1
|
Nagayama I, Takei Y, Takahashi S, Okada M, Maeshima A. The activin-follistatin system: Key regulator of kidney development, regeneration, inflammation, and fibrosis. Cytokine Growth Factor Rev 2024:S1359-6101(24)00092-3. [PMID: 39581798 DOI: 10.1016/j.cytogfr.2024.11.004] [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: 11/02/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Activins, multifunctional cytokines of the transforming growth factor-beta superfamily, play critical roles in the regulation of growth and differentiation in multiple biological systems. Activin activity is finely regulated by the endogenous antagonist follistatin. Early studies reported that activins are involved in renal organogenesis, but subsequent research demonstrated that activins also play a significant role in kidney regeneration following injury. The results of more recent studies suggest activins play roles in both inflammatory kidney diseases and renal fibrosis, conditions that often culminate in end-stage renal disease. Given these findings, the inhibition of activin activity represents a promising therapeutic approach for treating a range of kidney disorders. This review discusses the latest discoveries concerning the role of the activin-follistatin system in renal development and pathophysiology and explores the potential therapeutic implications of targeting this system in the management of kidney diseases.
Collapse
Affiliation(s)
- Izumi Nagayama
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | | | - Shunsuke Takahashi
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | - Mari Okada
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | - Akito Maeshima
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan.
| |
Collapse
|
2
|
Džepina P, Ćorić M, Kovačić Perica M, Aničić MN, Grizelj R, Vuković J. Expression of activin A in liver tissue and the outcome of patients with biliary atresia. Front Pediatr 2024; 12:1457837. [PMID: 39618695 PMCID: PMC11604446 DOI: 10.3389/fped.2024.1457837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/25/2024] [Indexed: 01/03/2025] Open
Abstract
Biliary atresia (BA) is a rare disease of unknown etiology which leads to cirrhosis and death if left untreated. The standard of care is an early hepatoportoenterostomy (HPE). Long-term follow-up is mandatory, during which most patients will require a liver transplant. Activin A belongs to the transforming growth factor-β (TGF-β) superfamily. TGF-β is a central regulator in chronic liver disease. We have studied the expression of activin A in liver tissue collected intraoperatively during the HPE. We included patients who underwent HPE in a single medical center. Clinical, ultrasonographic, and pathohistological data were collected. Activin A immunostaining was performed. Expression in the bile duct epithelium and hepatocytes was scored as either weakly positive, moderately positive, or strongly positive. Patients were then divided into three groups accordingly. We observed the outcome after the HPE at 3 months, 2 years, and at the end of follow-up. The study encompassed 37 patients. At 3 months after HPE, 92.3% of those with a weakly positive activin A reaction (group A) achieved good jaundice clearance, whereas only 44.4% of those with a moderately (group B) and 40% of those with a strongly positive reaction (group C) achieved good jaundice clearance (p = 0.008). Furthermore, 2 years after the HPE, 92.3% of those in group A survived with native liver (SNL), but only 33.3% of those in group B and 46.7% of those in group C had SNL (p = 0.007). At the end of follow-up, 83.3% of those in group A survived with native liver, as did 33.3% in group B and 40% in group C. Activin A is a valuable pathohistological predictor of the outcome of BA after an HPE.
Collapse
Affiliation(s)
- Petra Džepina
- Department of Pediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Marijana Ćorić
- Department of Pathology and Cytology, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Matea Kovačić Perica
- Department of Pediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Mirna Natalija Aničić
- Department of Pediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ruža Grizelj
- Department of Pediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jurica Vuković
- Department of Pediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
3
|
Jönsson C, Bergram M, Kechagias S, Nasr P, Ekstedt M. Activin A levels in metabolic dysfunction-associated steatotic liver disease associates with fibrosis and the PNPLA3 I148M variant. Scand J Gastroenterol 2024; 59:737-741. [PMID: 38563432 DOI: 10.1080/00365521.2024.2334804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver condition worldwide. There is an urgent need to develop new biomarkers to assess disease severity and to define patients with a progressive phenotype. Activin A is a new promising biomarker with conflicting results about liver fibrosis. In this study we investigate levels of Activin A in patients with biopsy proven MASLD. We assess levels of Activin A in regard to fibrosis stage and genetic variant I148M in the patatin-like phospholipase domain-containing protein 3 (PNPLA3). METHODS Activin A levels were assessed in plasma samples from patients with biopsy-proven MASLD in a cross-sectional study. All patients were clinically evaluated and the PNPLA3 I148M genotype of the cohort was assessed. FINDINGS 41 patients were included and 27% of these had advanced fibrosis. In MASLD patients with advanced fibrosis, Activin A levels was higher (p < 0.001) and could classify advanced fibrosis with an AUROC for activin A of 0.836 (p < 0.001). Patients homozygous for PNPLA3 I148M G/G had higher levels of activin A than non-homozygotes (p = 0.027). CONCLUSIONS Circulating activin A levels were associated with advanced fibrosis and could be a potential blood biomarker for identifying advanced fibrosis in MASLD. Patients with the risk genotype PNPLA3 I148M G/G had higher levels of activin A proposing activin A as a contributor of the transition from simple steatosis to a fibrotic phenotype.
Collapse
Affiliation(s)
- Cecilia Jönsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Martin Bergram
- Division of Prevention, Rehabilitation and Community Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Stergios Kechagias
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Patrik Nasr
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Swan J, Szabó Z, Peters J, Kummu O, Kemppi A, Rahtu-Korpela L, Konzack A, Hakkola J, Pasternack A, Ritvos O, Kerkelä R, Magga J. Inhibition of activin receptor 2 signalling ameliorates metabolic dysfunction-associated steatotic liver disease in western diet/L-NAME induced cardiometabolic disease. Biomed Pharmacother 2024; 175:116683. [PMID: 38705130 DOI: 10.1016/j.biopha.2024.116683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE Blockade of activin 2 receptor (ACVR2) signaling has been shown to improve insulin sensitivity and aid in weight loss. Inhibition of ACVR2 signaling restores cardiac function in multiple heart failure models. However, its potential in the treatment of obesity-related cardiometabolic disease remains unknown. Here, we investigated targeting ACVR2 signaling in cardiometabolic disease manifested with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Mice were fed a high-fat, high-sugar diet combined with the administration of nitric oxide synthase inhibitor L-NAME in drinking water, which causes hypertensive stress. For the last eight weeks, the mice were treated with the soluble ACVR2B decoy receptor (sACVR2B-Fc). RESULTS sACVR2B-Fc protected against the development of comorbidities associated with cardiometabolic disease. This was most pronounced in the liver where ACVR2 blockade attenuated the development of MASLD including cessation of pro-fibrotic activation. It also significantly reduced total plasma cholesterol levels, impeded brown adipose tissue whitening, and improved cardiac diastolic function. In vitro, ACVR2 ligands activin A, activin B and GDF11 induced profibrotic signaling and the proliferation of human cardiac fibroblasts. CONCLUSIONS Blockade of ACVR2B exerts broad beneficial effects for therapy of cardiometabolic disease. By reducing obesity, ameliorating cardiovascular deterioration and restraining MASLD, blockade of ACVR2B signaling proves a potential target in MASLD and its comorbidities.
Collapse
Affiliation(s)
- Julia Swan
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Juliana Peters
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anna Kemppi
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Lea Rahtu-Korpela
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anja Konzack
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Johanna Magga
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| |
Collapse
|
5
|
Zeplin PH. Activin A and Follistatin Serum Concentrations in Breast Augmentation Patients. Plast Surg (Oakv) 2023; 31:377-382. [PMID: 37915344 PMCID: PMC10617457 DOI: 10.1177/22925503211051120] [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: 04/14/2021] [Revised: 08/17/2021] [Accepted: 08/28/2021] [Indexed: 11/03/2023] Open
Abstract
Background: Capsular contracture is caused by an excessive fibrotic reaction similar as observed in other progressive fibrotic disorders. For their pathogenesis, several studies confirmed the importance of activins and follistatin. The aim of this study was to determine and analyze serum levels of Activin A and follistatin in patients with capsular contracture after aesthetic breast augmentation. Methods: The study included 361 female patients who underwent primary aesthetic breast augmentation, came for control examination after breast augmentation or for revision operation because of capsular contracture. Blood samples were taken and using a specific ELISA to determine the serum concentration levels of Activin A and Follistatin. Results: Ninety-six patients (n = 96), who developed a capsular contracture Baker ≥°III and underwent revision surgery were collected (capsular fibrosis group). One-hundred and fourteen patients (n = 114) were asymptomatic for capsular fibrosis Baker ≥°III after primary breast augmentation and 33 (n = 33) of them had developed no capsular fibrosis after more than 10 years (long-term group). For control group, blood samples were taken from 167 patients (n = 167) before primary aesthetic breast augmentation. Serum Activin A levels were significantly higher in the long-term Group compared with those in the capsular fibrosis- and the control groups. Follistatin levels were significantly lower in the capsular fibrosis group compared to the control- and the long-term groups. A small amount of control group patients (n = 16) developed a capsular fibrosis within 2 years after primary breast augmentation with significant lower follistatin levels. Retrospectively, they showed significantly lower serum follistatin levels than the control group even before the onset of capsular contracture. Conclusions: Capsular fibrosis has no effect on Activin A serum levels. In contrast, follistatin serum levels are lower in patients with capsular fibrosis. These results show that besides many other factors, a dysregulation of the Activin-follistatin axis may have importance on the pathogenesis of capsular contracture.
Collapse
Affiliation(s)
- Philip H. Zeplin
- Schlosspark Klinik Ludwigsburg, Privatklinik für Plastische und Ästhetische Chirurgie, Ludwigsburg, Germany
| |
Collapse
|
6
|
Zheng L, Wu J, Hu H, Cao H, Xu N, Chen K, Wen B, Wang H, Yuan H, Xie L, Jiang Y, Li Z, Liang C, Yuan J, Li Z, Yuan X, Xiao W, Wang J. Single-cell RNA transcriptome landscape of murine liver following systemic administration of mesoporous silica nanoparticles. J Control Release 2023; 361:427-442. [PMID: 37487929 DOI: 10.1016/j.jconrel.2023.07.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 07/22/2023] [Indexed: 07/26/2023]
Abstract
Due to the unique physicochemical properties, mesoporous silica nanoparticles (MONs) have been widely utilized in biomedical fields for drug delivery, gene therapy, disease diagnosis and imaging. With the extensive applications and large-scale production of MONs, the potential effects of MONs on human health are gaining increased attention. To better understand the cellular and molecular mechanisms underlying the effects of MONs on the mouse liver, we profiled the transcriptome of 63,783 single cells from mouse livers following weekly intravenous administration of MONs for 2 weeks. The results showed that the proportion of endothelial cells and CD4+ T cells was increased, whereas that of Kupffer cells was decreased, in a dose-dependent manner after MONs treatment in the mouse liver. We also observed that the proportion of inflammation-related Kupffer cell subtype and wound healing-related hepatocyte subtype were elevated, but the number of hepatocytes with detoxification characteristics was reduced after MONs treatment. The cell-cell communication network revealed that there was more crosstalk between cholangiocytes and Kupffer cells, liver capsular macrophages, hepatic stellate cells, and endothelial cells following MONs treatment. Furthermore, we identified key ligand-receptor pairs between crucial subtypes after MONs treatment that are known to promote liver fibrosis. Collectively, our study explored the effects of MONs on mouse liver at a single-cell level and provides comprehensive information on the potential hepatotoxicity of MONs.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jiangpeng Wu
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Hong Hu
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Hua Cao
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Nan Xu
- Division of Thyroid surgery, Department of General Surgery, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Kun Chen
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Bowen Wen
- College of Natural Resources and Environment, South China Agricultural University, 483 Wushan Road, Guangzhou, Guangdong 510642, China
| | - Huifang Wang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Haitao Yuan
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Lulin Xie
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Yuke Jiang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Zhifen Li
- School of Chemistry and Chemical Engineering, Shanxi Datong University, Xing Yun Street, Pingcheng District, Datong, Shanxi Province 037009, PR China
| | - Cailing Liang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Jimin Yuan
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Zhijie Li
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| | - Xiaopeng Yuan
- Department of Laboratory Medicine, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China.
| | - Jigang Wang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
7
|
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
|
8
|
Santol J, Pereyra D, Haegele S, Ammon D, Ortmayr G, Pirabe A, Jonas JP, Schuster S, Kim S, Nguyen T, Gruenberger T, Assinger A, Starlinger P. The Ratio of Activin A and Follistatin-Like 3 Is Associated With Posthepatectomy Liver Failure and Morbidity in Patients Undergoing Liver Resection. GASTRO HEP ADVANCES 2023; 2:642-651. [PMID: 39129875 PMCID: PMC11307668 DOI: 10.1016/j.gastha.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/28/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Activin A is a key regulator in liver regeneration, but data evaluating its role in humans after hepatic surgery are limited. In this study we explore the predictive role of circulating activin A, its antagonist follistatin-like 3 (FSTL-3), and their ratio for posthepatectomy liver failure (PHLF) and monitor their levels after surgery, to evaluate their role in human liver regeneration. Methods Activin A and FSTL-3 levels were assessed in 59 patients undergoing liver surgery. Using receiver operating characteristic analysis, we evaluated the predictive potential of activin A, FSTL-3, and their ratio. Results While perioperative dynamics of activin A and FSTL3 were significantly affected by hepatic resection (activin A P = .045, FSTL-3 P = .005), their functionally relevant ratio did not significantly change (P = .528). Neither activin A nor FSTL-3 alone but only their ratio exhibited a significant predictive potential for PHLF (area under the curve: 0.789, P = .038). Patients with low preoperative activin A/FSTL-3 ratio were found to more frequently suffer from PHLF (0.017) and morbidity (0.005). Conclusion Activin A/FSTL-3 ratio predicts PHLF and morbidity. Its significance in preoperative patient assessment needs to be further validated in larger, independent cohorts.
Collapse
Affiliation(s)
- Jonas Santol
- Department of Surgery, Vienna Health Network, HPB Center, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - David Pereyra
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefanie Haegele
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Daphni Ammon
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Gregor Ortmayr
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jan Philipp Jonas
- Department of Surgery, Vienna Health Network, HPB Center, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
- Department of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Stefan Schuster
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Sarang Kim
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Toni Nguyen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Gruenberger
- Department of Surgery, Vienna Health Network, HPB Center, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Patrick Starlinger
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Yuan C, Ni L, Wu X. Activin A activation drives renal fibrosis through the STAT3 signaling pathway. Int J Biochem Cell Biol 2021; 134:105950. [PMID: 33609746 DOI: 10.1016/j.biocel.2021.105950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/27/2022]
Abstract
The present study investigated whether TGF-β1 promotes fibrotic changes in HK-2 cells through the Activin A and STAT3 signaling pathways in vitro. Bioinformatics analysis of microarray profiles (GSE20247 and GSE23338) and a protein-protein interaction (PPI) analysis were performed to select hub genes. For the in vitro study, HK-2 cells were exposed to TGF-β1. The expression of Activin A and STAT3 was assayed, and the effect of Activin A and STAT3 expression on fibrosis was assessed (Collagen I and Fibronectin). The bioinformatics study revealed TGF-β1 and Activin A as hub genes. The in vitro study showed that Activin A expression was significantly increased after TGF-β1 incubation. Blocking Activin A attenuated TGF-β1-induced fibrosis. In addition, Activin A blockade attenuated TGF-β1-induced STAT3 signaling pathway activation and related fibrosis. More importantly, STAT3 inhibition by S3I-201 alleviated TGF-β1-induced fibrosis. Activin A promoted cellular fibrotic changes through the STAT3 signaling pathway. Attenuating Activin A expression to mediate the STAT3 signaling pathway might be a strategy for potent renal fibrosis treatment.
Collapse
Affiliation(s)
- Cheng Yuan
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
10
|
Zhang H, Ju B, Nie Y, Song B, Xu Y, Gao P. Adenovirus‑mediated knockdown of activin A receptor type 2A attenuates immune‑induced hepatic fibrosis in mice and inhibits interleukin‑17‑induced activation of primary hepatic stellate cells. Int J Mol Med 2018; 42:279-289. [PMID: 29620144 PMCID: PMC5979935 DOI: 10.3892/ijmm.2018.3600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/08/2018] [Indexed: 01/20/2023] Open
Abstract
Fibrosis induces a progressive loss of liver function, thus leading to organ failure. Activins are secreted proteins that belong to the transforming growth factor (TGF)-β superfamily, which initiate signaling by binding to their two type II receptors: Activin A receptor type 2A (ACVR2A) and activin A receptor type 2B. Previous studies that have explored the mechanisms underlying immune-induced hepatic fibrosis have mainly focused on TGF-β signaling, not activin signaling. To investigate the role of the activin pathway in this disease, adenovirus particles containing short hairpin (sh)RNA targeting ACVR2A mRNA (Ad-ACVR2A shRNA) were administered to mice, which were chronically treated with concanavalin A (Con A). The pathological changes in the liver were evaluated with hematoxylin/eosin staining, Masson trichrome staining and immunohistochemical assay. The results detected an increase in serum activin A and liver ACVR2A in Con A-treated animals. Conversely, liver function was partially restored and fibrotic injury was attenuated when activin signaling was blocked. In addition, the activation of hepatic stellate cells (HSCs) in response to Con A was suppressed by Ad-ACVR2A shRNA, as evidenced by decreased α-smooth muscle actin, and type I and IV collagen expression. Furthermore, primary mouse HSCs (mHSCs) were activated when exposed to interleukin (IL)-17A or IL-17F, which are two major cytokines produced by cluster of differentiation 4+ T helper 17 cells. The levels of activin A, type I and IV collagen were determined with ELISA kits and the expression of fibrotic molecules was determined with western blot analysis. Conversely, blocking activin/ACVR2A impaired the potency of HSCs to produce collagens in response to IL-17s. In addition, C terminus phosphorylation of Smad2 on Ser465 and Ser467, induced by either Con A in the liver or by IL-17s in mHSCs, was partly inhibited when activin A/ACVR2A signaling was suppressed. Collectively, the present study demonstrated an involvement of activated activin A/ACVR2A/Smad2 signaling in immune-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Hongjun Zhang
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Baoling Ju
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ying Nie
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Baohui Song
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yuanhong Xu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ping Gao
- Department of Gastroenterology, Mudanjiang Forestry Central Hospital, Mudanjiang, Heilongjiang 157000, P.R. China
| |
Collapse
|
11
|
Islam MS, Akhtar MM, Segars JH, Castellucci M, Ciarmela P. Molecular targets of dietary phytochemicals for possible prevention and therapy of uterine fibroids: Focus on fibrosis. Crit Rev Food Sci Nutr 2018; 57:3583-3600. [PMID: 28609115 DOI: 10.1080/10408398.2016.1245649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Uterine fibroids (myomas or leiomyomas) are common benign tumors of reproductive aged women. Fibroids are clinically apparent in 20-50% of women, and cause abnormal uterine bleeding, abdominal pain and discomfort, pregnancy complications and infertility. Unfortunately, limited numbers of medical treatment are available but no effective preventive strategies exist. Moreover, the benefits of medical treatments are tempered by lack of efficacy or serious adverse side effects. Fibrosis has recently been recognized as a key pathological event in leiomyoma development and growth. It is defined by the excessive deposition of extracellular matrix (ECM). ECM plays important role in making bulk structure of leiomyoma, and ECM-rich rigid structure is believed to be a cause of abnormal bleeding and pelvic pain/pressure. Dietary phytochemicals are known to regulate fibrotic process in different biological systems, and being considered as potential tool to manage human health. At present, very few dietary phytochemicals have been studied in uterine leiomyoma, and they are mostly known for their antiproliferative effects. Therefore, in this review, our aim was to introduce some dietary phytochemicals that could target fibrotic processes in leiomyoma. Thus, this review could serve as useful resource to develop antifibrotic drugs for possible prevention and treatment of uterine fibroids.
Collapse
Affiliation(s)
- Md Soriful Islam
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,b Biotechnology and Microbiology Laboratory, Department of Botany , University of Rajshahi , Rajshahi , Bangladesh
| | - Most Mauluda Akhtar
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,c Department of Clinical and Molecular Sciences , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy
| | - James H Segars
- d Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences, Department of Gynecology and Obstetrics , Johns Hopkins School of Medicine , Baltimore , Maryland , USA
| | - Mario Castellucci
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy
| | - Pasquapina Ciarmela
- a Department of Experimental and Clinical Medicine , Faculty of Medicine, Università Politecnica delle Marche , Ancona , Italy.,e Department of Information Engineering , Università Politecnica delle Marche , Ancona , Italy
| |
Collapse
|
12
|
Activin-A causes Hepatic stellate cell activation via the induction of TNFα and TGFβ in Kupffer cells. Biochim Biophys Acta Mol Basis Dis 2017; 1864:891-899. [PMID: 29287776 DOI: 10.1016/j.bbadis.2017.12.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/06/2017] [Accepted: 12/22/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS TGFβ superfamily member Activin-A is a multifunctional hormone/cytokine expressed in multiple tissues and cells, where it regulates cellular differentiation, proliferation, inflammation and tissue architecture. High activin-A levels have been reported in alcoholic cirrhosis and non-alcoholic steatohepatitis (NASH). Our aim was to identify the cell types involved in the fibrotic processes induced by activin-A in liver and verify the liver diseases that this molecule can be found increased. METHODS We studied the effect of activin-A on mouse primary Kupffer cells (KCs) and Hepatic Stellate cells (HSCs) and the levels of activin-A and its inhibitor follistatin in the serum of patients from a large panel of liver diseases. RESULTS Activin-A is expressed by mouse hepatocytes, HSCs and Liver Sinusoid Endothelial cells but not KCs. Each cell type expresses different activin receptor combinations. HSCs are unresponsive to activin-A due to downregulation/desensitization of type-II activin receptors, while KCs respond by increasing the expression/production of TNFα και TGFβ1. In the presence of KCs or conditioned medium from activin-A treated KCs, HSCs switch to a profibrogenic phenotype, including increased collagen and αSMA expression and migratory capacity. Incubation of activin-A treated KC conditioned medium with antibodies against TNFα and TGFβ1 partially blocks its capacity to activate HSCs. Only patients with alcoholic liver diseases and NASH cirrhosis have significantly higher activin-A levels and activin-A/follistatin ratio. CONCLUSIONS Activin-A may induce fibrosis in NASH and alcoholic cirrhosis via activation of KCs to express pro-inflammatory molecules that promote HSC-dependent fibrogenesis and could be a target for future anti-fibrotic therapies.
Collapse
|
13
|
Walton KL, Johnson KE, Harrison CA. Targeting TGF-β Mediated SMAD Signaling for the Prevention of Fibrosis. Front Pharmacol 2017; 8:461. [PMID: 28769795 PMCID: PMC5509761 DOI: 10.3389/fphar.2017.00461] [Citation(s) in RCA: 405] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/27/2017] [Indexed: 01/18/2023] Open
Abstract
Fibrosis occurs when there is an imbalance in extracellular matrix (ECM) deposition and degradation. Excessive ECM deposition results in scarring and thickening of the affected tissue, and interferes with tissue and organ homeostasis – mimicking an exaggerated “wound healing” response. Many transforming growth factor-β (TGF-β) ligands are potent drivers of ECM deposition, and additionally, have a natural affinity for the ECM, creating a concentrated pool of pro-fibrotic factors at the site of injury. Consequently, TGF-β ligands are upregulated in many human fibrotic conditions and, as such, are attractive targets for fibrosis therapy. Here, we will discuss the contribution of TGF-β proteins in the pathogenesis of fibrosis, and promising anti-fibrotic approaches that target TGF-β ligands.
Collapse
Affiliation(s)
- Kelly L Walton
- Growth Factor Therapeutics Laboratory, Department of Physiology, Monash University, ClaytonVIC, Australia
| | - Katharine E Johnson
- Growth Factor Therapeutics Laboratory, Department of Physiology, Monash University, ClaytonVIC, Australia
| | - Craig A Harrison
- Growth Factor Therapeutics Laboratory, Department of Physiology, Monash University, ClaytonVIC, Australia
| |
Collapse
|
14
|
Chang HM, Cheng JC, Liu Y, Klausen C, Xu C, Leung PCK. Activin A-induced increase in LOX activity in human granulosa-lutein cells is mediated by CTGF. Reproduction 2016; 152:293-301. [PMID: 27530347 DOI: 10.1530/rep-16-0254] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/04/2016] [Indexed: 01/03/2025]
Abstract
Lysyl oxidase (LOX) is the key enzyme involved in the crosslinking of collagen and elastin that is essential for the formation of extracellular matrix (ECM). LOX-mediated ECM remodeling plays a critical role in follicle development, oocyte maturation and corpus luteum formation. To date, the regulation of LOX in human ovary has never been elucidated. Activin A and its functional receptors are highly expressed in ovarian follicles from an early developmental stage. They locally regulate follicle progression. The aim of this study was to investigate the effects of activin A on the expression of LOX and its extracellular enzyme activity in primary and immortalized human granulosa-lutein cells obtained from patients undergoing an in vitro fertilization procedure. We demonstrated that activin A significantly upregulated the expression of connective tissue growth factor (CTGF) and LOX via an activin/TGF-β type I receptor mediated-signaling pathway. Using a target depletion small interfering RNA knockdown approach, we further confirmed that the upregulation of CTGF expression resulted in an activin-A-induced increases in LOX expression and activity. These findings may provide insight into the mechanisms by which intrafollicular growth factors regulate the expression of LOX for ECM formation and tissue remodeling in the human ovary.
Collapse
Affiliation(s)
- Hsun-Ming Chang
- Department of Obstetrics and GynaecologyChild and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and GynaecologyChild and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yingtao Liu
- Department of Obstetrics and GynaecologyChild and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada Obstetrics and Gynecology Hospital of Fudan UniversityShanghai, China
| | - Christian Klausen
- Department of Obstetrics and GynaecologyChild and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Congjian Xu
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghai, China
| | - Peter C K Leung
- Department of Obstetrics and GynaecologyChild and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
15
|
Ding ZY, Jin GN, Wang W, Sun YM, Chen WX, Chen L, Liang HF, Datta PK, Zhang MZ, Zhang B, Chen XP. Activin A-Smad Signaling Mediates Connective Tissue Growth Factor Synthesis in Liver Progenitor Cells. Int J Mol Sci 2016; 17:408. [PMID: 27011166 PMCID: PMC4813263 DOI: 10.3390/ijms17030408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 01/19/2023] Open
Abstract
Liver progenitor cells (LPCs) are activated in chronic liver damage and may contribute to liver fibrosis. Our previous investigation reported that LPCs produced connective tissue growth factor (CTGF/CCN2), an inducer of liver fibrosis, yet the regulatory mechanism of the production of CTGF/CCN2 in LPCs remains elusive. In this study, we report that Activin A is an inducer of CTGF/CCN2 in LPCs. Here we show that expression of both Activin A and CTGF/CCN2 were upregulated in the cirrhotic liver, and the expression of Activin A positively correlates with that of CTGF/CCN2 in liver tissues. We go on to show that Activin A induced de novo synthesis of CTGF/CCN2 in LPC cell lines LE/6 and WB-F344. Furthermore, Activin A contributed to autonomous production of CTGF/CCN2 in liver progenitor cells (LPCs) via activation of the Smad signaling pathway. Smad2, 3 and 4 were all required for this induction. Collectively, these results provide evidence for the fibrotic role of LPCs in the liver and suggest that the Activin A-Smad-CTGF/CCN2 signaling in LPCs may be a therapeutic target of liver fibrosis.
Collapse
Affiliation(s)
- Ze-Yang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Guan-Nan Jin
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yi-Min Sun
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei-Xun Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui-Fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiao-Ping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
16
|
Refaat B, Ashshi AM, El-Shemi AG, Azhar E. Activins and Follistatin in Chronic Hepatitis C and Its Treatment with Pegylated-Interferon-α Based Therapy. Mediators Inflamm 2015; 2015:287640. [PMID: 25969625 PMCID: PMC4417604 DOI: 10.1155/2015/287640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 12/12/2022] Open
Abstract
Pegylated-interferon-α based therapy for the treatment of chronic hepatitis C (CHC) is considered suboptimal as not all patients respond to the treatment and it is associated with several side effects that could lead to dose reduction and/or termination of therapy. The currently used markers to monitor the response to treatment are based on viral kinetics and their performance in the prediction of treatment outcome is moderate and does not combine accuracy and their values have several limitations. Hence, the development of new sensitive and specific predictor markers could provide a useful tool for the clinicians and healthcare providers, especially in the new era of interferon-free therapy, for the classification of patients according to their response to the standard therapy and only subscribing the novel directly acting antiviral drugs to those who are anticipated not to respond to the conventional therapy and/or have absolute contraindications for its use. The importance of activins and follistatin in the regulation of immune system, liver biology, and pathology has recently emerged. This review appraises the up-to-date knowledge regarding the role of activins and follistatin in liver biology and immune system and their role in the pathophysiology of CHC.
Collapse
Affiliation(s)
- Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-'Abdiyah Campus, P. O. Box 7607, Makkah, Saudi Arabia
| | - Ahmed Mohamed Ashshi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-'Abdiyah Campus, P. O. Box 7607, Makkah, Saudi Arabia
| | - Adel Galal El-Shemi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-'Abdiyah Campus, P. O. Box 7607, Makkah, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut 6515, Egypt
| | - Esam Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Islam MS, Protic O, Ciavattini A, Giannubilo SR, Tranquilli AL, Catherino WH, Castellucci M, Ciarmela P. Tranilast, an orally active antiallergic compound, inhibits extracellular matrix production in human uterine leiomyoma and myometrial cells. Fertil Steril 2014; 102:597-606. [PMID: 24934492 DOI: 10.1016/j.fertnstert.2014.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 02/06/2023]
|
18
|
Jin CH, Krishnaiah M, Sreenu D, Subrahmanyam VB, Rao KS, Lee HJ, Park SJ, Park HJ, Lee K, Sheen YY, Kim DK. Discovery of N-((4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)methyl)-2-fluoroaniline (EW-7197): a highly potent, selective, and orally bioavailable inhibitor of TGF-β type I receptor kinase as cancer immunotherapeutic/antifibrotic agent. J Med Chem 2014; 57:4213-38. [PMID: 24786585 DOI: 10.1021/jm500115w] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A series of 2-substituted-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)imidazoles was synthesized and evaluated to optimize a prototype inhibitor of TGF-β type I receptor kinase (ALK5), 6. Combination of replacement of a quinoxalin-6-yl moiety of 6 with a [1,2,4]triazolo[1,5-a]pyridin-6-yl moiety, insertion of a methyleneamino linker, and a o-F substituent in the phenyl ring markedly increased ALK5 inhibitory activity, kinase selectivity, and oral bioavailability. The 12b (EW-7197) inhibited ALK5 with IC50 value of 0.013 μM in a kinase assay and with IC50 values of 0.0165 and 0.0121 μM in HaCaT (3TP-luc) stable cells and 4T1 (3TP-luc) stable cells, respectively, in a luciferase assay. Selectivity profiling of 12b using a panel of 320 protein kinases revealed that it is a highly selective ALK5/ALK4 inhibitor. Pharmacokinetic study with 12b·HCl in rats showed an oral bioavailability of 51% with high systemic exposure (AUC) of 1426 ng × h/mL and maximum plasma concentration (Cmax) of 1620 ng/mL. Rational optimization of 6 has led to the identification of a highly potent, selective, and orally bioavailable ALK5 inhibitor 12b.
Collapse
Affiliation(s)
- Cheng Hua Jin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University , 11-1 Daehyun-dong, Seodaemun-gu, Seoul 120-750, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Follistatin, an activin antagonist, ameliorates renal interstitial fibrosis in a rat model of unilateral ureteral obstruction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:376191. [PMID: 24883308 PMCID: PMC4026945 DOI: 10.1155/2014/376191] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/14/2014] [Indexed: 02/04/2023]
Abstract
Activin, a member of the TGF-β superfamily, regulates cell growth and differentiation in various cell types. Activin A acts as a negative regulator of renal development as well as tubular regeneration after renal injury. However, it remains unknown whether activin A is involved in renal fibrosis. To clarify this issue, we utilized a rat model of unilateral ureteral obstruction (UUO). The expression of activin A was significantly increased in the UUO kidneys compared to that in contralateral kidneys. Activin A was detected in glomerular mesangial cells and interstitial fibroblasts in normal kidneys. In UUO kidneys, activin A was abundantly expressed by interstitial α-SMA-positive myofibroblasts. Administration of recombinant follistatin, an activin antagonist, reduced the fibrotic area in the UUO kidneys. The number of proliferating cells in the interstitium, but not in the tubules, was significantly lower in the follistatin-treated kidneys. Expression of α-SMA, deposition of type I collagen and fibronectin, and CD68-positive macrophage infiltration were significantly suppressed in the follistatin-treated kidneys. These data suggest that activin A produced by interstitial fibroblasts acts as a potent profibrotic factor during renal fibrosis. Blockade of activin A action may be a novel approach for the prevention of renal fibrosis progression.
Collapse
|
20
|
Voumvouraki A, Notas G, Koulentaki M, Georgiadou M, Klironomos S, Kouroumalis E. Increased serum activin-A differentiates alcoholic from cirrhosis of other aetiologies. Eur J Clin Invest 2012; 42:815-22. [PMID: 22304651 DOI: 10.1111/j.1365-2362.2012.02647.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Activin-A is a molecule of the TGF superfamily, implicated in liver fibrosis, regeneration and stem cell differentiation. However, data on activins in liver diseases are few. We therefore studied serum levels of activin-A in chronic liver diseases. To identify the origin of activin-A, levels in the hepatic vein were also estimated. MATERIALS AND METHODS Nineteen controls and 162 patients participated in the study: 39 with hepatocellular carcinoma (HCC: 19 viral associated and 20 alcohol associated), 18 with chronic hepatitis C (CHC), 47 with primary biliary cirrhosis (26 PBC stage I-II and 21 stage IV), 22 with alcoholic cirrhosis (AC, hepatic vein blood available in 16), 20 with HCV cirrhosis (hepatic vein blood available in 18) and 16 patients with alcoholic fatty liver with mild to moderate fibrosis but no cirrhosis. RESULTS Activin-A levels were significantly increased (P < 0·001) in serum of patients with AC (median 673 pg/mL, range 449-3279), compared with either controls (149 pg/mL, 91-193) or patients with viral cirrhosis (189 pg/mL, 81-480), CHC (142 pg/mL, 65-559) PBC stage I-II (100 pg/mL, 59-597) and PBC stage IV (104 pg/mL, 81-579). Only patients with AC-associated HCC had significantly increased levels of activin-A (2403 pg/mL, 1561-7220 pg/mL). Activin-A serum levels could accurately discriminate AC from cirrhosis of other aetiologies and noncirrhotic alcoholic fatty liver with fibrosis. CONCLUSIONS Increased serum levels of activin-A only in patients with alcohol-related cirrhosis or HCC suggest a possible role of this molecule in the pathophysiology of AC. Further research is warranted to elucidate its role during the profibrotic process and its possible clinical applications.
Collapse
Affiliation(s)
- Argyro Voumvouraki
- University Hospital Department of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | |
Collapse
|
21
|
The Monocyte Locomotion Inhibitory Factor (MLIF) Produced by Entamoeba histolytica Alters the Expression of Genes Related to the Wound-Healing Process. Int J Pept Res Ther 2012. [DOI: 10.1007/s10989-012-9315-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
22
|
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
|
23
|
Menthena A, Koehler CI, Sandhu JS, Yovchev MI, Hurston E, Shafritz DA, Oertel M. Activin A, p15INK4b signaling, and cell competition promote stem/progenitor cell repopulation of livers in aging rats. Gastroenterology 2011; 140:1009-20. [PMID: 21147108 PMCID: PMC3087123 DOI: 10.1053/j.gastro.2010.12.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 11/17/2010] [Accepted: 12/02/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Highly proliferative fetal liver stem/progenitor cells (FLSPCs) repopulate livers of normal recipients by cell competition. We investigated the mechanisms by which FLSPCs repopulate livers of older compared with younger rats. METHODS Fetal liver cells were transplanted from DPPIV(+) F344 rats into DPPIV(-) rats of different ages (2, 6, 14, or 18 months); liver tissues were analyzed 6 months later. Cultured cells and liver tissues were analyzed by reverse transcription polymerase chain reaction, immunoblot, histochemistry, laser-capture microscopy, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling analyses. RESULTS We observed 4- to 5-fold increases in liver repopulation when FLSPCs were transplanted into older compared with younger rats. Messenger RNA levels of cyclin-dependent kinase inhibitors increased progressively in livers of older rats; hepatocytes from 20-month-old rats had 6.1-fold higher expression of p15INK4b and were less proliferative in vitro than hepatocytes from 2-month-old rats. Expression of p15INK4b in cultured hepatocytes was up-regulated by activin A, which increased in liver during aging. Activin A inhibited proliferation of adult hepatocytes, whereas FLSPCs were unresponsive because they had reduced expression of activin receptors (eg, ALK-4). In vivo, expanding cell clusters derived from transplanted FLSPCs had lower levels of ALK-4 and p15INK4b and increased levels of Ki-67 compared with the host parenchyma. Liver tissue of older rats had 3-fold more apoptotic cells than that of younger rats. CONCLUSIONS FLSPCs, resistant to activin A signaling, repopulate livers of older rats; hepatocytes in older rats have less proliferation because of increased activin A and p15INK4b levels and increased apoptosis than younger rats. These factors and cell types might be manipulated to improve liver cell transplantation strategies in patients with liver diseases in which activin A levels are increased.
Collapse
|
24
|
Mantel PY, Schmidt-Weber CB. Transforming growth factor-beta: recent advances on its role in immune tolerance. Methods Mol Biol 2011; 677:303-38. [PMID: 20941619 DOI: 10.1007/978-1-60761-869-0_21] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor (TGF-β1) is a pleiotropic cytokine, secreted by immune and nonhematopoietic cells. TGF-β is involved in many different critical processes, such as embryonal development, cellular maturation and differentiation, wound healing, and immune regulation. It maintains immune homeostasis by acting as a potent immune suppressor through inhibition of proliferation, differentiation, activation, and effector function of immune cells. Paradoxically, depending on the context, it displays proinflammatory properties by being a potent chemoattractant for neutrophils and promoting inflammation. In addition, it does not only induce differentiation into the anti-inflammatory Treg cells, but also into the proinflammatory Th17 and Th9 cells and inhibits Th22 differentiation. TGF-β has been demonstrated to be involved in multiple pathologies. In infections, it protects against collateral damages caused by the immune system, but it also promotes immune evasion and chronic infections. In autoimmune diseases, a TGF-β dysfunction leads to the loss of tolerance to self-antigens. In cancer, TGF-β is a potent inhibitor of cell proliferation and acts as a tumor suppressor at the beginning of tumorogenesis. However, once the cells become resistant to TGF-β, it mainly supports tumor growth and metastasis by promoting immune evasion and angiogenesis. In asthma, it is assumed to promote allergen tolerance, but plays a detrimental role in irreversible remodeling of the airways. Despite the high numbers of TGF-β-targeted pathways, it is a promising drug target for treatment of autoimmunity, cancer, fibrosis, if cell specificity can be achieved.This review summarizes the progresses that have been accomplished on the understanding of TGF-β's signaling in the immune homeostasis and its role in pathogenesis.
Collapse
Affiliation(s)
- Pierre-Yves Mantel
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA, USA.
| | | |
Collapse
|
25
|
Anti-inflammatory defense mechanisms of Entamoeba histolytica. Inflamm Res 2010; 60:111-7. [PMID: 20938711 DOI: 10.1007/s00011-010-0261-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 09/02/2010] [Accepted: 09/22/2010] [Indexed: 01/23/2023] Open
Abstract
The monocyte locomotion inhibitory factor (MLIF), a heat-stable oligopeptide found in the supernatant fluid of Entamoeba histolytica axenic cultures, may contribute to the delayed inflammation observed in amoebic hepatic abscess. This factor was isolated by ultra-filtration and high powered liquid chromatography, obtaining a primary Met-Gln-Cys-Asn-Ser structure, identified afterwards as the carboxyl-terminal (…Cys-Asn-Ser) active site. The selective anti-inflammatory effects of the pentapeptide have been observed in both in vitro and in vivo models, using a synthetic pentapeptide to maintain the same anti-inflammatory conditions during the experimental assays. Anti-inflammatory effects observed include inhibition of human monocyte locomotion and the respiratory burst in monocytes and neutrophils, increasing expression of anti-inflammatory cytokines and inhibiting expression of the adhesion molecules VLA-4 and VCAM, among others. In this review, we will describe the effects of MLIF detected so far and how it might be used as a therapeutical agent against inflammatory diseases.
Collapse
|
26
|
Gregory LG, Mathie SA, Walker SA, Pegorier S, Jones CP, Lloyd CM. Overexpression of Smad2 drives house dust mite-mediated airway remodeling and airway hyperresponsiveness via activin and IL-25. Am J Respir Crit Care Med 2010; 182:143-54. [PMID: 20339149 DOI: 10.1164/rccm.200905-0725oc] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Airway hyperreactivity and remodeling are characteristic features of asthma. Interactions between the airway epithelium and environmental allergens are believed to be important in driving development of pathology, particularly because altered epithelial gene expression is common in individuals with asthma. OBJECTIVES To investigate the interactions between a modified airway epithelium and a common aeroallergen in vivo. METHODS We used an adenoviral vector to generate mice overexpressing the transforming growth factor-beta signaling molecule, Smad2, in the airway epithelium and exposed them to house dust mite (HDM) extract intranasally. MEASUREMENTS AND MAIN RESULTS Smad2 overexpression resulted in enhanced airway hyperreactivity after allergen challenge concomitant with changes in airway remodeling. Subepithelial collagen deposition was increased and smooth muscle hyperplasia was evident resulting in thickening of the airway smooth muscle layer. However, there was no increase in airway inflammation in mice given the Smad2 vector compared with the control vector. Enhanced airway hyperreactivity and remodeling did not correlate with elevated levels of Th2 cytokines, such as IL-13 or IL-4. However, mice overexpressing Smad2 in the airway epithelium showed significantly enhanced levels of IL-25 and activin A after HDM exposure. Blocking activin A with a neutralizing antibody prevented the increase in lung IL-25 and inhibited subsequent collagen deposition and also the enhanced airway hyperreactivity observed in the Smad2 overexpressing HDM-exposed mice. CONCLUSIONS Epithelial overexpression of Smad2 can specifically alter airway hyperreactivity and remodeling in response to an aeroallergen. Moreover, we have identified novel roles for IL-25 and activin A in driving airway hyperreactivity and remodeling.
Collapse
Affiliation(s)
- Lisa G Gregory
- Leukocyte Biology Section, NHLI, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | | | |
Collapse
|
27
|
Kreidl E, Oztürk D, Metzner T, Berger W, Grusch M. Activins and follistatins: Emerging roles in liver physiology and cancer. World J Hepatol 2009; 1:17-27. [PMID: 21160961 PMCID: PMC2999257 DOI: 10.4254/wjh.v1.i1.17] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 09/10/2009] [Accepted: 09/17/2009] [Indexed: 02/06/2023] Open
Abstract
Activins are secreted proteins belonging to the TGF-β family of signaling molecules. Activin signals are crucial for differentiation and regulation of cell proliferation and apoptosis in multiple tissues. Signal transduction by activins relies mainly on the Smad pathway, although the importance of crosstalk with additional pathways is increasingly being recognized. Activin signals are kept in balance by antagonists at multiple levels of the signaling cascade. Among these, follistatin and FLRG, two members of the emerging family of follistatin-like proteins, can bind secreted activins with high affinity, thereby blocking their access to cell surface-anchored activin receptors. In the liver, activin A is a major negative regulator of hepatocyte proliferation and can induce apoptosis. The functions of other activins expressed by hepatocytes have yet to be more clearly defined. Deregulated expression of activins and follistatin has been implicated in hepatic diseases including inflammation, fibrosis, liver failure and primary cancer. In particular, increased follistatin levels have been found in the circulation and in the tumor tissue of patients suffering from hepatocellular carcinoma as well as in animal models of liver cancer. It has been argued that up-regulation of follistatin protects neoplastic hepatocytes from activin-mediated growth inhibition and apoptosis. The use of follistatin as biomarker for liver tumor development is impeded, however, due to the presence of elevated follistatin levels already during preceding stages of liver disease. The current article summarizes our evolving understanding of the multi-faceted activities of activins and follistatins in liver physiology and cancer.
Collapse
Affiliation(s)
- Emanuel Kreidl
- Emanuel Kreidl, Deniz Öztürk, Thomas Metzner, Walter Berger, Michael Grusch, Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, Vienna A-1090, Austria
| | | | | | | | | |
Collapse
|
28
|
Role of activins and inducible nitric oxide in the pathogenesis of ectopic pregnancy in patients with or without Chlamydia trachomatis infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1493-503. [PMID: 19692623 DOI: 10.1128/cvi.00221-09] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Chlamydia trachomatis infection can lead to pelvic inflammatory disease, ectopic pregnancy (EP), infertility, and chronic pelvic pain in women. Activins and inducible nitric oxide synthase (iNOS) are produced by the human fallopian tube, and we speculate that tubal activins and iNOS may be involved in the immune response to C. trachomatis in humans and their pathological alteration may result in tubal pathology and the development of EP. Blood and fallopian tubes were collected from 14 women with EP. Sera were analyzed by enzyme-linked immunosorbent assay to detect antibodies against chlamydial heat shock protein 60 (chsp60) and the major outer membrane protein of C. trachomatis. Confirmation of C. trachomatis serology was made using the microimmunofluorescence test. The patients were classified into three groups according to their serological results, and immunohistochemistry and quantitative reverse transcription-PCR were performed to investigate the expression of candidate molecules by tubal epithelial cells among the three groups. This is the first study to show an increase in the expression of activin betaA subunit, type II receptors, follistatin, and iNOS within the human fallopian tube of EP patients who were serologically positive for C. trachomatis. A similar expression profile was observed in the fallopian tubes with detectable antibodies only against chsp60. These results were shown at the mRNA and protein levels. We suggest that tubal activin A, its type II receptors, follistatin, and NO could be involved in the microbial-mediated immune response within the fallopian tube, and their pathological expression may lead to tubal damage and the development of EP.
Collapse
|
29
|
Gressner OA, Lahme B, Siluschek M, Rehbein K, Weiskirchen R, Gressner AM. Intracrine signalling of activin A in hepatocytes upregulates connective tissue growth factor (CTGF/CCN2) expression. Liver Int 2008; 28:1207-16. [PMID: 18397232 DOI: 10.1111/j.1478-3231.2008.01729.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND/AIMS Up to now, the effect of activin A on the expression of the important transforming growth factor (TGF)-beta downstream modulator connective tissue growth factor (CTGF) is not known, but might be of relevance for the functional effects of this cytokine on several liver cell types. METHODS In this study, activin A-dependent CTGF expression in hepatocytes (PC) primed by exogenous activin A and in PC maintained under complete activin-free culture conditions was analysed by Western blots, metabolic labelling, gene silencing, reverse transcriptase-polymerase chain reaction (RT-PCR) and CTGF reporter gene assays. This study was supplemented by immunocytochemical staining of activin A and CTGF in PC of injured liver. RESULTS Using alkaline phosphatase alpha-alkaline phosphatase staining, it is demonstrated that activin A becomes increasingly detectable during the course of CCl(4)-liver damage. Addition of activin A to cultured PC induced CTGF protein expression via phosphorylation of Smad2 and Smad3. This induction can be inhibited by the antagonist follistatin and alpha-activin A antibody respectively. When PC were cultured under serum(i.e. activin A)-free culture conditions, a time-dependent increase of activin expression during the course of the culture was proven by RT-PCR. Silencing of inhibin beta(A) gene expression under serum-free conditions by small interfering RNAs greatly suppressed CTGF synthesis and the phosphorylations of Smad2 and Smad3. However, both the extracellularly acting follistatin and the alpha-activin A antibody could not inhibit spontaneous CTGF expression, which, however, was achieved by the cell-permeable TGF-beta Alk4/Alk5 receptor-kinase-inhibitor SB431542. CONCLUSIONS In conclusion, the results point to activin A as an inducer of CTGF synthesis in PC. Intracellular activin A contributes to spontaneous CTGF expression in PC independent of exogenous activin A, which is proposed to occur via Alk4/Alk5-receptors. The findings might be important for many actions of activin A on the liver.
Collapse
Affiliation(s)
- Olav A Gressner
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH-University Hospital, Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
30
|
Keophiphath M, Achard V, Henegar C, Rouault C, Clément K, Lacasa D. Macrophage-secreted factors promote a profibrotic phenotype in human preadipocytes. Mol Endocrinol 2008; 23:11-24. [PMID: 18945811 DOI: 10.1210/me.2008-0183] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
White adipose tissue (WAT) in obese humans is characterized by macrophage accumulation the effects of which on WAT biology are not fully understood. We previously demonstrated that macrophage-secreted factors impair preadipocyte differentiation and induce inflammation, and we described the excessive fibrotic deposition in WAT from obese individuals. Microarray analysis revealed significant overexpression of extracellular matrix (ECM) genes in inflammatory preadipocytes. We show here an organized deposition of fibronectin, collagen I, and tenascin-C and clustering of the ECM receptor alpha5 integrin, characterizing inflammatory preadipocytes. Anti-alpha5 integrin-neutralizing antibody decreased proliferation of these cells, underlining the importance of the fibronectin/integrin partnership. Fibronectin-cultured preadipocytes exhibited increased proliferation and expression of both nuclear factor-kappaB and cyclin D1. Small interfering RNA deletion of nuclear factor-kappaB and cyclin D1 showed that these factors link preadipocyte proliferation with inflammation and ECM remodeling. Macrophage-secreted molecules increased preadipocyte migration through an increase in active/phosphorylated focal adhesion kinase. Gene expression and neutralizing antibody experiments suggest that inhibin beta A, a TGF-beta family member, is a major fibrotic factor. Interactions between preadipocytes and macrophages were favored in a three-dimensional collagen I matrix mimicking the fibrotic context of WAT. Cell-rich regions were immunostained for preadipocytes, proliferation, and macrophages in the vicinity of fibrotic WAT from obese individuals. In conclusion, an inflammatory environment leads to profound modifications of the human preadipocyte phenotype, producing fibrotic components with increased migration and proliferation. This phenomenon might play a role in facilitating the constitution of quiescent preadipocyte pools and eventually in the maintenance and aggravation of increased fat mass in obesity.
Collapse
Affiliation(s)
- Mayoura Keophiphath
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 872, Centre de recherche des Cordeliers, 15 rue de l'école de Médecine, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
31
|
Neuman MG, Sha K, Esguerra R, Zakhari S, Winkler RE, Hilzenrat N, Wyse J, Cooper CL, Seth D, Gorrell MD, Haber PS, McCaughan GW, Leo MA, Lieber CS, Voiculescu M, Buzatu E, Ionescu C, Dudas J, Saile B, Ramadori G. Inflammation and repair in viral hepatitis C. Dig Dis Sci 2008; 53:1468-87. [PMID: 17994278 DOI: 10.1007/s10620-007-0047-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 09/26/2007] [Indexed: 02/07/2023]
Abstract
Hepatitis C viral infection (HCV) results in liver damage leading to inflammation and fibrosis of the liver and increasing rates of hepatic decompensation and hepatocellular carcinoma (HCC). However, the host's immune response and viral determinants of liver disease progression are poorly understood. This review will address the determinants of liver injury in chronic HCV infection and the risk factors leading to rapid disease progression. We aim to better understand the factors that distinguish a relatively benign course of HCV from one with progression to cirrhosis. We will accomplish this task by discussion of three topics: (1) the role of cytokines in the adaptive immune response against the HCV infection; (2) the progression of fibrosis; and (3) the risk factors of co-morbidity with alcohol and human immunodeficiency virus (HIV) in HCV-infected individuals. Despite recent improvements in treating HCV infection using pegylated interferon alpha (PEGIFN-alpha) and ribavirin, about half of individuals infected with some genotypes, for example genotypes 1 and 4, will not respond to treatment or cannot be treated because of contraindications. This review will also aim to describe the importance of IFN-alpha-based therapies in HCV infection, ways of monitoring them, and associated complications.
Collapse
Affiliation(s)
- Manuela G Neuman
- In Vitro Drug Safety and Biotechnology, Department of Pharmacology, Biophysics and Global Health, Institute of Drug Research, University of Toronto, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Deli A, Kreidl E, Santifaller S, Trotter B, Seir K, Berger W, Schulte-Hermann R, Rodgarkia-Dara C, Grusch M. Activins and activin antagonists in hepatocellular carcinoma. World J Gastroenterol 2008; 14:1699-709. [PMID: 18350601 PMCID: PMC2695910 DOI: 10.3748/wjg.14.1699] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In many parts of the world hepatocellular carcinoma (HCC) is among the leading causes of cancer-related mortality but the underlying molecular pathology is still insufficiently understood. There is increasing evidence that activins, which are members of the transforming growth factor β (TGFβ) superfamily of growth and differentiation factors, could play important roles in liver carcinogenesis. Activins are disulphide-linked homo- or heterodimers formed from four different β subunits termed βA, βB, βC, and βE, respectively. Activin A, the dimer of two βA subunits, is critically involved in the regulation of cell growth, apoptosis, and tissue architecture in the liver, while the hepatic function of other activins is largely unexplored so far. Negative regulators of activin signals include antagonists in the extracellular space like the binding proteins follistatin and FLRG, and at the cell membrane antagonistic co-receptors like Cripto or BAMBI. Additionally, in the intracellular space inhibitory Smads can modulate and control activin activity. Accumulating data suggest that deregulation of activin signals contributes to pathologic conditions such as chronic inflammation, fibrosis and development of cancer. The current article reviews the alterations in components of the activin signaling pathway that have been observed in HCC and discusses their potential significance for liver tumorigenesis.
Collapse
|
33
|
Zhang HJ, Liu ZH, Chen FF, Ma D, Zhou J, Tai GX. Activin receptor-interacting protein 2 expression and its biological function in mouse hepatocytes. Shijie Huaren Xiaohua Zazhi 2008; 16:350-355. [DOI: 10.11569/wcjd.v16.i4.350] [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] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the activin receptor-interacting protein 2 (ARIP2) expression and its biological function in hepatocytes.
METHODS: Expression of ARIP2 in mouse liver tissue and hepatoma cell line Hepal-6 cells was detected by Western blot, immunohistochemistry and cytochemical staining. Effect of ARIP2 on activin-induced gene transcription was analyzed using CAGA-lux plasmid. Effect of over-expression of ARIP2 on the proliferation of Hepal-6 cells was assayed with MTT method.
RESULTS: ARIP2 was expressed in mouse liver tissue and Hepal-6 cells. The expression of ARIP2 in activin A-stimulated Hepal-6 cells was increased in a time-dependent manner, and peaked at 24 h. There was a significant difference in the expression level of ARIP2 on Hepal-6 cells at 12 and 24 h in contrast with the control group (1.01 ± 0.16, 1.62 ± 0.26 vs 0.82 ± 0.11, P < 0.05, P < 0.01). pcDNA3-ARIP2-transfected Hepal-6 cells obviously suppressed the gene transcription induced by activin A. MTT assay displayed that activin A (5 μg/L and 10 μg/L) remarkably inhibited the proliferation of Hepal-6 cells, the A570 nm value was 1.59 ± 0.03 and 1.49 ± 0.04 vs 1.79±0.07, respectively (P < 0.05, P < 0.01). ARIP2 over-expression in Hepal-6 cells significantly blocked the inhibitory effects of activin A (5 μg/L and 10 μg/L) on the proliferation of Hepal-6 cells.
CONCLUSION: ARIP2 can be expected to become a regulation target of genes in treatment of liver injury induced by activin.
Collapse
|
34
|
McLean CA, Cleland H, Moncrieff NJ, Barton RJ, de Kretser DM, Phillips DJ. Temporal expression of activin in acute burn wounds—From inflammatory cells to fibroblasts. Burns 2008; 34:50-5. [PMID: 17644256 DOI: 10.1016/j.burns.2007.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Accepted: 01/29/2007] [Indexed: 11/24/2022]
Abstract
Activin A is a member of the transforming growth factor-beta (TGF-beta) family of cytokines and growth factors and upregulation of this protein has been linked with a number of disease processes associated with chronic inflammation and fibrosis. Its potential involvement in burns has not yet been investigated. We therefore studied the localization of activin in tissue sections from excised mid- and deep dermal and full thickness cutaneous burn by immunohistochemistry. There was cell-specific temporal expression in tissues with prominent expression from day 4 onwards in lymphocytes and histiocytes and expression from day 8 onwards in reactive fibroblasts and endothelial cells. Immunopositivity over the first 18 days persisted in reactive fibroblasts and lymphocytes although the latter were in most circumstances decreasing in number. These data are consistent with activin A being central to the inflammatory and repair phases occurring in burnt skin and early scar formation. Modulation of activin expression and actions may, therefore, be a target for the management of burns.
Collapse
Affiliation(s)
- Catriona A McLean
- Department of Anatomical Pathology, The Alfred Hospital, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
35
|
Fu K, Corbley MJ, Sun L, Friedman JE, Shan F, Papadatos JL, Costa D, Lutterodt F, Sweigard H, Bowes S, Choi M, Boriack-Sjodin PA, Arduini RM, Sun D, Newman MN, Zhang X, Mead JN, Chuaqui CE, Cheung HK, Zhang X, Cornebise M, Carter MB, Josiah S, Singh J, Lee WC, Gill A, Ling LE. SM16, an orally active TGF-beta type I receptor inhibitor prevents myofibroblast induction and vascular fibrosis in the rat carotid injury model. Arterioscler Thromb Vasc Biol 2008; 28:665-71. [PMID: 18202322 DOI: 10.1161/atvbaha.107.158030] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE TGF-beta plays a significant role in vascular injury-induced stenosis. This study evaluates the efficacy of a novel, small molecule inhibitor of ALK5/ALK4 kinase, in the rat carotid injury model of vascular fibrosis. METHODS AND RESULTS The small molecule, SM16, was shown to bind with high affinity to ALK5 kinase ATP binding site using a competitive binding assay and biacore analysis. SM16 blocked TGF-beta and activin-induced Smad2/3 phosphorylation and TGF-beta-induced plasminogen activator inhibitor (PAI)-luciferase activity in cells. Good overall selectivity was demonstrated in a large panel of kinase assays, but SM16 also showed nanomolar inhibition of ALK4 and weak (micromolar) inhibition of Raf and p38. In the rat carotid injury model, SM16 dosed once daily orally at 15 or 30 mg/kg SM16 for 14 days caused significant inhibition of neointimal thickening and lumenal narrowing. SM16 also prevented induction of adventitial smooth muscle alpha-actin-positive myofibroblasts and the production of intimal collagen, but did not decrease the percentage of proliferative cells. CONCLUSIONS These results are the first to demonstrate the efficacy of an orally active, small-molecule ALK5/ALK4 inhibitor in a vascular fibrosis model and suggest the potential therapeutic application of these inhibitors in vascular fibrosis.
Collapse
Affiliation(s)
- Kai Fu
- Department of Pharmacology, Biogen Idec, 14 Cambridge Center, Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fumagalli M, Musso T, Vermi W, Scutera S, Daniele R, Alotto D, Cambieri I, Ostorero A, Gentili F, Caposio P, Zucca M, Sozzani S, Stella M, Castagnoli C. Imbalance between activin A and follistatin drives postburn hypertrophic scar formation in human skin. Exp Dermatol 2007; 16:600-10. [PMID: 17576240 DOI: 10.1111/j.1600-0625.2007.00571.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hypertrophic scarring is a skin disorder characterized by persistent inflammation and fibrosis that may occur after wounding or thermal injury. Altered production of cytokines and growth factors, such as TGF-beta, play an important role in this process. Activin A, a member of the TGF-beta family, shares the same intra-cellular Smad signalling pathway with TGF-beta, but binds to its own specific transmembrane receptors and to follistatin, a secreted protein that inhibits activin by sequestration. Recent studies provide evidences of a novel role of activin A in inflammatory and repair processes. The aim of this study was to evaluate the importance of activin A and follistatin expression in the different phases of scar evolution. Immunostaining of sections obtained from active phase hypertrophic scars (AHS) revealed the presence of a high number of alpha-SMA(+) myofibroblasts and DC-SIGN(+) dendritic cells coexpressing activin A. Ex-vivo AHS fibroblasts produced more activin and less follistatin than normal skin or remission phase hypertrophic scar (HS) fibroblasts, both in basal conditions and upon TGF-betas stimulation. We demonstrate that fibroblasts do express activin receptors, and that this expression is not affected by TGF-betas. Treatment of HS fibroblasts with activin A induced Akt phosphorylation, promoted cell proliferation, and enhanced alpha-SMA and type I collagen expression. Follistatin reduced proliferation and suppressed activin-induced collagen expression. These results indicate that the activin/follistatin interplay has a role in HS formation and evolution. The impact of these observations on the understanding of wound healing and on the identification of new therapeutic targets is discussed.
Collapse
Affiliation(s)
- Mara Fumagalli
- Department of Plastic Surgery and Burn Unit Skin Bank, CTO Hospital, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Park MK, Han C, Lee KH, Hong SH, Kim HS, Lee YJ, Jeong IK, Noh JH, Yang TY, Lee MS, Kim KW, Lee MK. Effects of activin A on pancreatic ductal cells in streptozotocin-induced diabetic rats. Transplantation 2007; 83:925-30. [PMID: 17460564 DOI: 10.1097/01.tp.0000259978.62139.9d] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The shortage of islets for transplantation has led to find alternative insulin producing cells. Pancreatic progenitor cells in the duct have the potential to grow and differentiate into endocrine cells. In this study, we examined whether activin A can promote the expansion and/or differentiation of ductal cells into insulin-producing cells. METHODS Pancreatic ductal cells were treated with activin A for differentiation into endocrine cells, and transplanted into the renal subcapsular space of streptozotocin (STZ)-induced diabetic rats. The identity of the endocrine cells was confirmed by immunostaining and analysis of the expression of transcription factors and endocrine genes by reverse-transcriptase polymerase chain reaction. RESULTS Activin A treatment significantly increased the DNA synthesis and the expression of insulin I, insulin II, PDX-1, Nkx 6.1, Glut-2, Pax-4, Pax-6, and Ngn-3. De novo synthesis of insulin in activin A-treated ductal cells was observed by the immunocytochemical detection of C-peptide and the differentiated ductal cells secreted significantly increased amount of insulin compared to nontreated ductal cells in response to glucose stimulation. When activin A-treated ductal cells were transplanted on STZ-induced diabetic rats, blood glucose levels were normalized and the removal of the transplanted kidney resulted in return to hyperglycemia. CONCLUSIONS The pancreatic ductal cells could be efficiently differentiated into insulin secreting cells by activin A treatment in vitro and normalize hyperglycemia in vivo.
Collapse
Affiliation(s)
- Mi-Kyung Park
- Division of Endocrinology and Metabolism, Samsung Biomedical Research Institute, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Allen JW, Khetani SR, Johnson RS, Bhatia SN. In vitro liver tissue model established from transgenic mice: role of HIF-1alpha on hypoxic gene expression. ACTA ACUST UNITED AC 2007; 12:3135-47. [PMID: 17518628 DOI: 10.1089/ten.2006.12.3135] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The instability of the hepatocyte phenotype in vitro has limited the ability to quantitatively investigate regulation of stress responses of the liver. Here, we adopt a tissue-engineering approach to form stable liver tissue in vitro by forming collagen "sandwich" cultures of transgenic murine hepatocytes harboring a regulatory gene of interest flanked by loxP sites. The floxed gene is excised in a subset of cultures by transfection with adenovirus carrying the gene for Cre-recombinase, thereby generating wild-type and null liver tissues from a single animal. In this study, we specifically investigated the role of hypoxia inducible factor 1 alpha (HIF-1alpha) in the hepatocellular response to hypoxia. Using high-density oligonucleotide arrays, we examined genome-wide gene expression after 8 h of hypoxia in wild-type and HIF- 1alpha null hepatocyte cultures. We identified more than 130 genes differentially expressed under hypoxia involved in metabolic adaptation, angiogenic signaling, immediate early response, and cell cycle regulation. Real-time polymerase chain reaction analysis verified that known hypoxia-responsive genes such as glucose transporter-1 and vascular endothelial growth factor were induced in a HIF-1alpha-dependent manner under hypoxia. Our results demonstrate the potential to integrate in vitro tissue models with transgenic and microarray technologies for the study of physiologic stress responses.
Collapse
Affiliation(s)
- Jared W Allen
- Department of Bioengineering, University of California at San Diego, La Jolla, California, USA
| | | | | | | |
Collapse
|
39
|
Hattori S, Dhar DK, Hara N, Tonomoto Y, Onoda T, Ono T, Yamanoi A, Tachibana M, Tsuchiya M, Nagasue N. FR-167653, a selective p38 MAPK inhibitor, exerts salutary effect on liver cirrhosis through downregulation of Runx2. J Transl Med 2007; 87:591-601. [PMID: 17334410 DOI: 10.1038/labinvest.3700539] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Liver cirrhosis remains a difficult-to-treat disease with a substantial morbidity and mortality rate. There is an emerging body of data purporting a pivotal role of the activated p38 mitogen-activated protein kinase (MAPK) in the process of cirrhosis. Several anticirrhotic agents have been developed over the past few years, and most of them exert their effects by indirectly inhibiting the p38 pathway. Effect of a selective p38 inhibitor is yet to be reported. In this study, we evaluated the salutary effect of FR-167653 (FR), a selective p38 inhibitor, in a carbon tetrachloride (CCl(4))-induced rat cirrhotic model. Twenty rats were assigned into four groups: Sham, olive oil only; Control, CCl(4) in olive oil; FR50, FR 50 mg/kg/day and CCl(4); and FR100, FR 100 mg/kg/day and CCl(4). FR dose-dependently inhibited activation of p38 and had an ameliorating effect on cirrhosis formation. Significant dose-dependent reduction in alpha-smooth muscle actin immunostaining and hydroxyproline content of the liver was noticed in the FR-treated rats. Also densitometric analysis showed a significant reduction in azan-stained area in the FR-treated rats. These fibrotic changes were observed in the myofibroblasts including the hepatic stellate cells and portal fibroblasts. mRNA expression of runt-related protein 2 (Runx2), a profibrogenic transcription factor, was significantly low in FR-treated livers, indicating that Runx2 might be a key downstream regulator of the p38 pathway. A similar reduction in expression of Smad4 and tissue inhibitor of metalloproteinase-1 was noticed in the FR-treated rats. In conclusion, FR treatment exerted a significant beneficial effect in a CCl(4)-induced rat cirrhotic model. The ameliorating effect of FR could be partially attributable to an inhibition of the Smad4/p38/Runx2 axis in the cirrhotic liver.
Collapse
Affiliation(s)
- Shinji Hattori
- Department of Digestive and General Surgery, Shimane University, Izumo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mukhopadhyay A, Chan SY, Lim IJ, Phillips DJ, Phan TT. The role of the activin system in keloid pathogenesis. Am J Physiol Cell Physiol 2007; 292:C1331-8. [PMID: 16971493 DOI: 10.1152/ajpcell.00373.2006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Keloid scars represent a pathological response to cutaneous injury under the regulation of many growth factors. Activin-A, a dimeric protein and a member of the transforming growth factor-β superfamily, has been shown to regulate various aspects of cell growth and differentiation in the repair of the skin mesenchyme and the epidermis. Thus our aim was to study the role of activin and its antagonist, follistatin, in keloid pathogenesis. Increased mRNA expression for activin was observed in keloid scar tissue by performing RNase protection assay. Immunohistochemistry showed increased localization of both activin-A and follistatin in the basal layer of epidermis of keloid tissue compared with normal tissue. ELISA demonstrated a 29-fold increase in concentration of activin-A and an ∼5-fold increase in follistatin in conditioned media in keloid fibroblasts compared with normal fibroblasts. Although keloid keratinocytes produced 25% more follistatin than normal keratinocytes, the amounts of activin-A, in contrast, was ∼77% lower. Proliferation of fibroblasts was stimulated when treated with exogenous activin-A (46% increase in keloids fibroblasts) or following co-culture with hβAHaCaT cells (66% increase). Activin-A upregulated key extracellular matrix components, namely collagen, fibronectin, and α-smooth muscle actin, in normal and keloid fibroblasts. Co-treatment of follistatin with activin-A blocked the stimulatory effects of activin on extracellular matrix components. These findings emphasize the importance of the activin system in keloid biology and pathogenesis and suggest a possible therapeutic potential of follistatin in the prevention and treatment of keloids.
Collapse
|
41
|
Andreev K, Zenkel M, Kruse F, Jünemann A, Schlötzer-Schrehardt U. Expression of bone morphogenetic proteins (BMPs), their receptors, and activins in normal and scarred conjunctiva: Role of BMP-6 and activin-A in conjunctival scarring? Exp Eye Res 2006; 83:1162-70. [PMID: 16879818 DOI: 10.1016/j.exer.2006.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 05/19/2006] [Accepted: 06/02/2006] [Indexed: 11/30/2022]
Abstract
Bone morphogenetic proteins (BMPs) and activins are multifunctional growth factors, which also affect wound healing and tissue fibrosis. To determine their putative role in conjunctival wound healing and scarring, we investigated the expression of various BMPs, BMP receptors, and activins in normal and scarred human conjunctival tissue and in cultured human Tenon's capsule fibroblasts on the mRNA and protein level. Messenger RNA expression of BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, the BMP receptors type I (BMPR-IA, BMPR-IB) and II (BMPR-II), and of activin A and B was investigated by semi-quantitative RT-PCR in normal conjunctival specimens and in scarred filtering blebs as well as in cultured Tenon's capsule fibroblasts obtained from patients with primary open-angle glaucoma (POAG), pseudoexfoliation (PEX) glaucoma and cataract. Immunohistochemistry was used to study the protein expression of BMP-2, BMP-4, BMP-6, BMP-7, and activin A in normal and scarred conjunctival tissue as well as in cultured Tenon's capsule fibroblasts. BMP-2, BMP-3, BMP-4, BMP-6, BMP-7, all BMP receptors, and activin A were expressed on the mRNA and protein level in conjunctival biopsies without showing any differences between groups of patients. The mRNA and protein expression of both BMP-6 and activin A was found to be significantly increased in scar tissue compared with normal conjunctiva and could be immunolocalized to epithelial cells, vascular endothelia, stromal fibroblasts, and macrophage-like cells. However, no significant increase in receptor gene expression was observed in scar tissue. With the exception of BMP-7, all growth factors and receptors were also expressed in cultured Tenon's fibroblasts without showing any differences between cultures derived from normal and scarred conjunctival specimens. These findings suggest various BMPs and activin A as components of the conjunctival cytokine meshwork regulating tissue homeostasis and wound healing and provide evidence that alterations in the expression of BMP-6 and activin A, in particular, are associated with conjunctival scarring. Modulation of BMP/activin activities may, therefore, be explored as new approaches for managing postoperative conjunctival scarring responses in glaucoma patients.
Collapse
|
42
|
Rodgarkia-Dara C, Vejda S, Erlach N, Losert A, Bursch W, Berger W, Schulte-Hermann R, Grusch M. The activin axis in liver biology and disease. Mutat Res 2006; 613:123-37. [PMID: 16997617 DOI: 10.1016/j.mrrev.2006.07.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 07/27/2006] [Accepted: 07/27/2006] [Indexed: 12/22/2022]
Abstract
Activins are a closely related subgroup within the TGFbeta superfamily of growth and differentiation factors. They consist of two disulfide-linked beta subunits. Four mammalian activin beta subunits termed beta(A), beta(B), beta(C), and beta(E), respectively, have been identified. Activin A, the homodimer of two beta(A) subunits, has important regulatory functions in reproductive biology, embryonic development, inflammation, and tissue repair. Several intra- and extracellular antagonists, including the activin-binding proteins follistatin and follistatin-related protein, serve to fine-tune activin A activity. In the liver there is compelling evidence that activin A is involved in the regulation of cell number by inhibition of hepatocyte replication and induction of apoptosis. In addition, activin A stimulates extracellular matrix production in hepatic stellate cells and tubulogenesis of sinusoidal endothelial cells, and thus contributes to restoration of tissue architecture during liver regeneration. Accumulating evidence from animal models and from patient data suggests that deregulation of activin A signaling contributes to pathologic conditions such as hepatic inflammation and fibrosis, acute liver failure, and development of liver cancer. Increased production of activin A was suggested to be a contributing factor to impaired hepatocyte regeneration in acute liver failure and to overproduction of extracellular matrix in liver fibrosis. Recent evidence suggests that escape of (pre)neoplastic hepatocytes from growth control by activin A through overexpression of follistatin and reduced activin production contributes to hepatocarcinogenesis. The role of the activin subunits beta(C) and beta(E), which are both highly expressed in hepatocytes, is still quite incompletely understood. Down-regulation in liver tumors and a growth inhibitory function similar to that of beta(A) has been shown for beta(E). Contradictory results with regard to cell proliferation have been reported for beta(C). The profound involvement of the activin axis in liver biology and in the pathogenesis of severe hepatic diseases suggests activin as potential target for therapeutic interventions.
Collapse
Affiliation(s)
- Chantal Rodgarkia-Dara
- Department of Medicine I, Division: Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Schmidt-Weber CB, Blaser K. The role of TGF-beta in allergic inflammation. Immunol Allergy Clin North Am 2006; 26:233-44, vi-vii. [PMID: 16701142 DOI: 10.1016/j.iac.2006.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The transforming growth factor beta (TGF-beta) plays a dual role in allergic disease. It is important in suppressing T cells and also mediates repair responses that lead to unwanted remodeling of tissues. Advances in the immunology of allergy indicate that allergens cause overreactions in the lymphocyte compartment because of the lack or decreased number of suppressive, regulatory T cells. TGF-beta was shown to induce regulatory T cells and participate directly in suppression of effector T cells. Therefore, TGF-beta may help return reactivity to allergens to normal subsymptomatic activity. Whether chronic inflammatory diseases such as asthma profit from TGF-beta-mediated suppression of specific immune responses or whether the TGF-beta-mediated tissue remodeling aggravates diseases more than it helps control immune reactions is unclear. This article addresses these issues and future strategies in this field.
Collapse
Affiliation(s)
- Carsten B Schmidt-Weber
- Swiss Institute of Allergy and Asthma Research (SIAF), Obere Strasse 22, CH-7270 Davos, Switzerland.
| | | |
Collapse
|
44
|
Shi ZH, Liu H, Liu S, Zhang JM, Tu JW. Roles of activin A and hepatocellular apoptosis in the anti-liver fibrosis process induced by Ginkgo biloba extract in rats. Shijie Huaren Xiaohua Zazhi 2006; 14:2060-2066. [DOI: 10.11569/wcjd.v14.i21.2060] [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] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Ginkgo biloba extract (GBE) on CCl4-induced liver fibrosis as well as the underlying mechanisms.
METHODS: Thirty adult male Sprague Dawley rats were randomized into 3 groups: control group (n = 10), model group (n = 10) and treatment group (n = 10). Except the rats in the control group, the others were intraperitoneally injected with 500 mL/L CCl4 (1 mL/kg) to induce liver cirrhosis (twice a week, for 8 weeks). Moreover, the rats in treatment group were intragastrically administered with GBE (0.4 g/kg) for 8 weeks. At the end of the 8th week, all the rats were sacrificed. Blood samples were collected for the determination of biochemical indicators. Tissue samples were used for histopathological examinations. The expression of activin A was determined by immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR). Hepatocellular apoptosis was determined by the method of terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) method.
RESULTS: The grade of liver fibrosis in treatment group was lower than that in the model group (P < 0.05). The serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and albumin (ALB) in treatment group were significantly improved as compared with those in the model group (ALT: 2806.9 ±576.1 nkat/L vs 4452.9 ± 709.5 nkat/L; AST: 5314.2 ± 1042 nkat/L vs 15 743.4 ± 625.8 nkat/L; ALB: 31.0 ± 2.1 g/L vs 21.7 ± 1.8 g/L; all P < 0.05). GBE treatment markedly reduced mRNA and protein levels of activin A (mRNA: 0.42 ± 0.09 vs 0.78 ± 0.15; protein: 4.2 ± 0.8 vs 11.4 ± 1.2; both P < 0.01). In comparison with that in the model group, the apoptosis index was decreased in treatment groups (7.56 ± 3.36 vs 16.06 ± 8.84, P < 0.01).
CONCLUSION: GBE can markedly attenuate the degrees of hepatic fibrosis, and the mechanism may be correlated with the expression of activin A and hepatocellular apoptosis.
Collapse
|
45
|
Elsammak MY, Amin GM, Khalil GM, Ragab WS, Abaza MM. Possible contribution of serum activin A and IGF-1 in the development of hepatocellular carcinoma in Egyptian patients suffering from combined hepatitis C virus infection and hepatic schistosomiasis. Clin Biochem 2006; 39:623-9. [PMID: 16624274 DOI: 10.1016/j.clinbiochem.2006.01.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 12/06/2005] [Accepted: 01/13/2006] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The present study evaluated the role of activin A, insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein 3 (IGFBP-3) in Egyptian patients suffering from combined hepatitis C virus (HCV) infection and hepatic schistosomiasis. DESIGN AND METHODS Four groups were included in the present study. Group I: 30 healthy subjects were included as controls; Group II (HCV): 30 patients with chronic liver disease due to HCV infection without evidence of schistosomiasis; Group III (SHF + HCV): 30 patients with combined disease, chronic schistosomal hepatic fibrosis (SHF) and chronic hepatitis C infection; Group IV (HCC): 30 patients with hepatocellular carcinoma associated with chronic hepatitis C virus and schistosomal infection. RESULTS Patients with HCV, HCV + SHF and those with HCC had a significantly higher serum activin A compared with the control group (P < 0.001). Serum activin A level (mean +/- SD) was 5.7 +/- 2.76, 10.59 +/- 3.59, 15.39 +/- 4.61 and 19.93 +/- 5.43 ng/mL in controls, HCV patients, HCV + SHF patients and HCC patients, respectively. Serum IGF-1 was significantly lower in HCV patients, HCV + SHF patients and HCC patients compared to the control group (P < 0.001). Serum IGF-1 was 121.7 +/- 73.4, 76.7 +/- 23.5, 35.7 +/- 17.6 and 39.9 +/- 25.9 ng/mL in controls, HCV patients, HCV + SHF patients and HCC patients, respectively. Similarly, serum IGFBP-3 was significantly lower in HCV patients, HCV + SHF patients and HCC patients compared to the control group (P < 0.001). Furthermore, serum insulin-like growth factor binding protein 3 (IGFBP-3) was significantly lower in patients with HCC compared to patients with HCV or those with HCV + SHF (P < 0.01 and P = 0.024, respectively). The median (range) of serum IGFBP-3 was 4452 (352.2-8965), 3457 (1114-6000), 2114 (867-5901) and 1202 (576-3994) ng/mL in controls, HCV patients, HCV + SHF patients and HCC patients, respectively. Serum activin A correlated positively with Child-Pugh scoring in patients with HCV, HCV + SHF and those with HCC. The correlation coefficient was significant, at 0.001, in total cases. CONCLUSIONS We conclude that patients with HCV, HCV + SHF and those with HCC have a significantly higher serum activin A when compared with controls. Serum activin A level was significantly higher in patients with HCV + SHF compared to those with HCV alone (P < 0.01) with a significant positive correlation between the serum activin A level and Child-Pugh scoring in patients with HCV, HCV + SHF and those with HCC. Furthermore, serum IGF-1 and IGFBP-3 levels were significantly reduced in patients with HCV, HCV + SHF and those with HCC compared to the control group. We suggest that this pattern (high activin A and low IGF-1 and its binding protein 3) may play a role in development of HCC in Egyptian patients suffering from combined hepatitis C virus infection and hepatic schistosomiasis.
Collapse
Affiliation(s)
- Mohamed Yousry Elsammak
- Department of Chemical Pathology, Medical Research Instituitre, Alexandria University, Alexandria, Egypt.
| | | | | | | | | |
Collapse
|
46
|
Patella S, Phillips DJ, Tchongue J, de Kretser DM, Sievert W. Follistatin attenuates early liver fibrosis: effects on hepatic stellate cell activation and hepatocyte apoptosis. Am J Physiol Gastrointest Liver Physiol 2006; 290:G137-44. [PMID: 16123203 DOI: 10.1152/ajpgi.00080.2005] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Activin A, a member of the transforming growth factor-beta superfamily, is constitutively expressed in hepatocytes and regulates liver mass through tonic inhibition of hepatocyte DNA synthesis. Follistatin is the main biological inhibitor of activin bioactivity. These molecules may be involved in hepatic fibrogenesis, although defined roles remain unclear. We studied activin and follistatin gene and protein expression in cultured rat hepatic stellate cells (HSCs) and in rats given CCl4 for 8 wk and examined the effect of follistatin administration on the development of hepatic fibrosis. In activated HSCs, activin mRNA was upregulated with high expression levels, whereas follistatin mRNA expression was unchanged from baseline. Activin A expression in normal lobular hepatocytes redistributed to periseptal hepatocytes and smooth muscle actin-positive HSCs in the fibrotic liver. A 32% reduction in fibrosis, maximal at week 4, occurred in CCl4-exposed rats treated with follistatin. Hepatocyte apoptosis decreased by 87% and was maximal at week 4 during follistatin treatment. In conclusion, activin is produced by activated HSCs in vitro and in vivo. Absence of simultaneous upregulation of follistatin gene expression in HSCs suggests that HSC-derived activin is biologically active and unopposed by follistatin. Our in vivo and in vitro results demonstrate that activin-mediated events contribute to hepatic fibrogenesis and that follistatin attenuates early events in fibrogenesis by constraining HSC proliferation and inhibiting hepatocyte apoptosis.
Collapse
Affiliation(s)
- Shane Patella
- Centre for Inflammatory Diseases, Monash Institute of Medical Research, Monash University, Melbourne, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
47
|
Gaedeke J, Boehler T, Budde K, Neumayer HH, Peters H. Glomerular activin A overexpression is linked to fibrosis in anti-Thy1 glomerulonephritis. Nephrol Dial Transplant 2005; 20:319-28. [PMID: 15673690 DOI: 10.1093/ndt/gfh653] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Activin A, a member of the transforming growth factor-beta (TGF-beta) superfamily of proteins, shares many biological features with the pro-fibrotic cytokine TGF-beta1, which is primarily responsible for the accumulation of extracellular matrix proteins in renal disease. This study was designed to identify regulators of activin A production in glomerular mesangial cells and test if activin A acts as a pro-fibrotic cytokine in mesangial cells. METHODS The effect of inflammatory cytokines on activin A production and the effect of exogenous activin A on mediators of fibrosis were analysed in cultured rat mesangial cells. Expression of activin A and of established mediators of fibrosis was analysed in a rat model of glomerular fibrosis (anti-Thy1 glomerulonephritis). RESULTS In cultured mesangial cells, interleukin-1 and basic fibroblast growth factor, both mediators of glomerular inflammatory injury, dose-dependently increased activin A expression. Incubation with activin A significantly stimulated TGF-beta1, PAI-1 and connective tissue growth factor RNA expression and increased production of extracellular matrix proteins in mesangial cells. In rats with anti-Thy1 glomerulonephritis, expression of glomerular activin A mRNA and protein paralled the expression of TGF-beta and other indices of fibrosis, showing little change from normal on day 1, a marked, 70-fold increase of activin protein production on day 6, and a subsequent decrease at day 12. Antifibrotic therapy with the angiotensin-converting enzyme inhibitor enalapril significantly reduced glomerular activin A production. CONCLUSION Taken together, the results of this study link overexpression of activin A to glomerular matrix protein expansion in vivo and in vitro, suggesting that activin A acts as pro-fibrotic cytokine in renal disease.
Collapse
Affiliation(s)
- Jens Gaedeke
- Department of Nephrology, Universitätsklinikum Charité, Berlin, Germany.
| | | | | | | | | |
Collapse
|
48
|
Schmidt-Weber CB, Blaser K. Regulation and role of transforming growth factor-beta in immune tolerance induction and inflammation. Curr Opin Immunol 2005; 16:709-16. [PMID: 15511662 DOI: 10.1016/j.coi.2004.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is known to mediate pleiotropic functions both inside and outside the immune system. Recent progress in this field underlines the role of TGF-beta in regulatory T (Treg) cells, where it participates in both suppression and differentiation. In addition, recent information highlights the role of TGF-beta in repair responses that lead to matrix deposition and tissue remodelling. Many chronic inflammatory diseases, such as asthma, profit from the suppression of specific immune responses by TGF-beta; however, TGF-beta-mediated tissue remodelling can be a serious complication in such diseases.
Collapse
Affiliation(s)
- Carsten B Schmidt-Weber
- Swiss Institute of Allergy and Asthma Research, Obere Strasse 22, CH-7270 Davos, Switzerland.
| | | |
Collapse
|
49
|
Abstract
Inflammation is a complex process regulated by a cascade of cytokines and growth factors. This review summarizes the emerging evidence implicating activin A and follistatin in the inflammatory process. Our recent studies have highlighted that activin A is released early in the process as part of the circulatory cytokine cascade during acute systemic inflammation. This release occurs concurrently with tumor necrosis factor (TNF)-alpha and prior to that of interleukin (IL)-6 and follistatin. Although, the cellular source(s) of activin A are yet to be established, circulating blood cells and the vascular endothelium are candidates for this rapid release of activin A into the circulation. The release of activin A and follistatin is also observed in the clinical setting, in particular in sepsis. Furthermore activin A is released into cerebrospinal fluid in a model of meningitis in rabbits. The role of activin A in the inflammatory response is poorly understood, however, in vitro data has highlighted that activin A can have both pro- and anti-inflammatory actions on key mediators of the inflammatory response such as TNF-alpha, IL-1beta and IL-6. Furthermore, emerging data would suggest that activin A induction is restricted to certain types of inflammation and its release is dependant upon the inflammatory setting.
Collapse
Affiliation(s)
- Kristian L Jones
- Center for Molecular Reproduction and Endocrinology, Monash Institute of Reproduction and Development, 27-31 Wright Street, Clayton 3168, Victoria, Australia
| | | | | | | |
Collapse
|
50
|
Marra F, Delogu W, Petrai I, Pastacaldi S, Bonacchi A, Efsen E, Aleffi S, Bertolani C, Pinzani M, Gentilini P. Differential requirement of members of the MAPK family for CCL2 expression by hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol 2004; 287:G18-26. [PMID: 15016614 DOI: 10.1152/ajpgi.00336.2003] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic stellate cells (HSC) coordinate the liver wound-healing response through secretion of several cytokines and chemokines, including CCL2 (formerly known as monocyte chemoattractant protein-1). In this study, we evaluated the role of different proteins of the MAPK family (ERK, p38(MAPK), and JNK) in the regulation of CCL2 expression by HSC, as an index of their proinflammatory activity. Several mediators activated all three MAPK, including TNF, IL-1, and PDGF. To assess the relative role of the different MAPKs, specific pharmacological inhibitors were used; namely, SB203580 (p38(MAPK)), SP600125 (JNK), and PD98059 (MEK/ERK). The efficacy and specificity of the different inhibitors in our cellular system were verified analyzing the enzymatic activity of the different MAPKs using in vitro kinase assays and/or testing the inhibition of phosphorylation of downstream substrates. SB203580 and SP600125 dose-dependently inhibited CCL2 secretion and gene expression induced by IL-1 or TNF. In contrast, inhibition of ERK did not affect the upregulation of CCL2 induced by the two cytokines. Finally, activin A was also found to stimulate CCL2 expression and to activate ERK, JNK, p38, and their downstream targets. Unlike in cells exposed to proinflammatory cytokines, all three MAPKs were required to induce CCL2 secretion in response to activin. We conclude that members of the MAPK family differentially regulate cytokine-induced chemokine expression in human HSC.
Collapse
Affiliation(s)
- Fabio Marra
- Dipartimento di Medicina Interna, University of Florence, Viale Morgagni 85, I-50137 Florence, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|