placental cells. We wished to determine whether infectious HBV can traverse a polarized trophoblast monolayer. We used a human placenta-derived cell line, BeWo, cultured on membranes as polarized monolayers, to model the maternal-fetal barrier. We assessed the effects of placental maturity and maternal immunoglobulin on viral transport. Intracellular viral trafficking pathways were investigated by confocal microscopy. Free HBV (and infectious duck hepatitis B virus) transcytosed across trophoblastic cells at a rate of 5% in 30 min. Viral transport occurred in microtubule-dependent endosomal vesicles. Additionally, confocal microscopy showed that the internalized virus traverses a monensin-sensitive endosomal compartment. Differentiation of the cytotrophoblasts to syncytiotrophoblasts resulted in a 25% reduction in viral transcytosis, suggesting that placental maturity may protect the fetus. Virus translocation was also reduced in the presence of HBV immunoglobulin. We show for the first time that transcytosis of infectious hepadnavirus can occur across a trophoblastic barrier early in gestation, with the risk of transmission being reduced by placental maturity and specific maternal antibody. This study suggests a mechanism by which mother-infant transmission may occur.
Collapse
Affiliation(s)
- Purnima Bhat
- School of Biomedical Sciences, The University of Queensland, St. Lucia 4072, Australia.
| | | |
Collapse
|
34
|
Fant M, Barerra-Saldana H, Dubinsky W, Poindexter B, Bick R. The PLAC1 protein localizes to membranous compartments in the apical region of the syncytiotrophoblast. Mol Reprod Dev 2007; 74:922-9. [PMID: 17186554 DOI: 10.1002/mrd.20673] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
PLAC1 is a trophoblast-specific gene that maps to a locus on the X-chromosome important to placental development. We have previously shown that PLAC1 gene expression is linked to trophoblast differentiation. The objective of this study was to define the localization of the PLAC1 polypeptide as a prerequisite to understanding its function. Polyclonal antibodies specific for the putative PLAC1 polypeptide were generated. The subcellular localization of PLAC1 in the trophoblast was examined by immunohistochemical analysis of human placenta complemented by immunoblot analysis of subcellular fractions. Brightfield immunohistochemical analysis of placental tissue indicated that the PLAC1 protein localizes to the differentiated syncytiotrophoblast in the apical region of the cell. Deconvlution immunofluorescence microscopy confirmed localization to the apical region of the syncytiotrophoblast. Its distribution included both intracellular compartments as well as loci in close association with the maternal-facing, microvillous brush border membrane (MVM). These findings were supported by immunoblot analysis of subcellular fractions. A 30 kDa band was associated with the microsomal fraction of placental lysates but not the mitochondrial, nuclear, or soluble fractions, suggesting PLAC1 is targeted to a membrane location. Plasma membranes were obtained from the fetal-facing, basal surface (BM) and the maternal-facing, MVM of the syncytiotrophoblast membrane. PLAC1 immunoreactivity was only detected in membrane fractions derived from the apical MVM consistent with immunohistochemical analyses. These data demonstrate that the PLAC1 protein is restricted primarily to the differentiated trophoblast, localizing to intracellular membranous compartment(s) in the apical region of the syncytiotrophoblast and associated with its apical, microvillous membrane surface.
Collapse
Affiliation(s)
- Michael Fant
- Department of Pediatrics, University of Texas Health Science Center, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
35
|
Abstract
The mammalian genome contains multiple genetic factions with distinct interests in the outcomes of interactions among kin. In the context of an offspring's relations with its mother, these factions are proposed to align into two 'parties', one favoring increased demand by offspring and the other favoring reduced demand. A possible alignment has inhibitors of demand located on the X chromosome and enhancers of demand located on autosomes, because X-linked loci are maternally derived two-thirds of the time by contrast to autosomal loci which are maternally derived half of the time.
Collapse
Affiliation(s)
- D Haig
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|