1
|
Karimnia N, Wilson AL, Green E, Matthews A, Jobling TW, Plebanski M, Bilandzic M, Stephens AN. Chemoresistance is mediated by ovarian cancer leader cells in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:276. [PMID: 34470672 PMCID: PMC8408956 DOI: 10.1186/s13046-021-02086-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
Background Leader cells are a subset of cancer cells that coordinate the complex cell-cell and cell-matrix interactions required for ovarian cancer migration, invasion, tumour deposition and are negatively associated with progression-free survival and response to therapy. Emerging evidence suggests leader cells may be enriched in response to chemotherapy, underlying disease recurrence following treatment. Methods CRISPR was used to insert a bicistronic T2A-GFP cassette under the native KRT14 (leader cell) promoter. 2D and 3D drug screens were completed in the presence of chemotherapies used in ovarian cancer management. Leader cell; proliferative (Ki67); and apoptotic status (Cleaved Caspase 3) were defined by live cell imaging and flow cytometry. Quantitative real-time PCR defined “stemness” profiles. Proliferation was assessed on the xCELLigence real time cell analyser. Statistical Analysis was performed using unpaired non-parametric t-tests or one-way ANOVA and Tukey’s multiple comparison post hoc. Results Leader cells represent a transcriptionally plastic subpopulation of ovarian cancer cells that arise independently of cell division or DNA replication, and exhibit a “stemness” profile that does not correlate with epithelial-to-mesenchymal transition. Chemotherapeutics increased apoptosis-resistant leader cells in vitro, who retained motility and expressed known chemo-resistance markers including ALDH1, Twist and CD44v6. Functional impairment of leader cells restored chemosensitivity, with leader cell-deficient lines failing to recover following chemotherapeutic intervention. Conclusions Our data demonstrate that ovarian cancer leader cells are resistant to a diverse array of chemotherapeutic agents, and are likely to play a critical role in the recurrence of chemo-resistant disease as drivers of poor treatment outcomes. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02086-3.
Collapse
Affiliation(s)
- Nazanin Karimnia
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| | - Amy L Wilson
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| | - Emma Green
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Amelia Matthews
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Thomas W Jobling
- Monash Medical Centre, Department of Gynaecology Oncology, Monash Health, Moorabbin, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.
| | - Andrew N Stephens
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| |
Collapse
|
2
|
Whiteley SL, Holleley CE, Wagner S, Blackburn J, Deveson IW, Marshall Graves JA, Georges A. Two transcriptionally distinct pathways drive female development in a reptile with both genetic and temperature dependent sex determination. PLoS Genet 2021; 17:e1009465. [PMID: 33857129 PMCID: PMC8049264 DOI: 10.1371/journal.pgen.1009465] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
How temperature determines sex remains unknown. A recent hypothesis proposes that conserved cellular mechanisms (calcium and redox; 'CaRe' status) sense temperature and identify genes and regulatory pathways likely to be involved in driving sexual development. We take advantage of the unique sex determining system of the model organism, Pogona vitticeps, to assess predictions of this hypothesis. P. vitticeps has ZZ male: ZW female sex chromosomes whose influence can be overridden in genetic males by high temperatures, causing male-to-female sex reversal. We compare a developmental transcriptome series of ZWf females and temperature sex reversed ZZf females. We demonstrate that early developmental cascades differ dramatically between genetically driven and thermally driven females, later converging to produce a common outcome (ovaries). We show that genes proposed as regulators of thermosensitive sex determination play a role in temperature sex reversal. Our study greatly advances the search for the mechanisms by which temperature determines sex.
Collapse
Affiliation(s)
- Sarah L. Whiteley
- Institute for Applied Ecology, University of Canberra, Canberra, Australia
- Australian National Wildlife Collection CSIRO National Research Collections Australia, Canberra, Australia
| | - Clare E. Holleley
- Australian National Wildlife Collection CSIRO National Research Collections Australia, Canberra, Australia
| | - Susan Wagner
- Institute for Applied Ecology, University of Canberra, Canberra, Australia
| | - James Blackburn
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent’s Clinical School, UNSW, Sydney, Australia
| | - Ira W. Deveson
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent’s Clinical School, UNSW, Sydney, Australia
| | - Jennifer A. Marshall Graves
- Institute for Applied Ecology, University of Canberra, Canberra, Australia
- Latrobe University, Melbourne, Australia
| | - Arthur Georges
- Institute for Applied Ecology, University of Canberra, Canberra, Australia
| |
Collapse
|
3
|
Moffitt LR, Bilandzic M, Wilson AL, Chen Y, Gorrell MD, Oehler MK, Plebanski M, Stephens AN. Hypoxia Regulates DPP4 Expression, Proteolytic Inactivation, and Shedding from Ovarian Cancer Cells. Int J Mol Sci 2020; 21:ijms21218110. [PMID: 33143089 PMCID: PMC7672561 DOI: 10.3390/ijms21218110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/28/2020] [Indexed: 01/03/2023] Open
Abstract
The treatment of ovarian cancer has not significantly changed in decades and it remains one of the most lethal malignancies in women. The serine protease dipeptidyl peptidase 4 (DPP4) plays key roles in metabolism and immunity, and its expression has been associated with either pro- or anti-tumour effects in multiple tumour types. In this study, we provide the first evidence that DPP4 expression and enzyme activity are uncoupled under hypoxic conditions in ovarian cancer cells. Whilst we identified strong up-regulation of DPP4 mRNA expression under hypoxic growth, the specific activity of secreted DPP4 was paradoxically decreased. Further investigation revealed matrix metalloproteinases (MMP)-dependent inactivation and proteolytic shedding of DPP4 from the cell surface, mediated by at least MMP10 and MMP13. This is the first report of uncoupled DPP4 expression and activity in ovarian cancer cells, and suggests a previously unrecognized, cell- and tissue-type-dependent mechanism for the regulation of DPP4 in solid tumours. Further studies are necessary to identify the functional consequences of DPP4 processing and its potential prognostic or therapeutic value.
Collapse
Affiliation(s)
- Laura R. Moffitt
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Maree Bilandzic
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Amy L. Wilson
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Yiqian Chen
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
- Robinson Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia;
| | - Andrew N. Stephens
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-3-8572-2686
| |
Collapse
|
4
|
New markers of human cumulus oophorus cells cultured in vitro – transcriptomic profile. ACTA ACUST UNITED AC 2020. [DOI: 10.2478/acb-2020-0007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Abstract
The presence of CCs around the oocyte after ovulation is one of the key elements contributing to oocyte developmental competence. In the presented study, we used CCs from 12 patients aged 18-40 diagnosed with infertility. After harvesting cells on day 1, 7, 15 and 30 of culture, total RNA was isolated and transcriptomic analysis was performed. The DAVID software indicated the following GO BP terms: “cell junction organization”, “cell migration”, “cell morphogenesis involved in differentiation”, “cell morphogenesis” and “cell motility”. Of the genes belonging to all ontological groups, the most downregulated were: SLC7A8, DFNB31, COL1A1, CDC42SE1, TGFBR3, HMGB1, with the most upregulated genes being: ANXA3, KIAA1199, HTR2B, VCAM1, DKK1.
While many studies focus on attempts to obtain fully competent oocytes, scientists still have difficulty attaining adequate results in vitro. Lack of adequate knowledge often results in low in vitro fertilization efficiency. Therefore, our research focuses on CCs cells, thanks to which the oocyte most likely acquires developmental competence. The main purpose of the study was to identify the potential molecular markers responsible for cell junction organization, migration, differentiation, morphogenesis and motility.
Running title: New markers of human cumulus oophorus cells cultured in vitro
Collapse
|
5
|
Chermuła B, Jeseta M, Sujka-Kordowska P, Konwerska A, Jankowski M, Kranc W, Kocherova I, Celichowski P, Antosik P, Bukowska D, Milakovic I, Machatkova M, Pawelczyk L, Iżycki D, Zabel M, Mozdziak P, Kempisty B, Piotrowska-Kempisty H. Genes regulating hormone stimulus and response to protein signaling revealed differential expression pattern during porcine oocyte in vitro maturation, confirmed by lipid concentration. Histochem Cell Biol 2020; 154:77-95. [PMID: 32189110 PMCID: PMC7343741 DOI: 10.1007/s00418-020-01866-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2020] [Indexed: 01/12/2023]
Abstract
Genes influencing oocyte maturation may be valuable for predicting their developmental potential, as well as discerning the mechanistic pathways regulating oocyte development. In the presented research microarray gene expression analysis of immature and in vitro matured porcine oocytes was performed. Two groups of oocytes were compared in the study: before (3 × n = 50) and after in vitro maturation (3 × n = 50). The selection of viable oocytes was performed using the brilliant cresyl blue (BCB) test. Furthermore, microarrays and RT-qPCR was used to analyze the transcriptome of the oocytes before and after IVM. The study focused on the genes undergoing differential expression in two gene-ontology groups: “Cellular response to hormone stimulus” and “Cellular response to unfolded protein”, which contain genes that may directly or indirectly be involved in signal transduction during oocyte maturation. Examination of all the genes of interest showed a lower level of their expression after IVM. From the total number of genes in these gene ontologies ten of the highest change in expression were identified: FOS, ID2, BTG2, CYR61, ESR1, AR, TACR3, CCND2, EGR2 and TGFBR3. The successful maturation of the oocytes was additionally confirmed with the use of lipid droplet assay. The genes were briefly described and related to the literature sources, to investigate their potential roles in the process of oocyte maturation. The results of the study may serve as a basic molecular reference for further research aimed at improving the methods of oocyte in vitro maturation, which plays an important role in the procedures of assisted reproduction.
Collapse
Affiliation(s)
- Błażej Chermuła
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland
| | - Aneta Konwerska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland
| | - Maurycy Jankowski
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiesława Kranc
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Ievgeniia Kocherova
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland
| | - Paweł Antosik
- Department of Veterinary Surgery, Nicolaus Copernicus University in Torun, Toruń, Poland
| | - Dorota Bukowska
- Department of Elementary and Preclinical Sciences, Nicolaus Copernicus University in Torun, Toruń, Poland
| | | | | | - Leszek Pawelczyk
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Dariusz Iżycki
- Chair of Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Zabel
- Department of Histology and Embryology, Wroclaw Medical University, Wrocław, Poland
- Division of Anatomy and Histology, University of Zielona Gora, Zielona Gora, Poland
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, USA
| | - Bartosz Kempisty
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic.
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland.
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland.
- Department of Veterinary Surgery, Nicolaus Copernicus University in Torun, Toruń, Poland.
| | | |
Collapse
|
6
|
Schoonderwoerd MJA, Goumans MJTH, Hawinkels LJAC. Endoglin: Beyond the Endothelium. Biomolecules 2020; 10:biom10020289. [PMID: 32059544 PMCID: PMC7072477 DOI: 10.3390/biom10020289] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keywords: endoglin; CD105 TGF-β; BMP9; ALK-1; TRC105; tumor microenvironment.
Collapse
Affiliation(s)
- Mark J. A. Schoonderwoerd
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Lukas J. A. C. Hawinkels
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
7
|
Betaglycan (TβRIII) is a Key Factor in TGF-β2 Signaling in Prepubertal Rat Sertoli Cells. Int J Mol Sci 2019; 20:ijms20246214. [PMID: 31835434 PMCID: PMC6941059 DOI: 10.3390/ijms20246214] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-βs (TGF-βs) signal after binding to the TGF-β receptors TβRI and TβRII. Recently, however, betaglycan (BG) was identified as an important co-receptor, especially for TGF-β2. Both proteins are involved in several testicular functions. Thus, we analyzed the importance of BG for TGF-β1/2 signaling in Sertoli cells with ELISAs, qRT-PCR, siRNA silencing and BrdU assays. TGF-β1 as well as TGF-β2 reduced shedding of membrane-bound BG (mBG), thus reducing the amount of soluble BG (sBG), which is often an antagonist to TGF-β signaling. Treatment of Sertoli cells with GM6001, a matrix metalloproteinases (MMP) inhibitor, also counteracted BG shedding, thus suggesting MMPs to be mainly involved in shedding. Interestingly, TGF-β2 but not TGF-β1 enhanced secretion of tissue inhibitor of metalloproteinases 3 (TIMP3), a potent inhibitor of MMPs. Furthermore, recombinant TIMP3 attenuated BG shedding. Co-stimulation with TIMP3 and TGF-β1 reduced phosphorylation of Smad3, while a combination of TIMP3/TGF-β2 increased it. Silencing of BG as well as TIMP3 reduced TGF-β2-induced phosphorylation of Smad2 and Smad3 significantly, once more highlighting the importance of BG for TGF-β2 signaling. In contrast, this effect was not observed with TIMP3/TGF-β1. Silencing of BG and TIMP3 decreased significantly Sertoli cell proliferation. Taken together, BG shedding serves a major role in TGF-β2 signaling in Sertoli cells.
Collapse
|
8
|
Fonseca PADS, dos Santos FC, Lam S, Suárez-Vega A, Miglior F, Schenkel FS, Diniz LDAF, Id-Lahoucine S, Carvalho MRS, Cánovas A. Genetic mechanisms underlying spermatic and testicular traits within and among cattle breeds: systematic review and prioritization of GWAS results. J Anim Sci 2018; 96:4978-4999. [PMID: 30304443 PMCID: PMC6276581 DOI: 10.1093/jas/sky382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022] Open
Abstract
Reduced bull fertility imposes economic losses in bovine herds. Specifically, testicular and spermatic traits are important indicators of reproductive efficiency. Several genome-wide association studies (GWAS) have identified genomic regions associated with these fertility traits. The aims of this study were as follows: 1) to perform a systematic review of GWAS results for spermatic and testicular traits in cattle and 2) to identify key functional candidate genes for these traits. The identification of functional candidate genes was performed using a systems biology approach, where genes shared between traits and studies were evaluated by a guilt by association gene prioritization (GUILDify and ToppGene software) in order to identify the best functional candidates. These candidate genes were integrated and analyzed in order to identify overlapping patterns among traits and breeds. Results showed that GWAS for testicular-related traits have been developed for beef breeds only, whereas the majority of GWAS for spermatic-related traits were conducted using dairy breeds. When comparing traits measured within the same study, the highest number of genes shared between different traits was observed, indicating a high impact of the population genetic structure and environmental effects. Several chromosomal regions were enriched for functional candidate genes associated with fertility traits. Moreover, multiple functional candidate genes were enriched for markers in a species-specific basis, taurine (Bos taurus) or indicine (Bos indicus). For the different candidate regions identified in the GWAS in the literature, functional candidate genes were detected as follows: B. Taurus chromosome X (BTX) (TEX11, IRAK, CDK16, ATP7A, ATRX, HDAC6, FMR1, L1CAM, MECP2, etc.), BTA17 (TRPV4 and DYNLL1), and BTA14 (MOS, FABP5, ZFPM2). These genes are responsible for regulating important metabolic pathways or biological processes associated with fertility, such as progression of spermatogenesis, control of ciliary activity, development of Sertoli cells, DNA integrity in spermatozoa, and homeostasis of testicular cells. This study represents the first systematic review on male fertility traits in cattle using a system biology approach to identify key candidate genes for these traits.
Collapse
Affiliation(s)
- Pablo Augusto de Souza Fonseca
- Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Stephanie Lam
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Aroa Suárez-Vega
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Filippo Miglior
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Flavio S Schenkel
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Samir Id-Lahoucine
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Angela Cánovas
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
9
|
TGF-β receptors: In and beyond TGF-β signaling. Cell Signal 2018; 52:112-120. [PMID: 30184463 DOI: 10.1016/j.cellsig.2018.09.002] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/07/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
Abstract
Transforming growth factor β (TGF-β) plays an important role in normal development and homeostasis. Dysregulation of TGF-β responsiveness and its downstream signaling pathways contribute to many diseases, including cancer initiation, progression, and metastasis. TGF-β ligands bind to three isoforms of the TGF-β receptor (TGFBR) with different affinities. TGFBR1 and 2 are both serine/threonine and tyrosine kinases, but TGFBR3 does not have any kinase activity. They are necessary for activating canonical or noncanonical signaling pathways, as well as for regulating the activation of other signaling pathways. Another prominent feature of TGF-β signaling is its context-dependent effects, temporally and spatially. The diverse effects and context dependency are either achieved by fine-tuning the downstream components or by regulating the expressions and activities of the ligands or receptors. Focusing on the receptors in events in and beyond TGF-β signaling, we review the membrane trafficking of TGFBRs, the kinase activity of TGFBR1 and 2, the direct interactions between TGFBR2 and other receptors, and the novel roles of TGFBR3.
Collapse
|
10
|
Han YL, Yin JJ, Cong JJ. Downregulation of microRNA-193-3p inhibits the progression of intrahepatic cholangiocarcinoma cells by upregulating TGFBR3. Exp Ther Med 2018; 15:4508-4514. [PMID: 29725386 DOI: 10.3892/etm.2018.5958] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/16/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to investigate the function of microRNA (miR)-193-3p in human intrahepatic cholangiocarcinoma (ICC) tissues and cells. To evaluate whether miR-193-3p was aberrantly upregulated, we used reverse transcription-quantitative polymerase chain reaction to detect the level of miR-193-3p in ICC tissues and ICC-9810 cells. The effects of miR-193-3p downregulation on ICC cell proliferation, migration and invasion were also measured by MTT, wound-healing and Transwell assays. Additionally, transforming growth factor-β receptor type 3 (TGFBR3) was investigated as a direct target of miR-193-3p by dual-luciferase reporter assays and western blot analyses. The results demonstrated that miR-193-3p was aberrantly upregulated in ICC tissues and cell lines. Furthermore, TGFBR3 was confirmed to be a target of miR-193-3p in ICC and was notably upregulated by miR-193-3p knockdown in the ICC-9810 cells. The knockdown of miR-193-3p also exerted direct inhibitory effects on the proliferation, migration and invasion of the ICC-9810 cells. Therefore, we present evidence that miR-193-3p plays a key role in promoting human ICC by regulating TGFBR3.
Collapse
Affiliation(s)
- Yu-Long Han
- Department of Hepatobiliary Laparoscopic Surgery Ward, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Jia-Jun Yin
- Department of Hepatobiliary Laparoscopic Surgery Ward, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Jian-Jun Cong
- Department of Hepatobiliary Laparoscopic Surgery Ward, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
11
|
Shima Y, Morohashi KI. Leydig progenitor cells in fetal testis. Mol Cell Endocrinol 2017; 445:55-64. [PMID: 27940302 DOI: 10.1016/j.mce.2016.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/18/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Testicular Leydig cells play pivotal roles in masculinization of organisms by producing androgens. At least two distinct Leydig cell populations sequentially emerge in the mammalian testis. Leydig cells in the fetal testis (fetal Leydig cells) appear just after initial sex differentiation and induce masculinization of male fetuses. Although there has been a debate on the fate of fetal Leydig cells in the postnatal testis, it has been generally believed that fetal Leydig cells regress and are completely replaced by another Leydig cell population, adult Leydig cells. Recent studies revealed that gene expression patterns are different between fetal and adult Leydig cells and that the androgens produced in fetal Leydig cells are different from those in adult Leydig cells in mice. Although these results suggested that fetal and adult Leydig cells have distinct origins, several recent studies of mouse models support the hypothesis that fetal and adult Leydig cells arise from a common progenitor pool. In this review, we first provide an overview of previous knowledge, mainly from mouse studies, focusing on the cellular origins of fetal Leydig cells and the regulatory mechanisms underlying fetal Leydig cell differentiation. In addition, we will briefly discuss the functional differences of fetal Leydig cells between human and rodents. We will also discuss recent studies with mouse models that give clues for understanding how the progenitor cells in the fetal testis are subsequently destined to become fetal or adult Leydig cells.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | - Ken-Ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
12
|
Wang Y, Bilandzic M, Ooi GT, Findlay JK, Stenvers KL. Endogenous inhibins regulate steroidogenesis in mouse TM3 Leydig cells by altering SMAD2 signalling. Mol Cell Endocrinol 2016; 436:68-77. [PMID: 27465829 DOI: 10.1016/j.mce.2016.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/28/2022]
Abstract
This study tested the hypothesis that inhibins act in an autocrine manner on Leydig cells using a pre-pubertal Leydig cell line, TM3, as a model of immature Leydig cells. The expression of Inha, Inhba, and Inhbb in TM3 cells was determined by RT-PCR and the production of the inhibin-alpha subunit was confirmed by western blot. Knockdown of Inha expression resulted in significant decreases in the expression of Leydig cell markers Cyp17a1, Cyp11a1, Nr5a1, and Insl3. Western blot showed that activin A, TGFβ1 and TGFβ2 activated SMAD2, and that knockdown of Inha expression in TM3 cells enhanced both activin A- and TGFβ-induced SMAD2 activation. SB431542, a chemical inhibitor of the TGFβ/activin type I receptors, blocked ligand-induced SMAD2 activation and the downregulation of Cyp17a1 expression. Our findings demonstrate that TGFβs and activin A negatively regulate steroidogenic gene expression in TM3 cells via ALK4/5 and SMAD2 and endogenous inhibins can counter this regulation.
Collapse
Affiliation(s)
- Yao Wang
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia.
| | - Maree Bilandzic
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia
| | - Guck T Ooi
- Sun BioMedical Technologies, 209 W. Ridgecrest Blvd, Suite A, Ridgecrest, CA, 93555, USA
| | - Jock K Findlay
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia
| | - Kaye L Stenvers
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3168, Australia
| |
Collapse
|
13
|
Wen Q, Cheng CY, Liu YX. Development, function and fate of fetal Leydig cells. Semin Cell Dev Biol 2016; 59:89-98. [PMID: 26968934 PMCID: PMC5016207 DOI: 10.1016/j.semcdb.2016.03.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/21/2022]
Abstract
During fetal testis development, fetal Leydig cells (FLCs) are found to be originated from multiple progenitor cells. FLC specification and function are under tight regulation of specific genes and signaling proteins. Furthermore, Sertoli cells play a crucial role to regulate FLC differentiation during fetal testis development. FLC progenitor- and FLC-produced biomolecules are also involved in the differentiation and activity of rodent FLCs. The main function of FLCs is to produce androgens to masculinize XY embryos. However, FLCs are capable of producing androstenedione but not testosterone due to the lack of 17β-HSD (17β-hydroxysteroid dehydrogenase), but fetal Sertoli cells express 17β-HSD which thus transforms androstenedione to testosterone in the fetal testis. On the other hand, FLCs produce activin A to regulate Sertoli cell proliferation, and Sertoli cells in turn modulate testis cord expansion. It is now generally accepted that adult Leydig cells (ALCs) gradually replace FLCs during postnatal development to produce testosterone to support spermatogenesis as FLCs undergo degeneration in neonatal and pre-pubertal testes. However, based on studies using genetic tracing mouse models, FLCs are found to persist in adult testes, making up ∼20% of total Leydig cells. In this review, we evaluate the latest findings regarding the development, function and fate of FLCs during fetal and adult testis development.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York 10065, United States.
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Wang C, Li C, Li H, Ma W, Chen S, Zhao Y, Rao J, Zhou X. Downregulation of the expression of inhibin α subunit and betaglycan in porcine cystic follicles. J Vet Med Sci 2015; 77:1419-25. [PMID: 26097017 PMCID: PMC4667659 DOI: 10.1292/jvms.14-0617] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inhibins, as members of the transforming growth factor beta (TGF-β)
superfamily, downregulate the synthesis and secretion of follicle-stimulating hormone
(FSH) in an endocrine manner. The role of inhibin/betaglycan in the ovary regulation
recently gained attention. To date, no data exist on the function of inhibin α subunit and
betaglycan in cystic follicles. In this study, the expressions of inhibin α subunit and
betaglycan in cystic follicles were investigated using immunohistochemistry, real-time PCR
and Western blot analysis. Both inhibin α subunit and betaglycan immunoreactivities were
mainly localized in the granulosa cells of follicles. Expression of inhibin α subunit and
betaglycan was inferior in cystic follicles compared with that in normal large follicles.
However, the result of enzyme-linked immunosorbent assay showed no significant difference
in the decreasing in concentration of inhibin α subunit in cystic follicular fluid
compared with the control (P>0.05). In this study, we explored the
effects of FSH on betaglycan expression in granulosa cells in vitro. As
expected, a significant increase in the expressions of betaglycan mRNA and protein in
granulosa cells was observed in response to exogenous FSH (30
ng/ml) (P<0.05) compared with the
control. Consequently, this study provides evidence that the expressions of inhibin α
subunit and betaglycan are inferior in cystic follicles, and this may be caused by the
decrease in FSH in the presence of a cystic follicle.
Collapse
Affiliation(s)
- Chunqiang Wang
- College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, 5333 Xian Road, Changchun, Jilin 130062, P.R. of China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Itman C, Bielanowicz A, Goh H, Lee Q, Fulcher AJ, Moody SC, Doery JCG, Martin J, Eyre S, Hedger MP, Loveland KL. Murine Inhibin α-Subunit Haploinsufficiency Causes Transient Abnormalities in Prepubertal Testis Development Followed by Adult Testicular Decline. Endocrinology 2015; 156:2254-68. [PMID: 25781564 DOI: 10.1210/en.2014-1555] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activin production and signaling must be strictly regulated for normal testis development and function. Inhibins are potent activin inhibitors; mice lacking the inhibin-α gene (Inha-/- mice) cannot make inhibin and consequently have highly elevated activin and FSH serum concentrations and excessive activin signaling, resulting in somatic gonadal tumors and infertility. Dose-dependent effects of activin in testicular biology have been widely reported; hence, we hypothesized that male mice lacking one copy of the Inha gene would produce less inhibin and have an abnormal reproductive phenotype. To test this, we compared hormone concentrations, testis development, and sperm production in Inha+/+ and Inha+/- mice. Serum and testicular inhibin-α concentrations in adult Inha+/- mice were approximately 33% lower than wild type, whereas activin A, activin B, FSH, LH, and T were normal. Sixteen-day-old Inha+/- mice had a mixed phenotype, with tubules containing extensive germ cell depletion juxtaposed to tubules with advanced Sertoli and germ cell development. This abnormal phenotype resolved by day 28. By 8 weeks, Inha+/- testes were 11% larger than wild type and supported 44% greater daily sperm production. By 26 weeks of age, Inha+/- testes had distinct abnormalities. Although still fertile, Inha+/- mice had a 27% reduction in spermatogenic efficiency, a greater proportion of S-phase Sertoli cells and lower Leydig cell CYP11A1 expression. This study is the first to identify an intratesticular role for inhibin/inhibin-α subunit, demonstrating that a threshold level of this protein is required for normal testis development and to sustain adult somatic testicular cell function.
Collapse
Affiliation(s)
- Catherine Itman
- Priority Research Centres for Reproductive Science (C.I., A.B., J.M., S.E.) and Chemical Biology (C.I.), School of Environmental and Life Sciences, Faculty of Science and Information Technology, University of Newcastle, Callaghan, New South Wales 2308, Australia; Departments of Anatomy and Developmental Biology (H.G., Q.L., K.L.L.) and Biochemistry and Molecular Biology (S.C.M., K.L.L.) and Monash Micro Imaging (A.J.F.), Monash University, Clayton, Victoria 3800, Australia; and Faculty of Medicine, Nursing, and Health Sciences (J.C.G.D.), Department of Medicine, Monash Medical Centre, and Monash Institute of Medical Research-Prince Henry's Institute of Medical Research (M.P.H.), Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Barrionuevo F, Burgos M, Jiménez R. Origin and function of embryonic Sertoli cells. Biomol Concepts 2015; 2:537-47. [PMID: 25962053 DOI: 10.1515/bmc.2011.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 09/16/2011] [Indexed: 11/15/2022] Open
Abstract
In the adult testis, Sertoli cells (SCs) are the epithelial supporting cells of the seminiferous tubules that provide germ cells (GCs) with the required nutrients and structural and regulatory support to complete spermatogenesis. SCs also form the blood-testis barrier, phagocytose apoptotic spermatocytes and cell debris derived from spermiogenesis, and produce and secrete numerous paracrine and endocrine signals involved in different regulatory processes. In addition to their essential functions in the adult testis, SCs play a pivotal role during testis development. They are the first cells to differentiate in the embryonic XY gonadal primordium and are involved in the regulation of testis-specific differentiation processes, such as prevention of GC entry into meiosis, Leydig and peritubular myoid cell differentiation, and regression of the Müllerian duct, the anlagen of the uterus, oviducts, and the upper part of the vagina. Expression of the Y-linked gene SRY in pre-SCs initiates a genetic cascade that leads to SC differentiation and subsequently to testis development. Since the identification of the SRY gene, many Sertoli-specific transcription factors and signals underlying the molecular mechanisms of early testis differentiation have been identified. Here, we review the state of the art of the molecular interactions that commit the supporting cell lineage of the gonadal primordium to differentiate as SCs and the subsequent Sertoli-specific signaling pathways involved in early testis differentiation.
Collapse
|
17
|
Barthold JS, Wang Y, Kolon TF, Kollin C, Nordenskjöld A, Olivant Fisher A, Figueroa TE, BaniHani AH, Hagerty JA, Gonzalez R, Noh PH, Chiavacci RM, Harden KR, Abrams DJ, Kim CE, Mateson AB, Robbins AK, Li J, Akins RE, Hakonarson H, Devoto M. Phenotype specific association of the TGFBR3 locus with nonsyndromic cryptorchidism. J Urol 2014; 193:1637-45. [PMID: 25390077 DOI: 10.1016/j.juro.2014.10.097] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2014] [Indexed: 11/19/2022]
Abstract
PURPOSE Based on a genome-wide association study of testicular dysgenesis syndrome showing a possible association with TGFBR3, we analyzed data from a larger, phenotypically restricted cryptorchidism population for potential replication of this signal. MATERIALS AND METHODS We excluded samples based on strict quality control criteria, leaving 844 cases and 2,718 controls of European ancestry that were analyzed in 2 separate groups based on genotyping platform (ie Illumina® HumanHap550, version 1 or 3, or Human610-Quad, version 1 BeadChip in group 1 and Human OmniExpress 12, version 1 BeadChip platform in group 2). Analyses included genotype imputation at the TGFBR3 locus, association analysis of imputed data with correction for population substructure, subsequent meta-analysis of data for groups 1 and 2, and selective genotyping of independent cases (330) and controls (324) for replication. We also measured Tgfbr3 mRNA levels and performed TGFBR3/betaglycan immunostaining in rat fetal gubernaculum. RESULTS We identified suggestive (p ≤ 1× 10(-4)) association of markers in/near TGFBR3, including rs9661103 (OR 1.40; 95% CI 1.20, 1.64; p = 2.71 × 10(-5)) and rs10782968 (OR 1.58; 95% CI 1.26, 1.98; p = 9.36 × 10(-5)) in groups 1 and 2, respectively. In subgroup analyses we observed strongest association of rs17576372 (OR 1.42; 95% CI 1.24, 1.60; p = 1.67 × 10(-4)) with proximal and rs11165059 (OR 1.32; 95% CI 1.15, 1.38; p = 9.42 × 10(-4)) with distal testis position, signals in strong linkage disequilibrium with rs9661103 and rs10782968, respectively. Association of the prior genome-wide association study signal (rs12082710) was marginal (OR 1.13; 95% CI 0.99, 1.28; p = 0.09 for group 1), and we were unable to replicate signals in our independent cohort. Tgfbr3/betaglycan was differentially expressed in wild-type and cryptorchid rat fetal gubernaculum. CONCLUSIONS These data suggest complex or phenotype specific association of cryptorchidism with TGFBR3 and the gubernaculum as a potential target of TGFβ signaling.
Collapse
Affiliation(s)
- Julia S Barthold
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware; Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| | - Yanping Wang
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Thomas F Kolon
- Division of Urology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Claude Kollin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordenskjöld
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Alicia Olivant Fisher
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - T Ernesto Figueroa
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Ahmad H BaniHani
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Jennifer A Hagerty
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Ricardo Gonzalez
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Paul H Noh
- Division of Urology, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Rosetta M Chiavacci
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kisha R Harden
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Debra J Abrams
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cecilia E Kim
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Abigail B Mateson
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Alan K Robbins
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Jin Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert E Akins
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marcella Devoto
- Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
18
|
Sarraj MA, Escalona RM, Western P, Findlay JK, Stenvers KL. Effects of TGFbeta2 on Wild-Type and Tgfbr3 Knockout Mouse Fetal Testis1. Biol Reprod 2013; 88:66. [DOI: 10.1095/biolreprod.112.102194] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
19
|
Bilandzic M, Chu S, Wang Y, Tan HL, Fuller PJ, Findlay JK, Stenvers KL. Betaglycan alters NFκB-TGFβ2 cross talk to reduce survival of human granulosa tumor cells. Mol Endocrinol 2013; 27:466-79. [PMID: 23322721 DOI: 10.1210/me.2012-1239] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The molecular pathways controlling granulosa cell tumor (GCT) survival are poorly understood. In many cell types, nuclear factor-κB (NFκB) and TGFβ coordinately regulate cell survival to maintain tissue homeostasis. Because GCT cell lines exhibit constitutively activated NFκB, we hypothesized that NFκB blocks TGFβ-mediated cell death in GCT cells. To test this hypothesis, we used the human GCT cell line KGN, which exhibits loss of betaglycan, a TGFβ co-receptor. After inhibition of NFκB in KGN cells, re-expression of betaglycan resulted in a decrease in cell viability, which was further decreased by TGFβ2. Intriguingly, TGFβ2 increased NFκB reporter activity in control cells, but betaglycan expression suppressed both basal and TGFβ2-stimulated NFκB activity. Chemical inhibition of Mothers against decapentaplegic homolog 2/3 (SMAD2/3) signaling or SMAD2/3 gene silencing revealed that both SMADs contributed to cell survival. Furthermore, inhibiting NFκB activity resulted in a specific reduction in SMAD3 expression. Conversely, overexpression of SMAD3 increased basal NFκB activity and countered betaglycan-mediated suppression of NFκB activity. Finally, ERK1/2 activation emerged as the point of convergence of NFκB, SMAD3, and TGFβ2/betaglycan governance of GCT cell viability. Key findings in KGN cells were reproduced in a second GCT cell line, COV434. Collectively, our data establish that both SMAD2/3 and NFκB signaling pathways support GCT cell viability and suggest the existence of a positive feedback loop between NFκB and SMAD3 signaling in late-stage GCT. Furthermore, our data suggest that loss of betaglycan during tumor progression in GCT alters the functional outcomes generated by NFκB and TGFβ pathway cross talk.
Collapse
Affiliation(s)
- Maree Bilandzic
- Prince Henry’s Institute, Department of Developmental Biology and Anatomy, Monash University, Clayton, Victoria 3168, Australia.
| | | | | | | | | | | | | |
Collapse
|
20
|
Morohashi K, Baba T, Tanaka M. Steroid Hormones and the Development of Reproductive Organs. Sex Dev 2013; 7:61-79. [DOI: 10.1159/000342272] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
21
|
Ricci G, Guglielmo MC, Caruso M, Ferranti F, Canipari R, Galdieri M, Catizone A. Hepatocyte Growth Factor Is a Mouse Fetal Leydig Cell Terminal Differentiation Factor1. Biol Reprod 2012; 87:146. [DOI: 10.1095/biolreprod.112.104638] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
22
|
Aleman-Muench GR, Mendoza V, Stenvers K, Garcia-Zepeda EA, Lopez-Casillas F, Raman C, Soldevila G. Betaglycan (TβRIII) is expressed in the thymus and regulates T cell development by protecting thymocytes from apoptosis. PLoS One 2012; 7:e44217. [PMID: 22952931 PMCID: PMC3430661 DOI: 10.1371/journal.pone.0044217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/03/2012] [Indexed: 11/18/2022] Open
Abstract
TGF-β type III receptor (TβRIII) is a coreceptor for TGFβ family members required for high-affinity binding of these ligands to their receptors, potentiating their cellular functions. TGF-β [1]–[3], bone morphogenetic proteins (BMP2/4) and inhibins regulate different checkpoints during T cell differentiation. Although TβRIII is expressed on hematopoietic cells, the role of this receptor in the immune system remains elusive. Here, we provide the first evidence that TβRIII is developmentally expressed during T cell ontogeny, and plays a crucial role in thymocyte differentiation. Blocking of endogenous TβRIII in fetal thymic organ cultures led to a delay in DN-DP transition. In addition, in vitro development of TβRIII−/− thymic lobes also showed a significant reduction in absolute thymocyte numbers, which correlated with increased thymocyte apoptosis, resembling the phenotype reported in Inhibin α −/− thymic lobes. These data suggest that Inhibins and TβRIII may function as a molecular pair regulating T cell development.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cytoprotection
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Fetus/metabolism
- Gene Expression Regulation, Developmental
- Mice
- Mice, Inbred C57BL
- Organ Culture Techniques
- Proteoglycans/antagonists & inhibitors
- Proteoglycans/deficiency
- Proteoglycans/genetics
- Proteoglycans/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/deficiency
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Stromal Cells/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/metabolism
- Thymocytes/cytology
- Thymocytes/metabolism
- Thymus Gland/cytology
- Thymus Gland/embryology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- German R. Aleman-Muench
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, México
| | - Valentin Mendoza
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, México
| | - Kaye Stenvers
- Reproductive Development and Cancer laboratory, Prince Henry′s Institute of Medical Research, Clayton, Victoria, Australia
| | - Eduardo A. Garcia-Zepeda
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, México
| | - Fernando Lopez-Casillas
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, México
| | - Chander Raman
- Departments of Medicine and Microbiology, Division of Clinical Immunology and Rheumatology University of Alabama at Birmingham, Alabama, United States of America
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, México
- * E-mail:
| |
Collapse
|
23
|
Bilandzic M, Stenvers KL. Reprint of: Betaglycan: a multifunctional accessory. Mol Cell Endocrinol 2012; 359:13-22. [PMID: 22521265 DOI: 10.1016/j.mce.2012.03.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 04/18/2011] [Accepted: 04/18/2011] [Indexed: 12/21/2022]
Abstract
Betaglycan is a co-receptor for the TGFβ superfamily, particularly important in establishing the potency of its ligands on their target cells. In recent years, new insights have been gained into the structure and function of betaglycan, expanding its role from that of a simple co-receptor to include additional ligand-dependent and ligand-independent roles. This review focuses on recent advances in the betaglycan field, with a particular emphasis on its newly discovered actions in mediating the trafficking of TGFβ superfamily receptors and as a determinant of the functional output of TGFβ superfamily signalling. In addition, this review encompasses a discussion of the emerging roles of the betaglycan/inhibin pathway in reproductive cancers and disease.
Collapse
Affiliation(s)
- Maree Bilandzic
- Prince Henry's Institute, PO Box 5152, Clayton, Victoria 3168, Australia.
| | | |
Collapse
|
24
|
Barakat B, Itman C, Mendis SH, Loveland KL. Activins and inhibins in mammalian testis development: new models, new insights. Mol Cell Endocrinol 2012; 359:66-77. [PMID: 22406273 DOI: 10.1016/j.mce.2012.02.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 01/15/2023]
Abstract
The discovery of activin and inhibins as modulators of the hypothalamic-pituitary-gonadal axis has set the foundation for understanding their central importance to many facets of development and disease. This review contains an overview of the processes and cell types that are central to testis development and spermatogenesis and then provides an update focussed on information gathered over the past five years to address new concepts about how these proteins function to control testis development in fetal and juvenile life. Current knowledge about the interactive nature of the transforming growth factor-β (TGFβ) superfamily signalling network is applied to recent findings about activins and inhibins in the testis. Information about the regulated synthesis of signalling components and signalling regulators in the testis is integrated with new concepts that demonstrate their functional significance. The importance of activin bioactivity levels or dosage in controlling balanced growth of spermatogonial cells and their niche at different stages of testis development is highlighted.
Collapse
Affiliation(s)
- B Barakat
- Monash Institute of Reproduction and Development, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
25
|
Bilezikjian LM, Justice NJ, Blackler AN, Wiater E, Vale WW. Cell-type specific modulation of pituitary cells by activin, inhibin and follistatin. Mol Cell Endocrinol 2012; 359:43-52. [PMID: 22330643 PMCID: PMC3367026 DOI: 10.1016/j.mce.2012.01.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 01/01/2023]
Abstract
Activins are multifunctional proteins and members of the TGF-β superfamily. Activins are expressed locally in most tissues and, analogous to the actions of other members of this large family of pleiotropic factors, play prominent roles in the regulation of diverse biological processes in both differentiated and embryonic stem cells. They have an essential role in maintaining tissue homeostasis in the adult and are known to contribute to the developmental programs in the embryo. Activins are further implicated in the growth and metastasis of tumor cells. Through distinct modes of action, inhibins and follistatins function as antagonists of activin and several other TGF-β family members, including a subset of BMPs/GDFs, and modulate cellular responses and the signaling cascades downstream of these ligands. In the pituitary, the activin pathway is known to regulate key aspects of gonadotrope functions and also exert effects on other pituitary cell types. As in other tissues, activin is produced locally by pituitary cells and acts locally by exerting cell-type specific actions on gonadotropes. These local actions of activin on gonadotropes are modulated by the autocrine/paracrine actions of locally secreted follistatin and by the feedback actions of gonadal inhibin. Knowledge about the mechanism of activin, inhibin and follistatin actions is providing information about their importance for pituitary function as well as their contribution to the pathophysiology of pituitary adenomas. The aim of this review is to highlight recent findings and summarize the evidence that supports the important functions of activin, inhibin and follistatin in the pituitary.
Collapse
Affiliation(s)
- Louise M Bilezikjian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
26
|
Bilandzic M, Stenvers KL. Betaglycan: a multifunctional accessory. Mol Cell Endocrinol 2011; 339:180-9. [PMID: 21550381 DOI: 10.1016/j.mce.2011.04.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 04/18/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
Abstract
Betaglycan is a co-receptor for the TGFβ superfamily, particularly important in establishing the potency of its ligands on their target cells. In recent years, new insights have been gained into the structure and function of betaglycan, expanding its role from that of a simple co-receptor to include additional ligand-dependent and ligand-independent roles. This review focuses on recent advances in the betaglycan field, with a particular emphasis on its newly discovered actions in mediating the trafficking of TGFβ superfamily receptors and as a determinant of the functional output of TGFβ superfamily signalling. In addition, this review encompasses a discussion of the emerging roles of the betaglycan/inhibin pathway in reproductive cancers and disease.
Collapse
Affiliation(s)
- Maree Bilandzic
- Prince Henry's Institute, P.O. Box 5152, Clayton, Victoria 3168, Australia.
| | | |
Collapse
|
27
|
Betaglycan is required for the establishment of nephron endowment in the mouse. PLoS One 2011; 6:e18723. [PMID: 21533152 PMCID: PMC3078907 DOI: 10.1371/journal.pone.0018723] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 03/08/2011] [Indexed: 01/24/2023] Open
Abstract
Betaglycan is an accessory receptor for the transforming growth factor-β (TGFβ) superfamily, many members of which play key roles in kidney development. The purpose of this study was to define the role of this co-receptor on fetal murine kidney development. Stereological examination of embryonic and adult betaglycan heterozygous kidneys revealed augmented nephron number relative to littermate controls. Fetal heterozygous kidneys exhibited accelerated ureteric branching, which correlated with augmented nephron development at embryonic day (e) 15.5. In contrast, betaglycan null kidneys exhibited renal hypoplasia from e13.5 and reduced nephron number at e15.5. Quantitative real-time PCR analysis of e11.5–e14.5 kidneys demonstrated that heterozygous kidneys exhibited a transient decrease in Bmp4 expression at e11.5 and a subsequent cascade of changes in the gene regulatory network that governs metanephric development, including significant increases in Pax2, Eya1, Gdnf, Ret, Wnt4, and Wt1 expression. Conversely, gene expression in null kidneys was normal until e13.5, when significant reductions were detected in the expression of Bmp4 as well as other key metanephric regulatory genes. Tgfb1 and Tgfb2 mRNA expression was down-regulated in both nulls and heterozygotes at e13.5 and e14.5. The opposing morphological and molecular phenotypes in betaglycan heterozygote and null mutants demonstrate that the levels of betaglycan must be tightly regulated for optimal kidney development.
Collapse
|
28
|
Blair CR, Stone JB, Wells RG. The type III TGF-β receptor betaglycan transmembrane-cytoplasmic domain fragment is stable after ectodomain cleavage and is a substrate of the intramembrane protease γ-secretase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:332-9. [PMID: 21167215 DOI: 10.1016/j.bbamcr.2010.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Revised: 12/02/2010] [Accepted: 12/07/2010] [Indexed: 11/20/2022]
Abstract
The Type III TGF-β receptor, betaglycan, is a widely expressed proteoglycan co-receptor for TGF-β superfamily ligands. The full-length protein undergoes ectodomain cleavage with release of a soluble ectodomain fragment. The fate of the resulting transmembrane-cytoplasmic fragment, however, has never been explored. We demonstrate here that the transmembrane-cytoplasmic fragment is stable in transfected cells and in cell lines expressing endogenous betaglycan. Production of this fragment is inhibited by the ectodomain shedding inhibitor TAPI-2. Treatment of cells with inhibitors of the intramembrane protease γ-secretase stabilizes this fragment, suggesting that it is a substrate of γ-secretase. Expression of the transmembrane-cytoplasmic fragment as well as γ-secretase inhibitor stabilization are independent of TGF-β1 or -β2 and are unaffected by mutation of the cytoplasmic domain serines that undergo phosphorylation. γ-Secretase inhibition or the expression of a transmembrane-cytoplasmic fragment in HepG2 cells blunted TGF-β2 signaling. Our findings thus suggest that the transmembrane-cytoplasmic fragment remaining after betaglycan ectodomain cleavage is stable and a substrate of γ-secretase, which may have significant implications for the TGF-β signaling response.
Collapse
Affiliation(s)
- Cheyne R Blair
- Cellular and Molecular Biology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
29
|
Stenvers KL, Findlay JK. Inhibins: from reproductive hormones to tumor suppressors. Trends Endocrinol Metab 2010; 21:174-80. [PMID: 20005735 DOI: 10.1016/j.tem.2009.11.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 11/23/2009] [Accepted: 11/23/2009] [Indexed: 11/30/2022]
Abstract
Inhibins are peptide hormones shown originally to be produced by the gonads to regulate the secretion of follicle stimulating hormone by pituitary gonadotropes. Although gonadotropes have been regarded as the canonical inhibin target cells, in recent years extrapituitary actions of inhibins have come into the spotlight. In particular, disruptions to the local actions of inhibins in peripheral tissues might underlie certain diseases, especially cancers of the reproductive tract. This review focuses on recent advances in the inhibin field, with a particular emphasis on the determinants of inhibin availability, mechanisms of inhibin action, and the physiological relevancy of local inhibin actions in the development and progression of reproductive cancers.
Collapse
Affiliation(s)
- Kaye L Stenvers
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168 Australia.
| | | |
Collapse
|
30
|
Sarraj MA, Escalona RM, Umbers A, Chua HK, Small C, Griswold M, Loveland K, Findlay JK, Stenvers KL. Fetal testis dysgenesis and compromised Leydig cell function in Tgfbr3 (beta glycan) knockout mice. Biol Reprod 2009; 82:153-62. [PMID: 19696014 DOI: 10.1095/biolreprod.109.078766] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Betaglycan (Tgfbr3) is a coreceptor for transforming growth factor-beta (TGFB) superfamily ligands. In the current study, a defect in seminiferous cord formation was detected in 12.5-13.5 days postcoitum (dpc) beta glycan null murine testis. Immunohistochemistry with antibodies against cell-specific markers revealed defects in somatic cell populations. To confirm these data, quantitative real-time PCR was performed to determine changes in the expression levels of genes involved in fetal testis cell differentiation and function. The expression levels of the Leydig cell markers Insl3, Cyp17a1, Cyp11a1, Star, and Hsd3b1 were reduced in knockout testis compared to wild-type testis, beginning at 12.5 dpc. Whole mount in situ hybridization confirmed that Cyp11a1 expression was reduced in the null testis, but its distribution pattern was unchanged. Apoptosis was not affected by the loss of beta glycan, but proliferation within the interstitium was reduced at 14.5 dpc. However, morphometric analysis showed no changes in Leydig cell counts between the wild-type and the knockout testes at 14.5 dpc, indicating that fetal Leydig function, rather than number, was affected by the loss of beta glycan. The expression levels of Sertoli cell markers Dhh, Sox9, and Amh were also reduced in the knockout testis at 14.5 dpc. However, the expression of fetal germ cell markers Pou5f1 and DDX4 were not changed across the genotypes at any age examined. Our data show that the presence of beta glycan is required for normal cord formation, normal fetal Leydig cell development, and the establishment of fetal testis endocrine function, thus implicating TGFB superfamily members as regulators of early fetal testis structure and function.
Collapse
Affiliation(s)
- Mai A Sarraj
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Escalona RM, Stenvers KL, Farnworth PG, Findlay JK, Ooi GT. Reducing betaglycan expression by RNA interference (RNAi) attenuates inhibin bioactivity in LbetaT2 gonadotropes. Mol Cell Endocrinol 2009; 307:149-56. [PMID: 19524135 DOI: 10.1016/j.mce.2009.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 10/20/2022]
Abstract
Betaglycan is an inhibin-binding protein co-receptor, the forced expression of which confers inhibin responsiveness on cells previously non-responsive to inhibin. The present study determines whether removal of betaglycan expression in otherwise inhibin-responsive cells will render the cells insensitive to inhibin. Small interfering RNAs (siRNAs) designed to the betaglycan gene were transfected into LbetaT2 gonadotrope cells to 'knock-down' betaglycan expression. To control for non-specific effects, siRNAs corresponding to an unrelated sequence (BF-1) were used. Two activin-responsive promoter constructs were used to assess inhibin bioactivity; an ovine FSHbeta promoter (oFSHbeta-lux), and a construct containing three copies of the activin-responsive sequence from the GnRHR promoter (3XpGRAS-PRL-lux). Activin stimulated the activity of both promoters 5-8-fold. Inhibin suppressed these activin-stimulated promoter activities by 52+/-11% and 51+/-7%, respectively. Similar inhibin suppression was also seen for cells co-transfected with the control BF-1 siRNAs. In contrast, inhibin's ability to suppress activin-stimulated activity was significantly reduced (33+/-3%, p<0.005 and 24+/-4%, p<0.045, respectively) in cells co-transfected with betaglycan siRNAs. These results demonstrated that endocrine effects of inhibin as a negative feedback controller of FSH production in gonadotropes are dependent on betaglycan expression.
Collapse
Affiliation(s)
- Ruth M Escalona
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
32
|
Dias VL, Rajpert-De Meyts E, McLachlan R, Loveland KL. Analysis of activin/TGFB-signaling modulators within the normal and dysfunctional adult human testis reveals evidence of altered signaling capacity in a subset of seminomas. Reproduction 2009; 138:801-11. [PMID: 19661148 DOI: 10.1530/rep-09-0206] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activin is a pleiotropic growth factor belonging to the transforming growth factor-beta (TGFB) superfamily of signaling molecules. Regulated activin signaling is known to influence several steps in rodent male gamete differentiation. TGFB ligand isoforms, TGFB1-B3, also influence germ cell survival in the rodent testis at the onset of spermatogenesis and around the time of puberty. Given the importance of regulated activin and TGFB signaling in testis development and function, we sought to investigate the cellular production sites of activin/TGFB-signaling modulators in normal and dysfunctional adult human testes samples. Signaling transducers phosphorylated SMAD2/3, and signaling modulators SMAD6, MAN-1, inhibin alpha (INHA), and beta-glycan were detected in Bouins fixed, paraffin-embedded adult human testis sections using immunohistochemistry. Additional samples examined were from testicular cancer patients and from normal men subjected to gonadotropin suppression with androgen-based contraceptives. Our findings identify distinct differences between normal and gonadotropin-deprived human testis in the expression and cellular localization of activin/TGFB-signaling modulators. The presence of a nuclear phosphorylated SMAD2/3 signal in all analyzed seminoma specimens indicated active activin/TGFB signaling. Moreover, a subset of seminoma specimens exhibited selective enhanced expression of beta-glycan (4 out of 28 seminoma tumors), INHA (6 out of 28), and MAN-1 (6 out of 28), highlighting potential functional differences between individual tumors in their capacity to regulate activin/TGFB signaling. Within the heterogenous nonseminomas, expression of signaling modulators was variable and reflected the degree of somatic differentiation. Thus, synthesis of activin and TGFB-signaling modulators may be affected by spermatogenic disruption and altered hormone levels in the testis.
Collapse
Affiliation(s)
- Vinali L Dias
- Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
33
|
Identification of novel oocyte and granulosa cell markers. Gene Expr Patterns 2009; 9:404-10. [PMID: 19539053 DOI: 10.1016/j.gep.2009.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Revised: 06/09/2009] [Accepted: 06/10/2009] [Indexed: 01/01/2023]
Abstract
Here we present novel gene expression patterns in the ovary as part of an ongoing assessment of published micro-array data from mouse oocytes and embryos. We present the expression patterns of 13 genes that had been determined by micro-array to be expressed in the mature egg, but not during subsequent preimplantation development. In-situ hybridization of sectioned ovaries revealed that these genes were expressed in one of two distinct patterns: (1) oocyte-specific or (2) expressed in both the oocyte and surrounding granulosa cells. Despite the fact that micro-array data demonstrated expression in the egg, several of these genes are expressed at low levels in the oocyte, but strongly expressed in granulosa cells. Eleven of these genes have no reported function or expression during oogenesis, indicating that this approach is a necessary step towards functional annotation of the genome. Also of note is that while some of these gene products have been well characterized in other tissues and cell types, others are relatively unstudied in the literature. Our results provide novel gene expression information that may provide insights into the molecular mechanisms of follicular recruitment, oocyte maturation and ovulation and will direct further experimentation into the role these genes play during oogenesis.
Collapse
|
34
|
Bilandzic M, Chu S, Farnworth PG, Harrison C, Nicholls P, Wang Y, Escalona RM, Fuller PJ, Findlay JK, Stenvers KL. Loss of betaglycan contributes to the malignant properties of human granulosa tumor cells. Mol Endocrinol 2009; 23:539-48. [PMID: 19164448 DOI: 10.1210/me.2008-0300] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Betaglycan is a type III TGFbeta receptor that modulates cellular sensitivity to inhibins and TGFbeta. Previous studies have suggested that betaglycan acts as a tumor suppressor in certain human epithelial cancers. However, the roles of betaglycan in ovarian granulosa cell tumors (GCTs) are poorly understood. The objective of this study was to determine whether human GCTs exhibit betaglycan expression and, if so, what impact this receptor has on tumor biology. Real-time PCR was used to quantify betaglycan transcripts in human GCTs (n = 17) and normal premenopausal ovaries (n = 11). This analysis established that GCTs exhibited a significant 2-fold lower mean betaglycan mRNA level as compared with the normal ovary (P < 0.05). Similarly, two human GCT cell lines, KGN and COV434, exhibited low betaglycan expression and poor responsiveness to TGFbeta and inhibin A in luciferase reporter assays, which was restored by stable transfection of wild-type betaglycan. Betaglycan significantly increased the adhesion of COV434 (P < 0.05) and KGN (P < 0.0001) cells, decreased cellular invasion through Matrigel, and inhibited wound healing. Expression of mutant forms of betaglycan that are defective in TGFbeta and/or inhibin binding in each GCT cell line revealed that the inhibitory effects of betaglycan on wound healing were most strongly linked to the inhibin-binding region of betaglycan. Furthermore, knockdown of INHA mRNA expression abrogated the betaglycan-mediated inhibition of wound healing and invasion, whereas both INHA silencing and TGFbeta neutralization abolished the betaglycan-mediated increase in adhesion to substrate. These data suggest that loss of betaglycan contributes to the pathogenesis of GCTs.
Collapse
|