1
|
Chaudhary R, Weiskirchen R, Ehrlich M, Henis YI. Dual signaling pathways of TGF-β superfamily cytokines in hepatocytes: balancing liver homeostasis and disease progression. Front Pharmacol 2025; 16:1580500. [PMID: 40260391 PMCID: PMC12009898 DOI: 10.3389/fphar.2025.1580500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
The transforming growth factor-β (TGF-β) superfamily (TGF-β-SF) comprises over 30 cytokines, including TGF-β, activins/inhibins, bone morphogenetic proteins (BMPs), and growth differentiation factors (GDFs). These cytokines play critical roles in liver function and disease progression. Here, we discuss Smad-dependent (canonical) and non-Smad pathways activated by these cytokines in a hepatocellular context. We highlight the connection between the deregulation of these pathways or the balance between them and key hepatocellular processes (e.g., proliferation, apoptosis, and epithelial-mesenchymal transition (EMT)). We further discuss their contribution to various chronic liver conditions, such as metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), and hepatocellular carcinoma (HCC). In MASLD and MASH, TGF-β signaling contributes to hepatocyte lipid accumulation, cell death and fibrosis progression through both Smad and non-Smad pathways. In HCC, TGF-β and other TGF-β-SF cytokines have a dual role, acting as tumor suppressors or promoters in early vs. advanced stages of tumor progression, respectively. Additionally, we review the involvement of non-Smad pathways in modulating hepatocyte responses to TGF-β-SF cytokines, particularly in the context of chronic liver diseases, as well as the interdependence with other key pathways (cholesterol metabolism, insulin resistance, oxidative stress and lipotoxicity) in MASLD/MASH pathogenesis. The perspectives and insights detailed in this review may assist in determining future research directions and therapeutic targets in liver conditions, including chronic liver diseases and cancer.
Collapse
Affiliation(s)
- Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Kentistou KA, Lim BEM, Kaisinger LR, Steinthorsdottir V, Sharp LN, Patel KA, Tragante V, Hawkes G, Gardner EJ, Olafsdottir T, Wood AR, Zhao Y, Thorleifsson G, Day FR, Ozanne SE, Hattersley AT, O'Rahilly S, Stefansson K, Ong KK, Beaumont RN, Perry JRB, Freathy RM. Rare variant associations with birth weight identify genes involved in adipose tissue regulation, placental function and insulin-like growth factor signalling. Nat Commun 2025; 16:648. [PMID: 39809772 PMCID: PMC11733218 DOI: 10.1038/s41467-024-55761-2] [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: 04/13/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Investigating the genetic factors influencing human birth weight may lead to biological insights into fetal growth and long-term health. We report analyses of rare variants that impact birth weight when carried by either fetus or mother, using whole exome sequencing data in up to 234,675 participants. Rare protein-truncating and deleterious missense variants are collapsed to perform gene burden tests. We identify 9 genes; 5 with fetal-only effects on birth weight, 1 with maternal-only effects, 3 with both, and observe directionally concordant associations in an independent sample. Four of the genes were previously implicated by GWAS of birth weight. IGF1R and PAPPA2 (fetal and maternal-acting) have known roles in insulin-like growth factor bioavailability and signalling. PPARG, INHBE and ACVR1C (fetal-acting) are involved in adipose tissue regulation, and the latter two also show associations with favourable adiposity patterns in adults. We highlight the dual role of PPARG (fetal-acting) in adipocyte differentiation and placental angiogenesis. NOS3 (fetal and maternal-acting), NRK (fetal), and ADAMTS8 (maternal-acting) have been implicated in placental function and hypertension. To conclude, our analysis of rare coding variants identifies regulators of fetal adipose tissue and fetoplacental angiogenesis as determinants of birth weight, and further evidence for the role of insulin-like growth factors.
Collapse
Affiliation(s)
- Katherine A Kentistou
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Brandon E M Lim
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Andrew R Wood
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Yajie Zhao
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., 102 Reykjavik, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ken K Ong
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John R B Perry
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
3
|
Loh NY, Rosoff DB, Richmond R, Noordam R, Smith GD, Ray D, Karpe F, Lohoff FW, Christodoulides C. Bidirectional Mendelian Randomization Highlights Causal Relationships Between Circulating INHBC and Multiple Cardiometabolic Diseases and Traits. Diabetes 2024; 73:2084-2094. [PMID: 39283655 PMCID: PMC11579406 DOI: 10.2337/db24-0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/05/2024] [Indexed: 11/22/2024]
Abstract
Human genetic and transgenic mouse studies have highlighted a potential liver-adipose tissue endocrine axis, involving activin C (Act-C) and/or Act-E and ALK7, influencing fat distribution and systemic metabolism. We investigated the bidirectional effects between circulating INHBC, which homodimerizes into Act-C, and adiposity traits, insulin resistance, inflammation, and cardiometabolic disease risk. Additionally, we examined whether Act-C is an ALK7 ligand in human adipocytes. We used Mendelian randomization and in vitro studies in immortalized human abdominal and gluteal adipocytes. Circulating INHBC was causally linked to reduced lower-body fat, dyslipidemia, and increased risks of coronary artery disease (CAD) and nonalcoholic fatty liver disease (NAFLD). Conversely, upper-body fat distribution, obesity, hypertriglyceridemia, subclinical inflammation, and type 2 diabetes positively impacted plasma INHBC levels. Mechanistically, an atherogenic lipid profile may partly explain the INHBC-CAD link, while inflammation and hypertriglyceridemia may partly explain how adiposity traits affect circulating INHBC. Phenome-wide Mendelian randomization showed weak causal relationships between higher plasma INHBC and impaired kidney function and higher gout risk. In human adipocytes, recombinant Act-C activated SMAD2/3 signaling via ALK7 and suppressed lipolysis. In summary, INHBC influences systemic metabolism by activating ALK7 in adipose tissue and may serve as a drug target for atherogenic dyslipidemia, CAD, and NAFLD. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Nellie Y. Loh
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Daniel B. Rosoff
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - Rebecca Richmond
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - David Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
| | - Falk W. Lohoff
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
| | - Constantinos Christodoulides
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
| |
Collapse
|
4
|
Tang W, Gu Z, Guo J, Lin M, Tao H, Jia D, Jia P. Activins and Inhibins in Cardiovascular Pathophysiology. Biomolecules 2024; 14:1462. [PMID: 39595638 PMCID: PMC11592067 DOI: 10.3390/biom14111462] [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: 09/25/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Activins and inhibins, members of the transforming growth factor β (TGFβ) superfamily, were initially recognized for their opposing effects on the secretion of follicle-stimulating hormone. Subsequent research has demonstrated their broader biological roles across various tissue types. Primarily, activins and inhibins function through the classical TGFβ SMAD signaling pathway, but studies suggest that they also act through other pathways, with their specific signaling being complex and context-dependent. Recent research has identified significant roles for activins and inhibins in the cardiovascular system. Their actions in other systems and their signaling pathways show strong correlations with the development and progression of cardiovascular diseases, indicating potential broader roles in the cardiovascular system. This review summarizes the progress in research on the biological functions and mechanisms of activins and inhibins and their signaling pathways in cardiovascular diseases, offering new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| |
Collapse
|
5
|
Wang XL, Yang M, Wang Y. Roles of transforming growth factor-β signaling in liver disease. World J Hepatol 2024; 16:973-979. [PMID: 39086528 PMCID: PMC11287609 DOI: 10.4254/wjh.v16.i7.973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/04/2024] [Accepted: 05/24/2024] [Indexed: 07/26/2024] Open
Abstract
In this editorial we expand the discussion on the article by Zhang et al published in the recent issue of the World Journal of Hepatology. We focus on the diagnostic and therapeutic targets identified on the basis of the current understanding of the molecular mechanisms of liver disease. Transforming growth factor-β (TGF-β) belongs to a structurally related cytokine super family. The family members display different time- and tissue-specific expression patterns associated with autoimmunity, inflammation, fibrosis, and tumorigenesis; and, they participate in the pathogenesis of many diseases. TGF-β and its related signaling pathways have been shown to participate in the progression of liver diseases, such as injury, inflammation, fibrosis, cirrhosis, and cancer. The often studied TGF-β/Smad signaling pathway has been shown to promote or inhibit liver fibrosis under different circumstances. Similarly, the early immature TGF-β molecule functions as a tumor suppressor, inducing apoptosis; but, its interaction with the mitogenic molecule epidermal growth factor alters this effect, activating anti-apoptotic signals that promote liver cancer development. Overall, TGF-β signaling displays contradictory effects in different liver disease stages. Therefore, the use of TGF-β and related signaling pathway molecules for diagnosis and treatment of liver diseases remains a challenge and needs further study. In this editorial, we aim to review the evidence for the use of TGF-β signaling pathway molecules as diagnostic or therapeutic targets for different liver disease stages.
Collapse
Affiliation(s)
- Xiao-Ling Wang
- Clinical Laboratory, Shanxi Academy of Traditional Chinese Medicine, Taiyuan 030012, Shanxi Province, China.
| | - Meng Yang
- Clinical Laboratory, Shanxi Academy of Traditional Chinese Medicine, Taiyuan 030012, Shanxi Province, China
| | - Ying Wang
- Clinical Laboratory, Shanxi Academy of Traditional Chinese Medicine, Taiyuan 030012, Shanxi Province, China
| |
Collapse
|
6
|
Kentistou KA, Lim BEM, Kaisinger LR, Steinthorsdottir V, Sharp LN, Patel KA, Tragante V, Hawkes G, Gardner EJ, Olafsdottir T, Wood AR, Zhao Y, Thorleifsson G, Day FR, Ozanne SE, Hattersley AT, O'Rahilly S, Stefansson K, Ong KK, Beaumont RN, Perry JRB, Freathy RM. Rare variant associations with birth weight identify genes involved in adipose tissue regulation, placental function and insulin-like growth factor signalling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.03.24305248. [PMID: 38633783 PMCID: PMC11023655 DOI: 10.1101/2024.04.03.24305248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Investigating the genetic factors influencing human birth weight may lead to biological insights into fetal growth and long-term health. Genome-wide association studies of birth weight have highlighted associated variants in more than 200 regions of the genome, but the causal genes are mostly unknown. Rare genetic variants with robust evidence of association are more likely to point to causal genes, but to date, only a few rare variants are known to influence birth weight. We aimed to identify genes that harbour rare variants that impact birth weight when carried by either the fetus or the mother, by analysing whole exome sequence data in UK Biobank participants. We annotated rare (minor allele frequency <0.1%) protein-truncating or high impact missense variants on whole exome sequence data in up to 234,675 participants with data on their own birth weight (fetal variants), and up to 181,883 mothers who reported the birth weight of their first child (maternal variants). Variants within each gene were collapsed to perform gene burden tests and for each associated gene, we compared the observed fetal and maternal effects. We identified 8 genes with evidence of rare fetal variant effects on birth weight, of which 2 also showed maternal effects. One additional gene showed evidence of maternal effects only. We observed 10/11 directionally concordant associations in an independent sample of up to 45,622 individuals (sign test P=0.01). Of the genes identified, IGF1R and PAPPA2 (fetal and maternal-acting) have known roles in insulin-like growth factor bioavailability and signalling. PPARG, INHBE and ACVR1C (all fetal-acting) have known roles in adipose tissue regulation and rare variants in the latter two also showed associations with favourable adiposity patterns in adults. We highlight the dual role of PPARG in both adipocyte differentiation and placental angiogenesis. NOS3, NRK, and ADAMTS8 (fetal and maternal-acting) have been implicated in both placental function and hypertension. Analysis of rare coding variants has identified regulators of fetal adipose tissue and fetoplacental angiogenesis as determinants of birth weight, as well as further evidence for the role of insulin-like growth factors.
Collapse
Affiliation(s)
- Katherine A Kentistou
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Brandon E M Lim
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Andrew R Wood
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Yajie Zhao
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., 102 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Ken K Ong
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John R B Perry
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
7
|
Vestal KA, Kattamuri C, Koyiloth M, Ongaro L, Howard JA, Deaton A, Ticau S, Dubey A, Bernard DJ, Thompson TB. Activin E is a TGFβ ligand that signals specifically through activin receptor-like kinase 7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559288. [PMID: 37808681 PMCID: PMC10557571 DOI: 10.1101/2023.09.25.559288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Activins are one of the three distinct subclasses within the greater Transforming Growth Factor β (TGFβ) superfamily. First discovered for their critical roles in reproductive biology, activins have since been shown to alter cellular differentiation and proliferation. At present, members of the activin subclass include activin A (ActA), ActB, ActC, ActE, and the more distant members myostatin and GDF11. While the biological roles and signaling mechanisms of most activins class members have been well-studied, the signaling potential of ActE has remained largely unknown. Here, we characterized the signaling capacity of homodimeric ActE. Molecular modeling of the ligand:receptor complexes showed that ActC and ActE shared high similarity in both the type I and type II receptor binding epitopes. ActE signaled specifically through ALK7, utilized the canonical activin type II receptors, ActRIIA and ActRIIB, and was resistant to the extracellular antagonists follistatin and WFIKKN. In mature murine adipocytes, ActE invoked a SMAD2/3 response via ALK7, similar to ActC. Collectively, our results establish ActE as an ALK7 ligand, thereby providing a link between genetic and in vivo studies of ActE as a regulator of adipose tissue.
Collapse
Affiliation(s)
- Kylie A Vestal
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Muhasin Koyiloth
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Quebec, Canada
| | - James A Howard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | | | | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Quebec, Canada
| | - Thomas B Thompson
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
8
|
Adam RC, Pryce DS, Lee JS, Zhao Y, Mintah IJ, Min S, Halasz G, Mastaitis J, Atwal GS, Aykul S, Idone V, Economides AN, Lotta LA, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Activin E-ACVR1C cross talk controls energy storage via suppression of adipose lipolysis in mice. Proc Natl Acad Sci U S A 2023; 120:e2309967120. [PMID: 37523551 PMCID: PMC10410708 DOI: 10.1073/pnas.2309967120] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/13/2023] [Indexed: 08/02/2023] Open
Abstract
Body fat distribution is a heritable risk factor for cardiovascular and metabolic disease. In humans, rare Inhibin beta E (INHBE, activin E) loss-of-function variants are associated with a lower waist-to-hip ratio and protection from type 2 diabetes. Hepatic fatty acid sensing promotes INHBE expression during fasting and in obese individuals, yet it is unclear how the hepatokine activin E governs body shape and energy metabolism. Here, we uncover activin E as a regulator of adipose energy storage. By suppressing β-agonist-induced lipolysis, activin E promotes fat accumulation and adipocyte hypertrophy and contributes to adipose dysfunction in mice. Mechanistically, we demonstrate that activin E elicits its effect on adipose tissue through ACVR1C, activating SMAD2/3 signaling and suppressing PPARG target genes. Conversely, loss of activin E or ACVR1C in mice increases fat utilization, lowers adiposity, and drives PPARG-regulated gene signatures indicative of healthy adipose function. Our studies identify activin E-ACVR1C as a metabolic rheostat promoting liver-adipose cross talk to restrain excessive fat breakdown and preserve fat mass during prolonged fasting, a mechanism that is maladaptive in obese individuals.
Collapse
Affiliation(s)
| | | | | | - Yuanqi Zhao
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | - Soo Min
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | | | | - Senem Aykul
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Hamang M, Yaden B, Dai G. Gastrointestinal pharmacology activins in liver health and disease. Biochem Pharmacol 2023; 214:115668. [PMID: 37364623 PMCID: PMC11234865 DOI: 10.1016/j.bcp.2023.115668] [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: 04/03/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Activins are a subgroup of the TGFβ superfamily of growth and differentiation factors, dimeric in nature and consisting of two inhibin beta subunits linked via a disulfide bridge. Canonical activin signaling occurs through Smad2/3, with negative feedback initiated by Smad6/7 following signal transduction, which binds activin type I receptor preventing phosphorylation of Smad2/3 and activation of downstream signaling. In addition to Smad6/7, other inhibitors of activin signaling have been identified as well, including inhibins (dimers of an inhibin alpha and beta subunit), BAMBI, Cripto, follistatin, and follistatin-like 3 (fstl3). To date, activins A, B, AB, C, and E have been identified and isolated in mammals, with activin A and B having the most characterization of biological activity. Activin A has been implicated as a regulator of several important functions of liver biology, including hepatocyte proliferation and apoptosis, ECM production, and liver regeneration; the role of other subunits of activin in liver physiology are less understood. There is mounting data to suggest a link between dysregulation of activins contributing to various hepatic diseases such as inflammation, fibrosis, and hepatocellular carcinoma, and emerging studies demonstrating the protective and regenerative effects of inhibiting activins in mouse models of liver disease. Due to their importance in liver biology, activins demonstrate utility as a therapeutic target for the treatment of hepatic diseases such as cirrhosis, NASH, NAFLD, and HCC; further research regarding activins may provide diagnostic or therapeutic opportunity for those suffering from various liver diseases.
Collapse
Affiliation(s)
- Matthew Hamang
- Department of Biology, School of Science, Indiana University - Purdue University Indianapolis, IN, United States.
| | - Benjamin Yaden
- Department of Biology, School of Science, Indiana University - Purdue University Indianapolis, IN, United States.
| | - Guoli Dai
- Department of Biology, School of Science, Indiana University - Purdue University Indianapolis, IN, United States.
| |
Collapse
|
10
|
Chan MKK, Chan ELY, Ji ZZ, Chan ASW, Li C, Leung KT, To KF, Tang PMK. Transforming growth factor-β signaling: from tumor microenvironment to anticancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:316-343. [PMID: 37205317 PMCID: PMC10185444 DOI: 10.37349/etat.2023.00137] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/09/2023] [Indexed: 05/21/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is an important pathway for promoting the pathogenesis of inflammatory diseases, including cancer. The roles of TGF-β signaling are heterogeneous and versatile in cancer development and progression, both anticancer and protumoral actions are reported. Interestingly, increasing evidence suggests that TGF-β enhances disease progression and drug resistance via immune-modulatory actions in the tumor microenvironment (TME) of solid tumors. A better understanding of its regulatory mechanisms in the TME at the molecular level can facilitate the development of precision medicine to block the protumoral actions of TGF-β in the TME. Here, the latest information about the regulatory mechanisms and translational research of TGF-β signaling in the TME for therapeutic development had been summarized.
Collapse
Affiliation(s)
- Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Emily Lok-Yiu Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Zoey Zeyuan Ji
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Chunjie Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Correspondence: Patrick Ming-Kuen Tang, Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
11
|
Wang Y, Hamang M, Culver A, Jiang H, Yanum J, Garcia V, Lee J, White E, Kusumanchi P, Chalasani N, Liangpunsakul S, Yaden BC, Dai G. Activin B promotes the initiation and progression of liver fibrosis. Hepatol Commun 2022; 6:2812-2826. [PMID: 35866567 PMCID: PMC9512478 DOI: 10.1002/hep4.2037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
The role of activin B, a transforming growth factor β (TGFβ) superfamily cytokine, in liver health and disease is largely unknown. We aimed to investigate whether activin B modulates liver fibrogenesis. Liver and serum activin B, along with its analog activin A, were analyzed in patients with liver fibrosis from different etiologies and in mouse acute and chronic liver injury models. Activin B, activin A, or both was immunologically neutralized in mice with progressive or established carbon tetrachloride (CCl4 )-induced liver fibrosis. Hepatic and circulating activin B was increased in human patients with liver fibrosis caused by several liver diseases. In mice, hepatic and circulating activin B exhibited persistent elevation following the onset of several types of liver injury, whereas activin A displayed transient increases. The results revealed a close correlation of activin B with liver injury regardless of etiology and species. Injured hepatocytes produced excessive activin B. Neutralizing activin B largely prevented, as well as improved, CCl4 -induced liver fibrosis, which was augmented by co-neutralizing activin A. Mechanistically, activin B mediated the activation of c-Jun-N-terminal kinase (JNK), the induction of inducible nitric oxide synthase (iNOS) expression, and the maintenance of poly (ADP-ribose) polymerase 1 (PARP1) expression in injured livers. Moreover, activin B directly induced a profibrotic expression profile in hepatic stellate cells (HSCs) and stimulated these cells to form a septa structure. Conclusions: We demonstrate that activin B, cooperating with activin A, mediates the activation or expression of JNK, iNOS, and PARP1 and the activation of HSCs, driving the initiation and progression of liver fibrosis.
Collapse
Affiliation(s)
- Yan Wang
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Matthew Hamang
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Alexander Culver
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Huaizhou Jiang
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Jennifer Yanum
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Veronica Garcia
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Joonyong Lee
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Emily White
- College of ScienceDepartment of Biological SciencesPurdue UniversityWest LafayetteIndianaUSA
| | - Praveen Kusumanchi
- Division of Gastroenterology and HepatologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Naga Chalasani
- Division of Gastroenterology and HepatologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and HepatologyDepartment of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Roudebush Veterans Administration Medical CenterIndianapolisIndianaUSA
| | - Benjamin C. Yaden
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Guoli Dai
- Department of BiologySchool of ScienceCenter for Developmental and Regenerative BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| |
Collapse
|
12
|
Goebel EJ, Ongaro L, Kappes EC, Vestal K, Belcheva E, Castonguay R, Kumar R, Bernard DJ, Thompson TB. The orphan ligand, activin C, signals through activin receptor-like kinase 7. eLife 2022; 11:78197. [PMID: 35736809 PMCID: PMC9224996 DOI: 10.7554/elife.78197] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/09/2022] [Indexed: 12/11/2022] Open
Abstract
Activin ligands are formed from two disulfide-linked inhibin β (Inhβ) subunit chains. They exist as homodimeric proteins, as in the case of activin A (ActA; InhβA/InhβA) or activin C (ActC; InhβC/InhβC), or as heterodimers, as with activin AC (ActAC; InhβA:InhβC). While the biological functions of ActA and activin B (ActB) have been well characterized, little is known about the biological functions of ActC or ActAC. One thought is that the InhβC chain functions to interfere with ActA production by forming less active ActAC heterodimers. Here, we assessed and characterized the signaling capacity of ligands containing the InhβC chain. ActC and ActAC activated SMAD2/3-dependent signaling via the type I receptor, activin receptor-like kinase 7 (ALK7). Relative to ActA and ActB, ActC exhibited lower affinity for the cognate activin type II receptors and was resistant to neutralization by the extracellular antagonist, follistatin. In mature murine adipocytes, which exhibit high ALK7 expression, ActC elicited a SMAD2/3 response similar to ActB, which can also signal via ALK7. Collectively, these results establish that ActC and ActAC are active ligands that exhibit a distinct signaling receptor and antagonist profile compared to other activins.
Collapse
Affiliation(s)
- Erich J Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Canada
| | - Emily C Kappes
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | - Kylie Vestal
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | | | | | | | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Canada
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
13
|
Knowles H, Santucci N, Studdert J, Goh HN, Kaufman-Francis K, Salehin N, Tam PPL, Osteil P. Differential impact of TGFβ/SMAD signaling activity elicited by Activin A and Nodal on endoderm differentiation of epiblast stem cells. Genesis 2022; 60:e23466. [PMID: 35104045 DOI: 10.1002/dvg.23466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/11/2022]
Abstract
Allocation of cells to an endodermal fate in the gastrulating embryo is driven by Nodal signaling and consequent activation of TGFβ pathway. In vitro methodologies striving to recapitulate the process of endoderm differentiation, however, use TGFβ family member Activin in place of Nodal. This is despite Activin not known to have an in vivo role in endoderm differentiation. In this study, five epiblast stem cell lines were subjected to directed differentiation using both Activin A and Nodal to induce endodermal fate. A reporter line harboring endoderm markers FoxA2 and Sox17 was further analyzed for TGFβ pathway activation and WNT response. We demonstrated that Activin A-treated cells remain more primitive streak-like when compared to Nodal-treated cells that have a molecular profile suggestive of more advanced differentiation. Activin A elicited a robust TGFβ/SMAD activity, enhanced WNT signaling activity and promoted the generation of DE precursors. Nodal treatment resulted in lower TGFβ/SMAD activity, and a weaker, sustained WNT response, and ultimately failed to upregulate endoderm markers. This is despite signaling response resembling more closely the activity seen in vivo. These findings emphasize the importance of understanding the downstream activities of Activin A and Nodal signaling in directing in vitro endoderm differentiation of primed-state epiblast stem cells.
Collapse
Affiliation(s)
- Hilary Knowles
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Nicole Santucci
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Joshua Studdert
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Hwee Ngee Goh
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Keren Kaufman-Francis
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Nazmus Salehin
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Patrick P L Tam
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Pierre Osteil
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia.,Swiss Cancer Research Institute (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
14
|
Xu ZB, Gan MF, Yu HY, Mo LC, Xia YH, Yu QX, Zheng JM. The Significance of INHBE Expression in the Cancer Cells of Clear-Cell Renal Cell Carcinoma. Urol Int 2021; 106:376-386. [PMID: 34515260 DOI: 10.1159/000518161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/22/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Activins and inhibins are structurally related dimeric glycoprotein hormones belonging to the transforming growth factor-β superfamily but whether they are also involved in malignancy is far from clear. No study has reported the expression of INHBE in kidney cancer. The purpose of this study was to examine the expressions of INHBE in the tumor tissue of patients with clear-cell renal cell carcinoma (ccRCC) and to explore the pathologic significance. METHODS The INHBE mRNA expression in the tumor tissue of ccRCC patients was analyzed by using RNA sequencing data from the TCGA database. To examine the expression of inhibin βE protein, 241 ccRCC patients were recruited and immunohistochemistry was performed on the tumor tissue of these patients along with 39 normal renal samples. The association between the inhibin βE expression level and patient's clinicopathological indices was evaluated. RESULTS In the normal renal tissue, inhibin βE was found to be expressed mainly by renal tubular epithelial cells. In the tumor tissue, inhibin βE was expressed mainly in cancer cells. The expressions of INHBE mRNA and protein in the tumor tissue of ccRCC patients increased significantly compared with those in normal renal samples. There was a significant correlation between the level of inhibin βE in the tumor tissue and tumor grade. Patients with a lower inhibin βE expression in the tumor tissue were found to have a longer overall survival and disease-specific survival. CONCLUSIONS INHBE might be involved in the pathogenesis of ccRCC and function as a tumor promoter.
Collapse
Affiliation(s)
- Zi-Bin Xu
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Mei-Fu Gan
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Hong-Yuan Yu
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Li-Cai Mo
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Yu-Hui Xia
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Qing-Xin Yu
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Jing-Min Zheng
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, China.,Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| |
Collapse
|
15
|
Watt MJ, Miotto PM, De Nardo W, Montgomery MK. The Liver as an Endocrine Organ-Linking NAFLD and Insulin Resistance. Endocr Rev 2019; 40:1367-1393. [PMID: 31098621 DOI: 10.1210/er.2019-00034] [Citation(s) in RCA: 389] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
The liver is a dynamic organ that plays critical roles in many physiological processes, including the regulation of systemic glucose and lipid metabolism. Dysfunctional hepatic lipid metabolism is a cause of nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disorder worldwide, and is closely associated with insulin resistance and type 2 diabetes. Through the use of advanced mass spectrometry "omics" approaches and detailed experimentation in cells, mice, and humans, we now understand that the liver secretes a wide array of proteins, metabolites, and noncoding RNAs (miRNAs) and that many of these secreted factors exert powerful effects on metabolic processes both in the liver and in peripheral tissues. In this review, we summarize the rapidly evolving field of "hepatokine" biology with a particular focus on delineating previously unappreciated communication between the liver and other tissues in the body. We describe the NAFLD-induced changes in secretion of liver proteins, lipids, other metabolites, and miRNAs, and how these molecules alter metabolism in liver, muscle, adipose tissue, and pancreas to induce insulin resistance. We also synthesize the limited information that indicates that extracellular vesicles, and in particular exosomes, may be an important mechanism for intertissue communication in normal physiology and in promoting metabolic dysregulation in NAFLD.
Collapse
Affiliation(s)
- Matthew J Watt
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Paula M Miotto
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - William De Nardo
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
16
|
Reader KL, Marino FE, Nicholson HD, Risbridger GP, Gold EJ. Role of activin C in normal ovaries and granulosa cell tumours of mice and humans. Reprod Fertil Dev 2019; 30:958-968. [PMID: 29207252 DOI: 10.1071/rd17250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/08/2017] [Indexed: 12/30/2022] Open
Abstract
Activins and inhibins play important roles in the development, growth and function of the ovary. Mice lacking inhibin develop granulosa cell tumours in their ovaries that secrete activin A, and these tumours are modulated by increased activin C expression. The aim of the present study was to identify where activin C is expressed in mouse and human ovaries and whether overexpression of activin C modulates normal follicular development in mice. Immunohistochemical staining for the activin βC subunit was performed on sections from mouse and human ovaries and human adult granulosa cell tumours. Stereology techniques were used to quantify oocyte and follicular diameters, and the percentage of different follicular types in ovaries from wild-type mice and those underexpressing inhibin α and/or overexpressing activin C. Staining for activin βC was observed in the oocytes, granulosa cells, thecal cells and surface epithelium of mouse and human ovaries, and in the granulosa-like cells of adult granulosa cell tumours. Overexpression of activin C in mice did not alter follicular development compared with wild-type mice, but it did modulate the development of abnormal early stage follicles in inhibin α-null mice. These results provide further evidence of a role for activin C in the ovary.
Collapse
Affiliation(s)
- Karen L Reader
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| | | | - Helen D Nicholson
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| | - Gail P Risbridger
- Consortium and Cancer Program Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Vic. 3800, Australia
| | - Elspeth J Gold
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
17
|
Hashimoto O, Funaba M, Sekiyama K, Doi S, Shindo D, Satoh R, Itoi H, Oiwa H, Morita M, Suzuki C, Sugiyama M, Yamakawa N, Takada H, Matsumura S, Inoue K, Oyadomari S, Sugino H, Kurisaki A. Activin E Controls Energy Homeostasis in Both Brown and White Adipose Tissues as a Hepatokine. Cell Rep 2018; 25:1193-1203. [DOI: 10.1016/j.celrep.2018.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/12/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023] Open
|
18
|
Sugiyama M, Kikuchi A, Misu H, Igawa H, Ashihara M, Kushima Y, Honda K, Suzuki Y, Kawabe Y, Kaneko S, Takamura T. Inhibin βE (INHBE) is a possible insulin resistance-associated hepatokine identified by comprehensive gene expression analysis in human liver biopsy samples. PLoS One 2018; 13:e0194798. [PMID: 29596463 PMCID: PMC5875797 DOI: 10.1371/journal.pone.0194798] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 02/11/2018] [Indexed: 12/15/2022] Open
Abstract
The liver plays a major role in whole-body energy homeostasis by releasing secretory factors, termed hepatokines. To identify novel target genes associated with insulin resistance, we performed a comprehensive analysis of gene expression profiles using a DNA chip method in liver biopsy samples from humans with varying degrees of insulin resistance. Inhibin βE (INHBE) was identified as a novel putative hepatokine with hepatic gene expression that positively correlated with insulin resistance and body mass index in humans. Quantitative real time-PCR analysis also showed an increase in INHBE gene expression in independent liver samples from insulin-resistant human subjects. Additionally, Inhbe gene expression increased in the livers of db/db mice, a rodent model of type 2 diabetes. To preliminarily screen the role of Inhbe in vivo in whole-body energy metabolic status, hepatic mRNA was knocked down with siRNA for Inhbe (siINHBE) in db/db mice. Treatment with siINHBE suppressed body weight gain during the two-week experimental period, which was attributable to diminished fat rather than lean mass. Additionally, treatment with siINHBE decreased the respiratory quotient and increased plasma total ketone bodies compared with treatment with non-targeting siRNA, both of which suggest enhanced whole-body fat utilization. Our study suggests that INHBE functions as a possible hepatokine to alter the whole-body metabolic status under obese insulin-resistant conditions.
Collapse
Affiliation(s)
- Masakazu Sugiyama
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Akihiro Kikuchi
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- * E-mail: (TT); (AK)
| | - Hirofumi Misu
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Hirobumi Igawa
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Motooki Ashihara
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Youichi Kushima
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Kiyofumi Honda
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Yoshiyuki Suzuki
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Yoshiki Kawabe
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Shuichi Kaneko
- Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
- * E-mail: (TT); (AK)
| |
Collapse
|
19
|
Suzuki A, Yoshida H, van Heeringen SJ, Takebayashi-Suzuki K, Veenstra GJC, Taira M. Genomic organization and modulation of gene expression of the TGF-β and FGF pathways in the allotetraploid frog Xenopus laevis. Dev Biol 2017; 426:336-359. [DOI: 10.1016/j.ydbio.2016.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/10/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
|
20
|
Ottley EC, Reader KL, Lee K, Marino FE, Nicholson HD, Risbridger GP, Gold E. Over-Expression of Activin-β C Is Associated with Murine and Human Prostate Disease. Discov Oncol 2017; 8:100-107. [PMID: 28116672 DOI: 10.1007/s12672-017-0283-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/09/2017] [Indexed: 11/25/2022] Open
Abstract
Activins are members of the TGF-β superfamily and have been linked to prostate cancer. There are four mammalian activin subunits (βA, βB, βC, and βE) that dimerize to form functional proteins. The role of activin-A (βA-βA) has been relatively well characterized and has been shown to generally inhibit growth in the prostate. In contrast, little is known about the biological function of the βC and βE subunits. Previous work indicated activin-C (βC-βC) to be an antagonist of activin-A. This is important because resistance to activin-A growth inhibition occurs during prostate cancer progression. This paradox is not currently well understood. Hence, we hypothesize that local expression of the activin-βC subunit antagonizes activin-A-dependent growth inhibition and represents a key factor contributing to acquired insensitivity to activin-A observed in prostate cancer progression. To test our hypothesis, we characterized the ventral prostate lobes of 9-month-old transgenic mice over-expressing activin-βC and examined the expression of activin-βA, activin-βC, and the activin intracellular signaling factor, Smad-2, in human prostate diseases. Prostate epithelial cell hyperplasia, low-grade prostatic intraepithelial neoplasia (PIN) lesions, alterations in cell proliferation, and reduced Smad-2 nuclear localization were evident in mice over-expressing activin-βC. Increased activin-βA and -βC subunit immunoreactive scores and decreased Smad-2 nuclear localization were also evident in human prostate cancer. This study suggests that over-expression of activin-βC is associated with murine and human prostate pathologies. We conclude that the activin-βC subunit may have therapeutic and/or diagnostic implications in human prostate disease.
Collapse
Affiliation(s)
- Edward C Ottley
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| | - Karen L Reader
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand.
| | - Kailun Lee
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| | - Francesco E Marino
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| | - Helen D Nicholson
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Elspeth Gold
- Department of Anatomy, University of Otago, PO Box 913, Dunedin, 9054, New Zealand
| |
Collapse
|
21
|
Gold E, Zellhuber-McMillan S, Risbridger G, Marino FE. Regional localization of activin-β A , activin-β C , follistatin, proliferation, and apoptosis in adult and developing mouse prostate ducts. Gene Expr Patterns 2017; 23-24:70-79. [DOI: 10.1016/j.gep.2017.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 01/04/2023]
|
22
|
Mouse Models for the Study of Synthesis, Secretion, and Action of Pituitary Gonadotropins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:49-84. [PMID: 27697204 DOI: 10.1016/bs.pmbts.2016.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gonadotropins play fundamental roles in reproduction. More than 30years ago, Cga transgenic mice were generated, and more than 20years ago, the phenotypes of Cga null mice were reported. Since then, numerous mouse strains have been generated and characterized to address several questions in reproductive biology involving gonadotropin synthesis, secretion, and action. More recently, extragonadal expression, and in some cases, functions of gonadotropins in nongonadal tissues have been identified. Several genomic and proteomic approaches including novel mouse genome editing tools are available now. It is anticipated that these and other emerging technologies will be useful to build an integrated network of gonadotropin signaling pathways in various tissues. Undoubtedly, research on gonadotropins will continue to provide new knowledge and allow us transcend from benchside to the bedside.
Collapse
|
23
|
Namwanje M, Brown CW. Activins and Inhibins: Roles in Development, Physiology, and Disease. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a021881. [PMID: 27328872 DOI: 10.1101/cshperspect.a021881] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since their original discovery as regulators of follicle-stimulating hormone (FSH) secretion and erythropoiesis, the TGF-β family members activin and inhibin have been shown to participate in a variety of biological processes, from the earliest stages of embryonic development to highly specialized functions in terminally differentiated cells and tissues. Herein, we present the history, structures, signaling mechanisms, regulation, and biological processes in which activins and inhibins participate, including several recently discovered biological activities and functional antagonists. The potential therapeutic relevance of these advances is also discussed.
Collapse
Affiliation(s)
- Maria Namwanje
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Chester W Brown
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030 Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
24
|
Mahgoub A, Steer CJ. MicroRNAs in the Evaluation and Potential Treatment of Liver Diseases. J Clin Med 2016; 5:E52. [PMID: 27171116 PMCID: PMC4882481 DOI: 10.3390/jcm5050052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 02/07/2023] Open
Abstract
Acute and chronic liver disease continue to result in significant morbidity and mortality of patients, along with increasing burden on their families, society and the health care system. This in part is due to increased incidence of liver disease associated factors such as metabolic syndrome; improved survival of patients with chronic predisposing conditions such as HIV; as well as advances in the field of transplantation and associated care leading to improved survival. The fact that one disease can result in different manifestations and outcomes highlights the need for improved understanding of not just genetic phenomenon predisposing to a condition, but additionally the role of epigenetic and environmental factors leading to the phenotype of the disease. It is not surprising that providers continue to face daily challenges pertaining to diagnostic accuracy, prognostication of disease severity, progression, and response to therapies. A number of these challenges can be addressed by incorporating a personalized approach of management to the current paradigm of care. Recent advances in the fields of molecular biology and genetics have paved the way to more accurate, individualized and precise approach to caring for liver disease. The study of microRNAs and their role in both healthy and diseased livers is one example of such advances. As these small, non-coding RNAs work on fine-tuning of cellular activities and organ function in a dynamic and precise fashion, they provide us a golden opportunity to advance the field of hepatology. The study of microRNAs in liver disease promises tremendous improvement in hepatology and is likely to lay the foundation towards a personalized approach in liver disease.
Collapse
Affiliation(s)
- Amar Mahgoub
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Veterans of Foreign Wars Cancer Research Center, 406 Harvard Street, S.E., Minneapolis, MN 55455, USA.
| | - Clifford J Steer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Veterans of Foreign Wars Cancer Research Center, 406 Harvard Street, S.E., Minneapolis, MN 55455, USA.
- Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Veterans of Foreign Wars Cancer Research Center, 406 Harvard Street, S.E., Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Bi X, Xia X, Fan D, Mu T, Zhang Q, Iozzo RV, Yang W. Oncogenic activin C interacts with decorin in colorectal cancer in vivo and in vitro. Mol Carcinog 2015; 55:1786-1795. [DOI: 10.1002/mc.22427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 10/09/2015] [Accepted: 10/18/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Xiuli Bi
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Xichun Xia
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Dongdong Fan
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Teng Mu
- School of Life Science; Liaoning University; Shenyang 110036 China
| | - Qiuhua Zhang
- Department of Pharmacology; Liaoning Traditional Chinese Medicine University; Liaoning 110036 China
| | - Renato V. Iozzo
- Department of Pathology; Anatomy and Cell Biology; Thomas Jefferson University; Philadelphia Pennsylvania 19107
| | - Wancai Yang
- Department of Pathology and Institute of Precision Medicine; Jining Medical University; Jining Shandong 272067 China
- Department of Pathology; University of Illinois at Chicago; Chicago Illinois 60612
| |
Collapse
|
26
|
Peng J, Fullerton PT, Monsivais D, Clementi C, Su GH, Matzuk MM. Uterine Activin-Like Kinase 4 Regulates Trophoblast Development During Mouse Placentation. Mol Endocrinol 2015; 29:1684-93. [PMID: 26484579 DOI: 10.1210/me.2015-1048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The placenta is the first organ to develop after fertilization. It forms an interface between the maternal uterus and growing fetus to allow nutrient uptake, waste elimination, and gas exchange for a successful pregnancy in both mice and humans. In the past 2 decades, in vivo and in vitro approaches have been used to show that several members of the TGF-β superfamily regulate embryo implantation and placental development. Nodal, a TGF-β superfamily ligand, is essential for mesendoderm formation and left-right axis patterning during embryogenesis, and Nodal null mutants exhibit abnormal placental organization with expansion of trophoblast giant cells and a decrease of spongiotrophoblast and labyrinth. To better understand the importance of Nodal signaling in the uterus, we established a mouse model to conditionally ablate activin-like kinase 4 (ALK4; the Nodal type 1 receptor) using Cre recombinase driven by the progesterone receptor promoter sequences (Pgr-Cre). Alk4 conditional knockout females are subfertile due to placental abnormalities and fetal loss in pregnancy, with a placental disorganization phenotype similar to what is observed in Nodal null mice. Thus, Nodal likely functions as an indirect regulator of placental development by binding to type 1 and type 2 receptors on maternal decidual cells to stimulate expression of unknown regulators of placental development. Our findings not only describe the generation of a mouse model that enables study of Nodal signaling in placentation but also provides insights into the pathogenesis of pregnancy complications in humans, including spontaneous abortion, preeclampsia, intrauterine growth restriction, and preterm birth.
Collapse
Affiliation(s)
- Jia Peng
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Paul T Fullerton
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Diana Monsivais
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Caterina Clementi
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Gloria H Su
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Martin M Matzuk
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| |
Collapse
|
27
|
Marino FE, Risbridger G, Gold E. Re-evaluating the role of activin-βC in cancer biology. Cytokine Growth Factor Rev 2015; 26:463-70. [DOI: 10.1016/j.cytogfr.2015.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/20/2015] [Indexed: 12/31/2022]
|
28
|
Zhong C, Jiang C, Xia X, Mu T, Wei L, Lou Y, Zhang X, Zhao Y, Bi X. Antihepatic Fibrosis Effect of Active Components Isolated from Green Asparagus (Asparagus officinalis L.) Involves the Inactivation of Hepatic Stellate Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:6027-6034. [PMID: 26089141 DOI: 10.1021/acs.jafc.5b01490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Green asparagus (Asparagus officinalis L.) is a vegetable with numerous nutritional properties. In the current study, a total of 23 compounds were isolated from green asparagus, and 9 of these compounds were obtained from this genus for the first time. Preliminary data showed that the ethyl acetate (EtOAc)-extracted fraction of green asparagus exerted a stronger inhibitory effect on the growth of t-HSC/Cl-6 cells, giving an IC50 value of 45.52 μg/mL. The biological activities of the different compounds isolated from the EtOAc-extracted fraction with respect to antihepatic fibrosis were investigated further. Four compounds, C3, C4, C10, and C12, exhibited profound inhibitory effect on the activation of t-HSC/Cl-6 cells induced by TNF-α. The activation t-HSC/Cl-6 cells, which led to the production of fibrotic matrix (TGF-β1, activin C) and accumulation of TNF-α, was dramatically decreased by these compounds. The mechanisms by which these compounds inhibited the activation of hepatic stellate cells appeared to be associated with the inactivation of TGF-β1/Smad signaling and c-Jun N-terminal kinases, as well as the ERK phosphorylation cascade.
Collapse
Affiliation(s)
- Chunge Zhong
- †College of Life Science, Liaoning University, Shenyang 110036, China
| | | | - Xichun Xia
- †College of Life Science, Liaoning University, Shenyang 110036, China
| | - Teng Mu
- †College of Life Science, Liaoning University, Shenyang 110036, China
| | | | | | | | | | - Xiuli Bi
- †College of Life Science, Liaoning University, Shenyang 110036, China
| |
Collapse
|
29
|
Marino FE, Risbridger G, Gold E. Activin-βC modulates gonadal, but not adrenal tumorigenesis in the inhibin deficient mice. Mol Cell Endocrinol 2015; 409:41-50. [PMID: 25869398 DOI: 10.1016/j.mce.2015.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/15/2015] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
Activins and inhibins are involved in the regulation of several biological processes, including reproduction, development and fertility. Deregulation of the inhibin/activin signaling pathway has been implicated in the progression of reproductive and adrenal cancers. Deletion of the inhibin α-subunit results in up-regulation of the circulating levels of activins and this leads to the development of sex-cord stromal tumors followed by a cancer associated-cachexia in mice. When gonadectomy is performed, development of adrenocortical carcinomas is observed. We previously showed that overexpression of activin-βC modulates the development of sex-cord stromal tumors and reduces cancer-cachexia in the inhibin-deficient mice by antagonizing the activin signaling pathway. The adrenal cortex and gonads share in common a large subset of genes, consistent with their common embryonic lineage. Additionally, it has been shown that adrenocortical carcinomas adopt an altered cellular identity resembling the ovary. Therefore, a study to assess the impact of overexpression of activin-βC on the onset of adrenocortical carcinoma in gonadectomized inhibin-deficient mice was warranted. Within the current study we evaluated markers of apoptosis, proliferation, tumor burden, survival analysis and serum levels of activin-A in gonadectomized mice versus sham operated controls. Results showed that overexpression of activin-βC modulated the development of reproductive tumors but had no effect on adrenal tumorigenesis. Our data reinforces the importance of activin-βC in reproductive biology and suggest that activin-βC is a tumor modulator with gonadal specificity.
Collapse
Affiliation(s)
| | - Gail Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria
| | - Elspeth Gold
- Department of Anatomy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
30
|
Abstract
Activin/Nodal growth factors control a broad range of biological processes, including early cell fate decisions, organogenesis and adult tissue homeostasis. Here, we provide an overview of the mechanisms by which the Activin/Nodal signalling pathway governs stem cell function in these different stages of development. We describe recent findings that associate Activin/Nodal signalling to pathological conditions, focusing on cancer stem cells in tumorigenesis and its potential as a target for therapies. Moreover, we will discuss future directions and questions that currently remain unanswered on the role of Activin/Nodal signalling in stem cell self-renewal, differentiation and proliferation.
Collapse
Affiliation(s)
- Siim Pauklin
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ludovic Vallier
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| |
Collapse
|
31
|
|
32
|
Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, Schwartz NB, Mayo KE, Woodruff TK. Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 2014; 35:747-94. [PMID: 25051334 PMCID: PMC4167436 DOI: 10.1210/er.2014-1003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
When it was initially discovered in 1923, inhibin was characterized as a hypophysiotropic hormone that acts on pituitary cells to regulate pituitary hormone secretion. Ninety years later, what we know about inhibin stretches far beyond its well-established capacity to inhibit activin signaling and suppress pituitary FSH production. Inhibin is one of the major reproductive hormones involved in the regulation of folliculogenesis and steroidogenesis. Although the physiological role of inhibin as an activin antagonist in other organ systems is not as well defined as it is in the pituitary-gonadal axis, inhibin also modulates biological processes in other organs through paracrine, autocrine, and/or endocrine mechanisms. Inhibin and components of its signaling pathway are expressed in many organs. Diagnostically, inhibin is used for prenatal screening of Down syndrome as part of the quadruple test and as a biochemical marker in the assessment of ovarian reserve. In this review, we provide a comprehensive summary of our current understanding of the biological role of inhibin, its relationship with activin, its signaling mechanisms, and its potential value as a diagnostic marker for reproductive function and pregnancy-associated conditions.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Department of Obstetrics and Gynecology (Y.M., J.Z., C.H., W.P.S.W., T.K.W.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60610; Center for Molecular Innovation and Drug Discovery (R.M., C.H.), Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208; and Department of Molecular Biosciences (N.B.S., K.E.M., T.K.W.), Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Marino FE, Risbridger G, Gold E. The inhibin/activin signalling pathway in human gonadal and adrenal cancers. ACTA ACUST UNITED AC 2014; 20:1223-37. [DOI: 10.1093/molehr/gau074] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
34
|
Suragani RNVS, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, Davies MV, Alexander MJ, Devine M, Loveday KS, Underwood KW, Grinberg AV, Quisel JD, Chopra R, Pearsall RS, Seehra J, Kumar R. Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med 2014; 20:408-14. [PMID: 24658078 DOI: 10.1038/nm.3512] [Citation(s) in RCA: 345] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/25/2014] [Indexed: 02/08/2023]
Abstract
Erythropoietin (EPO) stimulates proliferation of early-stage erythrocyte precursors and is widely used for the treatment of chronic anemia. However, several types of EPO-resistant anemia are characterized by defects in late-stage erythropoiesis, which is EPO independent. Here we investigated regulation of erythropoiesis using a ligand-trapping fusion protein (ACE-536) containing the extracellular domain of human activin receptor type IIB (ActRIIB) modified to reduce activin binding. ACE-536, or its mouse version RAP-536, produced rapid and robust increases in erythrocyte numbers in multiple species under basal conditions and reduced or prevented anemia in murine models. Unlike EPO, RAP-536 promoted maturation of late-stage erythroid precursors in vivo. Cotreatment with ACE-536 and EPO produced a synergistic erythropoietic response. ACE-536 bound growth differentiation factor-11 (GDF11) and potently inhibited GDF11-mediated Smad2/3 signaling. GDF11 inhibited erythroid maturation in mice in vivo and ex vivo. Expression of GDF11 and ActRIIB in erythroid precursors decreased progressively with maturation, suggesting an inhibitory role for GDF11 in late-stage erythroid differentiation. RAP-536 treatment also reduced Smad2/3 activation, anemia, erythroid hyperplasia and ineffective erythropoiesis in a mouse model of myelodysplastic syndromes (MDS). These findings implicate transforming growth factor-β (TGF-β) superfamily signaling in erythroid maturation and identify ACE-536 as a new potential treatment for anemia, including that caused by ineffective erythropoiesis.
Collapse
Affiliation(s)
| | | | | | - Dianne Sako
- Acceleron Pharma, Cambridge, Massachusetts, USA
| | | | - Robert Li
- Acceleron Pharma, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | - Rajesh Chopra
- Translational Development Department, Celgene, San Francisco, California, USA
| | | | | | | |
Collapse
|
35
|
Jückstock J, Kimmich T, Mylonas I, Friese K, Dian D. The inhibin-βC subunit is down-regulated, while inhibin-βE is up-regulated by interferon-β1a in Ishikawa carcinoma cell line. Arch Gynecol Obstet 2013; 288:883-8. [PMID: 23580013 DOI: 10.1007/s00404-013-2848-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/03/2013] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Inhibins are important regulators of the female reproductive system. Recently, two new inhibin-subunits βC and βE have been described, although, their function is still quite unclear. Interestingly, there is an association between interferon and TGF-β expression. Therefore, the aim of this study was to determine expression changes of inhibin-βC and -βE subunits in endometrial Ishikawa carcinoma cell line after stimulation with interferon-β1a. MATERIALS AND METHODS The Ishikawa cell line was cultured until confluence was observed (after 2 days). After adding interferon-β1a (1,000 IE/ml), Ishikawa cells were analyzed for inhibin-βC and -βE subunits by RT-PCR. The fibroblast cell line BJ6 served as negative control. Experiments were performed in triplicates. RESULTS The endometrial adenocarcinoma cell line Ishikawa synthesized the inhibin- βC and -βE subunits. The fibroblast cells BJ6 did not demonstrate an inhibin -βC and -βE mRNA expression, while inhibin-βC subunit is down-regulated and inhibin-βE is up-regulated in Ishikawa carcinoma cell line after stimulation with interferon-β1a in Ishikawa. DISCUSSION We demonstrated for the first time a functional relationship between interferon and the novel inhibin-βC and -βE subunits. It might be possible that interferon exerts a possible apoptotic function through the βE-subunit, while, by down-regulating the βC isoform, cell proliferation is inhibited. However, the precise function of the novel βC- and βE-subunits are still not known in human endometrial tissue and a possible association with interferon is still unclear and warrants further research.
Collapse
Affiliation(s)
- Julia Jückstock
- 1st Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University Munich, Maistrasse 11, 80337, Munich, Germany
| | | | | | | | | |
Collapse
|
36
|
Activin, neutrophils, and inflammation: just coincidence? Semin Immunopathol 2013; 35:481-99. [PMID: 23385857 PMCID: PMC7101603 DOI: 10.1007/s00281-013-0365-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/17/2013] [Indexed: 01/18/2023]
Abstract
During the 26 years that have elapsed since its discovery, activin-A, a member of the transforming growth factor β super-family originally discovered from its capacity to stimulate follicle-stimulating hormone production by cultured pituitary gonadotropes, has been established as a key regulator of various fundamental biological processes, such as development, homeostasis, inflammation, and tissue remodeling. Deregulated expression of activin-A has been observed in several human diseases characterized by an immuno-inflammatory and/or tissue remodeling component in their pathophysiology. Various cell types have been recognized as sources of activin-A, and plentiful, occasionally contradicting, functions have been described mainly by in vitro studies. Not surprisingly, both harmful and protective roles have been postulated for activin-A in the context of several disorders. Recent findings have further expanded the functional repertoire of this molecule demonstrating that its ectopic overexpression in mouse airways can cause pathology that simulates faithfully human acute respiratory distress syndrome, a disorder characterized by strong involvement of neutrophils. This finding when considered together with the recent discovery that neutrophils constitute an important source of activin-A in vivo and earlier observations of upregulated activin-A expression in diseases characterized by strong activation of neutrophils may collectively imply a more intimate link between activin-A expression and neutrophil reactivity. In this review, we provide an outline of the functional repertoire of activin-A and suggest that this growth factor functions as a guardian of homeostasis, a modulator of immunity and an orchestrator of tissue repair activities. In this context, a relationship between activin-A and neutrophils may be anything but coincidental.
Collapse
|
37
|
Gold E, Marino FE, Harrison C, Makanji Y, Risbridger G. Activin-βcreduces reproductive tumour progression and abolishes cancer-associated cachexia in inhibin-deficient mice. J Pathol 2013. [DOI: 10.1002/path.4142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Elspeth Gold
- Department of Anatomy; University of Otago; Dunedin New Zealand
| | | | | | | | - Gail Risbridger
- Department of Anatomy and Developmental Biology; Monash University; Clayton Victoria Australia
| |
Collapse
|
38
|
Fields SZ, Parshad S, Anne M, Raftopoulos H, Alexander MJ, Sherman ML, Laadem A, Sung V, Terpos E. Activin receptor antagonists for cancer-related anemia and bone disease. Expert Opin Investig Drugs 2012; 22:87-101. [PMID: 23127248 DOI: 10.1517/13543784.2013.738666] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Antagonists of activin receptor signaling may be beneficial for cancer-related anemia and bone disease caused by malignancies such as multiple myeloma and solid tumors. AREAS COVERED We review evidence of dysregulated signaling by activin receptor pathways in anemia, myeloma-associated osteolysis, and metastatic bone disease, as well as potential involvement in carcinogenesis. We then review properties of activin receptor antagonists in clinical development. EXPERT OPINION Sotatercept is a novel receptor fusion protein that functions as a soluble trap to sequester ligands of activin receptor type IIA (ActRIIA). Preclinically, the murine version of sotatercept increased red blood cells (RBC) in a model of chemotherapy-induced anemia, inhibited tumor growth and metastasis, and exerted anabolic effects on bone in diverse models of multiple myeloma. Clinically, sotatercept increases RBC markedly in healthy volunteers and patients with multiple myeloma. With a rapid onset of action differing from erythropoietin, sotatercept is in clinical development as a potential first-in-class therapeutic for cancer-related anemia, including those characterized by ineffective erythropoiesis as in myelodysplastic syndromes. Anabolic bone activity in early clinical studies and potential antitumor effects make sotatercept a promising therapeutic candidate for multiple myeloma and malignant bone diseases. Antitumor activity has been observed preclinically with small-molecule inhibitors of transforming growth factor-β receptor type I (ALK5) that also antagonize the closely related activin receptors ALK4 and ALK7. LY-2157299, the first such inhibitor to enter clinical studies, has shown an acceptable safety profile so far in patients with advanced cancer. Together, these data identify activin receptor antagonists as attractive therapeutic candidates for multiple diseases.
Collapse
Affiliation(s)
- Scott Z Fields
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Brüning A, Matsingou C, Brem GJ, Rahmeh M, Mylonas I. Inhibin beta E is upregulated by drug-induced endoplasmic reticulum stress as a transcriptional target gene of ATF4. Toxicol Appl Pharmacol 2012; 264:300-4. [PMID: 22935518 DOI: 10.1016/j.taap.2012.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/02/2012] [Accepted: 08/06/2012] [Indexed: 01/13/2023]
Abstract
Inhibins and activins are gonadal peptide hormones of the transforming growth factor-β super family with important functions in the reproductive system. By contrast, the recently identified inhibin βE subunit, primarily expressed in liver cells, appears to exert functions unrelated to the reproductive system. Previously shown downregulation of inhibin βE in hepatoma cells and anti-proliferative effects of ectopic inhibin βE overexpression indicated growth-regulatory effects of inhibin βE. We observed a selective re-expression of the inhibin βE subunit in HepG2 hepatoblastoma cells, MCF7 breast cancer cells, and HeLa cervical cancer cells under endoplasmic reticulum stress conditions induced by tunicamycin, thapsigargin, and nelfinavir. Analysis of XPB1 splicing and ATF4 activation revealed that inhibin βE re-expression was associated with induction of the endoplasmic reticulum stress reaction by these drugs. Transfection of an ATF4 expression plasmid specifically induced inhibin βE expression in HeLa cells and indicates inhibin βE as a hitherto unidentified target gene of ATF4, a key transcription factor of the endoplasmic reticulum stress response. Therefore, the inhibin βE subunit defines not only a new player but also a possible new marker for drug-induced endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ansgar Brüning
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University Munich, Munich, Germany.
| | | | | | | | | |
Collapse
|
40
|
Hedger MP, Winnall WR. Regulation of activin and inhibin in the adult testis and the evidence for functional roles in spermatogenesis and immunoregulation. Mol Cell Endocrinol 2012; 359:30-42. [PMID: 21964464 DOI: 10.1016/j.mce.2011.09.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 09/16/2011] [Accepted: 09/16/2011] [Indexed: 02/03/2023]
Abstract
Activin A provides a unique link between reproduction and immunity, which is especially significant in the adult testis. This cytokine, together with inhibin B and follistatin acting as regulators of activin A activity, is fundamentally involved in the regulation of spermatogenesis and testicular steroidogenesis. However, activin A also has a much broader role in control of inflammation, fibrosis and immunity. In the Sertoli cell, activin A is regulated by signalling pathways that normally regulate stress and inflammation, signalling pathways that intersect with the classical hormonal regulatory pathways mediated by FSH. Modulation of activin A production and activity during spermatogenesis is implicated in the fine control of the cycle of the seminiferous epithelium. The immunoregulatory properties of activin A also suggest that it may be involved in maintaining testicular immune privilege. Consequently, elevated activin A production within the testis during inflammation and infection may contribute to spermatogenic failure, fibrosis and testicular damage.
Collapse
Affiliation(s)
- Mark P Hedger
- Monash Institute of Medical Research, Monash University, Melbourne, Victoria, Australia.
| | | |
Collapse
|
41
|
Gold E, Risbridger G. Activins and activin antagonists in the prostate and prostate cancer. Mol Cell Endocrinol 2012; 359:107-12. [PMID: 21787836 DOI: 10.1016/j.mce.2011.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 07/01/2011] [Accepted: 07/01/2011] [Indexed: 10/17/2022]
Abstract
Activins are members of the TGF-β super-family. There are 4 mammalian activin subunits (β(A), β(B), β(C) and β(E)) that combine to form functional proteins. The role of activin A (β(A)β(A)) is well characterized and known to be a potent growth and differentiation factor. Two of the activin subunits (β(C) and β(E)) were discovered more recently and little is known about their biological functions. In this review the evidence that activin-β(C) is a significant regulator of activin A bioactivity is presented and discussed. It is concluded that activin-β(C), like other antagonists of activin A, is an important growth regulator in prostate health and disease.
Collapse
Affiliation(s)
- Elspeth Gold
- Department of Anatomy and Structural Biology, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
42
|
Shelling AN. Mutations in inhibin and activin genes associated with human disease. Mol Cell Endocrinol 2012; 359:113-20. [PMID: 21827823 DOI: 10.1016/j.mce.2011.07.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 07/13/2011] [Accepted: 07/14/2011] [Indexed: 11/18/2022]
Abstract
Inhibins and activins are members of the transforming growth factor (TGFβ) superfamily, that includes the TGFβs, inhibins and activins, bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs). The family members are expressed throughout the human body, and are involved in the regulation of a range of important functions. The precise regulation of the TGFβ pathways is critical, and mutations of individual molecules or even minor alterations of signalling will have a significant affect on function, that may lead to development of disease or predisposition to the development of disease. The inhibins and activins regulate aspects of the male and female reproductive system, therefore, it is not surprising that most of the diseases associated with abnormalities of the inhibin and activin genes are focused on reproductive disorders and reproductive cancers. In this review, I highlight the role of genetic variants in the development of conditions such as premature ovarian failure, pre-eclampsia, and various reproductive cancers. Given the recent advances in human genetic research, such as genome wide association studies and next generation sequencing, it is likely that inhibins and activins will be shown to play more important roles in a range of human genetic diseases in the future.
Collapse
Affiliation(s)
- Andrew N Shelling
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
43
|
Abstract
The liver is the body's most important detoxification organ and has an extreme ability to regenerate. The regeneration process can be divided into three stages: initiation, proliferation and termination. Most of previous studies focus on the initial stage and proliferative stage, while the mechanism for the proper termination of liver regeneration is still poorly understood. The termination stage involves a variety of cytokines and growth factors, which mainly function to inhibit mitogen-mediated liver cell growth-promoting effect and promote the apoptosis of excessively proliferating liver cells. In this paper we will discuss the major factors involved in the termination of liver regeneration.
Collapse
|
44
|
Liu XJ, Zhang FX, Liu H, Li KC, Lu YJ, Wu QF, Li JY, Wang B, Wang Q, Lin LB, Zhong YQ, Xiao HS, Bao L, Zhang X. Activin C expressed in nociceptive afferent neurons is required for suppressing inflammatory pain. Brain 2012; 135:391-403. [DOI: 10.1093/brain/awr350] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Xing-Jun Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang-Xiong Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kai-Cheng Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying-Jin Lu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing-Feng Wu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia-Yin Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiong Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Bo Lin
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Yan-Qing Zhong
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua-Sheng Xiao
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Lan Bao
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
45
|
Inhibin/activin betaE-subunit in uterine endometrioid adenocarcinoma and endometrial cancer cell lines: From immunohistochemistry to clinical testing? Gynecol Oncol 2011; 122:132-40. [DOI: 10.1016/j.ygyno.2011.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 03/11/2011] [Accepted: 03/18/2011] [Indexed: 01/07/2023]
|
46
|
Hedger MP, Winnall WR, Phillips DJ, de Kretser DM. The regulation and functions of activin and follistatin in inflammation and immunity. VITAMINS AND HORMONES 2011; 85:255-97. [PMID: 21353885 DOI: 10.1016/b978-0-12-385961-7.00013-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The activins are members of the transforming growth factor β superfamily with broad and complex effects on cell growth and differentiation. Activin A has long been known to be a critical regulator of inflammation and immunity, and similar roles are now emerging for activin B, with which it shares 65% sequence homology. These molecules and their binding protein, follistatin, are widely expressed, and their production is increased in many acute and chronic inflammatory conditions. Synthesis and release of the activins are stimulated by inflammatory cytokines, Toll-like receptor ligands, and oxidative stress. The activins interact with heterodimeric serine/threonine kinase receptor complexes to activate SMAD transcription factors and the MAP kinase signaling pathways, which mediate inflammation, stress, and immunity. Follistatin binds to the activins with high affinity, thereby obstructing the activin receptor binding site, and targets them to cell surface proteoglycans and lysosomal degradation. Studies on transgenic mice and those with gene knockouts, together with blocking studies using exogenous follistatin, have established that activin A plays critical roles in the onset of cachexia, acute and chronic inflammatory responses such as septicemia, colitis and asthma, and fibrosis. However, activin A also directs the development of monocyte/macrophages, myeloid dendritic cells, and T cell subsets to promote type 2 and regulatory immune responses. The ability of both endogenous and exogenous follistatin to block the proinflammatory and profibrotic actions of activin A has led to interest in this binding protein as a potential therapeutic for limiting the severity of disease and to improve subsequent damage associated with inflammation and fibrosis. However, the ability of activin A to sculpt the subsequent immune response as well means that the full range of effects that might arise from blocking activin bioactivity will need to be considered in any therapeutic applications.
Collapse
Affiliation(s)
- Mark P Hedger
- Monash Institute of Medical Research, Monash University, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
47
|
Inhibin/activin expression in human and rodent liver: subunits α and βB as new players in human hepatocellular carcinoma? Br J Cancer 2011; 104:1303-12. [PMID: 21407220 PMCID: PMC3078591 DOI: 10.1038/bjc.2011.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Activins and inhibins belong to the TGFβ-superfamily, which controls cell proliferation and differentiation in many organs. Activin A, the dimer of inhibin βA subunit, acts strongly anti-proliferative in hepatocytes. Little is known on the other activin/inhibin subunits in human liver and hepatocellular carcinoma (HCC). Methods: We studied the expression of the complete inhibin family α, βA, βB, βC, βE in normal liver, tumour-adjacent and HCC tissue, 12 additional organs and rodent liver. A total of 16 HCC and 10 disease-free livers were analysed. Expression of inhibin subunits was determined by qRT–PCR, normalised to RNA input and by geNorm algorithm, and confirmed by immunohistochemistry. Results: Remarkably, βA expression was not decreased in HCC. Similarly, βC and βE exhibited no major changes. In contrast, inhibin α, barely detectable in normal liver, was strongly increased in tumour-adjacent liver and dramatically enhanced in HCC. βB was strongly enhanced in some HCC. At variance with human liver, rodent liver showed higher inhibin α and βC expression, but βA was somewhat, and βB dramatically lower. Conclusions: Upregulation of inhibin α – and possibly of βB – may shield HCC cells from anti-proliferative effects of activin A. Dramatic variations between humans and rodents may reflect different functions of some inhibins/activins.
Collapse
|
48
|
|
49
|
Mylonas I, Makovitzky J, Kunze S, Brüning A, Kainer F, Schiessl B. Inhibin-betaC subunit expression in normal and pathological human placental tissues. Syst Biol Reprod Med 2010; 57:197-203. [DOI: 10.3109/19396368.2010.528505] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
50
|
Mylonas I, Brüning A, Shabani N, Kunze S, Kupka MS. Evidence of inhibin/activin subunit betaC and betaE synthesis in normal human endometrial tissue. Reprod Biol Endocrinol 2010; 8:143. [PMID: 21092084 PMCID: PMC3002354 DOI: 10.1186/1477-7827-8-143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 11/19/2010] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Inhibins are important regulators of the female reproductive system. Recently, two new inhibin subunits betaC and betaE have been described, although it is unclear if they are synthesized in normal human endometrium. METHODS Samples of human endometrium were obtained from 82 premenopausal, non-pregnant patients undergoing gynecological surgery for benign diseases. Endometrium samples were classified according to anamnestic and histological dating into proliferative (day 1-14, n = 46), early secretory (day 15-22, n = 18) and late secretory phase (day 23-28, n = 18). Immunohistochemical analyses were performed with specific antibodies against inhibin alpha (n = 81) as well as inhibin betaA (n = 82), betaB (n = 82), betaC (n = 74) and betaE (n = 76) subunits. RT-PCR was performed for all inhibin subunits. Correlation was assessed with the Spearman factor to assess the relationship of inhibin-subunits expression within the different endometrial samples. RESULTS The novel inhibin betaC and betaE subunits were found in normal human endometrium by immunohistochemical and molecular techniques. Inhibin alpha, betaA, betaB and betaE subunits showed a circadian expression pattern, being more abundant during the late secretory phase than during the proliferative phase. Additionally, a significant correlation between inhibin alpha and all inhibin beta subunits was observed. CONCLUSIONS The differential expression pattern of the betaC- and betaE-subunits in normal human endometrial tissue suggests that they function in endometrial maturation and blastocyst implantation. However, the precise role of these novel inhibin/activin subunits in human endometrium is unclear and warrants further investigation.
Collapse
Affiliation(s)
- Ioannis Mylonas
- Ludwig-Maximilians-University Munich, 1st Department of Obstetrics and Gynecology, Maistrasse 11, 80337 Munich, Germany
| | - Ansgar Brüning
- Ludwig-Maximilians-University Munich, 1st Department of Obstetrics and Gynecology, Maistrasse 11, 80337 Munich, Germany
| | - Naim Shabani
- Ludwig-Maximilians-University Munich, 1st Department of Obstetrics and Gynecology, Maistrasse 11, 80337 Munich, Germany
- Department of Obstetrics and Gynecology, Klinikum Neuperlach, Munich, Germany
| | - Susanne Kunze
- Ludwig-Maximilians-University Munich, 1st Department of Obstetrics and Gynecology, Maistrasse 11, 80337 Munich, Germany
| | - Markus S Kupka
- Ludwig-Maximilians-University Munich, 1st Department of Obstetrics and Gynecology, Maistrasse 11, 80337 Munich, Germany
| |
Collapse
|