1
|
Kato-Shinomiya M, Sugino H, Wang L, Saito Y, He J, Tanei ZI, Oda Y, Tanikawa S, Tanino M, Gong JP, Tsuda M, Tanaka S. SLC13A5 plays an essential role in the energy shift to oxidative phosphorylation in cisplatin-resistant mesothelioma stem cells. Pathol Int 2025; 75:151-165. [PMID: 39912507 DOI: 10.1111/pin.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025]
Abstract
Mesothelioma is a highly aggressive tumor affecting an increasing number of patients worldwide. Owing to the poor clinical outcomes associated with current therapies, the development of novel therapies that target cancer stem cells (CSCs) is desirable. Here, we examined the applicability of our previously established hydrogel-based rapid CSC generation method to human mesothelioma cell lines and further analyzed the characteristics of the induced mesothelioma stem cell (MesoSC) -like cells. Human mesothelioma cell lines cultured on hydrogels presented increased expression of pan-stem cell markers and acquired spheroid formation and early tumorigenicity, suggesting that MesoSC-like cells are highly malignant. Microarray analysis demonstrated that the expression of SLC13A5, a citrate transporter involved in TCA cycle, was significantly induced in the resulting MesoSC-like cells. The overexpression of SLC13A5 resulted in a metabolic shift toward oxidative phosphorylation, increased phosphorylation of ERK and YAP, and increased SOX2 expression, leading to increased cisplatin resistance. scRNA-seq database analysis revealed that clinical mesothelioma samples contained a small number of SLC13A5-expressing cells. Our findings suggest that the hydrogel-based CSC generation method is also effective for human mesothelioma cells and that SLC13A5 may contribute to MesoSC survival. The new properties of MesoSCs revealed in this study may provide clues for establishing future treatments.
Collapse
Affiliation(s)
- Marie Kato-Shinomiya
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Hirokazu Sugino
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yusuke Saito
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Jintao He
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Zen-Ichi Tanei
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Tanikawa
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Laboratory Medicine and Pathobiology, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Canada
| | - Mishie Tanino
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Jian Ping Gong
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
2
|
Du R, Wen L, Niu M, Zhao L, Guan X, Yang J, Zhang C, Liu H. Activin receptors in human cancer: Functions, mechanisms, and potential clinical applications. Biochem Pharmacol 2024; 222:116061. [PMID: 38369212 DOI: 10.1016/j.bcp.2024.116061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/18/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Activins are members of the transforming growth factor-β (TGF-β) superfamily and act as key regulators in various physiological processes, such as follicle and embryonic development, as well as in multiple human diseases, including cancer. They have been established to signal through three type I and two type II serine/threonine kinase receptors, which, upon ligand binding, form a final signal-transducing receptor complex that activates downstream signaling and governs gene expression. Recent research highlighted the dysregulation of the expression or activity of activin receptors in multiple human cancers and their critical involvement in cancer progression. Furthermore, expression levels of activin receptors have been associated with clinicopathological features and patient outcomes across different cancers. However, there is currently a paucity of comprehensive systematic reviews of activin receptors in cancer. Thus, this review aimed to consolidate existing knowledge concerning activin receptors, with a primary emphasis on their signaling cascade and emerging biological functions, regulatory mechanisms, and potential clinical applications in human cancers in order to provide novel perspectives on cancer prognosis and targeted therapy.
Collapse
Affiliation(s)
- Ruochen Du
- First Clinical Medical College of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Department of Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Liqi Wen
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Min Niu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Liting Zhao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Xiaoya Guan
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Jiao Yang
- Department of Anatomy, the Basic Medical School of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China.
| | - Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; First Clinical Medical College of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Department of Cell Biology and Genetics, the Basic Medical School of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China.
| |
Collapse
|
3
|
Schelch K, Eder S, Zitta B, Phimmachanh M, Johnson TG, Emminger D, Wenninger‐Weinzierl A, Sturtzel C, Poplimont H, Ries A, Hoetzenecker K, Hoda MA, Berger W, Distel M, Dome B, Reid G, Grusch M. YB-1 regulates mesothelioma cell migration via snail but not EGFR, MMP1, EPHA5 or PARK2. Mol Oncol 2024; 18:815-831. [PMID: 36550787 PMCID: PMC10994239 DOI: 10.1002/1878-0261.13367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/11/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Pleural mesothelioma (PM) is characterized by rapid growth, local invasion, and limited therapeutic options. The multifunctional oncoprotein Y-box-binding protein-1 (YB-1) is frequently overexpressed in cancer and its inhibition reduces aggressive behavior in multiple tumor types. Here, we investigated the effects of YB-1 on target gene regulation and PM cell behavior. Whereas siRNA-mediated YB-1 knockdown reduced cell motility, YB-1 overexpression resulted in scattering, increased migration, and intravasation in vitro. Furthermore, YB-1 stimulated PM cell spreading in zebrafish. Combined knockdown and inducible overexpression of YB-1 allowed bidirectional control and rescue of cell migration, the pattern of which was closely followed by the mRNA and protein levels of EGFR and the protein level of snail, whereas the mRNA levels of MMP1, EPHA5, and PARK2 showed partial regulation by YB-1. Finally, we identified snail as a critical regulator of YB-1-mediated cell motility in PM. This study provides insights into the mechanism underlying the aggressive nature of PM and highlights the important role of YB-1 in this cancer. In this context, we found that YB-1 closely regulates EGFR and snail, and, moreover, that YB-1-induced cell migration depends on snail.
Collapse
Affiliation(s)
- Karin Schelch
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
- Department of Thoracic SurgeryMedical University of ViennaAustria
| | - Sebastian Eder
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| | - Benjamin Zitta
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| | - Monica Phimmachanh
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
- University of Technology SydneyNSWAustralia
| | - Thomas G. Johnson
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
- The University of SydneyNSWAustralia
| | - Dominik Emminger
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| | | | - Caterina Sturtzel
- St. Anna Children's Cancer Research Institute, Innovative Cancer ModelsViennaAustria
| | - Hugo Poplimont
- St. Anna Children's Cancer Research Institute, Innovative Cancer ModelsViennaAustria
| | - Alexander Ries
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| | | | - Mir A. Hoda
- Department of Thoracic SurgeryMedical University of ViennaAustria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| | - Martin Distel
- St. Anna Children's Cancer Research Institute, Innovative Cancer ModelsViennaAustria
| | - Balazs Dome
- Department of Thoracic SurgeryMedical University of ViennaAustria
- National Koranyi Institute of PulmonologyBudapestHungary
- Department of Thoracic SurgerySemmelweis University and National Institute of OncologyBudapestHungary
| | - Glen Reid
- Department of PathologyDunedin School of MedicineNew Zealand
- The Maurice Wilkins CentreUniversity of OtagoDunedinNew Zealand
| | - Michael Grusch
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaAustria
| |
Collapse
|
4
|
Lan X, Guo L, Hu C, Zhang Q, Deng J, Wang Y, Chen ZJ, Yan J, Li Y. Fibronectin mediates activin A-promoted human trophoblast migration and acquisition of endothelial-like phenotype. Cell Commun Signal 2024; 22:61. [PMID: 38263146 PMCID: PMC10807102 DOI: 10.1186/s12964-023-01463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/27/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND During human early placentation, a proportion of extravillous trophoblasts (EVTs) migrate to the maternal decidua, differentiating into endovascular EVTs to remodel spiral arteries and ensure the establishment of blood circulation at the maternal-fetal interface. Inadequate EVT migration and endovascular differentiation are closely associated with adverse pregnancy outcomes such as miscarriage. Activin A and fibronectin are both secretory molecules abundantly expressed at the maternal-fetal interface. Activin A has been reported to regulate EVT biological functions. However, whether fibronectin mediates activin A-promoted EVT migration and acquisition of endothelial-like phenotype as well as the underlying molecular mechanisms remain unknown. Additionally, the role of fibronectin in pregnancy establishment and maintenance warrants further investigation. METHODS Primary and immortalized (HTR8/SVneo) human EVTs were used as in vitro study models. Cultured human first-trimester chorionic villous explants were utilized for ex vivo validation. A local fibronectin knockdown model in ICR mouse uteri, achieved by nonviral in vivo transfection with small interfering RNA (siRNA) targeting fibronectin 1 (si-Fn1), was employed to explore the roles of fibronectin in the establishment and maintenance of early pregnancy. RESULTS Our results showed that activin A treatment significantly induced fibronectin 1 (FN1) mRNA expression and fibronectin protein production, which is essential for human trophoblast migration and endothelial-like tube formation. Both basal and activin A-upregulated fibronectin expression were abolished by the TGF-β type I receptor inhibitor SB431542 or siRNA-mediated knockdown of activin receptor-like kinase (ALK4) or SMAD4. Moreover, activin A-increased trophoblast migration and endothelial-like tube formation were attenuated following the depletion of fibronectin. Fibronectin knockdown via intrauterine siRNA administration reduced CD31 and cytokeratin 8 (CK8) expression at the maternal-fetal interface, resulting in a decrease in the number of implantation sites and embryos. CONCLUSIONS Our study demonstrates that activin A promotes trophoblast cell migration and acquisition of endothelial-like phenotype via ALK4-SMAD2/3-SMAD4-mediated fibronectin upregulation. Furthermore, through a local fibronectin knockdown model in mouse uteri, we found that the absence of fibronectin at the maternal-fetal interface impedes endovascular migration of trophoblasts and decidual vascularization, thereby interfering with early embryo implantation and the maintenance of pregnancy. These findings provide novel insights into placental development during early pregnancy establishment and contribute to the advancement of therapeutic approaches for managing pregnancy complications related to trophoblast dysfunction.
Collapse
Affiliation(s)
- Xiangxin Lan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Ling Guo
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Cuiping Hu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Qian Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Jianye Deng
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Yufeng Wang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, 250012, Shandong, China
| | - Junhao Yan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China.
| | - Yan Li
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
5
|
Inactivating the Uninhibited: The Tale of Activins and Inhibins in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24043332. [PMID: 36834742 PMCID: PMC9963072 DOI: 10.3390/ijms24043332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Advances in technology and biomedical knowledge have led to the effective diagnosis and treatment of an increasing number of rare diseases. Pulmonary arterial hypertension (PAH) is a rare disorder of the pulmonary vasculature that is associated with high mortality and morbidity rates. Although significant progress has been made in understanding PAH and its diagnosis and treatment, numerous unanswered questions remain regarding pulmonary vascular remodeling, a major factor contributing to the increase in pulmonary arterial pressure. Here, we discuss the role of activins and inhibins, both of which belong to the TGF-β superfamily, in PAH development. We examine how these relate to signaling pathways implicated in PAH pathogenesis. Furthermore, we discuss how activin/inhibin-targeting drugs, particularly sotatercep, affect pathophysiology, as these target the afore-mentioned specific pathway. We highlight activin/inhibin signaling as a critical mediator of PAH development that is to be targeted for therapeutic gain, potentially improving patient outcomes in the future.
Collapse
|
6
|
Li F, Long Y, Yu X, Tong Y, Gong L. Different Immunoregulation Roles of Activin A Compared With TGF-β. Front Immunol 2022; 13:921366. [PMID: 35774793 PMCID: PMC9237220 DOI: 10.3389/fimmu.2022.921366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A, a critical member of the transforming growth factor-β (TGF-β) superfamily, is a pluripotent factor involved in allergies, autoimmune diseases, cancers and other diseases with immune disorder. Similar to its family member, TGF-β, activin A also transmits signals through SMAD2/SMAD3, however, they bind to distinct receptors. Recent studies have uncovered that activin A plays a pivotal role in both innate and adaptive immune systems. Here we mainly focus its effects on activation, differentiation, proliferation and function of cells which are indispensable in the immune system and meanwhile make some comparisons with those of TGF-β.
Collapse
Affiliation(s)
- Fanglin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiru Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- *Correspondence: Likun Gong,
| |
Collapse
|
7
|
Orozco Morales ML, Rinaldi CA, de Jong E, Lansley SM, Gummer JP, Olasz B, Nambiar S, Hope DE, Casey TH, Lee YCG, Leslie C, Nealon G, Shackleford DM, Powell AK, Grimaldi M, Balaguer P, Zemek RM, Bosco A, Piggott MJ, Vrielink A, Lake RA, Lesterhuis WJ. PPARα and PPARγ activation is associated with pleural mesothelioma invasion but therapeutic inhibition is ineffective. iScience 2022; 25:103571. [PMID: 34984327 PMCID: PMC8692993 DOI: 10.1016/j.isci.2021.103571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/16/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Mesothelioma is a cancer that typically originates in the pleura of the lungs. It rapidly invades the surrounding tissues, causing pain and shortness of breath. We compared cell lines injected either subcutaneously or intrapleurally and found that only the latter resulted in invasive and rapid growth. Pleural tumors displayed a transcriptional signature consistent with increased activity of nuclear receptors PPARα and PPARγ and with an increased abundance of endogenous PPAR-activating ligands. We found that chemical probe GW6471 is a potent, dual PPARα/γ antagonist with anti-invasive and anti-proliferative activity in vitro. However, administration of GW6471 at doses that provided sustained plasma exposure levels sufficient for inhibition of PPARα/γ transcriptional activity did not result in significant anti-mesothelioma activity in mice. Lastly, we demonstrate that the in vitro anti-tumor effect of GW6471 is off-target. We conclude that dual PPARα/γ antagonism alone is not a viable treatment modality for mesothelioma.
Collapse
Affiliation(s)
- M. Lizeth Orozco Morales
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
| | - Catherine A. Rinaldi
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
- Centre for Microscopy Characterisation and Analysis, Nedlands, WA 6009, Australia
| | - Emma de Jong
- Telethon Kids Institute, University of Western Australia, West Perth, WA 6872, Australia
| | | | - Joel P.A. Gummer
- School of Science, Department of Science, Edith Cowan University, Joondalup, WA 6027, Australia
- UWA Medical School, The University of Western Australia, Crawley, WA 6009, Australia
| | - Bence Olasz
- School of Molecular Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Shabarinath Nambiar
- School of Veterinary and Life Science, Murdoch University, Murdoch, WA 6150, Australia
| | - Danika E. Hope
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
| | - Thomas H. Casey
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
| | - Y. C. Gary Lee
- Institute for Respiratory Health, Nedlands, WA 6009, Australia
| | - Connull Leslie
- Department of Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Gareth Nealon
- School of Molecular Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - David M. Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Andrew K. Powell
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Marina Grimaldi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier 34090, France
| | - Patrick Balaguer
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier 34090, France
| | - Rachael M. Zemek
- Telethon Kids Institute, University of Western Australia, West Perth, WA 6872, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, West Perth, WA 6872, Australia
| | - Matthew J. Piggott
- School of Molecular Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Alice Vrielink
- School of Molecular Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Richard A. Lake
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
| | - W. Joost Lesterhuis
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA 6009, Australia
- Telethon Kids Institute, University of Western Australia, West Perth, WA 6872, Australia
| |
Collapse
|
8
|
Ramundo V, Zanirato G, Aldieri E. The Epithelial-to-Mesenchymal Transition (EMT) in the Development and Metastasis of Malignant Pleural Mesothelioma. Int J Mol Sci 2021; 22:ijms222212216. [PMID: 34830097 PMCID: PMC8621591 DOI: 10.3390/ijms222212216] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive tumor mainly associated with asbestos exposure and is characterized by a very difficult pharmacological approach. One of the molecular mechanisms associated with cancer onset and invasiveness is the epithelial-to-mesenchymal transition (EMT), an event induced by different types of inducers, such as transforming growth factor β (TGFβ), the main inducer of EMT, and oxidative stress. MPM development and metastasis have been correlated to EMT; On one hand, EMT mediates the effects exerted by asbestos fibers in the mesothelium, particularly via increased oxidative stress and TGFβ levels evoked by asbestos exposure, thus promoting a malignant phenotype, and on the other hand, MPM acquires invasiveness via the EMT event, as shown by an upregulation of mesenchymal markers or, although indirectly, some miRNAs or non-coding RNAs, all demonstrated to be involved in cancer onset and metastasis. This review aims to better describe how EMT is involved in driving the development and invasiveness of MPM, in an attempt to open new scenarios that are useful in the identification of predictive markers and to improve the pharmacological approach against this aggressive cancer.
Collapse
Affiliation(s)
- Valeria Ramundo
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.R.); (G.Z.)
| | - Giada Zanirato
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.R.); (G.Z.)
| | - Elisabetta Aldieri
- Department of Oncology, University of Torino, 10126 Torino, Italy; (V.R.); (G.Z.)
- Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates “G. Scansetti”, University of Torino, 10126 Torino, Italy
- Correspondence:
| |
Collapse
|
9
|
Gandhi M, Nair S. New vistas in malignant mesothelioma: MicroRNA architecture and NRF2/MAPK signal transduction. Life Sci 2020; 257:118123. [PMID: 32710945 DOI: 10.1016/j.lfs.2020.118123] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022]
Abstract
Malignant mesothelioma (MM) is a cancer of the mesothelial lining of the pleura, peritoneum, pericardium and testes. The most common form is asbestos-linked MM that is etiologically linked to repeated asbestos exposure with a long latency period, although non-asbestos MM has also been reported. Late diagnosis, poor survival rates, lack of diagnostic and prognostic markers act as major impediments in the clinical management of MM. Despite advances in immune checkpoint inhibition and CAR T-cell-based therapies, MM which is of different histologic subtypes remains challenging to treat. We review microRNAs (miRNAs) and the miRNA interactome implicated in MM which can be useful as circulating miRNA biomarkers for early diagnosis of MM and as biomarkers for prognostication in MM. Further, we underscore the relevance of the NRF2/MAPK signal transduction pathway that has been implicated in MM which may be useful as druggable targets or as biomarkers of predictive response. In addition, since MM is driven partly by inflammation, we elucidate chemopreventive phytochemicals that are beneficial in MM, either via crosstalk with the NRF2/MAPK pathway or via concerted anticancer mechanisms, and may be of benefit as adjuvants in chemotherapy. Taken together, a multifactorial approach comprising identification of miRNA target hubs and NRF2/MAPK biomarkers along with appropriately designed clinical trials may enable early detection and faster intervention in MM translating into better patient outcomes for this aggressive cancer.
Collapse
Affiliation(s)
- Manav Gandhi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, VL Mehta Road, Vile Parle (West), Mumbai 400 056, India
| | - Sujit Nair
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, VL Mehta Road, Vile Parle (West), Mumbai 400 056, India.
| |
Collapse
|
10
|
Cakiroglu E, Senturk S. Genomics and Functional Genomics of Malignant Pleural Mesothelioma. Int J Mol Sci 2020; 21:ijms21176342. [PMID: 32882916 PMCID: PMC7504302 DOI: 10.3390/ijms21176342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, aggressive cancer of the mesothelial cells lining the pleural surface of the chest wall and lung. The etiology of MPM is strongly associated with prior exposure to asbestos fibers, and the median survival rate of the diagnosed patients is approximately one year. Despite the latest advancements in surgical techniques and systemic therapies, currently available treatment modalities of MPM fail to provide long-term survival. The increasing incidence of MPM highlights the need for finding effective treatments. Targeted therapies offer personalized treatments in many cancers. However, targeted therapy in MPM is not recommended by clinical guidelines mainly because of poor target definition. A better understanding of the molecular and cellular mechanisms and the predictors of poor clinical outcomes of MPM is required to identify novel targets and develop precise and effective treatments. Recent advances in the genomics and functional genomics fields have provided groundbreaking insights into the genomic and molecular profiles of MPM and enabled the functional characterization of the genetic alterations. This review provides a comprehensive overview of the relevant literature and highlights the potential of state-of-the-art genomics and functional genomics research to facilitate the development of novel diagnostics and therapeutic modalities in MPM.
Collapse
Affiliation(s)
- Ece Cakiroglu
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir 35340, Turkey;
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir 35340, Turkey
- Correspondence:
| |
Collapse
|
11
|
Ries A, Schelch K, Falch D, Pany L, Hoda MA, Grusch M. Activin A: an emerging target for improving cancer treatment? Expert Opin Ther Targets 2020; 24:985-996. [PMID: 32700590 DOI: 10.1080/14728222.2020.1799350] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Activin A is involved in the regulation of a surprisingly broad number of processes that are relevant for cancer development and treatment; it is implicated in cell autonomous functions and multiple regulatory functions in the tumor microenvironment. AREAS COVERED This article summarizes the current knowledge about activin A in cell growth and death, migration and metastasis, angiogenesis, stemness and drug resistance, regulation of antitumor immunity, and cancer cachexia. We explore the role of activin A as a biomarker and discuss strategies for using it as target for cancer therapy. Literature retrieved from Medline until 25 June 2020 was considered. EXPERT OPINION While many functions of activin A were investigated in preclinical models, there is currently limited experience from clinical trials. Activin A has growth- and migration-promoting effects, contributes to immune evasion and cachexia and is associated with shorter survival in several cancer types. Targeting activin A could offer the chance to simultaneously limit tumor growth and spreading, improve drug response, boost antitumor immune responses and improve cancer-associated or treatment-associated cachexia, bone loss, and anemia. Nevertheless, defining which patients have the highest likelihood of benefiting from these effects is challenging and will require further work.
Collapse
Affiliation(s)
- Alexander Ries
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Karin Schelch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - David Falch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Laura Pany
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Mir Alireza Hoda
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna , Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| |
Collapse
|
12
|
Yuan J, Xie A, Cao Q, Li X, Chen J. INHBB Is a Novel Prognostic Biomarker Associated with Cancer-Promoting Pathways in Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6909672. [PMID: 33083477 PMCID: PMC7563060 DOI: 10.1155/2020/6909672] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Inhibin subunit beta B (INHBB) is a protein-coding gene that participated in the synthesis of the transforming growth factor-β (TGF-β) family members. The study is aimed at exploring the clinical significance of INHBB in patients with colorectal cancer (CRC) by bioinformatics analysis. METHODS Real-time PCR and analyses of Oncomine, Gene Expression Omnibus (GEO), and The Cancer Genome Atlas (TCGA) databases were utilized to evaluate the INHBB gene transcription level of colorectal cancer (CRC) tissue. We evaluated the INHBB methylation level and the relationship between expression and methylation levels of CpG islands in CRC tissue. The corresponding clinical data were obtained to further explore the association of INHBB with clinical and survival features. In addition, Gene Set Enrichment Analysis (GSEA) was performed to explore the gene ontology and signaling pathways of INHBB involved. RESULTS INHBB expression was elevated in CRC tissue. Although the promoter of INHBB was hypermethylated in CRC, methylation did not ultimately correlate with the expression of INHBB. Overexpression of INHBB was significantly and positively associated with invasion depth, distant metastasis, and TNM stage. Cox regression analyses and Kaplan-Meier survival analysis indicated that high expression of INHBB was correlated with worse overall survival (OS) and disease-free survival (DFS). GSEA showed that INHBB was closely correlated with 5 cancer-promoting signaling pathways including the Hedgehog signaling pathway, ECM receptor interaction, TGF-β signaling pathway, focal adhesion, and pathway in cancer. INHBB expression significantly promoted macrophage infiltration and inhibited memory T cell, mast cell, and dendritic cell infiltration. INHBB expression was positively correlated with stromal and immune scores of CRC samples. CONCLUSION INHBB might be a potential prognostic biomarker and a novel therapeutic target for CRC.
Collapse
Affiliation(s)
- Jinpeng Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
- Shantou University Medical College, China
| | - Aosi Xie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Qiangjian Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Xinxin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| | - Juntian Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, China
| |
Collapse
|
13
|
Paajanen J, Ilonen I, Lauri H, Järvinen T, Sutinen E, Ollila H, Rouvinen E, Lemström K, Räsänen J, Ritvos O, Koli K, Myllärniemi M. Elevated Circulating Activin A Levels in Patients With Malignant Pleural Mesothelioma Are Related to Cancer Cachexia and Reduced Response to Platinum-based Chemotherapy. Clin Lung Cancer 2019; 21:e142-e150. [PMID: 31734071 DOI: 10.1016/j.cllc.2019.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/14/2019] [Accepted: 10/18/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Previous preclinical studies have shown that activin A is overexpressed in malignant pleural mesothelioma (MPM), associates with cancer cachexia, and is observed in in vitro resistance to platinum-based chemotherapy. We evaluated circulating activin levels and their endogenous antagonists' follistatin/follistatin-like 3 in intrathoracic tumors. MATERIALS AND METHODS Patients suspected of thoracic malignancy were recruited prior to surgery. Serum samples were collected from 21 patients with MPM, 59 patients with non-small-cell lung cancer (NSCLC), and 22 patients with benign lung lesions. Circulating activin/follistatin levels were measured using enzyme-linked immunosorbent assay and compared with clinicopathologic parameters. RESULTS Circulating activin A levels were elevated in patients with MPM when compared with patients with NSCLC or benign lung lesion samples (P < .0001). Also, follistatin and follistatin-like 3 levels were the highest in MPM, although with less difference compared with activin A. Receiver operating characteristic analysis for activin A for separating NSCLC from benign lung lesion showed an area under the curve of 0.856 (95% confidence interval, 0.77-0.94). Activin A levels were higher in patients with cachexia (P < .001). In patients with MPM, activin A levels correlated positively with computed tomography-based baseline tumor size (R = 0.549; P = .010) and the change in tumor size after chemotherapy (R = 0.743; P = .0006). Patients with partial response or stable disease had lower circulating activin A levels than the ones with progressive disease (P = .028). CONCLUSION Activin A serum level could be used as a biomarker in differentiating malignant and benign lung tumors. Circulating activin A levels were elevated in MPM and associates with cancer cachexia and reduced chemotherapy response.
Collapse
Affiliation(s)
- Juuso Paajanen
- Department of Pulmonary Medicine, Helsinki University Hospital, Helsinki, Finland; Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland.
| | - Ilkka Ilonen
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Helena Lauri
- Medical Imaging Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tommi Järvinen
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eva Sutinen
- Department of Pulmonary Medicine, Helsinki University Hospital, Helsinki, Finland; Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Hely Ollila
- Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Eeva Rouvinen
- Department of Pulmonary Medicine, Helsinki University Hospital, Helsinki, Finland; Transplantation Immunology Program, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Karl Lemström
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Transplantation Immunology Program, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Jari Räsänen
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katri Koli
- Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Marjukka Myllärniemi
- Department of Pulmonary Medicine, Helsinki University Hospital, Helsinki, Finland; Individualized Drug Therapy, Research Programs Unit, Medical Faculty, University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Mak G, Soria JC, Blagden SP, Plummer R, Fleming RA, Nebot N, Zhang J, Mazumdar J, Rogan D, Gazzah A, Rizzuto I, Greystoke A, Yan L, Tolson J, Auger KR, Arkenau HT. A phase Ib dose-finding, pharmacokinetic study of the focal adhesion kinase inhibitor GSK2256098 and trametinib in patients with advanced solid tumours. Br J Cancer 2019; 120:975-981. [PMID: 30992546 PMCID: PMC6735221 DOI: 10.1038/s41416-019-0452-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 11/10/2022] Open
Abstract
Background Combined focal adhesion kinase (FAK) and MEK inhibition may provide greater anticancer effect than FAK monotherapy. Methods This dose-finding phase Ib study (adaptive 3 + 3 design) determined the maximum tolerated dose (MTD) of trametinib and the FAK inhibitor GSK2256098 in combination. Eligible patients had mesothelioma or other solid tumours with probable mitogen activated protein kinase pathway activation. Adverse events (AEs), dose-limiting toxicities, disease progression and pharmacokinetics/pharmacodynamics were analysed. Results Thirty-four subjects were enrolled. The GSK2256098/trametinib MTDs were 500 mg twice daily (BID)/0.375 mg once daily (QD) (high/low) and 250 mg BID/0.5 mg QD (low/high). The most common AEs were nausea, diarrhoea, decreased appetite, pruritus, fatigue and rash; none were grade 4. Systemic exposure to trametinib increased when co-administered with GSK2256098, versus trametinib monotherapy; GSK2256098 pharmacokinetics were unaffected by concomitant trametinib. Median progression-free survival (PFS) was 11.8 weeks (95% CI: 6.1–24.1) in subjects with mesothelioma and was longer with Merlin-negative versus Merlin-positive tumours (15.0 vs 7.3 weeks). Conclusions Trametinib exposure increased when co-administered with GSK2256098, but not vice versa. Mesothelioma patients with loss of Merlin had longer PFS than subjects with wild-type, although support for efficacy with this combination was limited. Safety profiles were acceptable up to the MTD.
Collapse
Affiliation(s)
- Gabriel Mak
- Sarah Cannon Research Institute, London, UK.,Cancer Centre, University College London, London, UK
| | - Jean-Charles Soria
- Drug Development Department at Gustave Roussy Cancer Campus, University Paris-Sud, Paris, France
| | - Sarah P Blagden
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK.,Department of Oncology, University of Oxford, Oxford, UK
| | - Ruth Plummer
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Ronald A Fleming
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA
| | - Noelia Nebot
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA.,Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Jolly Mazumdar
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA.,Chimeron Bio, New York, NY, 10016, USA
| | - Debra Rogan
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA
| | - Anas Gazzah
- Drug Development Department at Gustave Roussy Cancer Campus, University Paris-Sud, Paris, France
| | - Ivana Rizzuto
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Alastair Greystoke
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Li Yan
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA
| | - Jerry Tolson
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA
| | - Kurt R Auger
- GlaxoSmithKline, Research Triangle Park, NC and Upper Providence, Collegeville, PA, USA
| | - Hendrik-Tobias Arkenau
- Sarah Cannon Research Institute, London, UK. .,Cancer Centre, University College London, London, UK.
| |
Collapse
|
15
|
Activin A regulates activities of peripheral blood natural killer cells of mouse in an autocrine and paracrine manner. Exp Cell Res 2019; 374:114-121. [DOI: 10.1016/j.yexcr.2018.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/16/2018] [Accepted: 11/17/2018] [Indexed: 12/19/2022]
|
16
|
|
17
|
Lu X, Wan X, Li X, Pan K, Maimaitiaili W, Zhang Y. Expression of TLR4 gene is downregulated in acquired immune deficiency syndrome-associated Kaposi's sarcoma. Exp Ther Med 2018; 17:27-34. [PMID: 30651761 PMCID: PMC6307526 DOI: 10.3892/etm.2018.6941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/12/2017] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the expression of Toll-like receptor 4 (TLR4) and proteins involved in its associated signaling pathways in patients with classic Kaposi's sarcoma (KS) and acquired immune deficiency syndrome (AIDS)-associated KS (AIDS-KS) in Xinjiang Autonomous Region of China. A total of 35 patients with KS were enrolled in the present study between May 2011 and July 2013, including 26 cases of AIDS-KS and 9 cases of classic KS. Another 10 healthy subjects of the Uygur ethnic group were included in the normal control group. KS tissues were subjected to hematoxylin and eosin staining and immunohistochemical staining. To measure the expression of mRNA, reverse-transcription quantitative polymerase chain reaction was performed. To determine protein expression, western blot analysis was employed. AIDS-KS was mainly distributed in the face and limbs, while classic KS was mainly distributed in the limbs. The histopathological characteristics of AIDS-KS and classic KS tissues were different from those of normal tissues. TLR4 was mainly distributed in the dermis of KS tissues. The mRNA expression levels of TLR4 were reduced in classic KS and AIDS-KS. The protein expression levels of RAS, RAF, nuclear factor (NF)-κB P65 and P50 as well as inhibitor of NF-κB-α of the TLR4 signaling pathway in AIDS-KS and KS tissues were higher than those in normal tissues. In conclusion, the expression of TLR4 gene in KS tissues was decreased, while the expression of proteins of the TLR4 signaling pathway was upregulated in KS. Downregulation of TLR4 may be associated with the occurrence and development of AIDS-KS, while its restoration may represent a novel therapeutic approach for AIDS-KS.
Collapse
Affiliation(s)
- Xiaobo Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Xuefeng Wan
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Xiaoran Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Kejun Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Wubuli Maimaitiaili
- Department of Infectious Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Yuexin Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
18
|
Portale F, Cricrì G, Bresolin S, Lupi M, Gaspari S, Silvestri D, Russo B, Marino N, Ubezio P, Pagni F, Vergani P, Kronnie GT, Valsecchi MG, Locatelli F, Rizzari C, Biondi A, Dander E, D'Amico G. ActivinA: a new leukemia-promoting factor conferring migratory advantage to B-cell precursor-acute lymphoblastic leukemic cells. Haematologica 2018; 104:533-545. [PMID: 30262563 PMCID: PMC6395324 DOI: 10.3324/haematol.2018.188664] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022] Open
Abstract
B-cell precursor-acute lymphoblastic leukemia modulates the bone marrow (BM) niche to become leukemia-supporting and chemo-protective by reprogramming the stromal microenvironment. New therapies targeting the interplay between leukemia and stroma can help improve disease outcome. We identified ActivinA, a TGF-β family member with a well-described role in promoting several solid malignancies, as a factor favoring leukemia that could represent a new potential target for therapy. ActivinA resulted over-expressed in the leukemic BM and its production was strongly induced in mesenchymal stromal cells after culture with leukemic cells. Moreover, MSCs isolated from BM of leukemic patients showed an intrinsic ability to secrete higher amounts of ActivinA compared to their normal counterparts. The pro-inflammatory leukemic BM microenvironment synergized with leukemic cells to induce stromal-derived ActivinA. Gene expression analysis of ActivinA-treated leukemic cells showed that this protein was able to significantly influence motility-associated pathways. Interestingly, ActivinA promoted random motility and CXCL12-driven migration of leukemic cells, even at suboptimal chemokine concentrations, characterizing the leukemic niche. Conversely, ActivinA severely impaired CXCL12-induced migration of healthy CD34+ cells. This opposite effect can be explained by the ability of ActivinA to increase intracellular calcium only in leukemic cells, boosting cytoskeleton dynamics through a higher rate of actin polymerization. Moreover, by stimulating the invasiveness of the leukemic cells, ActivinA was found to be a leukemia-promoting factor. Importantly, the ability of ActivinA to enhance BM engraftment and the metastatic potential of leukemic cells was confirmed in a xenograft mouse model of the disease. Overall, ActivinA was seen to be a key factor in conferring a migratory advantage to leukemic cells over healthy hematopoiesis within the leukemic niche.
Collapse
Affiliation(s)
- Federica Portale
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| | - Giulia Cricrì
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| | - Silvia Bresolin
- Department of Women's and Children's Health, University of Padova
| | - Monica Lupi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano
| | - Stefania Gaspari
- Department of Paediatric Haematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù and Sapienza University of Rome.,Medical Statistics Unit, Department of Clinical Medicine and Prevention, University of Milano-Bicocca, Monza
| | - Daniela Silvestri
- Medical Statistics Unit, Department of Clinical Medicine and Prevention, University of Milano-Bicocca.,School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | - Barbara Russo
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| | - Noemi Marino
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| | - Paolo Ubezio
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano
| | - Fabio Pagni
- School of Medicine and Surgery, University of Milano-Bicocca
| | - Patrizia Vergani
- Department of Obstetrics and Gynecology, University of Milano-Bicocca, Monza, Italy
| | | | - Maria Grazia Valsecchi
- Medical Statistics Unit, Department of Clinical Medicine and Prevention, University of Milano-Bicocca
| | - Franco Locatelli
- Department of Paediatric Haematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù and Sapienza University of Rome
| | - Carmelo Rizzari
- School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | - Andrea Biondi
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza.,School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | - Erica Dander
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| | - Giovanna D'Amico
- Centro Ricerca Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza
| |
Collapse
|
19
|
Differential regulation of the sphere formation and maintenance of cancer-initiating cells of malignant mesothelioma via CD44 and ALK4 signaling pathways. Oncogene 2018; 37:6357-6367. [PMID: 30061637 PMCID: PMC6283855 DOI: 10.1038/s41388-018-0405-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 06/03/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023]
Abstract
Malignant mesothelioma (MM) has a poor prognosis and is largely resistant to standard treatments, so it is important to seek novel therapeutic strategies for this disease. Cancer-initiating cells (CICs) were previously identified in MM using stem cell-associated markers in combination with spheroid cultures. However, the mechanisms underlying the induction and maintenance of CICs in MM remain to be fully explored. Here we showed that the CICs, which had high aldehyde dehydrogenase levels (ALDHbright) and stem cell-associated genes, were expanded in MM cells cultured under sphere-forming conditions. The MM spheroids also initiated tumors in immunodeficient mice more efficiently than did conventional adherent MM cells. In the MM spheroids, the expression of hyaluronan (HA) synthases was upregulated. Inhibiting the HA synthesis or CD44 functions by gene knockdown or neutralizing antibody abolished the formation of large-sized spheroids and the expansion of ALDHbright CICs. The expression of activin-A was also increased in the spheroids, and type I activin-A receptor subunit (ALK4) was upregulated in the ALDHbright CICs. The neutralization of activin-A or functional inactivation of ALK4 diminished the ALDHbright CICs without affecting spheroid formation. The knockdown of CD44 or ALK4 strongly suppressed the tumor growth in immunodeficient mice. These results together suggest that the HA–CD44 and activin-A–ALK4 pathways differentially regulate the spheroid formation and maintenance of ALDHbright CICs in MM cells, and that both pathways play critical roles in tumor growth in immunodeficient hosts. Our findings provide a novel therapeutic option for MM that targets signaling pathways that promote the CIC compartment through CD44 and ALK4.
Collapse
|
20
|
Li C, Rezov V, Joensuu E, Vartiainen V, Rönty M, Yin M, Myllärniemi M, Koli K. Pirfenidone decreases mesothelioma cell proliferation and migration via inhibition of ERK and AKT and regulates mesothelioma tumor microenvironment in vivo. Sci Rep 2018; 8:10070. [PMID: 29968778 PMCID: PMC6030186 DOI: 10.1038/s41598-018-28297-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
Malignant mesothelioma is an aggressive cancer with poor prognosis. It is characterized by prominent extracellular matrix, mesenchymal tumor cell phenotypes and chemoresistance. In this study, the ability of pirfenidone to alter mesothelioma cell proliferation and migration as well as mesothelioma tumor microenvironment was evaluated. Pirfenidone is an anti-fibrotic drug used in the treatment of idiopathic pulmonary fibrosis and has also anti-proliferative activities. Mesothelioma cell proliferation was decreased by pirfenidone alone or in combination with cisplatin. Pirfenidone also decreased significantly Transwell migration/invasion and 3D collagen invasion. This was associated with increased BMP pathway activity, decreased GREM1 expression and downregulation of MAPK/ERK and AKT/mTOR signaling. The canonical Smad-mediated TGF-β signaling was not affected by pirfenidone. However, pirfenidone blocked TGF-β induced upregulation of ERK and AKT pathways. Treatment of mice harboring mesothelioma xenografts with pirfenidone alone did not reduce tumor proliferation in vivo. However, pirfenidone modified the tumor microenvironment by reducing the expression of extracellular matrix associated genes. In addition, GREM1 expression was downregulated by pirfenidone in vivo. By reducing two major upregulated pathways in mesothelioma and by targeting tumor cells and the microenvironment pirfenidone may present a novel anti-fibrotic and anti-cancer adjuvant therapy for mesothelioma.
Collapse
Affiliation(s)
- Chang Li
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Soochow, China
| | - Veronika Rezov
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Emmi Joensuu
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Ville Vartiainen
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Mikko Rönty
- Department of Pathology, University of Helsinki and Fimlab laboratories, Pathology, Tampere, Finland
| | - Miao Yin
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Marjukka Myllärniemi
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Katri Koli
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
21
|
Xiong S, Klausen C, Cheng JC, Leung PCK. Activin B promotes endometrial cancer cell migration by down-regulating E-cadherin via SMAD-independent MEK-ERK1/2-SNAIL signaling. Oncotarget 2018; 7:40060-40072. [PMID: 27223076 PMCID: PMC5129992 DOI: 10.18632/oncotarget.9483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 04/24/2016] [Indexed: 01/03/2023] Open
Abstract
High-risk type II endometrial cancers account for ~30% of cases but ~75% of deaths due, in part, to their tendency to metastasize. Histopathological studies of type II endometrial cancers (non-endometrioid, mostly serous) suggest overproduction of activin B and down-regulation of E-cadherin, both of which are associated with reduced survival. Our previous studies have shown that activin B increases the migration of type II endometrial cancer cell lines. However, little is known about the relationship between activin B signaling and E-cadherin in endometrial cancer. We now demonstrate that activin B treatment significantly decreases E-cadherin expression in both a time- and concentration-dependent manner in KLE and HEC-50 cell lines. Interestingly, these effects were not inhibited by knockdown of SMAD2, SMAD3 or SMAD4. Rather, the suppressive effects of activin B on E-cadherin were mediated by MEK-ERK1/2-induced production of the transcription factor SNAIL. Importantly, activin B-induced cell migration was inhibited by forced-expression of E-cadherin or pre-treatment with the activin/TGF-β type I receptor inhibitor SB431542 or the MEK inhibitor U0126. We have identified a novel SMAD-independent pathway linking enhanced activin B signaling to reduced E-cadherin expression and increased migration in type II endometrial cancer.
Collapse
Affiliation(s)
- Siyuan Xiong
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
22
|
Gremlin-1 is a key regulator of the invasive cell phenotype in mesothelioma. Oncotarget 2017; 8:98280-98297. [PMID: 29228689 PMCID: PMC5716729 DOI: 10.18632/oncotarget.21550] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Malignant mesothelioma originates from mesothelial cells and is a cancer type that aggressively invades into the surrounding tissue, has poor prognosis and no effective treatment. Gremlin-1 is a cysteine knot protein that functions by inhibiting BMP-pathway activity during development. BMP-independent functions have also been described for gremlin-1. We have previously shown high gremlin-1 expression in mesothelioma tumor tissue. Here, we investigated the functions of gremlin-1 in mesothelioma cell migration and invasive growth. Gremlin-1 promoted mesothelioma cell sprouting and invasion into three dimensional collagen and Matrigel matrices. The expression level of gremlin-1 was linked to changes in the expression of SNAI2, integrins, matrix metalloproteinases (MMP) and TGF-β family signaling - all previously associated with a mesenchymal invasive phenotype. Small molecule inhibitors of MMPs completely blocked mesothelioma cell invasive growth. In addition, inhibitors of TGF-β receptors significantly reduced invasive growth. This was associated with reduced expression of MMP2 but not SNAI2, indicating that gremlin-1 has both TGF-β pathway dependent and independent mechanisms of action. Results of in vivo mesothelioma xenograft experiments indicated that gremlin-1 overexpressing tumors were more vascular and had a tendency to send metastases. This suggests that by inducing a mesenchymal invasive cell phenotype together with enhanced tumor vascularization, gremlin-1 drives mesothelioma invasion and metastasis. These data identify gremlin-1 as a potential therapeutic target in mesothelioma.
Collapse
|
23
|
Loomans HA, Arnold SA, Quast LL, Andl CD. Esophageal squamous cell carcinoma invasion is inhibited by Activin A in ACVRIB-positive cells. BMC Cancer 2016; 16:873. [PMID: 27829391 PMCID: PMC5101642 DOI: 10.1186/s12885-016-2920-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/01/2016] [Indexed: 01/05/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a global public health issue, as it is the eighth most common cancer worldwide. The mechanisms behind ESCC invasion and progression are still poorly understood, and warrant further investigation into these processes and their drivers. In recent years, the ligand Activin A has been implicated as a player in the progression of a number of cancers. The objective of this study was to investigate the role of Activin A signaling in ESCC. Methods To investigate the role Activin A plays in ESCC biology, tissue microarrays containing 200 cores from 120 ESCC patients were analyzed upon immunofluorescence staining. We utilized three-dimensional organotypic reconstruct cultures of dysplastic and esophageal squamous tumor cells lines, in the context of fibroblast-secreted Activin A, to identify the effects of Activin A on cell invasion and determine protein expression and localization in epithelial and stromal compartments by immunofluorescence. To identify the functional consequences of stromal-derived Activin A on angiogenesis, we performed endothelial tube formation assays. Results Analysis of ESCC patient samples indicated that patients with high stromal Activin A expression had low epithelial ACVRIB, the Activin type I receptor. We found that overexpression of stromal-derived Activin A inhibited invasion of esophageal dysplastic squamous cells, ECdnT, and TE-2 ESCC cells, both positive for ACVRIB. This inhibition was accompanied by a decrease in expression of the extracellular matrix (ECM) protein fibronectin and podoplanin, which is often expressed at the leading edge during invasion. Endothelial tube formation was disrupted in the presence of conditioned media from fibroblasts overexpressing Activin A. Interestingly, ACVRIB-negative TE-11 cells did not show the prior observed effects in the context of Activin A overexpression, indicating a dependence on the presence of ACVRIB. Conclusions We describe the first observation of an inhibitory role for Activin A in ESCC progression that is dependent on the expression of ACVRIB. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2920-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Holli A Loomans
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Shanna A Arnold
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura L Quast
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Claudia D Andl
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4110 Libra Drive, Building 20, BMS 223, Orlando, FL, 32816, USA.
| |
Collapse
|
24
|
Circulating activin A is a novel prognostic biomarker in malignant pleural mesothelioma – A multi-institutional study. Eur J Cancer 2016; 63:64-73. [DOI: 10.1016/j.ejca.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/19/2016] [Indexed: 12/28/2022]
|
25
|
Wijayarathna R, de Kretser DM. Activins in reproductive biology and beyond. Hum Reprod Update 2016; 22:342-57. [PMID: 26884470 DOI: 10.1093/humupd/dmv058] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/20/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Activins are members of the pleiotrophic family of the transforming growth factor-beta (TGF-β) superfamily of cytokines, initially isolated for their capacity to induce the release of FSH from pituitary extracts. Subsequent research has demonstrated that activins are involved in multiple biological functions including the control of inflammation, fibrosis, developmental biology and tumourigenesis. This review summarizes the current knowledge on the roles of activin in reproductive and developmental biology. It also discusses interesting advances in the field of modulating the bioactivity of activins as a therapeutic target, which would undoubtedly be beneficial for patients with reproductive pathology. METHODS A comprehensive literature search was carried out using PUBMED and Google Scholar databases to identify studies in the English language which have contributed to the advancement of the field of activin biology, since its initial isolation in 1987 until July 2015. 'Activin', 'testis', 'ovary', 'embryonic development' and 'therapeutic targets' were used as the keywords in combination with other search phrases relevant to the topic of activin biology. RESULTS Activins, which are dimers of inhibin β subunits, act via a classical TGF-β signalling pathway. The bioactivity of activin is regulated by two endogenous inhibitors, inhibin and follistatin. Activin is a major regulator of testicular and ovarian development. In the ovary, activin A promotes oocyte maturation and regulates granulosa cell steroidogenesis. It is also essential in endometrial repair following menstruation, decidualization and maintaining pregnancy. Dysregulation of the activin-follistatin-inhibin system leads to disorders of female reproduction and pregnancy, including polycystic ovary syndrome, ectopic pregnancy, miscarriage, fetal growth restriction, gestational diabetes, pre-eclampsia and pre-term birth. Moreover, a rise in serum activin A, accompanied by elevated FSH, is characteristic of female reproductive aging. In the male, activin A is an autocrine and paracrine modulator of germ cell development and Sertoli cell proliferation. Disruption of normal activin signalling is characteristic of many tumours affecting reproductive organs, including endometrial carcinoma, cervical cancer, testicular and ovarian cancer as well as prostate cancer. While activin A and B aid the progression of many tumours of the reproductive organs, activin C acts as a tumour suppressor. Activins are important in embryonic induction, morphogenesis of branched glandular organs, development of limbs and nervous system, craniofacial and dental development and morphogenesis of the Wolffian duct. CONCLUSIONS The field of activin biology has advanced considerably since its initial discovery as an FSH stimulating agent. Now, activin is well known as a growth factor and cytokine that regulates many aspects of reproductive biology, developmental biology and also inflammation and immunological mechanisms. Current research provides evidence for novel roles of activins in maintaining the structure and function of reproductive and other organ systems. The fact that activin A is elevated both locally as well as systemically in major disorders of the reproductive system makes it an important biomarker. Given the established role of activin A as a pro-inflammatory and pro-fibrotic agent, studies of its involvement in disorders of reproduction resulting from these processes should be examined. Follistatin, as a key regulator of the biological actions of activin, should be evaluated as a therapeutic agent in conditions where activin A overexpression is established as a contributing factor.
Collapse
Affiliation(s)
- R Wijayarathna
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31, Wright Street, Clayton, VIC 3168, Australia
| | - D M de Kretser
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31, Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|