1
|
Hou L, Hong H, Cao W, Wei L, Weng L, Yuan S, Xiao C, Zhang Q, Wang Q, Lai D. Identification and characterization of multipotential stem cells in immortalized normal ovarian surface epithelial cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:239-254. [PMID: 38243680 PMCID: PMC10984850 DOI: 10.3724/abbs.2023253] [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] [Received: 07/22/2023] [Accepted: 09/21/2023] [Indexed: 01/21/2024] Open
Abstract
The ovarian surface epithelium (OSE) is a single layer of squamous-to-cuboidal epithelial cells that experience repetitive ovulatory rupture and subsequent repair. However, the characteristics of human immortalized ovarian surface epithelial cells (IOSE80) remain elusive. This study aims to determine whether IOSE80 cells have the characteristics of stem cell proliferation and multilineage differentiation and their application in regenerative medicine. IOSE80 cells are sequenced by high-throughput transcriptome analysis, and 5 sets of public data are used to compare the differences between IOSE80 cells and bone marrow mesenchymal stem cells, pluripotent stem cells, and oocytes in transcriptome profiling. The IOSE80 cells present a cobblestone-like monolayer and express the epithelial cell marker KRT18; the stem cell markers IFITM3, ALDH1A1, and VIM; lowly express stem cell marker LGR5 and germ cell markers DDX4 and DAZL. In addition, the GO terms "regulation of stem cell proliferation", "epithelial cell proliferation", etc., are significantly enriched ( P<0.05). IOSE80 cells have the potential to act as mesenchymal stem cells to differentiate into adipocytes with lipid droplets, osteoblasts, and chondroblasts in vitro. IOSE80 cells express pluripotent stem cell markers, including OCT4, SSEA4, TRA-1-60, and TRA-1-81, and they can be induced into three germ layers in vitro. IOSE80 cells also form oocyte-like cells in vitro and in vivo. In addition, IOSE80 cells exhibit robust proliferation, migration, and ovarian repair functions after in vivo transplantation. This study demonstrates that IOSE80 cells have the characteristics of pluripotent/multipotent stem cells, indicating their important role in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lin Hou
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Hanqing Hong
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Wenjiao Cao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Liutong Wei
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Lichun Weng
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Shuang Yuan
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qian Wang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Dongmei Lai
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| |
Collapse
|
2
|
Chumduri C, Turco MY. Organoids of the female reproductive tract. J Mol Med (Berl) 2021; 99:531-553. [PMID: 33580825 PMCID: PMC8026429 DOI: 10.1007/s00109-020-02028-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Healthy functioning of the female reproductive tract (FRT) depends on balanced and dynamic regulation by hormones during the menstrual cycle, pregnancy and childbirth. The mucosal epithelial lining of different regions of the FRT-ovaries, fallopian tubes, uterus, cervix and vagina-facilitates the selective transport of gametes and successful transfer of the zygote to the uterus where it implants and pregnancy takes place. It also prevents pathogen entry. Recent developments in three-dimensional (3D) organoid systems from the FRT now provide crucial experimental models that recapitulate the cellular heterogeneity and physiological, anatomical and functional properties of the organ in vitro. In this review, we summarise the state of the art on organoids generated from different regions of the FRT. We discuss the potential applications of these powerful in vitro models to study normal physiology, fertility, infections, diseases, drug discovery and personalised medicine.
Collapse
Affiliation(s)
- Cindrilla Chumduri
- Department of Microbiology, University of Würzburg, Biocenter, Würzburg, Germany.
- Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Cambridge, UK.
| |
Collapse
|
3
|
Bharti D, Tikka M, Lee SY, Bok EY, Lee HJ, Rho GJ. Female Germ Cell Development, Functioning and Associated Adversities under Unfavorable Circumstances. Int J Mol Sci 2021; 22:1979. [PMID: 33671303 PMCID: PMC7922109 DOI: 10.3390/ijms22041979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/07/2023] Open
Abstract
In the present era, infertility is one of the major issues which restricts many couples to have their own children. Infertility is the inability to achieve a clinical pregnancy after regular unprotected sexual intercourse for the period of one year or more. Various factors including defective male or female germ cell development, unhealthy and improper lifestyles, diseases like cancer and associated chemo-or-radiation therapies, congenital disorders, etc., may be responsible for infertility. Therefore, it is highly important to understand the basic concepts of germ cell development including primordial germ cell (PGC) formation, specification, migration, entry to genital ridges and their molecular mechanisms, activated pathways, paracrine and autocrine signaling, along with possible alteration which can hamper germ cell development and can cause adversities like cancer progression and infertility. Knowing all these aspects in a proper way can be very much helpful in improving our understanding about gametogenesis and finding possible ways to cure related disorders. Here in this review, various aspects of gametogenesis especially female gametes and relevant factors causing functional impairment have been thoroughly discussed.
Collapse
Affiliation(s)
- Dinesh Bharti
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-Y.L.); (E.-Y.B.)
| | - Manisha Tikka
- Department of Zoology and Environmental Sciences, Punjabi University, Patiala 147002, India;
| | - Sang-Yun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-Y.L.); (E.-Y.B.)
| | - Eun-Yeong Bok
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-Y.L.); (E.-Y.B.)
| | - Hyeon-Jeong Lee
- Department of Medicine, University of California, San Diego, CA 92093-0021, USA;
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-Y.L.); (E.-Y.B.)
| |
Collapse
|
4
|
Yan D, Liu X, Xu H, Guo SW. Mesothelial Cells Participate in Endometriosis Fibrogenesis Through Platelet-Induced Mesothelial-Mesenchymal Transition. J Clin Endocrinol Metab 2020; 105:5894452. [PMID: 32813013 DOI: 10.1210/clinem/dgaa550] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT While fibrosis in endometriosis has recently loomed prominently, the sources of myofibroblasts, the principal effector cell in fibrotic diseases, remain largely obscure. Mesothelial cells (MCs) can be converted into myofibroblasts through mesothelial-mesenchymal transition (MMT) in many fibrotic diseases and adhesion. OBJECTIVE To evaluate whether MCs contribute to the progression and fibrogenesis in endometriosis through MMT. SETTING, DESIGN, PATIENTS, INTERVENTION, AND MAIN OUTCOME MEASURES Dual immunofluorescence staining and immunohistochemistry using antibodies against calretinin, Wilms' tumor-1 (WT-1), and α-smooth muscle actin (α-SMA) were performed on lesion samples from 30 patients each with ovarian endometrioma (OE) and deep endometriosis (DE), and 30 normal endometrial (NE) tissue samples. Human pleural and peritoneal MCs were co-cultured with activated platelets or control medium with and without neutralization of transforming growth factor β1 (TGF-β1) and/or platelet-derived growth factor receptor (PDGFR) and their morphology, proliferation, and expression levels of genes and proteins known to be involved in MMT were evaluated, along with their migratory and invasive propensity, contractility, and collagen production. RESULTS The number of calretinin/WT-1 and α-SMA dual-positive fibroblasts in OE/DE lesions was significantly higher than NE samples. The extent of lesional fibrosis correlated positively with the lesional α-SMA staining levels. Human MCs co-cultured with activated platelets acquire a morphology suggestive of MMT, concomitant with increased proliferation, loss of calretinin expression, and marked increase in expression of mesenchymal markers. These changes coincided with functional differentiation as reflected by increased migratory and invasive capacity, contractility, and collagen production. Neutralization of TGF-β1 and PDGFR signaling abolished platelet-induced MMT in MCs. CONCLUSIONS MCs contribute to lesional progression and fibrosis through platelet-induced MMT.
Collapse
Affiliation(s)
- Dingmin Yan
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Hong Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Silvestrini VC, Lanfredi GP, Masson AP, Poersch A, Ferreira GA, Thomé CH, Faça VM. A proteomics outlook towards the elucidation of epithelial-mesenchymal transition molecular events. Mol Omics 2020; 15:316-330. [PMID: 31429845 DOI: 10.1039/c9mo00095j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The main cause of death in cancer is the spread, or metastasis, of cancer cells to distant organs with consequent tumor formation. Additionally, metastasis is a process that demands special attention, as the cellular transformations make cancer at this stage very difficult or occasionally even impossible to be cured. The main process that converts epithelial tumor cells to mesenchymal-like metastatic cells is the Epithelial to Mesenchymal Transition (EMT). This process allows stationary and polarized epithelial cells, which are connected laterally to several types of junctions as well as the basement membrane, to undergo multiple biochemical changes that enable disruption of cell-cell adherence and apical-basal polarity. Moreover, the cells undergo important reprogramming to remodel the cytoskeleton and acquire mesenchymal characteristics such as enhanced migratory capacity, invasiveness, elevated resistance to apoptosis and a large increase in the production of ECM components. As expected, the alterations of the protein complement are extensive and complex, and thus exploring this by proteomic approaches is of particular interest. Here we review the overall findings of proteome modifications during EMT, mainly focusing on molecular signatures observed in multiple proteomic studies as well as coordinated pathways, cellular processes and their clinical relevance for altered proteins. As a result, an interesting set of proteins is highlighted as potential targets to be further investigated in the context of EMT, metastasis and cancer progression.
Collapse
Affiliation(s)
- Virgínia Campos Silvestrini
- Department of Biochemistry and Immunology - FMRP - University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil.
| | | | | | | | | | | | | |
Collapse
|
6
|
Mesothelial to mesenchyme transition as a major developmental and pathological player in trunk organs and their cavities. Commun Biol 2018; 1:170. [PMID: 30345394 PMCID: PMC6191446 DOI: 10.1038/s42003-018-0180-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022] Open
Abstract
The internal organs embedded in the cavities are lined by an epithelial monolayer termed the mesothelium. The mesothelium is increasingly implicated in driving various internal organ pathologies, as many of the normal embryonic developmental pathways acting in mesothelial cells, such as those regulating epithelial-to-mesenchymal transition, also drive disease progression in adult life. Here, we summarize observations from different animal models and organ systems that collectively point toward a central role of epithelial-to-mesenchymal transition in driving tissue fibrosis, acute scarring, and cancer metastasis. Thus, drugs targeting pathways of mesothelium’s transition may have broad therapeutic benefits in patients suffering from these diseases. Tim Koopmans and Yuval Rinkevich review recent findings linking the mesothelium’s embryonic programs that drive epithelial-to-mesenchyme transition with adult pathologies, such as fibrosis, acute scarring, and cancer metastasis. They highlight new avenues for drug development that would target pathways of the mesothelium’s mesenchymal transition.
Collapse
|
7
|
Tomono T, Machida T, Kamioka H, Shibasaki Y, Yano K, Ogihara T. Entinostat reverses P-glycoprotein activation in snail-overexpressing adenocarcinoma HCC827 cells. PLoS One 2018; 13:e0200015. [PMID: 29979729 PMCID: PMC6034804 DOI: 10.1371/journal.pone.0200015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) in cancer cells facilitates tumor progression by promoting invasion and metastasis. Snail is a transcriptional factor that induces EMT, while P-glycoprotein (P-gp) is an efflux transporter involved in anticancer drug resistance, and P-gp efflux activity is stimulated in Snail-overexpressing lung cancer cells with EMT characteristics. Since the histone deacetylase (HDAC) inhibitor entinostat (Ent) reverses EMT features, our aim in this study was to determine whether Ent also suppresses P-gp activation in Snail-induced cells. First, we confirmed that Ent treatment reduced migration activity, downregulated E-cadherin and upregulated vimentin at the mRNA level in Snail-overexpressing cells, thus inhibiting EMT. Efflux and uptake assays using rhodamine123 (Rho123), a fluorescent P-gp substrate, showed that Ent also inhibited Snail-induced activation of P-gp. Moreover, P-gp activity was more strongly inhibited by Ent in Snail-overexpressing cells than in Mock cells. When we evaluated the uptakes of Rho123 by LLC-PK1 cells and P-gp-overexpressing LLC-GA5COL150 cells, Rho123 accumulation in LLC-GA5COL150 cells was significantly decreased compared with that in LLC-PK1 cells. Coincubation with Ent had no effect on Rho123 accumulation in either of the cell lines. Thus, Ent appears to be an inhibitor, but not a substrate, of P-gp at low concentration. Our results suggest that Ent treatment might suppress not only Snail-induced cancer malignant alteration, but also P-gp-mediated multidrug resistance.
Collapse
Affiliation(s)
- Takumi Tomono
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
| | - Tatsuya Machida
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
| | - Hiroki Kamioka
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
| | - Yumi Shibasaki
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
| | - Kentaro Yano
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
| | - Takuo Ogihara
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan
- * E-mail:
| |
Collapse
|
8
|
Alwosaibai K, Abedini A, Al-Hujaily EM, Tang Y, Garson K, Collins O, Vanderhyden BC. PAX2 maintains the differentiation of mouse oviductal epithelium and inhibits the transition to a stem cell-like state. Oncotarget 2017; 8:76881-76897. [PMID: 29100356 PMCID: PMC5652750 DOI: 10.18632/oncotarget.20173] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/18/2017] [Indexed: 12/18/2022] Open
Abstract
Recent studies have provided evidence that the secretory cells of the fallopian tube (oviduct) are a probable origin for high-grade serous ovarian carcinoma. In addition to secretory cells, the fallopian tube epithelium consists of ciliated cells and CD44+ undifferentiated stem-like cells. Loss of PAX2 expression is recognized as an early event in epithelial transformation, but the specific role of PAX2 in this transition is unknown. The aim of this study was to define the role of PAX2 in oviductal epithelial (OVE) cells and its response to transforming growth factor β1 (TGFβ), characterizing specifically its potential involvement in regulating stem cell-like behaviors that may contribute to formation of cancer-initiating cells. Treatment of primary cultures of mouse OVE cells with TGFβ induced an epithelial-mesenchymal transition (EMT) associated with decreased expression of PAX2 and an increase in the fraction of cells expressing CD44. PAX2 knockdown in OVE cells and overexpression in ovarian epithelial cells confirmed that PAX2 inhibits stem cell characteristics and regulates the degree of epithelial differentiation of OVE cells. These results suggest that loss of PAX2, as occurs in serous tubal intraepithelial carcinomas, may shift secretory cells to a more mesenchymal phenotype associated with stem-like features.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada.,King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Atefeh Abedini
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada
| | - Ensaf M Al-Hujaily
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada
| | - Yong Tang
- Department of Urology, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, China
| | - Kenneth Garson
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada
| | - Olga Collins
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada
| | - Barbara C Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ontario, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Ottawa, Ontario, Canada
| |
Collapse
|
9
|
Rashed HE, Hussein S, Mosaad H, Abdelwahab MM, Abdelhamid MI, Mohamed SY, Mohamed AM, Fayed A. Prognostic significance of the genetic and the immunohistochemical expression of epithelial-mesenchymal-related markers in colon cancer. Cancer Biomark 2017; 20:107-122. [PMID: 28759954 DOI: 10.3233/cbm-170034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is one of the main events in colorectal cancer (CRC) spread. Snail-1 is a zinc transcription factor that mediates EMT in tumor cells probably by down-regulation of E-cadherin and claudin-1. OBJECTIVES To detect the expression of epithelial markers (claudin-1 and E-cadherin) and mesenchymal markers (snail-1 and vimentin) in primary cancer colon. Also, to select stage II cancer patients of a high risk that can benefit from postoperative adjuvant chemotherapy. METHODS Reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemical analysis were performed to investigate snail-1, claudin-1, E-cadherin and vimentin expressions at mRNA and protein levels in fresh tissues of cancer colon and normal colonic mucosa. The correlations between the expression of these markers and clinicopathological parameters were performed. RESULTS Normal colonic mucosa revealed complete membranous expression of claudin-1, preserved E-cadherin and negative snail-1 and vimentin expressions. Compared to control, the expression of snail-1 and vimentin mRNA in cancer colon was significantly up-regulated while the expression of claudin-1 and E-cadherin mRNA was significantly down-regulated. These changes were significantly associated with stage and lymph node involvement at both mRNA and protein levels (p< 0.05). There were significant negative correlations between vimentin and each of E-cadherin and claudin-1 gene expression and between snail-1 and each of E-cadherin and claudin-1 gene expression. Moreover, these changes were independent predictors of recurrence of stage II cancer colon cases. CONCLUSION There is a clinical significance of snail-1, claudin-1, E-cadherin and vimentin as possible markers for recognizing patients with lymph node involvement, advanced stage and high incidence of tumor recurrence in cancer colon.
Collapse
Affiliation(s)
- Hayam E Rashed
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samia Hussein
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hala Mosaad
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mai M Abdelwahab
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed I Abdelhamid
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Salem Y Mohamed
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abdel Motaleb Mohamed
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Alaa Fayed
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Bilyk O, Coatham M, Jewer M, Postovit LM. Epithelial-to-Mesenchymal Transition in the Female Reproductive Tract: From Normal Functioning to Disease Pathology. Front Oncol 2017; 7:145. [PMID: 28725636 PMCID: PMC5497565 DOI: 10.3389/fonc.2017.00145] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/21/2017] [Indexed: 12/15/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a physiological process that is vital throughout the human lifespan. In addition to contributing to the development of various tissues within the growing embryo, EMT is also responsible for wound healing and tissue regeneration later in adulthood. In this review, we highlight the importance of EMT in the development and normal functioning of the female reproductive organs (the ovaries and the uterus) and describe how dysregulation of EMT can lead to pathological conditions, such as endometriosis, adenomyosis, and carcinogenesis. We also summarize the current literature relating to EMT in the context of ovarian and endometrial carcinomas, with a particular focus on how molecular mechanisms and the tumor microenvironment can govern cancer cell plasticity, therapy resistance, and metastasis.
Collapse
Affiliation(s)
- Olena Bilyk
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Mackenzie Coatham
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Michael Jewer
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | | |
Collapse
|
11
|
Gutgold N, Davidson B, Catane LJ, Holth A, Hellesylt E, Tropé CG, Dørum A, Reich R. TGFβ splicing and canonical pathway activation in high-grade serous carcinoma. Virchows Arch 2017; 470:665-678. [PMID: 28432432 DOI: 10.1007/s00428-017-2127-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/05/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
The present study analyzed the expression and clinical role of the transforming growth factor-β (TGFβ) pathway in high-grade serous carcinoma (HGSC), with focus on malignant effusions. TGFβ1-3 and TGFβRI-III mRNA expression by qRT-PCR was analyzed in 70 HGSC effusions and 55 solid specimens (28 ovarian, 27 abdominal metastases). Protein expression of Smad2 and Smad3 and their phosphorylated forms by Western blotting was analyzed in 73 specimens (42 effusions, 13 ovarian carcinomas, 18 solid metastases). Expression was analyzed for association with anatomic site and clinical parameters, including survival. TGFβRI and TGFβRII mRNA was overexpressed in effusions and solid metastases, particularly the former, compared to that in the ovarian tumors (p < 0.001 to p = 0.05), with anatomic site-dependent expression of splice variants. Conversely, Smad2, p-Smad2, and p-Smad3 were overexpressed in solid specimens (ovarian and peritoneal) compared to those in effusions (p < 0.001 for all). In univariate survival analysis, higher TGFβRI variant 1 and TGFβRIII mRNA levels were associated with a trend for shorter overall survival in patients with post-chemotherapy effusions (p = 0.066 and p = 0.087, respectively), and the latter was an independent prognostic marker in Cox multivariate analysis (p = 0.041). Smad3 protein expression was associated with a trend for shorter overall survival in univariate survival analysis (p = 0.052). TGFβ receptor splice variant expression is anatomic site-dependent in HGSC. Elevated levels of TGFβ signaling pathway mRNAs are seen in metastatic HGSC, but are not accompanied by increased Smad expression and activation in HGSC effusions, evidence of failure to activate canonical TGFβ signaling. Assessment of the prognostic role of this pathway in HGSC effusions merits further research.
Collapse
Affiliation(s)
- Neriya Gutgold
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Ben Davidson
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Montebello, N-0310, Oslo, Norway. .,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, N-0316, Oslo, Norway.
| | - Liora Jacobs Catane
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Arild Holth
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Montebello, N-0310, Oslo, Norway
| | - Ellen Hellesylt
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Montebello, N-0310, Oslo, Norway
| | - Claes G Tropé
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Anne Dørum
- Department of Gynecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, N-0310, Oslo, Norway
| | - Reuven Reich
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel. .,David R. Bloom Center for Pharmacy and the Adolf and Klara Brettler Center for Research in Molecular Pharmacology and Therapeutics, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
12
|
Lv J, Sun B, Mai Z, Jiang M, Du J. CLDN-1 promoted the epithelial to migration and mesenchymal transition (EMT) in human bronchial epithelial cells via Notch pathway. Mol Cell Biochem 2017; 432:91-98. [PMID: 28316062 DOI: 10.1007/s11010-017-3000-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/04/2017] [Indexed: 01/11/2023]
Abstract
Claudin-1 (CLDN-1) is one of main tight junction components that play an important role in epithelial-mesenchymal transition (EMT). However, the effects of CLDN-1 on the migration and EMT induced by TGF-β1 in primary normal human bronchial epithelial (NHBE) and BEAS-2B cells have not been clear. The expression of CLDN-1 was quantified by Western blotting in NHBE and BEAS-2B cells. Cell migration and invasion were detected using transwell assays. The expression level of E-cadherin, N-cadherin, α-SMA, and Vimentin was evaluated by quantitative real-time PCR and Western blotting. Here we showed that the protein expression of CLDN-1 was increased exposed to TGF-β1 in a dose- and time-dependent manner. Knockdown of CLDN-1 using small interfering CLDN-1 RNA (siCLDN-1) prevented the migration and invasion in NHBE and BEAS-2B cells. Moreover, depletion of CLDN-1 promoted the E-cadherin expression and decreased the mRNA and protein levels of N-cadherin, α-SMA, and Vimentin induced by TGF-β1. Furthermore, CLDN-1 silencing resulted in the reduction of the Notch intracellular domain (NICD) and hairy enhancer of split-1 (Hes-1) in mRNA and protein level. Jagged-1, an activator of Notch signaling pathway, abrogated the protective function of siCLDN-1 in migration and EMT. In conclusion, CLDN-1 promoted the migration and EMT through the Notch signaling pathway.
Collapse
Affiliation(s)
- Jing Lv
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Baohua Sun
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China.
| | - Zhitao Mai
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Mingming Jiang
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Junfeng Du
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| |
Collapse
|
13
|
Tepekoy F, Ozturk S, Sozen B, Ozay RS, Akkoyunlu G, Demir N. CD90 and CD105 expression in the mouse ovary and testis at different stages of postnatal development. Reprod Biol 2015; 15:195-204. [PMID: 26679159 DOI: 10.1016/j.repbio.2015.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 10/10/2015] [Accepted: 10/13/2015] [Indexed: 11/16/2022]
Abstract
CD90 (i.e., THY1) and CD105 (i.e., endoglin) are glycoproteins known as mesenchymal stem cell markers that are expressed in various cell types including male and female gonadal cells. We aimed to determine ovarian and testicular expression of CD90 and CD105 in various cell types during postnatal development in mice. The present study was carried out on male (C57BL/6) and female (Balb/C) mice during critical stages of gonadal development. Immunohistochemical localization of CD90 and CD105 was determined in the ovaries obtained at postnatal days (PND) -1, -7, -21 and -60 and in the testes obtained at PND6, -8, -16, -20, -29, -32 and -88. The relative expression of CD90 and CD105 was evaluated by ImageJ software and data were analyzed by analysis of variance. The relative expression of CD90 and CD105 varied during postnatal development and increased significantly in the adult ovary (PND60) and testis (PND88) compared to the early postnatal gonads. In the ovaries, the expression of CD90 was significantly higher in somatic cells in comparison to germ cell compartments. In the testis, CD90 expression was greater in germ cells and Sertoli cells compared to other cell types. Expression of CD105 was higher in germ cells than somatic cells of both the ovary and testis. In addition to different expression of CD90 and CD105 during various developmental stages, also their altered expression in particular cell types suggests specific roles of these glycoproteins in physiological processes of mouse gonads.
Collapse
Affiliation(s)
- Filiz Tepekoy
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey
| | - Berna Sozen
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey
| | - Recep S Ozay
- Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey
| | - Gokhan Akkoyunlu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey
| | - Necdet Demir
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070, Campus, Antalya, Turkey.
| |
Collapse
|
14
|
Martínez-Ramírez AS, Garay E, García-Carrancá A, Vázquez-Cuevas FG. The P2RY2 Receptor Induces Carcinoma Cell Migration and EMT Through Cross-Talk With Epidermal Growth Factor Receptor. J Cell Biochem 2015; 117:1016-26. [PMID: 26443721 DOI: 10.1002/jcb.25390] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/05/2015] [Indexed: 11/11/2022]
Abstract
Extracellular nucleotides are signaling elements present in the tumor microenvironment; however, their role in tumor growth is not completely understood. In the present study, we asked whether nucleotides regulate cell migration in ovarian carcinoma-derived cells. We observed that 100 μM UTP induced migration in SKOV-3 cells (1.57 ± 0.08 fold over basal), and RT-PCR showed expression of transcripts for the P2RY2 and P2RY4 receptors. Knockdown of P2RY2 expression in SKOV-3 cells (P2RY2-KD) abolished the UTP-induced migration. The mechanism activated by UTP to induce migration involves transactivation of the epidermal growth factor receptor (EGFR) since we observed that the EGFR kinase inhibitor AG1478 and the PI3K inhibitor Wortmannin inhibit this response (to 0.76 ± 0.23 and 0.46 ± 0.14 relative to the control, respectively). In agreement with these observations, UTP was able to modify the phosphorylation state of the EGFR; likewise, the induction of ERK1/2 phosphorylation promoted by UTP was abolished by a 30-60 min treatment with AG1478. Our data also suggested that the enhanced cell migration involves the epithelium to mesenchymal transition (EMT) process, since a 12 h stimulation of SKOV-3 cells with 100 μM UTP showed an increase in vimentin and SNAIL protein levels (459.8 ± 132.4% over basal for SNAIL). Interestingly, treatment with apyrase (10 U/mL) reduces the migration of control cells and induces a considerable enrichment of E-cadherin in the cell-cell contacts, favoring an epithelial phenotype and strongly suggesting that the nucleotides released by tumor cells and acting through the P2RY2 receptor are potential regulators of invasiveness.
Collapse
Affiliation(s)
- A S Martínez-Ramírez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| | - E Garay
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| | - A García-Carrancá
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. San Fernando #22, Colonia Sección XVI, Tlalpan, CP 14080, Mexico.,División de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, México. Av. San Fernando #22, Colonia Sección XVI, Tlalpan, CP 14080, Mexico
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| |
Collapse
|
15
|
Abstract
The mammalian ovary is covered by a single-layered epithelium that undergoes rupture and remodelling following each ovulation. Although resident stem cells are presumed to be crucial for this cyclic regeneration, their identity and mode of action have been elusive. Surrogate stemness assays and in vivo fate-mapping studies using recently discovered stem cell markers have identified stem cell pools in the ovary and fimbria that ensure epithelial homeostasis. Recent findings provide insights into intrinsic mechanisms and local extrinsic cues that govern the function of ovarian and fimbrial stem cells. These discoveries have advanced our understanding of stem cell biology in the ovary and fimbria, and lay the foundations for evaluating the contribution of resident stem cells to the initiation and progression of human epithelial ovarian cancer.
Collapse
|
16
|
Gao J, Zhu Y, Nilsson M, Sundfeldt K. TGF-β isoforms induce EMT independent migration of ovarian cancer cells. Cancer Cell Int 2014; 14:72. [PMID: 25278811 PMCID: PMC4180856 DOI: 10.1186/s12935-014-0072-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 07/12/2014] [Indexed: 12/23/2022] Open
Abstract
Background Transforming growth factor beta (TGF-β) plays major roles in tumorigenesis by regulating cell growth, epithelial-to-mesenchymal transition (EMT), migration/invasion and metastasis. The epithelial markers E-cadherin, claudin-3 and claudin-4, commonly decreased in human adenocarcinomas are actually up regulated during ovarian carcinogenesis. In human ovarian cancer TGF-β1 may either suppress or promote tumor progression, but whether other TGF-β isoforms (TGF-β2 and TGF-β3) exert similar effects is not known. Methods In this study we investigated the ability of the TGF-β isoforms (TGF-β1-3) to induce proliferation and migration by BrdU labeling, scratch wound and trans-filter migration assays in the human serous adenocarcinoma cell-line NIH-OVCAR3. Transepithelial resistance was measured and EMT observed by light-microscopy. Expression of adherens-, tight-junction and EMT-related transcription factors was analyzed by qRT-PCR and immunoblotting. Results All TGF-β isoforms dose-dependently inhibited NIH-OVCAR3 cell growth, stimulated tumor cell migration with similar efficiency. The mesenchymal marker N-cadherin and claudin-1 expression was induced and occludin down regulated. However, migrating cells retained an epithelial shape and E-cadherin expression. The E-cadherin repressor SNAIL mRNA levels remained low independently of TGF-β1-3 treatment while ZEB1 expression was enhanced. Conclusions TGF-β1, TGF-β2 and TGF-β3 promote migration of NIH-OVCAR3 ovarian cancer cells independently of cell proliferation and without conversion to a complete EMT phenotype. Epithelial ovarian cancer commonly metastasis to the surrounding tissue or inside the peritoneum rather than invading blood vessels to set distance metastasis. Our result raises the question whether ovarian cancer primarily spread via collective migration than via single cell invasion.
Collapse
Affiliation(s)
- Jingfang Gao
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Goteborg, SE-40530 Sweden
| | - Yihong Zhu
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Goteborg, SE-40530 Sweden
| | - Mikael Nilsson
- Sahlgrenska Cancer Center, Institute of Biomedicine, University of Gothenburg, Goteborg, SE-40530 Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Goteborg, SE-40530 Sweden
| |
Collapse
|
17
|
Chen Z, Wang Y, Liu W, Zhao G, Lee S, Balogh A, Zou Y, Guo Y, Zhang Z, Gu W, Li C, Tigyi G, Yue J. Doxycycline inducible Krüppel-like factor 4 lentiviral vector mediates mesenchymal to epithelial transition in ovarian cancer cells. PLoS One 2014; 9:e105331. [PMID: 25137052 PMCID: PMC4138168 DOI: 10.1371/journal.pone.0105331] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/20/2014] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer presents therapeutic challenges due to its typically late detection, aggressive metastasis, and therapeutic resistance. The transcription factor Krüppel-like factor 4 (KLF4) has been implicated in human cancers as a tumor suppressor or oncogene, although its role depends greatly on the cellular context. The role of KLF4 in ovarian cancer has not been elucidated in mechanistic detail. In this study, we investigated the role of KLF4 in ovarian cancer cells by transducing the ovarian cancer cell lines SKOV3 and OVCAR3 with a doxycycline-inducible KLF4 lentiviral vector. Overexpression of KLF4 reduced cell proliferation, migration, and invasion. The epithelial cell marker gene E-cadherin was significantly upregulated, whereas the mesenchymal cell marker genes vimentin, twist1and snail2 (slug) were downregulated in both KLF4-expressing SKOV3 and OVCAR3 cells. KLF4 inhibited the transforming growth factor β (TGFβ)-induced epithelial to mesenchymal transition (EMT) in ovarian cancer cells. Taken together, our data demonstrate that KLF4 functions as a tumor suppressor gene in ovarian cancer cells by inhibiting TGFβ-induced EMT.
Collapse
Affiliation(s)
- Zixuan Chen
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Southern Medical University, Guangzhou, P. R. China
| | - Yinan Wang
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Wen Liu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Guannan Zhao
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, P. R. China
| | - Suechin Lee
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Andrea Balogh
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Yanan Zou
- The Second Affiliated Hospital of Harbin Medical University, Harbin, P. R. China
| | - Yuqi Guo
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, P. R. China
| | - Zhan Zhang
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, P. R. China
| | - Weiwang Gu
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Chengyao Li
- Southern Medical University, Guangzhou, P. R. China
- * E-mail: (JY); (CL)
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail: (JY); (CL)
| |
Collapse
|
18
|
Steinestel K, Eder S, Schrader AJ, Steinestel J. Clinical significance of epithelial-mesenchymal transition. Clin Transl Med 2014; 3:17. [PMID: 25050175 PMCID: PMC4094902 DOI: 10.1186/2001-1326-3-17] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 06/27/2014] [Indexed: 12/21/2022] Open
Abstract
The concept of epithelial-mesenchymal transition (EMT), a process where cells change their epithelial towards a mesenchymal phenotype, has gained overwhelming attention especially in the cancer research community. Thousands of scientific reports investigated changes in gene, mRNA and protein expression compatible with EMT and their possible correlation with tumor invasion, metastatic spread or patient prognosis; however, up to now, a proof of clinical significance of the concept is still missing. This review, with a main focus on the role of EMT in tumors, will summarize the basic molecular events underlying EMT including the signaling pathways capable of its induction as well as changes in EMT-associated protein expression and will very briefly touch the role of microRNAs in EMT. We then outline protein markers that are used most frequently for the assessment of EMT in research and diagnostic evaluation of tumor specimens and depict the link between EMT, a cancer stem cell (CSC) phenotype and resistance to conventional antineoplastic therapies. Furthermore, we evaluate a possible correlation between EMT marker expression and patient prognosis as well as current therapeutic concepts targeting the EMT process to slow down or prevent metastatic spread of malignant tumors.
Collapse
Affiliation(s)
- Konrad Steinestel
- Bundeswehr Institute of Radiobiology, Neuherbergstrasse 11, Munich 80937, Germany
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, Ulm 89081, Germany
| | - Stefan Eder
- Bundeswehr Institute of Radiobiology, Neuherbergstrasse 11, Munich 80937, Germany
| | - Andres Jan Schrader
- Department of Urology, Ulm University Medical Center, Prittwitzstrasse 43, Ulm 89075, Germany
| | - Julie Steinestel
- Department of Urology, Ulm University Medical Center, Prittwitzstrasse 43, Ulm 89075, Germany
| |
Collapse
|
19
|
Kristjánsdóttir B, Partheen K, Fung ET, Yip C, Levan K, Sundfeldt K. Early inflammatory response in epithelial ovarian tumor cyst fluids. Cancer Med 2014; 3:1302-12. [PMID: 24947406 PMCID: PMC4302680 DOI: 10.1002/cam4.282] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/21/2023] Open
Abstract
Mortality rates for epithelial ovarian cancer (EOC) are high, mainly due to late-stage diagnosis. The identification of biomarkers for this cancer could contribute to earlier diagnosis and increased survival rates. Given that chronic inflammation plays a central role in cancer initiation and progression, we selected and tested 15 cancer-related cytokines and growth factors in 38 ovarian cyst fluid samples. We used ovarian cyst fluid since it is found in proximity to the pathology and mined it for inflammatory biomarkers suitable for early detection of EOC. Immunoprecipitation and high-throughput sample fractionation were obtained by using tandem antibody libraries bead and mass spectrometry. Two proteins, monocyte chemoattractant protein-1 (MCP-1/CCL2) and interleucin-8 (IL-8/CXCL8), were significantly (P < 0.0001) higher in the malignant (n = 16) versus benign (n = 22) tumor cysts. Validation of MCP-1, IL-8, and growth-regulated protein-α (GROα/CXCL1) was performed with ELISA in benign, borderline, and malignant cyst fluids (n = 256) and corresponding serum (n = 256). CA125 was measured in serum from all patients and used in the algorithms performed. MCP-1, IL-8, and GROα are proinflammatory cytokines and promoters of tumor growth. From 5- to 100-fold higher concentrations of MCP-1, IL-8 and GROα were detected in the cyst fluids compared to the serum. Significant (P < 0.001) cytokine response was already established in borderline cyst fluids and stage I EOC. In serum a significant (P < 0.01) increase of IL-8 and GROα was found, but not until stage I and stage III EOC, respectively. These findings confirm that early events in tumorigenesis can be analyzed and detected in the tumor environment and we conclude that ovarian cyst fluid is a promising source in the search for new biomarkers for early ovarian tumors.
Collapse
Affiliation(s)
- Björg Kristjánsdóttir
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | |
Collapse
|
20
|
Stammler A, Müller D, Tabuchi Y, Konrad L, Middendorff R. TGFβs modulate permeability of the blood-epididymis barrier in an in vitro model. PLoS One 2013; 8:e80611. [PMID: 24236189 PMCID: PMC3827453 DOI: 10.1371/journal.pone.0080611] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/07/2013] [Indexed: 11/23/2022] Open
Abstract
The blood-epididymis barrier (BEB) is formed by epithelial tight junctions mediating selective permeability of the epididymal epithelium. Defective barrier function can disturb the balance of the epididymal milieu, which may result in infertility. The stroma of the epididymis contains high amounts of cytokines of the TGFβ family of unknown function. We screened possible effects of all three TGFβ isoforms on paracellular tightness in a BEB in vitro model based on the strongly polarized mouse epididymal epithelial MEPC5 cells in the transwell system. In this model we found a robust transepithelial electrical resistance (TER) of about 840 Ω x cm2. Effects on the paracellular permeability were evaluated by two methods, TER and FITC-Dextran-based tracer diffusion assays. Both assays add up to corresponding results indicating a time-dependent disturbance of the BEB differentially for the three TGFβ isoforms (TGFβ3>TGFβ1>TGFβ2) in a TGFβ-recetor-1 kinase- and Smad-dependent manner. The tight junction protein claudin-1 was found to be reduced by the treatment with TGFβs, whereas occludin was not influenced. Epididymal epithelial cells are predominantly responsive to TGFβs from the basolateral side, suggesting that TGFβ may have an impact on the epididymal epithelium from the stroma in vivo. Our data show for the first time that TGFβs decrease paracellular tightness in epididymal epithelial cells, thus establishing a novel mechanism of regulation of BEB permeability, which is elementary for sperm maturation and male fertility.
Collapse
Affiliation(s)
- Angelika Stammler
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
- Department of Gynecology and Obstetrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Dieter Müller
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, Graduate School of Medicine Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Lutz Konrad
- Department of Gynecology and Obstetrics, Justus-Liebig-University Giessen, Giessen, Germany
- * E-mail: (RM); (LK)
| | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
- * E-mail: (RM); (LK)
| |
Collapse
|
21
|
Stimpfel M, Skutella T, Cvjeticanin B, Meznaric M, Dovc P, Novakovic S, Cerkovnik P, Vrtacnik-Bokal E, Virant-Klun I. Isolation, characterization and differentiation of cells expressing pluripotent/multipotent markers from adult human ovaries. Cell Tissue Res 2013; 354:593-607. [PMID: 23820736 DOI: 10.1007/s00441-013-1677-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/28/2013] [Indexed: 01/19/2023]
Abstract
Pluripotent stem cells are still generally accepted not to exist in adult human ovaries, although increasing studies confirm the presence of pluripotent/multipotent stem cells in adult mammalian ovaries, including those of humans. The aim of this study is to isolate, characterize and differentiate in vitro stem cells that originate from the adult human ovarian cortex and that express markers of pluripotency/multipotency. After enzymatic degradation of small ovarian cortex biopsies retrieved from 18 women, ovarian cell cultures were successfully established from 17 and the formation of cell colonies was observed. The presence of cells/colonies expressing some markers of pluripotency (alkaline phosphatase, surface antigen SSEA-4, OCT4, SOX-2, NANOG, LIN28, STELLA), germinal lineage (DDX4/VASA) and multipotency (M-CAM/CD146, Thy-1/CD90, STRO-1) was confirmed by various methods. Stem cells from the cultures, including small round SSEA-4-positive cells with diameters of up to 4 μm, showed a relatively high degree of plasticity. We were able to differentiate them in vitro into various types of somatic cells of all three germ layers. However, these cells did not form teratoma when injected into immunodeficient mice. Our results thus show that ovarian tissue is a potential source of stem cells with a pluripotent/multipotent character for safe application in regenerative medicine.
Collapse
Affiliation(s)
- Martin Stimpfel
- Reproductive Unit, Department of Obstetrics and Gynaecology, University Medical Centre Ljubljana, Slajmerjeva 3, 1000, Ljubljana, SI, Slovenia
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kroepil F, Fluegen G, Vallböhmer D, Baldus SE, Dizdar L, Raffel AM, Hafner D, Stoecklein NH, Knoefel WT. Snail1 expression in colorectal cancer and its correlation with clinical and pathological parameters. BMC Cancer 2013; 13:145. [PMID: 23522088 PMCID: PMC3617032 DOI: 10.1186/1471-2407-13-145] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 03/14/2013] [Indexed: 11/14/2022] Open
Abstract
Background Snail1 is a transcription regulator of E-cadherin. The loss of E-cadherin seems to be a crucial step in the process of Epithelial-mesenchymal transition (EMT). EMT initiates invasion and proliferation in many tumours. Overexpression of Snail1 is known to be associated with poor outcome in several solid tumours. The aim of this study was to analyse its expression profile and prognostic significance in colorectal cancer. Methods Tissue microarrays (TMA) containing paraffin-embedded primary colorectal cancer (CRC) tissue samples from 251 patients were used in this study. The expression of Snail1 and E-cadherin was assessed by immunohistochemistry in different tumour compartments, corresponding lymph node metastases and normal colonic mucosa. Intensity of staining was classified according to the Remmele score (standardized scoring system) as well as the semiquantitative score established by Blechschmidt et al. Results Snail1 expression was observed in 76% of the CRC. Loss of E-cadherin was noted in 87% of the CRC. Snail1 positive tumours were significantly correlated with Snail1 positive lymph node metastases (p=0.03). There was no significant correlation between loss of E-cadherin and Snail1 expression, or between N-stage or grading and Snail1 expression. Kaplan-Meier survival analysis identified no prognostic impact of Snail1 expression on overall survival. Conclusion Snail1 expression was detectable in most of the CRC but showed no significant association with E-cadherin loss, clinical pathological characteristics or overall survival. The observed loss of E-cadherin could be explained by effects of other important EMT pathways, such as the Wnt-signalling cascade.
Collapse
Affiliation(s)
- Feride Kroepil
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Düsseldorf, 40225, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yang D, Sun Y, Hu L, Zheng H, Ji P, Pecot CV, Zhao Y, Reynolds S, Cheng H, Rupaimoole R, Cogdell D, Nykter M, Broaddus R, Rodriguez-Aguayo C, Lopez-Berestein G, Liu J, Shmulevich I, Sood AK, Chen K, Zhang W. Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell 2013; 23:186-99. [PMID: 23410973 PMCID: PMC3603369 DOI: 10.1016/j.ccr.2012.12.020] [Citation(s) in RCA: 321] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Revised: 11/08/2011] [Accepted: 12/28/2012] [Indexed: 12/23/2022]
Abstract
Integrated genomic analyses revealed a miRNA-regulatory network that further defined a robust integrated mesenchymal subtype associated with poor overall survival in 459 cases of serous ovarian cancer (OvCa) from The Cancer Genome Atlas and 560 cases from independent cohorts. Eight key miRNAs, including miR-506, miR-141, and miR-200a, were predicted to regulate 89% of the targets in this network. Follow-up functional experiments illustrate that miR-506 augmented E-cadherin expression, inhibited cell migration and invasion, and prevented TGFβ-induced epithelial-mesenchymal transition by targeting SNAI2, a transcriptional repressor of E-cadherin. In human OvCa, miR-506 expression was correlated with decreased SNAI2 and VIM, elevated E-cadherin, and beneficial prognosis. Nanoparticle delivery of miR-506 in orthotopic OvCa mouse models led to E-cadherin induction and reduced tumor growth.
Collapse
Affiliation(s)
- Da Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Sun
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Limei Hu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Ping Ji
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chad V. Pecot
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanrui Zhao
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | | | - Hanyin Cheng
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rajesha Rupaimoole
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Cogdell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matti Nykter
- Tampere University of Technology, Tampere 33101, Finland
| | - Russell Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Wei Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
24
|
Celik O, Celik E, Turkcuoglu I, Yilmaz E, Simsek Y, Tiras B. Germline cells in ovarian surface epithelium of mammalians: a promising notion. Reprod Biol Endocrinol 2012; 10:112. [PMID: 23245287 PMCID: PMC3566967 DOI: 10.1186/1477-7827-10-112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 12/14/2012] [Indexed: 01/03/2023] Open
Abstract
It is a long held doctrine in reproductive biology that women are born with a finite number of oocytes and there is no oogenesis during the postnatal period. However, recent evidence challenges this by showing the presence of germ line stem cells in the human ovarian surface epithelium (OSE), which can serve as a source of germ cells, and differentiate into oocyte like structures. Postnatal renewal of oocytes may have enormous therapeutic potential especially in women facing the risk of premature ovarian failure idiopathically or iatrogenically after exposure to gonadotoxic chemotherapy and radiation for cancer therapy.This article reviews current knowledge on germ line stem cells in human OSE.
Collapse
Affiliation(s)
- Onder Celik
- Department of Obstetrics and Gynecology, Inonu University, Medical Faculty, Malatya, Turkey
| | - Ebru Celik
- Department of Obstetrics and Gynecology, Inonu University, Medical Faculty, Malatya, Turkey
| | - Ilgin Turkcuoglu
- Department of Obstetrics and Gynecology, Inonu University, Medical Faculty, Malatya, Turkey
| | - Ercan Yilmaz
- Department of Obstetrics and Gynecology, Inonu University, Medical Faculty, Malatya, Turkey
| | - Yavuz Simsek
- Department of Obstetrics and Gynecology, Inonu University, Medical Faculty, Malatya, Turkey
| | - Bulent Tiras
- Department of Obstetric and Gynecology, Acibadem University, School of Medicine, Istanbul, Turkey
| |
Collapse
|
25
|
[Molecular characterization of corona radiata cells from patients with diminished ovarian reserve]. ACTA ACUST UNITED AC 2012; 40:500-6. [PMID: 22906700 DOI: 10.1016/j.gyobfe.2012.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/30/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Diminished ovarian reserve (DOR) is one of the causes of infertility. In this prospective study, gene expression profiling (GEP) of corona radiata cells (CRC) was performed to identify genes deregulated in DOR patients. PATIENTS AND METHODS Microarray-based GEP of CRC isolated from eight women undergoing IVF was performed to identify genes differentially expressed between patients with normal ovarian reserve and DOR patients. Microfluidic-based quantitative RT-PCR assay were used to validate selected transcripts on 40 independent patients. RESULTS Forty-eight transcripts were differentially expressed, including CXXC5 and FOXC1 down regulated in DOR, as well as CTGF, FSTL3, PTGS2 and SOCS2 up regulated in DOR. According to these transcripts, two DOR patients'subgroups (DOR Gr1 and Gr2) were identified. In DOR Gr2 patients, CITED2, CTGF, GAS-1, IRS2, PTGS2, SOCS2, VCAN were expressed at significantly higher levels, and CXXC5, FOXC1, GBP2 and ZMIZ1 at significantly lower level. Eleven of those genes are transcriptional targets of Estrogens and higher baseline oestradiol levels were observed in DOR Gr2 patients (P<0.006). DISCUSSION AND CONCLUSION Twelve genes deregulated in CRC of DOR patients were identified, which could be involved in DOR pathogenesis. The distinction of a particular subgroup of DOR patients suggests the possibility of deregulation of estrogen response genes.
Collapse
|
26
|
Davidson B, Tropé CG, Reich R. Epithelial-mesenchymal transition in ovarian carcinoma. Front Oncol 2012; 2:33. [PMID: 22655269 PMCID: PMC3356037 DOI: 10.3389/fonc.2012.00033] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 03/21/2012] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, with the majority of patients dying within 5 years of diagnosis. This poor survival of patients diagnosed with this malignancy is attributed to diagnosis at advanced stage, when the tumor has metastasized, and to chemotherapy resistance, either primary or developing along tumor progression. However, ovarian carcinomas, constituting the vast majority of ovarian cancers, additionally have unique biology, one aspect of which is the ability to co-express epithelial and mesenchymal determinants. epithelial–mesenchymal transition (EMT), a physiological process by which mesenchymal cells are formed and migrate to target organs during embryogenesis, is involved in cancer cell invasion and metastasis. However, these changes do not fully occur in ovarian carcinoma, and are even reversed in tumor cells present in malignant peritoneal and pleural effusions. This review summarizes current knowledge in this area, including the characteristics of EMT related to adhesion, transcriptional regulation and chemoresistance, and their clinical relevance, as well as the recently observed regulation of EMT by microRNA.
Collapse
Affiliation(s)
- Ben Davidson
- Division of Pathology, Norwegian Radium Hospital, Oslo University Hospital Oslo, Norway
| | | | | |
Collapse
|
27
|
Stem cell interaction with somatic niche may hold the key to fertility restoration in cancer patients. Obstet Gynecol Int 2012; 2012:921082. [PMID: 22548074 PMCID: PMC3324916 DOI: 10.1155/2012/921082] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 12/08/2011] [Accepted: 12/19/2011] [Indexed: 01/17/2023] Open
Abstract
The spontaneous return of fertility after bone marrow transplantation or heterotopic grafting of cryopreserved ovarian cortical tissue has surprised many, and a possible link with stem cells has been proposed. We have reviewed the available literature on ovarian stem cells in adult mammalian ovaries and presented a model that proposes that the ovary harbors two distinct populations of stem cells, namely, pluripotent, quiescent, very small embryonic-like stem cells (VSELs), and slightly larger “progenitor” ovarian germ stem cells (OGSCs). Besides compromising the somatic niche, oncotherapy destroys OGSCs since, like tumor cells, they are actively dividing; however VSELs persist since they are relatively quiescent. BMT or transplanted ovarian cortical tissue may help rejuvenate the ovarian niche, which possibly supports differentiation of persisting VSELs resulting in neo-oogenesis and follicular development responsible for successful pregnancies. Postnatal oogenesis in mammalian ovary from VSELs may be exploited for fertility restoration in cancer survivors including those who were earlier deprived of gametes and/or gonadal tissue cryopreservation options.
Collapse
|
28
|
Ma CIJ, Martin C, Ma Z, Hafiane A, Dai M, Lebrun JJ, Kiss RS. Engulfment protein GULP is regulator of transforming growth factor-β response in ovarian cells. J Biol Chem 2012; 287:20636-51. [PMID: 22451657 DOI: 10.1074/jbc.m111.314997] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a key regulatory molecule with pleiotropic effects on cell growth, migration, and invasion. As a result, impairment of proper TGF-β signaling is central to tumorigenesis and metastasis. The TGF-β receptor V (TGFBRV or LRP1) has been shown to be responsible for TGF-β-mediated cell growth inhibition in Chinese hamster ovary (CHO) cells. The LRP1 adapter protein GULP mediates internalization of the various LRP1-specific ligands, and we hypothesize that GULP acts as a novel regulator of TGF-β signaling in ovarian cells. CHO cells that overexpress exogenous GULP (FL) demonstrate enhancement in growth inhibition, migration, and invasion from TGF-β treatment, whereas cells that lack GULP (AS) show impairment of growth inhibition and decreased migration and invasion. The enhanced TGF-β response in FL cells was confirmed by a prolonged TGF-β-induced SMAD3 phosphorylation, whereas a shortening of the phosphorylation event is observed in AS cells. Mechanistically, the presence of GULP retains the TGF-β in a signaling-competent early endosome for enhanced signaling. To address this mechanism in a physiological setting, TGF-β insensitive ovarian adenocarcinoma cells (HEY) have a very low GULP expression level, similar to the observation made in a wide selection of human ovarian adenocarcinomas. Transfection of GULP into the HEY cells restored the TGF-β responsiveness, as measured by SMAD3 phosphorylation and impairment of cell growth. Because GULP expression positively regulates TGF-β signaling leading to growth inhibition, this may represent an attractive target to achieve TGF-β responsiveness in ovarian cells.
Collapse
Affiliation(s)
- Cheng-I J Ma
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Bezdekova M, Brychtova S, Sedlakova E, Langova K, Brychta T, Belej K. Analysis of Snail-1, E-cadherin and claudin-1 expression in colorectal adenomas and carcinomas. Int J Mol Sci 2012; 13:1632-1643. [PMID: 22408413 PMCID: PMC3291982 DOI: 10.3390/ijms13021632] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 01/29/2023] Open
Abstract
We report the expression of Snail-1, E-cadherin and claudin-1 by indirect immunohistochemistry in colonic neoplasia. Snail-1 is a zinc finger transcription factor expressed in cells that already have undergone almost complete epithelial-mesenchymal transition (EMT) and have already evaded from the tumor. The main mechanism by which Snail induces EMT is downregulation of E-cadherin, of which expression was shown to be frequently downregulated in many different types of tumors, where it accompanies the invasiveness and metastatic behavior of malignant cells. Moreover, Snail-1 may downregulate the expression of claudin-1, a cell-cell adhesion protein which plays a likely role in progression and dissemination during tumorigenesis. Snail-1 was expressed in both carcinoma and adenoma cells with histologically normal epithelium in the mucosa, adjacent to the tumors, without significant differences, and predominant strong intensity of staining. Statistically significant differences were revealed between normal and tumorous epithelium (p = 0.003) at the subcellular level, where the shift of the protein to the cytoplasm with combined cytoplasmic/nuclear or pure cytoplasmic expression was observed. E-cadherin expression was present in 100% of cases of both adenocarcinomas and adenomas, with prevailing strong membranous immunoreactivity and no differences between protein expression in tumors and normal mucosa. Predominating strong positivity of claudin-1 was detected in tumor cells of adenocarcinomas and adenomas. Marked differences were seen in protein localization, where membranous staining, typical for nontumorous epithelium, changed to combined membranous/cytoplasmic expression in adenocarcinomas (p = 0.0001) and adenomas (0.0002), in which cytoplasmic shift was associated with a higher degree of dysplasia. Furthermore, membranous/cytoplasmic localization was more frequent in the carcinoma group (87%) in comparison with adenomas (51%) (p = 0.0001). We conclude that dystopic subcellular localizations of Snail-1 and claudin-1 may participate in changes of cellular morphology and behavior which might be associated with altered effectory pathways of proteins and thus substantially contribute to the cancer development.
Collapse
Affiliation(s)
- Michala Bezdekova
- Laboratory of Molecular Pathology, Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic; E-Mails: (M.B.); (E.S.)
| | - Svetlana Brychtova
- Laboratory of Molecular Pathology, Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic; E-Mails: (M.B.); (E.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +420-58-563-9547; Fax: +420-58-563-2966
| | - Eva Sedlakova
- Laboratory of Molecular Pathology, Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic; E-Mails: (M.B.); (E.S.)
| | - Katerina Langova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic; E-Mail:
| | - Tomas Brychta
- Department of Recreology, Faculty of Physical Culture, Palacky University, tr. Miru 115, 771 11 Olomouc, Czech Republic; E-Mail:
| | - Kamil Belej
- Department of Urology, Central Military Hospital, U Vojenské nemocnice 1200, 169 02 Prague 6, Czech Republic; E-Mail:
| |
Collapse
|
30
|
May-Panloup P, Ferré-L'Hôtellier V, Morinière C, Marcaillou C, Lemerle S, Malinge MC, Coutolleau A, Lucas N, Reynier P, Descamps P, Guardiola P. Molecular characterization of corona radiata cells from patients with diminished ovarian reserve using microarray and microfluidic-based gene expression profiling. Hum Reprod 2012; 27:829-43. [DOI: 10.1093/humrep/der431] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
31
|
Oh JE, Kim RH, Shin KH, Park NH, Kang MK. DeltaNp63α protein triggers epithelial-mesenchymal transition and confers stem cell properties in normal human keratinocytes. J Biol Chem 2011; 286:38757-38767. [PMID: 21880709 DOI: 10.1074/jbc.m111.244939] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
p63 is a p53 family protein required for morphogenesis and postnatal regeneration of epithelial tissues. Here we demonstrate that ΔNp63α, a p63 isoform lacking the N-terminal transactivation domain, induces epithelial-mesenchymal transition (EMT) in primary human keratinocytes in a TGF-β-dependent manner. Rapidly proliferating normal human epidermal keratinocytes (NHEK) were infected with retroviral vector expressing ΔNp63α or empty vector and serially subcultured until replicative senescence. No phenotypic changes were observed until the culture reached senescence. Then the ΔNp63α-transduced cells underwent morphological changes resembling mesenchymal cells and acquired the EMT phenotype. Treatment with exogenous TGF-β accelerated EMT in presenescent ΔNp63α-transduced cells, whereas the inhibition of TGF-β signaling reversed the EMT phenotype. TGF-β treatment alone led to growth arrest in control NHEK with no evidence of EMT, indicating that ΔNp63α altered the cellular response to TGF-β treatment. ΔNp63α-transduced cells acquiring EMT gained the ability to be differentiated to osteo-/odontogenic and adipogenic pathways, resembling mesenchymal stem cells. Furthermore, these cells expressed enhanced levels of Nanog and Lin28, which are transcription factors associated with pluripotency. These data indicate that EMT required ΔNp63α transduction and intact TGF-β signaling in NHEK.
Collapse
Affiliation(s)
- Ju-Eun Oh
- School of Dentistry, UCLA, Los Angeles, California 90095
| | - Reuben H Kim
- School of Dentistry, UCLA, Los Angeles, California 90095; Dental Research Institute, UCLA, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| | - Ki-Hyuk Shin
- School of Dentistry, UCLA, Los Angeles, California 90095; Dental Research Institute, UCLA, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095
| | - No-Hee Park
- School of Dentistry, UCLA, Los Angeles, California 90095; Dental Research Institute, UCLA, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095; David Geffen School of Medicine, UCLA, Los Angeles, California 90095
| | - Mo K Kang
- School of Dentistry, UCLA, Los Angeles, California 90095; Dental Research Institute, UCLA, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095.
| |
Collapse
|
32
|
Bukovsky A. Ovarian stem cell niche and follicular renewal in mammals. Anat Rec (Hoboken) 2011; 294:1284-306. [PMID: 21714105 DOI: 10.1002/ar.21422] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 04/28/2011] [Indexed: 12/24/2022]
Abstract
Stem cell niche consists of perivascular compartment, which connects the stem cells to the immune and vascular systems. During embryonic period, extragonadal primordial germ cells colonize coelomic epithelium of developing gonads. Subsequently, ovarian stem cells (OSC) produce secondary germ cells under the influence of OSC niche, including immune system-related cells and hormonal signaling. The OSC in fetal and adult human ovaries serve as a source of germ and granulosa cells. Lack of either granulosa or germ cell niche will result in premature ovarian failure in spite of the presence of OSC. During perinatal period, the OSC transdifferentiate into fibroblast-like cells forming the ovarian tunica albuginea resistant to environmental threats. They represent mesenchymal precursors of epithelial OSC during adulthood. The follicular renewal during the prime reproductive period (PRP) ensures that there are fresh eggs available for a healthy progeny. End of PRP is followed by exponentially growing fetal genetic abnormalities. The OSC are present in adult, aging, and postmenopausal ovaries, and differentiate in vitro into new oocytes. During in vitro development of large isolated oocytes reaching 200 μm in diameter, an ancestral mechanism of premeiotic nurse cells, which operates during oogenesis in developing ovaries from invertebrates to mammalian species, is utilized. In vitro developed eggs could be used for autologous IVF treatment of premature ovarian failure. Such eggs are also capable to produce parthenogenetic embryos like some cultured follicular oocytes. The parthenotes produce embryonic stem cells derived from inner cell mass, and these cells can serve as autologous pluripotent stem cells.
Collapse
Affiliation(s)
- Antonin Bukovsky
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
33
|
Clelland ES, Kelly SP. Exogenous GDF9 but not Activin A, BMP15 or TGFβ alters tight junction protein transcript abundance in zebrafish ovarian follicles. Gen Comp Endocrinol 2011; 171:211-7. [PMID: 21291886 DOI: 10.1016/j.ygcen.2011.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/14/2011] [Accepted: 01/27/2011] [Indexed: 11/26/2022]
Abstract
The tight junction (TJ) complex plays an important role in regulating paracellular permeability and provides mechanical stability in vertebrate epithelia and endothelia. In zebrafish ovarian follicles, TJ complexes in the follicular envelope degenerate as the follicles develop towards maturation. In the current study, transcript abundance of claudins (cldn d, g, h, 1, and 12) and occludins (ocln, and ocln b) were assessed in mid-vitellogenic follicles in response to treatment with exogenous growth factors that are reported to be involved in zebrafish follicle development (i.e. Activin A, BMP15, GDF9 and TGFβ). Exogenous GDF9 reduced the transcript abundance of cldn g, ocln and ocln b in mid-vitellogenic follicles, whereas Activin A, BMP15, and TGFβ had no effect. Subsequent studies with GDF9 revealed that this factor did not alter TJ protein transcript abundance in pre-vitellogenic follicles but did increase the abundance of ocln b in fully grown (maturing) follicles. GDF9 was also seen to increase the abundance of StAR mRNA in all but primary stage follicles. These data suggest a role for GDF9 in the regulation of TJ integrity in zebrafish ovarian follicles, perhaps in the facilitation of ovulation, and support a previously postulated role for GDF9 in zebrafish ovarian follicle development. In addition, data also support the idea that endocrine factors play an important role in the regulation of TJ proteins during ovarian follicle development.
Collapse
Affiliation(s)
- Eric S Clelland
- Department of Biology, York University, Toronto, ON, Canada M3J 1P3.
| | | |
Collapse
|
34
|
Wright JW, Pejovic T, Jurevic L, Bishop CV, Hobbs T, Stouffer RL. Ovarian surface epitheliectomy in the non-human primate: continued cyclic ovarian function and limited epithelial replacement. Hum Reprod 2011; 26:1422-30. [PMID: 21421661 DOI: 10.1093/humrep/der061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The fifth leading cause of cancer deaths among women is ovarian cancer (OC), which originates primarily in the ovarian surface epithelium (OSE) that surrounds the ovary. Permanent removal of the OSE could provide a novel strategy to substantially reduce OC risk, while retaining the benefits of ovarian function, including gameto- and steroidogenesis. It must be determined whether ovarian surface epitheliectomy (OSEx) carries deleterious side effects, including loss of menstrual cyclicity, infertility or scarring (e.g. adhesions), prior to any clinical application of this strategy. To achieve this, we selected the non-human primate, rhesus macaque, for long-term (12 month) studies on the effects of OSEx. METHODS Rhesus macaque females underwent OSEx by detergent treatment and were then monitored for menstrual cyclicity (menstruation, steroidogenesis and follicle development) and adverse side effects (tissue scarring or adhesions). Ovaries were collected at 6 or 12 months and examined for evidence of tissue damage, follicle rupture and regression of the corpus luteum. The ovarian surface was examined immunohistologically for signs of epithelial replacement, using markers for OSE and fimbrial epithelium (FE), a possible alternative source of pelvic tumors diagnosed as OC. RESULTS After OSEx, menstrual cycle length, estrogen and progesterone production, follicle rupture and luteal regression appeared normal. No evidence of adhesions was seen. At 6 and 12 months post-OSEx, the ovarian surface was sparsely populated by cells expressing OSE and FE markers. Proliferative activity in this population was notably low. CONCLUSIONS OSEx may provide a novel method to reduce the risk of OC, without sacrificing ovarian function, although the effects on fertility remain to be tested. The absence of epithelial replacement via enhanced proliferation suggests OSEx does not increase malignant potential. Complete and permanent OSEx may be feasible.
Collapse
Affiliation(s)
- Jay W Wright
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Virant-Klun I, Skutella T, Stimpfel M, Sinkovec J. Ovarian surface epithelium in patients with severe ovarian infertility: a potential source of cells expressing markers of pluripotent/multipotent stem cells. J Biomed Biotechnol 2011; 2011:381928. [PMID: 22187524 PMCID: PMC3237017 DOI: 10.1155/2011/381928] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 08/31/2011] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to confirm the presence of stem cells in the ovarian surface epithelium of patients with premature ovarian failure and no mature follicles and oocytes. In these patients, small round cells of unknown origin expressing SOX-2 marker of pluripotency were observed among the epithelial cells just after the ovarian surface epithelium scraping. These cells were an integral part of the ovarian surface epithelium. When the scraped cells were cultured in a medium with added follicular fluid to provide some ovarian niche, primitive oocyte-like cells and typical round-shaped cell clusters positively stained on alkaline phosphatase, and markers of pluripotency, such as SOX-2 and SSEA-4, were developed. These markers were expressed early and also later in the culture. Single oocyte-like cells expressed genes OCT4A, SOX-2, NANOG, NANOS, STELLA, CD9, LIN28, KLF4, GDF3, and MYC, characteristic for pluripotent stem cells. The results of this study confirmed the presence of putative stem cells in the ovarian surface epithelium of these patients and provided some basis to create a stem cell line in the future.
Collapse
Affiliation(s)
- Irma Virant-Klun
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia.
| | | | | | | |
Collapse
|
36
|
Brown TJ, Shathasivam P. Maintaining mesenchymal properties of ovarian surface epithelial cells: a potential early protective role for TGF-beta in ovarian carcinogenesis. Endocrinology 2010; 151:5092-4. [PMID: 20962057 DOI: 10.1210/en.2010-0938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Theodore J Brown
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Ontario, Canada.
| | | |
Collapse
|