1
|
Yu B, Huang Y, Yang Y, Hu H, Yang J. Effect of CTP-mediated PTEN on 5637 bladder cancer cells and the underlying molecular mechanism. BMC Urol 2022; 22:200. [PMID: 36496361 PMCID: PMC9741776 DOI: 10.1186/s12894-022-01152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The aim of the present study was to explore the effect of cytoplasmic transduction peptide (CTP)-phosphatase and tensin homolog (PTEN) on the proliferation, cell cycle, apoptosis, migration and invasion of bladder cancer cells and the underlying molecular mechanism. METHODS A eukaryotic expression vector, pTT5-CTP-PTEN, was constructed. The constructed vector was transfected into HEK 293-6E cells to express a fusion protein, CTP-PTEN. The fusion protein was purified. 5637 bladder cancer cells were cocultured with purified CTP-PTEN fusion protein. Target gene expression, protein expression, cell proliferation, cell cycle, apoptosis, cell invasion and cell migration were examined by reverse transcription polymerase chain reaction (RT-PCR), western blot, MTT assay, flow cytometry, Transwell assay, and cell scratch assay, respectively. RESULTS Both PTEN and CTP-PTEN fusion protein inhibited the proliferation, cell cycle, invasion and migration of bladder cancer cells and promoted the apoptosis of bladder cancer cells. The effect of CTP-PTEN was more significant. CONCLUSIONS The fused expression of CTP and PTEN significantly increased the penetrability of the tumor suppressor gene PTEN into cancer cells. The CTP-PTEN fusion protein exhibited a significant carcinostatic effect on 5637 bladder cancer cells.
Collapse
Affiliation(s)
- Bei Yu
- grid.411292.d0000 0004 1798 8975Urological Department, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan China
| | - Yuan Huang
- grid.411292.d0000 0004 1798 8975Department of Clinical Laboratory, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan China
| | - Yue Yang
- grid.411292.d0000 0004 1798 8975Urological Department, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan China
| | - Haifeng Hu
- grid.411292.d0000 0004 1798 8975Urological Department, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan China
| | - Jin Yang
- grid.411292.d0000 0004 1798 8975Urological Department, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan China
| |
Collapse
|
2
|
Zhuang Y, Zhang S, Liu Y, Yang H. Can adjuvant chemotherapy improve the prognosis of adult ovarian granulosa cell tumors?: A narrative review. Medicine (Baltimore) 2022; 101:e29062. [PMID: 35356927 PMCID: PMC10513366 DOI: 10.1097/md.0000000000029062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/24/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Adult granulosa cell tumors (aGCTs) are rare ovarian neoplasms with a relatively favorable prognosis. They follow an indolent course, characterized by a prolonged natural history and a tendency to late recurrences, Around a quarter of patients develop recurrence and More than 70% of women with recurrence die from their disease, The percentage of patients received chemotherapy increases over time, whether adjuvant chemotherapy improve the prognosis of aGCTs is equivocal? The purpose of this review is to summarize the previously published evidence to evaluate whether adjuvant chemotherapy improve the prognosis of aGCTs to provide guidance for clinical practice. EMBASE, PubMed, Web of Science, WanFang Data and Chinese National Knowledge Infrastructure are searched up to December 2020, used the search strategy of ovar* and granulosa cell* and (tumor* or tumour* or malignan* or cancer* or carcinom* or neoplasm*) and chemotherapy. The screening process was conducted strictly based on inclusion and exclusion criteria. Clinical studies based on human including randomized controlled trial, quasi-randomised controlled trials, nonrandomised trials cohort study and case control study were included without restriction of time. The percentage of patients received chemotherapy increases over time, but the benefit of adjuvant chemotherapy is lack of high-grade evidence of prospective study, based on the current retrospective studies, we still do not have the evidence to confirm the survival benefit of adjuvant chemotherapy in early stage, advanced stage or recurrent aGCT with no residual tumor, but for inoperable disseminated disease or disease with suboptimal cytoreduction, adjuvant chemotherapy maybe an Optable options. Multinational prospective randomised controlled trials are urgently needed to validate the role of adjuvant chemotherapy. Further research on molecular mechanisms and developing novel targeted medicines may improve the survival of aGCTs.
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China,Sun Yat-sen University, Zhuhai, China
| | | | | | | |
Collapse
|
3
|
Bildik G, Akin N, Senbabaoglu F, Esmalian Y, Sahin GN, Urman D, Karahuseyinoglu S, Ince U, Palaoglu E, Taskiran C, Arvas M, Guzel Y, Yakin K, Oktem O. Endogenous c-Jun N-terminal kinase (JNK) activity marks the boundary between normal and malignant granulosa cells. Cell Death Dis 2018; 9:421. [PMID: 29549247 PMCID: PMC5856777 DOI: 10.1038/s41419-018-0459-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/21/2022]
Abstract
Granulosa cell tumor of the ovary (GCT) is a very rare tumor, accounting for only 2% of all ovarian tumors. It originates from sex cords in the ovary and can be divided into adult (95%) and juvenile (5%) types based on histologic findings. To date, no clear etiologic process has been identified other than a missense point mutation in the FOXL2 gene. Our previous works showed that c-Jun N-terminal kinase (JNK) pathway plays critical role in cell cycle progression and mitosis of normal and immortalized granulosa cells and follicle growth in rodent ovaries. These findings led us to investigate the role of JNK pathway in the granulosa cell tumor of the ovary. We used two different GCT cell lines (COV434 and KGN) and fresh GCT samples of adult and juvenile types obtained from the patients during surgery. We have discovered that endogenous kinase activity of JNK is markedly enhanced in the GCT samples and cell lines, whereas it was almost undetectable in mitotic non-malignant human granulosa cells. The inhibition of JNK pathway in GCT cell lines with two different pharmacologic inhibitors (SP600125 and AS601245) or siRNA resulted in a dose-dependent reduction in in vitro cell growth, increased apoptosis and diminished estradiol and AMH productions. JNK inhibition was also associated with a decrease in the number of cells positive for mitosis marker phospho-histone H3Ser 10 in the asynchronous cells; and diminished EdU uptake during S phase and cell cycle arrest at G2/M-phase transition in the synchronized cells. Ex vivo treatment of patient-derived GCT samples with JNK inhibitors for 24 h significantly decreased their in vitro growth and estradiol and AMH productions. Furthermore, in human GCT xenograft model, in vivo tumor growth was significantly reduced and plasma AMH levels were significantly decreased in SCID mice after administration of JNK inhibitors and siRNA. These findings suggest that targeting JNK pathway may provide therapeutic benefit in the treatment of granulosa cell tumors for which currently no curative therapy exists beyond surgery.
Collapse
Affiliation(s)
- Gamze Bildik
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Nazli Akin
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Filiz Senbabaoglu
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Yashar Esmalian
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Gizem Nur Sahin
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Defne Urman
- Graduate School of Health Sciences and School of Medicines, Koc University, Istanbul, Turkey
| | - Sercin Karahuseyinoglu
- Department of Histology and Embryology, School of Medicine, Koc University, Istanbul, Turkey
| | - Umit Ince
- Department of Pathology, School of Medicine, Acibadem University, Istanbul, Turkey
| | - Erhan Palaoglu
- American Hospital Clinical Biochemistry Laboratories, Istanbul, Turkey
| | - Cagatay Taskiran
- Department of Obstetrics and Gynecology, Gynecologic Oncology Division, School of Medicine, Koc University, Istanbul, Turkey
| | - Macit Arvas
- Women's Health Center, American Hospital, Istanbul, Turkey
| | - Yilmaz Guzel
- Women's Health Center, American Hospital, Istanbul, Turkey
| | - Kayhan Yakin
- Department of Obstetrics and Gynecology, The Division of Reproductive Endocrinology and Infertility, Translational Research Laboratories, School of Medicine, Koc University, Istanbul, Turkey
| | - Ozgur Oktem
- Department of Obstetrics and Gynecology, The Division of Reproductive Endocrinology and Infertility, Translational Research Laboratories, School of Medicine, Koc University, Istanbul, Turkey.
| |
Collapse
|
4
|
Guzel Y, Oktem O. Understanding follicle growth in vitro: Are we getting closer to obtaining mature oocytes from in vitro-grown follicles in human? Mol Reprod Dev 2017; 84:544-559. [PMID: 28452156 DOI: 10.1002/mrd.22822] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/06/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
Obtaining and fertilizing mature oocytes from immature follicles that were grown outside the body has conceptually attracted scientists for centuries, with initial attempts first documented in the 19th century. Significant progress has been made since then, due in part to a better understanding of folliculogenesis and improved techniques of in vitro follicle growth. Indeed, in vitro growth is now considered a reasonable approach to preserve or restore fertility when immature follicles and their oocytes need to be grown and matured outside the body. Certain patients would benefit from in vitro follicle growth, particularly those who carry a risk of cancer re-seeding after grafting of frozen-thawed ovarian tissue or who are at the risk of premature ovarian failure due to several intrinsic ovarian defects and genetic mutations that lead to accelerated follicle atresia and early exhaustion of the ovarian reserve. This review provides an update on the current status of in vitro growth of preantral human follicles, from initial efforts to the most recent achievements.
Collapse
Affiliation(s)
- Yilmaz Guzel
- Department of Obstetrics and Gynecology, Istanbul Aydin University School of Medicine, Istanbul, Turkey
| | - Ozgur Oktem
- Division Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey.,Women's Health Center, Assisted Reproduction Unit, American Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Fuller P, Leung D, Chu S. Genetics and genomics of ovarian sex cord-stromal tumors. Clin Genet 2017; 91:285-291. [DOI: 10.1111/cge.12917] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022]
Affiliation(s)
- P.J. Fuller
- Centre for Endocrinology and Metabolism; Hudson Institute of Medical Research; Clayton Australia
- Department of Molecular and Translational Science; Monash University; Clayton Australia
| | - D. Leung
- Centre for Endocrinology and Metabolism; Hudson Institute of Medical Research; Clayton Australia
- Department of Molecular and Translational Science; Monash University; Clayton Australia
| | - S. Chu
- Centre for Endocrinology and Metabolism; Hudson Institute of Medical Research; Clayton Australia
- Department of Molecular and Translational Science; Monash University; Clayton Australia
| |
Collapse
|
6
|
Alexiadis M, Chu S, Leung D, Gould JA, Jobling T, Fuller PJ. Transcriptomic analysis of stage 1 versus advanced adult granulosa cell tumors. Oncotarget 2016; 7:14207-19. [PMID: 26893359 PMCID: PMC4924709 DOI: 10.18632/oncotarget.7422] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/29/2016] [Indexed: 01/14/2023] Open
Abstract
Ovarian granulosa cell tumors (GCT) are hormonally-active neoplasms characterized, in the adult-subtype, by a mutation in the FOXL2 gene (C134W). They exhibit an indolent course with an unexplained propensity for late recurrence; ~80% of patients with aggressive, advanced stage tumors die from their disease; aside from surgery, therapeutic options are limited. To identify the molecular basis of advanced stage disease we have used whole transcriptome analysis of FOXL2 C134W mutation positive adult (a)GCT to identify genes that are differentially expressed between early (stage 1) and advanced (stage 3) aGCT. Transcriptome profiles for early (n = 6) and stage 3 (n = 6) aGCT, and for the aGCT-derived KGN, cell line identified 24 genes whose expression significantly differs between the early and stage 3 aGCT. Of these, 16 were more abundantly expressed in the stage 3 aGCT and 8 were higher in the stage 1 tumors. These changes were further examined for the genes which showed the greatest fold change: the cytokine CXCL14, microfibrillar-associated protein 5, insulin-like 3 and desmin. Gene Set Enrichment Analysis identified overexpression of genes on chromosome 7p15 which includes the homeobox A gene locus. The analysis therefore identifies a small number of genes with clearly discriminate patterns of expression arguing that the clinicopathological-derived distinction of the tumor stage is robust, whilst confirming the relative homogeneity of expression for many genes across the cohort and hence of aGCT. The expression profiles do however identify several overexpressed genes in both stage 1 and/or stage 3 aGCT which warrant further study as possible therapeutic targets.
Collapse
Affiliation(s)
- Maria Alexiadis
- Hudson Institute of Medical Research (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
| | - Simon Chu
- Hudson Institute of Medical Research (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
- Monash University Department of Biochemistry and Molecular Biology, Clayton, Victoria 3168, Australia
| | - Dilys Leung
- Hudson Institute of Medical Research (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
- Monash University Department of Biochemistry and Molecular Biology, Clayton, Victoria 3168, Australia
| | - Jodee A. Gould
- Hudson Institute of Medical Research (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
- MHTP Medical Genomics Facility, Clayton, Victoria 3168, Australia
| | - Tom Jobling
- Department of Gynecology Oncology, Monash Health, Clayton, Victoria 3168, Australia
| | - Peter J. Fuller
- Hudson Institute of Medical Research (formerly Prince Henry's Institute of Medical Research), Clayton, Victoria 3168, Australia
- Monash University Department of Biochemistry and Molecular Biology, Clayton, Victoria 3168, Australia
| |
Collapse
|
7
|
Liu Z, Ren YA, Pangas SA, Adams J, Zhou W, Castrillon DH, Wilhelm D, Richards JS. FOXO1/3 and PTEN Depletion in Granulosa Cells Promotes Ovarian Granulosa Cell Tumor Development. Mol Endocrinol 2015; 29:1006-24. [PMID: 26061565 DOI: 10.1210/me.2015-1103] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The forkhead box (FOX), FOXO1 and FOXO3, transcription factors regulate multiple functions in mammalian cells. Selective inactivation of the Foxo1 and Foxo3 genes in murine ovarian granulosa cells severely impairs follicular development and apoptosis causing infertility, and as shown here, granulosa cell tumor (GCT) formation. Coordinate depletion of the tumor suppressor Pten gene in the Foxo1/3 strain enhanced the penetrance and onset of GCT formation. Immunostaining and Western blot analyses confirmed FOXO1 and phosphatase and tensin homolog (PTEN) depletion, maintenance of globin transcription factor (GATA) 4 and nuclear localization of FOXL2 and phosphorylated small mothers against decapentaplegic (SMAD) 2/3 in the tumor cells, recapitulating results we observed in human adult GCTs. Microarray and quantitative PCR analyses of mouse GCTs further confirmed expression of specific genes (Foxl2, Gata4, and Wnt4) controlling granulosa cell fate specification and proliferation, whereas others (Emx2, Nr0b1, Rspo1, and Wt1) were suppressed. Key genes (Amh, Bmp2, and Fshr) controlling follicle growth, apoptosis, and differentiation were also suppressed. Inhbb and Grem1 were selectively elevated, whereas reduction of Inha provided additional evidence that activin signaling and small mothers against decapentaplegic (SMAD) 2/3 phosphorylation impact GCT formation. Unexpectedly, markers of Sertoli/epithelial cells (SRY [sex determining region Y]-box 9/keratin 8) and alternatively activated macrophages (chitinase 3-like 3) were elevated in discrete subpopulations within the mouse GCTs, indicating that Foxo1/3/Pten depletion not only leads to GCTs but also to altered granulosa cell fate decisions and immune responses. Thus, analyses of the Foxo1/3/Pten mouse GCTs and human adult GCTs provide strong evidence that impaired functions of the FOXO1/3/PTEN pathways lead to dramatic changes in the molecular program within granulosa cells, chronic activin signaling in the presence of FOXL2 and GATA4, and tumor formation.
Collapse
Affiliation(s)
- Zhilin Liu
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Yi A Ren
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Stephanie A Pangas
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Jaye Adams
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Wei Zhou
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Diego H Castrillon
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Dagmar Wilhelm
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - JoAnne S Richards
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| |
Collapse
|
8
|
Caburet S, Anttonen M, Todeschini AL, Unkila-Kallio L, Mestivier D, Butzow R, Veitia RA. Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC Cancer 2015; 15:251. [PMID: 25884336 PMCID: PMC4407711 DOI: 10.1186/s12885-015-1283-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 03/27/2015] [Indexed: 12/23/2022] Open
Abstract
Background Ovarian granulosa cell tumors (GCTs) are the most frequent sex cord-stromal tumors. Several studies have shown that a somatic mutation leading to a C134W substitution in the transcription factor FOXL2 appears in more than 95% of adult-type GCTs. Its pervasive presence suggests that FOXL2 is the main cancer driver gene. However, other mutations and genomic changes might also contribute to tumor formation and/or progression. Methods We have performed a combined comparative genomic hybridization and transcriptomic analyses of 10 adult-type GCTs to obtain a picture of the genomic landscape of this cancer type and to identify new candidate co-driver genes. Results Our results, along with a review of previous molecular studies, show the existence of highly recurrent chromosomal imbalances (especially, trisomy 14 and monosomy 22) and preferential co-occurrences (i.e. trisomy 14/monosomy 22 and trisomy 7/monosomy 16q). In-depth analyses showed the presence of recurrently broken, amplified/duplicated or deleted genes. Many of these genes, such as AKT1, RUNX1 and LIMA1, are known to be involved in cancer and related processes. Further genomic explorations suggest that they are functionally related. Conclusions Our combined analysis identifies potential candidate genes, whose alterations might contribute to adult-type GCT formation/progression together with the recurrent FOXL2 somatic mutation. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1283-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandrine Caburet
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France. .,Université Paris-Diderot & Institut Jacques Monod, CNRS-UMR 7592, Bâtiment Buffon, 15 Rue Hélène Brion, Paris, Cedex 13, France.
| | - Mikko Anttonen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland. .,Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Anne-Laure Todeschini
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France.
| | - Leila Unkila-Kallio
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Denis Mestivier
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France.
| | - Ralf Butzow
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland. .,Department of pathology, University of Helsinki, and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland.
| | - Reiner A Veitia
- Institut Jacques Monod, Paris, France. .,Université Paris Diderot/Paris, Paris, France. .,Université Paris-Diderot & Institut Jacques Monod, CNRS-UMR 7592, Bâtiment Buffon, 15 Rue Hélène Brion, Paris, Cedex 13, France.
| |
Collapse
|
9
|
Andersson N, Anttonen M, Färkkilä A, Pihlajoki M, Bützow R, Unkila-Kallio L, Heikinheimo M. Sensitivity of human granulosa cell tumor cells to epidermal growth factor receptor inhibition. J Mol Endocrinol 2014; 52:223-34. [PMID: 24463098 DOI: 10.1530/jme-13-0286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidermal growth factor receptor (EGFR) is implicated in the progression of many human cancers, but its significance in ovarian granulosa cell tumor (GCT) pathobiology remains poorly understood. We assessed the EGFR gene copy number, surveyed the mRNA and protein expression patterns of EGFR in 90 adult GCTs, and assessed the in vitro sensitivity of GCT cells to EGFR inhibition. Low-level amplification of EGFR gene was observed in five GCTs and high-level amplification in one sample. EGFR mRNA was robustly expressed in GCTs. Most tumors expressed both unphosphorylated and phosphorylated EGFR protein, but the protein expression did not correlate with clinical parameters, including the risk of recurrence. Small-molecule EGFR inhibitors reduced the EGF-induced activation of EGFR and its downstream signaling molecules at nanomolar doses, but cell viability was reduced, and caspase-3/7 was activated in GCT cells only at micromolar doses. Based on the present results, EGFR is active and abundantly expressed in the majority of GCTs, but probably has only minor contribution to GCT cell growth. Given the high doses of EGFR inhibitors required to reduce GCT cell viability in vitro, they are not likely to be effective for GCT treatment as single agents; they should rather be tested as part of combination therapies for these malignancies.
Collapse
Affiliation(s)
- Noora Andersson
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, PO Box 20, 00014 University of Helsinki, Finland Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, PO Box 140, 00290 Helsinki, Finland Department of Pathology, Helsinki University Central Hospital, University of Helsinki and HUSlab, Haartmaninkatu 3, 00290 Helsinki, Finland Department of Pediatrics, Washington University School of Medicine, St Louis Children's Hospital, St Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Van Nieuwenhuysen E, Lambrechts S, Lambrechts D, Leunen K, Amant F, Vergote I. Genetic changes in nonepithelial ovarian cancer. Expert Rev Anticancer Ther 2014; 13:871-82. [DOI: 10.1586/14737140.2013.811174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
11
|
Francis JC, Kolomeyevskaya N, Mach CM, Dietrich JE, Anderson ML. MicroRNAs and Recent Insights into Pediatric Ovarian Cancers. Front Oncol 2013; 3:95. [PMID: 23641362 PMCID: PMC3639433 DOI: 10.3389/fonc.2013.00095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/07/2013] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is the most common pediatric gynecologic malignancy. When diagnosed in children, ovarian cancers present unique challenges that differ dramatically from those faced by adults. Here, we review the spectrum of ovarian cancers found in young women and girls and discuss the biology of these diseases. A number of advances have recently shed significant new understanding on the potential causes of ovarian cancer in this unique population. Particular emphasis is placed on understanding how altered expression of non-coding RNA transcripts known as microRNAs play a key role in the etiology of ovarian germ cell and sex cord-stromal tumors. Emerging transgenic models for these diseases are also reviewed. Lastly, future challenges and opportunities for understanding pediatric ovarian cancers, delineating clinically useful biomarkers, and developing targeted therapies are discussed.
Collapse
Affiliation(s)
- Jessica C Francis
- Department of Obstetrics and Gynecology, Baylor College of Medicine Houston, TX, USA
| | | | | | | | | |
Collapse
|
12
|
Rico C, Laguë MN, Lefèvre P, Tsoi M, Dodelet-Devillers A, Kumar V, Lapointe E, Paquet M, Nadeau MÈ, Boerboom D. Pharmacological targeting of mammalian target of rapamycin inhibits ovarian granulosa cell tumor growth. Carcinogenesis 2012; 33:2283-92. [PMID: 22871496 DOI: 10.1093/carcin/bgs263] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Few targeted therapies have been developed for ovarian granulosa cell tumor (GCT), even though it represents 5% of all malignant ovarian tumors in women. As misregulation of PI3K/AKT signaling has been implicated in GCT development, we hypothesized that the AKT signaling effector mammalian target of rapamycin (mTOR) may play a role in the pathogenesis of GCT and could represent a therapeutic target. Analyses of human GCT samples showed an increase in protein levels of mTOR and its downstream effectors RPS6KB1, RPS6, eIF4B and PPARG relative to normal granulosa cells, suggestive of an increase in mTOR pathway activity and increased translational activity and/or protein stability. We next sought to evaluate mTOR as a GCT therapeutic target using the Pten (tm1Hwu/tmiHwu);Ctnnb1 (tm1Mmt/+);Amhr2 (tm3(cre)Bhr/+) (PCA) mouse model, in which mTOR, RPS6KB1, eIF4B and PPARG are upregulated in tumor cells in a manner similar to human GCT. Treatment of PCA mice with the mTOR-specific inhibitor everolimus reduced tumor growth rate (1.5-fold; P < 0.05) and also reduced total tumor burden (4.7-fold; P < 0.05) and increased survival rate (78 versus 44% in the vehicle group) in a PCA surgical model of GCT peritoneal carcinomatosis. Everolimus decreased tumor cell proliferation and tumor cell volume relative to controls (P < 0.05), whereas apoptosis was unaffected. Phosphorylation of RPS6KB1 and RPS6 were decreased (P < 0.05) by everolimus, but RPS6KB1, RPS6, eIF4B and PPARG expressions were not affected. These results suggest that mTOR is a valid and clinically useful pharmacological target for the treatment of GCT, although its inhibition does not reverse all consequences of aberrant PI3K/AKT signaling in the PCA model.
Collapse
Affiliation(s)
- Charlène Rico
- Centre de Recherche en Reproduction animale, Université de Montréal, St-Hyacinthe, Québec J2S 7C6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zheng W, Nagaraju G, Liu Z, Liu K. Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary. Mol Cell Endocrinol 2012; 356:24-30. [PMID: 21684319 DOI: 10.1016/j.mce.2011.05.027] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/10/2011] [Indexed: 12/31/2022]
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling is a fundamental pathway for the regulation of cell proliferation, survival, migration, and metabolism in a variety of physiological and pathological processes. In recent years information provided by genetically modified mouse models has revealed that PI3K signaling plays vital roles in oogenesis, folliculogenesis, ovulation, and carcinogenesis in mouse ovary. In this review, we summarize (1) the physiological function of intra-oocyte PI3K signaling in regulation of primordial follicle survival and activation; (2) intra-granulosa cell PI3K signaling in regulation of cyclic follicular recruitment and ovulation; (3) intra-oocyte PI3K signaling in regulation of meiosis resumption and early embryogenesis; and also (4) the pathological function of PI3K signaling in ovarian diseases such as premature ovarian failure, granulosa cell tumors, and ovarian surface epithelium carcinomas. This updated info hopefully will lead to a better understanding of the human ovary and provide potential therapies for treating human infertility.
Collapse
Affiliation(s)
- Wenjing Zheng
- Department of Cell and Molecular Biology, University of Gothenburg, Gothenburg SE-40530, Sweden.
| | | | | | | |
Collapse
|
14
|
Fleming NI, Knower KC, Lazarus KA, Fuller PJ, Simpson ER, Clyne CD. Aromatase is a direct target of FOXL2: C134W in granulosa cell tumors via a single highly conserved binding site in the ovarian specific promoter. PLoS One 2010; 5:e14389. [PMID: 21188138 PMCID: PMC3004790 DOI: 10.1371/journal.pone.0014389] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 11/30/2010] [Indexed: 12/14/2022] Open
Abstract
Background Granulosa cell tumors (GCT) of the ovary often express aromatase and synthesize estrogen, which in turn may influence their progression. Recently a specific point mutation (C134W) in the FOXL2 protein was identified in >94% of adult-type GCT and it is likely to contribute to their development. A number of genes are known to be regulated by FOXL2, including aromatase/CYP19A1, but it is unclear which are direct targets and whether the C134W mutation alters their regulation. Recently, it has been reported that FOXL2 forms a complex with steroidogenic factor 1 (SF-1) which is a known regulator of aromatase in granulosa cells. Methodology/Principal Findings In this work, the human GCT-derived cell lines KGN and COV434 were heterozygous and wildtype for the FOXL2:C134W mutation, respectively. KGN had abundant FOXL2 mRNA expression but it was not expressed in COV434. Expression of exogenous FOXL2:C134W in COV434 cells induced higher expression of a luciferase reporter for the ovarian specific aromatase promoter, promoter II (PII) (−516bp) than expression of wildtype FOXL2, but did not alter induction of a similar reporter for the steroidogenic acute regulatory protein (StAR) promoter (−1300bp). Co-immunoprecipitation confirmed that FOXL2 bound SF-1 and that it also bound its homologue, liver receptor homologue 1 (LRH-1), however, the C134W mutation did not alter these interactions or induce a selective binding of the proteins. A highly conserved putative binding site for FOXL2 was identified in PII. FOXL2 was demonstrated to bind the site by electrophoretic mobility shift assays (EMSA) and site-directed mutagenesis of this element blocked its differential induction by wildtype FOXL2 and FOXL2:C134W. Conclusions/Significance These findings suggest that aromatase is a direct target of FOXL2:C134W in adult-type GCT via a single distinctive and highly conserved binding site in PII and therefore provide insight into the pathogenic mechanism of this mutation.
Collapse
Affiliation(s)
| | - Kevin C. Knower
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | - Kyren A. Lazarus
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | - Peter J. Fuller
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Evan R. Simpson
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Colin D. Clyne
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
- * E-mail:
| |
Collapse
|
15
|
Abstract
Granulosa cell tumors of the ovary represent ∼5% of malignant ovarian cancers. It has recently been reported that 95-97% of adult granulosa cell tumors carry a unique somatic mutation in the FOXL2 gene. We undertook this study to verify the presence of the FOXL2 Cys134Trp mutation in two geographically independent cohorts of granulosa cell tumors and to examine the expression pattern of FOXL2 in these tumors. A total of 56 tumors with the histological diagnosis of adult granulosa cell tumor from two centers, Melbourne and Helsinki, were examined for the presence of the mutation using direct sequence analysis. Two granulosa cell tumor-derived cell lines, COV434 and KGN, three juvenile granulosa cell tumors and control tissues were also examined. The expression of the FOXL2 gene was determined using quantitative RT-PCR and/or immunohistochemistry. We found that 52 of the 56 adult granulosa cell tumors harbor the mutation, of which three were hemi/homozygous. Of the four cases with wild-type FOXL2 sequence, reappraisal suggests that three may have been misclassified at primary diagnosis. The KGN cells were heterozygous for the mutation, whereas the COV434 cells had a wild-type FOXL2 genotype. The expression levels of FOXL2 were similar across the adult granulosa cell tumors and the normal ovary controls; one mutation-negative granulosa cell tumor had high FOXL2 mRNA levels, whereas the COV434 cells and two of the three juvenile granulosa cell tumors lacked the expression of FOXL2. Our data provide confirmation of the frequent presence of the FOXL2 C134W mutation in adult granulosa cell tumors and demonstrate that the mutation is not associated with altered FOXL2 expression. The mutation analysis may be a useful tool to differentiate particularly between cell-rich diffuse granulosa cell tumors and mitotically active sex cord-stromal tumors. This unique FOXL2 mutation appears to be characteristic of adult granulosa cell tumors.
Collapse
|
16
|
Middlebrook BS, Eldin K, Li X, Shivasankaran S, Pangas SA. Smad1-Smad5 ovarian conditional knockout mice develop a disease profile similar to the juvenile form of human granulosa cell tumors. Endocrinology 2009; 150:5208-17. [PMID: 19819941 PMCID: PMC2819741 DOI: 10.1210/en.2009-0644] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Granulosa cell tumors (GCTs) of the ovary are rare sex cord stromal tumors. Although generally indolent, GCTs recur, and if not diagnosed and treated in early stages, survival rates are significantly shortened. Very little is known regarding GCT etiology. Because of the low incidence of cases and lack of standard diagnostics, mouse models for granulosa cell tumors are a valuable tool for studying GCTs and provide models for developing diagnostic and treatment strategies. We recently developed a novel mouse model of metastatic granulosa cell tumors by genetic deletion of the bone morphogenetic protein signaling transcription factors (SMADs) in granulosa cells of the ovary. Histological and serum hormone analyses reveal that this mouse model most closely resembles the juvenile form of GCT. We further analyzed samples of human juvenile GCT (JGCT) for expression of anti-Müllerian hormone and activation of two major signaling pathways: TGFbeta/SMAD2/3 and wingless-related mouse mammary tumor virus integration site (Wnt)/beta-catenin. The TGFbeta family is active in mouse Smad1-Smad5 double knockout tumors, and here we show that this pathway, but not the beta-catenin pathway, is activated in samples of human JGCT. These data suggest that the SMAD family, possibly through disruption of SMAD1/5 or activation of SMAD2/3 may contribute to the pathogenesis of JGCT in humans.
Collapse
|