1
|
Chia ZJ, Kumarapperuma H, Zhang R, Little PJ, Kamato D. Smad transcription factors as mediators of 7 transmembrane G protein-coupled receptor signalling. Acta Pharmacol Sin 2025; 46:795-804. [PMID: 39506064 PMCID: PMC11950520 DOI: 10.1038/s41401-024-01413-6] [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: 07/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
The Smad transcription factors are well known for their role at the core of transforming growth factor-β (TGF-β) signalling. However, recent evidence shows that the Smad transcription factors play a vital role downstream of other classes of receptors including G protein-coupled receptors (GPCR). The versatility of Smad transcription factors originated from the two regions that can be differently activated by the TGF-β receptor superfamily or through the recruitment of intracellular kinases stimulated by other receptors classes such as GPCRs. The classic GPCR signalling cascade is further expanded to conditional adoption of the Smad transcription factor under the stimulation of Akt, demonstrating the unique involvement of the Smad transcription factor in GPCR signalling pathways in disease environments. In this review, we provide a summary of the signalling pathways of the Smad transcription factors as an important downstream mediator of GPCRs, presenting exciting opportunities for discovering new therapeutic targets for diseases.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hirushi Kumarapperuma
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Ruizhi Zhang
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
2
|
Singh P, Sun J, Cavalera M, Al-Sharify D, Matthes F, Barghouth M, Tengryd C, Dunér P, Persson A, Sundius L, Nitulescu M, Bengtsson E, Rattik S, Engelbertsen D, Orho-Melander M, Nilsson J, Monaco C, Goncalves I, Edsfeldt A. Dysregulation of MMP2-dependent TGF-ß2 activation impairs fibrous cap formation in type 2 diabetes-associated atherosclerosis. Nat Commun 2024; 15:10464. [PMID: 39653743 PMCID: PMC11628557 DOI: 10.1038/s41467-024-50753-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/18/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 diabetes is associated with cardiovascular disease, possibly due to impaired vascular fibrous repair. Yet, the mechanisms are elusive. Here, we investigate alterations in the fibrous repair processes in type 2 diabetes atherosclerotic plaque extracellular matrix by combining multi-omics from the human Carotid Plaque Imaging Project cohort and functional studies. Plaques from type 2 diabetes patients have less collagen. Interestingly, lower levels of transforming growth factor-ß distinguish type 2 diabetes plaques and, in these patients, lower levels of fibrous repair markers are associated with cardiovascular events. Transforming growth factor-ß2 originates mostly from contractile vascular smooth muscle cells that interact with synthetic vascular smooth muscle cells in the cap, leading to collagen formation and vascular smooth muscle cell differentiation. This is regulated by free transforming growth factor-ß2 which is affected by hyperglycemia. Our findings underscore the importance of transforming growth factor-ß2-driven fibrous repair in type 2 diabetes as an area for future therapeutic strategies.
Collapse
MESH Headings
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Humans
- Transforming Growth Factor beta2/metabolism
- Matrix Metalloproteinase 2/metabolism
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Female
- Animals
- Middle Aged
- Aged
- Collagen/metabolism
- Extracellular Matrix/metabolism
- Mice
- Cell Differentiation
Collapse
Affiliation(s)
- Pratibha Singh
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Jiangming Sun
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Michele Cavalera
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Dania Al-Sharify
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Frank Matthes
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Mohammad Barghouth
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Christoffer Tengryd
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Ana Persson
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Lena Sundius
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Mihaela Nitulescu
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Biomedical Science, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Sara Rattik
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Isabel Goncalves
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden
- Department of Cardiology, University Hospital of Skåne, Lund/Malmö, Sweden
| | - Andreas Edsfeldt
- Cardiovascular Research-Translational Studies, Lund University, Malmö, Sweden.
- Department of Cardiology, University Hospital of Skåne, Lund/Malmö, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
3
|
Kumarapperuma H, Chia ZJ, Malapitan SM, Wight TN, Little PJ, Kamato D. Response to retention hypothesis as a source of targets for arterial wall-directed therapies to prevent atherosclerosis: A critical review. Atherosclerosis 2024; 397:118552. [PMID: 39180958 DOI: 10.1016/j.atherosclerosis.2024.118552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
The subendothelial retention of circulating lipoproteins on extracellular matrix proteins and proteoglycans is one of the earliest events in the development of atherosclerosis. Multiple factors, including the size, type, composition, surrounding pH, and chemical modifications to lipoproteins, influence the electrostatic interactions between relevant moieties of the apolipoproteins on lipoproteins and the glycosaminoglycans of proteoglycans. The length and chemical composition of glycosaminoglycan chains attached to proteoglycan core proteins determine the extent of initial lipoprotein binding and retention in the artery wall. The phenomena of hyperelongation of glycosaminoglycan chains is associated with initial lipid retention and later atherosclerotic plaque formation. This review includes a summary of the current literature surrounding cellular mechanisms leading to GAG chain modification and lipid retention and discusses potential therapeutic strategies to target lipoprotein:proteoglycan interactions to prevent the development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Hirushi Kumarapperuma
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, 4102, Australia; Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia; Discovery Biology, School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, 4102, Australia; Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia; Discovery Biology, School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Sanchia Marie Malapitan
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia; Discovery Biology, School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, 4102, Australia; Department of Pharmacy, Guangzhou Xinhua University, Tianhe District, Guangzhou, Guangdong Pr., 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, 4102, Australia; Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland, 4111, Australia; Discovery Biology, School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia.
| |
Collapse
|
4
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
5
|
Mohammed KAK, Madeddu P, Avolio E. MEK inhibitors: a promising targeted therapy for cardiovascular disease. Front Cardiovasc Med 2024; 11:1404253. [PMID: 39011492 PMCID: PMC11247000 DOI: 10.3389/fcvm.2024.1404253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
Collapse
Affiliation(s)
- Khaled A K Mohammed
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paolo Madeddu
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
6
|
Kmieć P, Rosenkranz S, Odenthal M, Caglayan E. Differential Role of Aldosterone and Transforming Growth Factor Beta-1 in Cardiac Remodeling. Int J Mol Sci 2023; 24:12237. [PMID: 37569619 PMCID: PMC10419155 DOI: 10.3390/ijms241512237] [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: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Angiotensin II, a major culprit in cardiovascular disease, activates mediators that are also involved in pathological cardiac remodeling. In this context, we aimed at investigating the effects of two of them: aldosterone (Ald) and transforming growth factor beta-1 (TGF-β1) in an in vivo model. Six-week-old male wild-type (WT) and TGF-β1-overexpressing transgenic (TGF-β1-TG) mice were infused with subhypertensive doses of Ald for 2 weeks and/or treated orally with eplerenone from postnatal day 21. Thehearts' ventricles were examined by morphometry, immunoblotting to assess the intracellular signaling pathways and RT qPCR to determine hypertrophy and fibrosis marker genes. The TGF-β1-TG mice spontaneously developed cardiac hypertrophy and interstitial fibrosis and exhibited a higher baseline phosphorylation of p44/42 and p38 kinases, fibronectin and ANP mRNA expression. Ald induced a comparable increase in the ventricular-heart-weight-to-body-weight ratio and cardiomyocyte diameter in both strains, but a less pronounced increase in interstitial fibrosis in the transgenic compared to the WT mice (23.6% vs. 80.9%, p < 0.005). Ald increased the phosphorylation of p44/42 and p38 in the WT but not the TGF-β1-TG mice. While the eplerenone-enriched chow partially prevented Ald-induced cardiac hypertrophy in both genotypes and interstitial fibrosis in the WT controls, it completely protected against additional fibrosis in transgenic mice. Ald appears to induce cardiac hypertrophy independently of TGF-β1, while in the case of fibrosis, the downstream signaling pathways of these two factors probably converge.
Collapse
Affiliation(s)
- Piotr Kmieć
- Department of Endocrinology and Internal Medicine, Medical University of Gdańsk, 80214 Gdańsk, Poland;
| | - Stephan Rosenkranz
- Clinic for Internal Medicine III and Cologne Cardiovascular Research Center, Cologne University Heart Center, 50937 Köln, Germany;
| | - Margarete Odenthal
- Institute of Pathology, University Hospital of Cologne and Center for Molecular Medicine, University of Cologne, 50937 Köln, Germany;
| | - Evren Caglayan
- Department of Cardiology, University-Medicine Rostock, 18057 Rostock, Germany
| |
Collapse
|
7
|
Zarezade V, Mohammadtaghvaei N, Rashidi M, Babaahmadi-Rezaei H. Cardioprotective effect of tamoxifen and raloxifene: Preventing proteoglycan synthesis by modulating non-canonical TGF-β signalling through NADPH oxidase and ERK phosphorylation. Biochem Biophys Res Commun 2023; 671:263-269. [PMID: 37307710 DOI: 10.1016/j.bbrc.2023.06.031] [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: 05/28/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
Atherosclerosis, a leading cause of cardiovascular disease, remains a significant global health concern. Tamoxifen and raloxifene, selective estrogen receptor modulators (SERMs), have demonstrated potential cardioprotective effects. However, the underlying molecular mechanisms by which these SERMs modulate Transforming Growth Factor-β (TGF-β) signaling in human vascular smooth muscle cells (VSMCs) remain largely unexplored. This study sought to investigate the impact of tamoxifen and raloxifene on TGF-β-induced CHSY1 expression and Smad2 linker region phosphorylation in VSMCs and to elucidate the role of reactive oxygen species (ROS), NADPH oxidase (NOX), and kinase pathways in mediating these effects. Employing a comprehensive experimental strategy, VSMCs were treated with TGF-β in the presence or absence of tamoxifen, raloxifene, and various pharmacological inhibitors. Subsequently, CHSY1 mRNA expression, Smad2C and Smad2L phosphorylation, ROS production, p47phox and ERK 1/2 phosphorylation were assessed. Our results revealed that tamoxifen and raloxifene significantly attenuated TGF-β-mediated CHSY1 mRNA expression and Smad2 linker region phosphorylation, without affecting the canonical TGF-β-Smad2C pathway. Furthermore, these compounds effectively inhibited ROS production, p47phox and ERK 1/2 phosphorylation, implicating the involvement of the TGF-β-NOX-ERK-Smad2L signaling cascade in their cardioprotective properties. This study provides a comprehensive understanding of the molecular mechanisms underlying the cardioprotective effects of tamoxifen and raloxifene in VSMCs, offering valuable insights for the development of targeted therapeutic strategies aimed at atherosclerosis prevention and the promotion of cardiovascular health.
Collapse
Affiliation(s)
- Vahid Zarezade
- Hyperlipidemia Research Center, Department of Medical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Mohammadtaghvaei
- Hyperlipidemia Research Center, Department of Laboratory Sciences, School of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Rashidi
- Hyperlipidemia Research Center, Department of Medical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Medical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Lv Y, Wang W, Yao L, He J, Bai G, Lin C, Tu C. Sodium Fluoride and Sulfur Dioxide Derivatives Induce TGF-β1-Mediated NBCe1 Downregulation Causing Acid-Base Disorder of LS8 Cells. Biol Trace Elem Res 2023; 201:828-842. [PMID: 35304687 DOI: 10.1007/s12011-022-03169-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/16/2022] [Indexed: 01/25/2023]
Abstract
The aim of the present work was to assess whether the combination of sodium fluoride (NaF) and sulfur dioxide derivatives (SO2 derivatives) affects the expression of the electrogenic sodium bicarbonate cotransporter NBCe1 (SLC4A4), triggering an acid-base imbalance during enamel development, leading to enamel damage. LS8 cells was taken as the research objects and fluorescent probes, quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and factorial analysis were used to clarify the nature of the fluoro-sulfur interaction and the potential signaling pathway involved in the regulation of NBCe1. The results showed that exposure to fluoride or SO2 derivatives resulted in an acid-base imbalance, and these changes were accompanied by inhibited expression of NBCe1 and TGF-β1; these effects were more significant after fluoride exposure as compared to exposure to SO2 derivatives. Interestingly, in most cases, the toxic effects during combined exposure were significantly reduced compared to the effects observed with fluoride or sulfur dioxide derivatives alone. The results also indicated that activation of TGF-β1 signaling significantly upregulated the expression of NBCe1, and this effect was suppressed after the Smad, ERK, and JNK signals were blocked. Furthermore, fluoride and SO2 derivative-dependent NBCe1 regulation was found to require TGF-β1. In conclusion, this study indicates that the combined effect of fluorine and sulfur on LS8 cells is mainly antagonistic. TGF-β1 may regulate NBCe1 and may participate in the occurrence of dental fluorosis through the classic TGF-β1/Smad pathway and the unconventional ERK and JNK pathways.
Collapse
Affiliation(s)
- Ying Lv
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China
| | - Wentai Wang
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China
| | - Lili Yao
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China
| | - Jiaojiao He
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China
| | - Guohui Bai
- Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Changhu Lin
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China
| | - Chenglong Tu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guizhou, China.
- The Toxicity Testing Center of Guizhou Medical University, Guizhou Medical University, Guizhou, China.
| |
Collapse
|
9
|
Tan R, You Q, Cui J, Wang M, Song N, An K, Lin L, Adu-Amankwaah J, Yuan J, Sun H. Sodium houttuyfonate against cardiac fibrosis attenuates isoproterenol-induced heart failure by binding to MMP2 and p38. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154590. [PMID: 36610170 DOI: 10.1016/j.phymed.2022.154590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/17/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Heart failure (HF), caused by stress cardiomyopathy, is a major cause of mortality. Cardiac fibrosis is an essential structural remodeling associated with HF; therefore, preventing cardiac fibrosis is crucial to decelerating the progression of HF. Sodium houttuyfonate (SH), an extract of Houttuynia cordata, has a potent therapeutic effect on hypoxic cardiomyocytes in a myocardial infarction model. PURPOSE To investigate the preventative and therapeutic effects of SH during isoproterenol (ISO)-induced HF and explore the pharmacological mechanism of SH in alleviating HF. METHODS We analyzed the overlapping target genes between SH and cardiac fibrosis or HF using a network pharmacology analytical method. We verified the suppressive effect of SH on ISO-induced proliferation and activation of cardiac fibroblasts by immunohistochemical staining and histological analysis in an isoproterenol-induced HF mouse model. Additionally, we investigated the effect of SH by evaluating fibrosis and cardiac remodeling markers. To further decipher the pharmacological mechanism of SH against cardiac fibrosis and HF, we performed a molecular docking analysis between SH and hub common target genes. RESULTS There were 20 overlapping target genes between SH and cardiac fibrosis and 32 overlapping target genes between SH and HF. The 16 common target genes of SH against cardiac fibrosis and HF included MMP2 (matrix metalloproteinase 2), and p38. SH significantly inhibited the ISO- or TGF-β-induced expression of Col1α (collagen 1), α-SMA (smooth muscle actin), MMP2, TIMP2 (tissue inhibitor of metalloproteinase 2), TGF-β (transforming growth factor), and Smad2 phosphorylation. Moreover, both ISO- and TGF-β-induced p38 phosphorylation was inhibited. Molecular docking analysis showed that SH forms a stable complex with MMP2 and p38. CONCLUSIONS In addition to protecting cardiomyocytes, SH directly inhibits cardiac fibroblast activation and proliferation by binding to MMP2 and p38, subsequently delaying cardiac fibrosis and HF progression. Our prevention- and intervention-based approaches in this study showed that SH inhibited the development of stress cardiomyopathy-mediated cardiac fibrosis and HF when SH was administered before or after the initiation of cardiac stress.
Collapse
Affiliation(s)
- Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Qiang You
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Cui
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Mingming Wang
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Nina Song
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Ke An
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Lili Lin
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining 272000, China
| | - Hong Sun
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
10
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
11
|
Sarohi V, Chakraborty S, Basak T. Exploring the cardiac ECM during fibrosis: A new era with next-gen proteomics. Front Mol Biosci 2022; 9:1030226. [PMID: 36483540 PMCID: PMC9722982 DOI: 10.3389/fmolb.2022.1030226] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/31/2022] [Indexed: 10/24/2023] Open
Abstract
Extracellular matrix (ECM) plays a critical role in maintaining elasticity in cardiac tissues. Elasticity is required in the heart for properly pumping blood to the whole body. Dysregulated ECM remodeling causes fibrosis in the cardiac tissues. Cardiac fibrosis leads to stiffness in the heart tissues, resulting in heart failure. During cardiac fibrosis, ECM proteins get excessively deposited in the cardiac tissues. In the ECM, cardiac fibroblast proliferates into myofibroblast upon various kinds of stimulations. Fibroblast activation (myofibroblast) contributes majorly toward cardiac fibrosis. Other than cardiac fibroblasts, cardiomyocytes, epithelial/endothelial cells, and immune system cells can also contribute to cardiac fibrosis. Alteration in the expression of the ECM core and ECM-modifier proteins causes different types of cardiac fibrosis. These different components of ECM culminated into different pathways inducing transdifferentiation of cardiac fibroblast into myofibroblast. In this review, we summarize the role of different ECM components during cardiac fibrosis progression leading to heart failure. Furthermore, we highlight the importance of applying mass-spectrometry-based proteomics to understand the key changes occurring in the ECM during fibrotic progression. Next-gen proteomics studies will broaden the potential to identify key targets to combat cardiac fibrosis in order to achieve precise medicine-development in the future.
Collapse
Affiliation(s)
- Vivek Sarohi
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| | - Sanchari Chakraborty
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)- Mandi, Himachal Pradesh, India
| |
Collapse
|
12
|
Guo S, Zhou Y, Xie X. Resveratrol inhibiting TGF/ERK signaling pathway can improve atherosclerosis: backgrounds, mechanisms and effects. Biomed Pharmacother 2022; 155:113775. [DOI: 10.1016/j.biopha.2022.113775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/16/2022] [Accepted: 09/28/2022] [Indexed: 11/02/2022] Open
|
13
|
Luo G, Wang C, Li J, Zhang X, Sun Z, Song S, Fan C. Thrombin improves diabetic wound healing by ERK dependent and independent Smad2/3 linker region phosphorylation. Curr Pharm Des 2022; 28:1433-1443. [PMID: 35546767 DOI: 10.2174/1381612828666220511125237] [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: 11/03/2021] [Accepted: 03/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Impaired wound healing is one of the most noteworthy features and troublesome complications of diabetes mellitus, which arouse a rising global health concern but without potent remedies. Thrombin is the major hemostatic agent applied at wound healing initiation and recently gained therapeutic credits in later phases. However, a rare investigation achieved prolonged use of thrombin and probed the detailed mechanism. OBJECTIVE To investigate the effects and mechanism of thrombin on diabetic skin wound healing. METHODS The effect of thrombin on fibroblast proliferation, α-SMA, and Collagen I expression was firstly studied in vitro by Cell Counting Kit 8 (CCK8) and western blotting. Then, the specific phosphorylation site of SMAD2/3 and their ERK1/2 dependence during thrombin treatment were assessed by western blotting for mechanism exploration. After that, full-thickness wound defects were established in diabetic male SD rats and treated with thrombin in the presence or absence of PD98059 to observe the in vivo effects of thrombin and to confirm its ERK dependence. RESULTS We found that thrombin promoted fibroblast proliferation and their α-SMA and Collagen I production. Mechanistically, thrombin induced phosphorylation of Smad2 linker region (Ser245/250/255) through ERK1/2 phosphorylation but promoted phosphorylation of Smad3 linker region (Ser204) independent of ERK1/2. Histological results showed that thrombin facilitated wound healing by promoting α-SMA and Collagen I expression, which was not abolished by inhibiting ERK phosphorylation. CONCLUSION Collectively, this study validated the therapeutic efficacy of thrombin on diabetic wound healing and identified both ERK-dependent and -independent Smad2/3 linker region phosphorylation as the essential signaling events in this process.
Collapse
Affiliation(s)
- Gang Luo
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Chongyang Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Xuancheng Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Ziyang Sun
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Sa Song
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People\'s Hospital, 600 Yishan Rd, Shanghai 200233, PR China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, China
| |
Collapse
|
14
|
Babaahmadi-Rezaei H, Mohamed R, Dayati P, Mehr RN, Seif F, Sharifat N, Khedri A, Kamato D, Little PJ. Endothelin-1 dependent expression of GAG genes involves NOX and p38 mediated Smad linker region phosphorylation. Clin Exp Pharmacol Physiol 2022; 49:710-718. [PMID: 35527471 PMCID: PMC9322435 DOI: 10.1111/1440-1681.13650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
Abstract
Endothelin-1 (ET-1) is implicated in the development of atherosclerosis and mediates glycosaminoglycan (GAG) chain hyperelongation on proteoglycans. Our aim was to identify the ET-1-mediated signalling pathway involving NADPH oxidase (NOX), p38 MAP kinsae and Smad2 linker region phosphorylation (phospho-Smad2L) regulate GAG synthesizing enzymes mRNA expression (C4ST-1 and ChSy1) involved in GAG chains hyperelongation in human vascular smooth muscle cells (VSMCs). Signalling intermediates were detected and quantified by Western blotting and the mRNA levels of GAG synthesizing enzymes were assessed by quantitative real-time polymerase chain reaction (qRT-PCR). ET-1 treatment of human VSMCs resulted in an increase in phospho-Smad2L level. The TGF-β receptor antagonist, SB431542 and the mixed ETA and ETB receptor antagonist bosentan, inhibited ET-1-mediated phospho-Smad2L level. In the presence of apocynin and diphenyleneiodonium chloride (DPI) (NOX inhibitors) and SB239063 (p38 inhibitor) ET-1-mediated phospho-Smad2L levels were inhibited. The gene expression levels of GAG synthesizing enzymes post-ET-1 treatment were increased compared to untreated controls (P<0.01). The ET-mediated the mRNA levels of these enzymes were blocked by the bosentan, SB431542, SB239063, DPI, apocynin and antioxidant N-acetyl-L-cysteine (NAC). ET-1-mediated signalling to GAG synthesizing enzymes gene expression occurs via transactivation-dependent pathway involving NOX, p38 MAP kinsae and Smad2 linker region phosphorylation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Rafat Mohamed
- The University of Queensland, , Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, QLD, Australia
| | - Parisa Dayati
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reyhaneh Niayesh Mehr
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Faezeh Seif
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Sharifat
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azam Khedri
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Dannii Kamato
- The University of Queensland, , Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, QLD, Australia
| | - Peter J Little
- The University of Queensland, , Pharmacy Australia Centre of Excellence, 20 Cornwall St, Woolloongabba, QLD, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| |
Collapse
|
15
|
Zieba J, Forlenza KN, Heard K, Martin JH, Bosakova M, Cohn DH, Robertson SP, Krejci P, Krakow D. Intervertebral disc degeneration is rescued by TGFβ/BMP signaling modulation in an ex vivo filamin B mouse model. Bone Res 2022; 10:37. [PMID: 35474298 PMCID: PMC9042866 DOI: 10.1038/s41413-022-00200-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/01/2021] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
Spondylocarpotarsal syndrome (SCT) is a rare musculoskeletal disorder characterized by short stature and vertebral, carpal, and tarsal fusions resulting from biallelic nonsense mutations in the gene encoding filamin B (FLNB). Utilizing a FLNB knockout mouse, we showed that the vertebral fusions in SCT evolved from intervertebral disc (IVD) degeneration and ossification of the annulus fibrosus (AF), eventually leading to full trabecular bone formation. This resulted from alterations in the TGFβ/BMP signaling pathway that included increased canonical TGFβ and noncanonical BMP signaling. In this study, the role of FLNB in the TGFβ/BMP pathway was elucidated using in vitro, in vivo, and ex vivo treatment methodologies. The data demonstrated that FLNB interacts with inhibitory Smads 6 and 7 (i-Smads) to regulate TGFβ/BMP signaling and that loss of FLNB produces increased TGFβ receptor activity and decreased Smad 1 ubiquitination. Through the use of small molecule inhibitors in an ex vivo spine model, TGFβ/BMP signaling was modulated to design a targeted treatment for SCT and disc degeneration. Inhibition of canonical and noncanonical TGFβ/BMP pathway activity restored Flnb-/- IVD morphology. These most effective improvements resulted from specific inhibition of TGFβ and p38 signaling activation. FLNB acts as a bridge for TGFβ/BMP signaling crosstalk through i-Smads and is key for the critical balance in TGFβ/BMP signaling that maintains the IVD. These findings further our understanding of IVD biology and reveal new molecular targets for disc degeneration as well as congenital vertebral fusion disorders.
Collapse
Affiliation(s)
- Jennifer Zieba
- Department of Orthopedic Surgery, Los Angeles, CA, 90095, USA
| | | | - Kelly Heard
- Department of Orthopedic Surgery, Los Angeles, CA, 90095, USA
| | - Jorge H Martin
- Department of Orthopedic Surgery, Los Angeles, CA, 90095, USA
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
| | - Daniel H Cohn
- Department of Orthopedic Surgery, Los Angeles, CA, 90095, USA
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
| | - Deborah Krakow
- Department of Orthopedic Surgery, Los Angeles, CA, 90095, USA.
- Department of Human Genetics, Los Angeles, CA, 90095, USA.
- Department of Obstetrics and Gynecology, Los Angeles, CA, 90095, USA.
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
16
|
Afroz R, Kumarapperuma H, Nguyen QVN, Mohamed R, Little PJ, Kamato D. Lipopolysaccharide acting via toll-like receptor 4 transactivates the TGF-β receptor in vascular smooth muscle cells. Cell Mol Life Sci 2022; 79:121. [PMID: 35122536 PMCID: PMC8817999 DOI: 10.1007/s00018-022-04159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) recognise pathogen‑associated molecular patterns, which allow the detection of microbial infection by host cells. Bacterial-derived toxin lipopolysaccharide activates TLR4 and leads to the activation of the Smad2 transcription factor. The phosphorylation of the Smad2 transcription factor is the result of the activation of the transforming growth factor-β receptor 1 (TGFBR1). Therefore, we sought to investigate LPS via TLR4-mediated Smad2 carboxy terminal phosphorylation dependent on the transactivation of the TGFBR1. The in vitro model used human aortic vascular smooth muscle cells to assess the implications of TLR4 transactivation of the TGFBR1 in vascular pathophysiology. We show that LPS-mediated Smad2 carboxy terminal phosphorylation is inhibited in the presence of TGFBR1 inhibitor, SB431542. Treatment with MyD88 and TRIF pathway antagonists does not affect LPS-mediated phosphorylation of Smad2 carboxy terminal; however, LPS-mediated Smad2 phosphorylation was inhibited in the presence of MMP inhibitor, GM6001, and unaffected in the presence of ROCK inhibitor Y27632 or ROS/NOX inhibitor DPI. LPS via transactivation of the TGFBR1 stimulates PAI-1 mRNA expression. TLRs are first in line to respond to exogenous invading substances and endogenous molecules; our findings characterise a novel signalling pathway in the context of cell biology. Identifying TLR transactivation of the TGFBR1 may provide future insight into the detrimental implications of pathogens in pathophysiology.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD, 4111, Australia
| | - Hirushi Kumarapperuma
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Quang V N Nguyen
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Raafat Mohamed
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Peter J Little
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Tianhe District, Guangzhou, 510520, China.,Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Danielle Kamato
- School of Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
17
|
Thielen N, Neefjes M, Wiegertjes R, van den Akker G, Vitters E, van Beuningen H, Blaney Davidson E, Koenders M, van Lent P, van de Loo F, van Caam A, van der Kraan P. Osteoarthritis-Related Inflammation Blocks TGF-β's Protective Effect on Chondrocyte Hypertrophy via (de)Phosphorylation of the SMAD2/3 Linker Region. Int J Mol Sci 2021; 22:ijms22158124. [PMID: 34360888 PMCID: PMC8347103 DOI: 10.3390/ijms22158124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/13/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by irreversible cartilage damage, inflammation and altered chondrocyte phenotype. Transforming growth factor-β (TGF-β) signaling via SMAD2/3 is crucial for blocking hypertrophy. The post-translational modifications of these SMAD proteins in the linker domain regulate their function and these can be triggered by inflammation through the activation of kinases or phosphatases. Therefore, we investigated if OA-related inflammation affects TGF-β signaling via SMAD2/3 linker-modifications in chondrocytes. We found that both Interleukin (IL)-1β and OA-synovium conditioned medium negated SMAD2/3 transcriptional activity in chondrocytes. This inhibition of TGF-β signaling was enhanced if SMAD3 could not be phosphorylated on Ser213 in the linker region and the inhibition by IL-1β was less if the SMAD3 linker could not be phosphorylated at Ser204. Our study shows evidence that inflammation inhibits SMAD2/3 signaling in chondrocytes via SMAD linker (de)-phosphorylation. The involvement of linker region modifications may represent a new therapeutic target for OA.
Collapse
Affiliation(s)
- Nathalie Thielen
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Margot Neefjes
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Renske Wiegertjes
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Guus van den Akker
- Department of Orthopedic Surgery, Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Elly Vitters
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Henk van Beuningen
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Esmeralda Blaney Davidson
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Marije Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Peter van Lent
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Fons van de Loo
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Arjan van Caam
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
| | - Peter van der Kraan
- Department of Experimental Rheumatology, Radboud University Medical Center, 6500 MD Nijmegen, The Netherlands; (N.T.); (M.N.); (R.W.); (E.V.); (H.v.B.); (E.B.D.); (M.K.); (P.v.L.); (F.v.d.L.); (A.v.C.)
- Correspondence:
| |
Collapse
|
18
|
Hussain H, Cao Y, Mohamad R, Afroz R, Zhou Y, Moyle P, Bansal N, Wattoo FH, Kamato D, Little PJ. YY-11, a camel milk-derived peptide, inhibits TGF-β-mediated atherogenic signaling in human vascular smooth muscle cells. J Food Biochem 2021; 46:e13882. [PMID: 34312884 DOI: 10.1111/jfbc.13882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/17/2021] [Accepted: 07/15/2021] [Indexed: 11/28/2022]
Abstract
Atherosclerosis, the major underlying pathology of cardiovascular disease, commences with the binding and trapping of lipids on modified proteoglycans, with hyperelongated glycosaminoglycan chains. Transforming growth factor (TGF)-β stimulates glycosaminoglycan elongation in vascular smooth muscle cells. We have recently shown that this TGF-β signaling pathway involves reactive oxygen species (ROS). YY-11 is a dodecapeptide derived from camel milk and it has antioxidant activity. We have investigated the role of YY-11 in blocking ROS signaling and downstream atherogenic responses. YY-11 inhibited TGF-β stimulated ROS production and inhibited the expression of genes for glycosaminoglycan chain elongation as a component of an in vitro model of atherosclerosis. This study provides a biochemical mechanism for the role of camel milk as a potential nutritional product to contribute to the worldwide amelioration of cardiovascular disease. PRACTICAL APPLICATIONS: The identification of readily accessible foods with antioxidant properties would provide a convenient and cost-effective approach community wide reducing oxidative stress induced pathologies such as atherosclerosis. We demonstrate that camel milk-derived peptide is an antioxidant that can inhibit growth factor-mediated proteoglycan modification in vitro. As proteoglycan modification is being recognized as one of the earliest atherogenic responses, these data support the notion of camel milk as a suitable nutritional product to contribute to the prevention of early stage of atherosclerosis development.
Collapse
Affiliation(s)
- Humaira Hussain
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.,Department of Biochemistry and Biotechnology, Arid Agriculture University, Rawalpindi, Pakistan
| | - Yingnan Cao
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Raafat Mohamad
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Rizwana Afroz
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Ying Zhou
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Peter Moyle
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Nidhi Bansal
- School of Agriculture and Food Sciences, Faculty of Science, University of Queensland, St. Lucia, QLD, Australia
| | - Feroza Hamid Wattoo
- Department of Biochemistry and Biotechnology, Arid Agriculture University, Rawalpindi, Pakistan
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Decato BE, Ammar R, Reinke-Breen L, Thompson JR, Azzara AV. Transcriptome analysis reveals key genes modulated by ALK5 inhibition in a bleomycin model of systemic sclerosis. Rheumatology (Oxford) 2021; 61:1717-1727. [PMID: 34289031 PMCID: PMC8996787 DOI: 10.1093/rheumatology/keab580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 07/09/2021] [Indexed: 11/14/2022] Open
Abstract
Objective SSc is a rheumatic autoimmune disease affecting roughly 20 000 people worldwide and characterized by excessive collagen accumulation in the skin and internal organs. Despite the high morbidity and mortality associated with SSc, there are no approved disease-modifying agents. Our objective in this study was to explore transcriptomic and model-based drug discovery approaches for SSc. Methods In this study, we explored the molecular basis for SSc pathogenesis in a well-studied mouse model of scleroderma. We profiled the skin and lung transcriptomes of mice at multiple timepoints, analysing the differential gene expression that underscores the development and resolution of bleomycin-induced fibrosis. Results We observed shared expression signatures of upregulation and downregulation in fibrotic skin and lung tissue, and observed significant upregulation of key pro-fibrotic genes including GDF15, Saa3, Cxcl10, Spp1 and Timp1. To identify changes in gene expression in responses to anti-fibrotic therapy, we assessed the effect of TGF-β pathway inhibition via oral ALK5 (TGF-β receptor I) inhibitor SB525334 and observed a time-lagged response in the lung relative to skin. We also implemented a machine learning algorithm that showed promise at predicting lung function using transcriptome data from both skin and lung biopsies. Conclusion This study provides the most comprehensive look at the gene expression dynamics of an animal model of SSc to date, provides a rich dataset for future comparative fibrotic disease research, and helps refine our understanding of pathways at work during SSc pathogenesis and intervention.
Collapse
Affiliation(s)
- Benjamin E Decato
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Ron Ammar
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Lauren Reinke-Breen
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - John R Thompson
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Anthony V Azzara
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| |
Collapse
|
20
|
Zhou Y, Little PJ, Xu S, Kamato D. Curcumin Inhibits Lysophosphatidic Acid Mediated MCP-1 Expression via Blocking ROCK Signalling. Molecules 2021; 26:2320. [PMID: 33923651 PMCID: PMC8073974 DOI: 10.3390/molecules26082320] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Curcumin is a natural compound that has been widely used as a food additive and medicine in Asian countries. Over several decades, diverse biological effects of curcumin have been elucidated, such as anti-inflammatory and anti-oxidative activities. Monocyte chemoattractant protein-1 (MCP-1) is a key inflammatory marker during the development of atherosclerosis, and curcumin blocks MCP-1 expression stimulated by various ligands. Hence, we studied the action of curcumin on lysophosphatidic acid (LPA) mediated MCP-1 expression and explored the specific underlying mechanisms. In human vascular smooth muscle cells, LPA induces Rho-associated protein kinase (ROCK) dependent transforming growth factor receptor (TGFBR1) transactivation, leading to glycosaminoglycan chain elongation. We found that LPA also signals via the TGFBR1 transactivation pathway to regulate MCP-1 expression. Curcumin blocks LPA mediated TGFBR1 transactivation and subsequent MCP-1 expression by blocking the ROCK signalling. In the vasculature, ROCK signalling regulates smooth muscle cell contraction, inflammatory cell recruitment, endothelial dysfunction and vascular remodelling. Therefore, curcumin as a ROCK signalling inhibitor has the potential to prevent atherogenesis via multiple ways.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; (Y.Z.); (D.K.)
| | - Peter J. Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; (Y.Z.); (D.K.)
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD 4575, Australia
| | - Suowen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230037, China;
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; (Y.Z.); (D.K.)
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| |
Collapse
|
21
|
Tao XM, Li D, Zhang C, Wen GH, Wu C, Xu YY, Kan Y, Lu WP, Ding HY, Yang Y. Salvianolic acid B protects against acute and chronic liver injury by inhibiting Smad2C/L phosphorylation. Exp Ther Med 2021; 21:341. [PMID: 33732314 PMCID: PMC7903446 DOI: 10.3892/etm.2021.9772] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/19/2020] [Indexed: 12/17/2022] Open
Abstract
Salvianolic acid B (Sal B) has strong antioxidant and anti-fibrosis effects, which are related to the transforming growth factor β/Smad signaling pathway. However, how Sal B affects this antioxidant pathway and the phosphorylation (p-) of Smad2 at both the COOH-terminal (pSmad2C) and linker region (pSmad2L) are unknown. The aims of the present study were to investigate the underlying mechanisms of Sal B on acute and chronic liver injury induced by CCl4 and H2O2, and its effects on p-Smad2C/L. In in vivo experiments, acute and chronic liver injury models were induced by CCl4, and the oxidative damage cell model was established in vitro with H2O2. Liver histopathology was assessed using hematoxylin and eosin and Van Gieson's staining. Moreover, serum biochemical indicators were analyzed using specific assay kits. Furthermore, the present study evaluated the oxidant/antioxidant status in acute and chronic liver injury models by oxidative stress parameters such as malondialdehyde, glutathione and superoxide dismutase. In addition, western blot analysis was performed to analyze the protein expression levels of pSmad2C, pSmad2L, nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). It was found that Sal B improved liver histology, decreased the levels of aminotransferase and attenuated oxidative stress in acute and chronic liver injury models. Additionally, the protein expression levels of pSmad2C and pSmad2L were decreased, but Nrf2 and HO-1 expression levels were increased both in vivo and in vitro. Collectively, the present results suggested that Sal B may protect against acute and chronic liver injury via inhibition of Smad2C/L phosphorylation, and the Nrf2/HO-1 signaling pathway may play an important role in this process.
Collapse
Affiliation(s)
- Xiang-Ming Tao
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Dong Li
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Chong Zhang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Guang-Hua Wen
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Chao Wu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Yuan-Yuan Xu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Yue Kan
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Wan-Peng Lu
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Han-Yan Ding
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Yan Yang
- Department of Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
22
|
Zhou Y, Little PJ, Cao Y, Ta HT, Kamato D. Lysophosphatidic acid receptor 5 transactivation of TGFBR1 stimulates the mRNA expression of proteoglycan synthesizing genes XYLT1 and CHST3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118848. [PMID: 32920014 DOI: 10.1016/j.bbamcr.2020.118848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
Lysophosphatidic acid (LPA) via transactivation dependent signalling pathways contributes to a plethora of physiological and pathophysiological responses. In the vasculature, hyperelongation of glycosaminoglycan (GAG) chains on proteoglycans leads to lipid retention in the intima resulting in the early pathogenesis of atherosclerosis. Therefore, we investigated and defined the contribution of transactivation dependent signalling in LPA mediated GAG chain hyperelongation in human vascular smooth muscle cells (VSMCs). LPA acting via the LPA receptor 5 (LPAR5) transactivates the TGFBR1 to stimulate the mRNA expression of GAG initiation and elongation genes xylosyltransferase-1 (XYLT1) and chondroitin 6-sulfotransferase-1 (CHST3), respectively. We found that LPA stimulates ROS and Akt signalling in VSMCs, however they are not associated in LPAR5 transactivation of the TGFBR1. We observed that LPA via ROCK dependent pathways transactivates the TGFBR1 to stimulate genes associated with GAG chain elongation. We demonstrate that GPCR transactivation of the TGFBR1 occurs via a universal biochemical mechanism and the identified effectors represent potential therapeutic targets to inhibit pathophysiological effects of GPCR transactivation of the TGFBR1.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia.
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China.
| | - Yingnan Cao
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Hang T Ta
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; School of Environment and Science, Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia.
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China.
| |
Collapse
|
23
|
Zhou Y, Kumarapperuma H, Sichone S, Chia ZJ, Little PJ, Xu S, Kamato D. Artemisinin inhibits glycosaminoglycan chain synthesizing gene expression but not proliferation of human vascular smooth muscle cells. Biochem Biophys Res Commun 2020; 532:239-243. [PMID: 32868072 DOI: 10.1016/j.bbrc.2020.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
Pleotropic growth factor, transforming growth factor (TGF)-β drives the modification and elongation of glycosaminoglycan (GAG) chains on proteoglycans. Hyperelongated GAG chains bind and trap lipoproteins in the intima leading to the formation of atherosclerotic plaques. We have identified that phosphorylation of Smad2 linker region drives GAG chain modification. The identification of an inhibitor of Smad2 linker region phosphorylation and GAG chain modification signifies a potential therapeutic for cardiovascular diseases. Artemisinin renowned for its potent anti-malarial effects possesses a broad range of biological effects. Our aim was to characterise the anti-atherogenic role of artemisinin in vascular smooth muscle cells (VSMCs). We demonstrate that TGF-β mediated Smad2 linker region phosphorylation and GAG chain elongation was attenuated by artemisinin; however, we observed no effect on VSMC proliferation. Our data demonstrates the potential for artemisinin to be developed as a therapy to inhibit the development of atherosclerosis by prevention of lipid deposition in the vessel wall without affecting the proliferation of VSMCs.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia.
| | - Hirushi Kumarapperuma
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia.
| | - Salifya Sichone
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia.
| | - Zheng Jie Chia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia.
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, China.
| | - Suowen Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology, Hefei, 230037, China.
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, China.
| |
Collapse
|
24
|
Kamato D, Little PJ. Smad2 linker region phosphorylation is an autonomous cell signalling pathway: Implications for multiple disease pathologies. Biomed Pharmacother 2020; 124:109854. [DOI: 10.1016/j.biopha.2020.109854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/18/2022] Open
|
25
|
Afroz R, Zhou Y, Little PJ, Xu S, Mohamed R, Stow J, Kamato D. Toll-like Receptor 4 Stimulates Gene Expression via Smad2 Linker Region Phosphorylation in Vascular Smooth Muscle Cells. ACS Pharmacol Transl Sci 2020; 3:524-534. [PMID: 32566917 DOI: 10.1021/acsptsci.9b00113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis begins in the vessel wall with the retention of low density lipoproteins to modified proteoglycans with hyperelongated glycosaminoglycan (GAG) chains. Bacterial infections produce endotoxins such as lipopolysaccharide that exacerbate the outcome of atherosclerosis by generating a heightened state of inflammation. Lipopolysaccharide (LPS) via its toll-like receptor (TLR) is well-known for its role in mediating an inflammatory response in the body. Emerging evidence demonstrates that TLRs are involved in regulating vascular functions. In this study we sought to investigate the role of LPS in proteoglycan modification and GAG chain elongation, and we hypothesize that LPS will signal via Smad2 dependent pathways to regulate GAG chain elongation. The in vitro model used human aortic vascular smooth muscle cells. GAG gene expression was assessed by quantitative real-time polymerase chain reaction. Western blotting was performed using whole-cell protein lysates to assess the signaling pathway. LPS via TLR4 stimulates the expression of GAG synthesizing enzymes to an equal extent to traditional cardiovascular agonists. LPS phosphorylates the Smad2 linker region via TAK-1/MAPK dependent pathways which correlated with genes associated with GAG chain initiation and elongation. The well-characterized role of LPS in inflammation and our data on GAG gene expression demonstrates that GAG chain elongation is the earliest marker of the inflammatory cascade in atherosclerosis development.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Suowen Xu
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Raafat Mohamed
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Jennifer Stow
- Institute of Molecular Bioscience, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| |
Collapse
|
26
|
Kamato D, Do BH, Osman N, Ross BP, Mohamed R, Xu S, Little PJ. Smad linker region phosphorylation is a signalling pathway in its own right and not only a modulator of canonical TGF-β signalling. Cell Mol Life Sci 2020; 77:243-251. [PMID: 31407020 PMCID: PMC11104920 DOI: 10.1007/s00018-019-03266-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/16/2019] [Accepted: 08/05/2019] [Indexed: 01/01/2023]
Abstract
Transforming growth factor (TGF)-β signalling pathways are intensively investigated because of their diverse association with physiological and pathophysiological states. Smad transcription factors are the key mediators of TGF-β signalling. Smads can be directly phosphorylated in the carboxy terminal by the TGF-β receptor or in the linker region via multiple intermediate serine/threonine kinases. Growth factors in addition to hormones and TGF-β can activate many of the same kinases which can phosphorylate the Smad linker region. Historically, Smad linker region phosphorylation was shown to prevent nuclear translocation of Smads and inhibit TGF-β signalling pathways; however, it was subsequently shown that Smad linker region phosphorylation can be a driver of gene expression. This review will cover the signalling pathways of Smad linker region phosphorylation that drive the expression of genes involved in pathology and pathophysiology. The role of Smad signalling in cell biology is expanding rapidly beyond its role in TGF-β signalling and many signalling paradigms need to be re-evaluated in terms of Smad involvement.
Collapse
Affiliation(s)
- Danielle Kamato
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia.
- Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Tianhe District, Guangzhou, 510520, China.
| | - Bich Hang Do
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam
| | - Narin Osman
- School of Medical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Benjamin P Ross
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Raafat Mohamed
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
- Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Peter J Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
- Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Tianhe District, Guangzhou, 510520, China
| |
Collapse
|
27
|
Zhou Y, Little PJ, Ta HT, Xu S, Kamato D. Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease. Pharmacol Ther 2019; 204:107404. [DOI: 10.1016/j.pharmthera.2019.107404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
|
28
|
Liu W, Zhang D, Li X, Zheng L, Cui C, Cui Y, Sun J, Xie J, Zhou X. TGF-β1 facilitates cell-cell communication in osteocytes via connexin43- and pannexin1-dependent gap junctions. Cell Death Discov 2019; 5:141. [PMID: 31666990 PMCID: PMC6814792 DOI: 10.1038/s41420-019-0221-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/04/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023] Open
Abstract
Connexins and pannexins are two families of channel forming proteins that are able to pass small molecules to achieve communication between cells. While connexins have been recognized to mediate gap junctional intercellular communication (GJIC), pannexins are far less known. Our previous study reported the potential role of TGF-β1 in mediating of connexins in osteocytes in vitro. Herein, we aimed to elucidate the influence of TGF-β1 on cell-cell communication based on gap junctions assembled by connexins and pannexins in vitro and ex vivo. We first showed that TGF-β1 positively affected the elongation of dendritic processes of osteocytes. Our data indicated that TGF-β1 increased expressions of connexin43 (Cx43) and pannexin1 (panx1), which are indispensable for hemichannel formation in gap junctions, in osteocytes in vitro and ex vivo. TGF-β1 enhanced gap junction formation and impacted cell-cell communication in living osteocytes, as indicated by the scrape loading and Lucifer yellow transfer assays. TGF-β1 enhanced the expressions of Cx43 and panx1 via activation of ERK1/2 and Smad3/4 signalling. The TGF-β1-restored expressions of Cx43 and panx1 in osteocytes in the presence of an ERK inhibitor, U0126, further demonstrated the direct participation of Smad3/4 signalling. TGF-β1 increased the accumulation of Smad3 in the nuclear region (immunofluorescence assay) and promoted the enrichment of Smad3 at the binding sites of the promoters of Gja1 (Cx43) and Panx1 (ChIP assay), thereby initiating the enhanced gene expression. These results provide a deep understanding of the molecular mechanisms involved in the modulation of cell-cell communication in osteocytes induced by TGF-β1.
Collapse
Affiliation(s)
- Wenjing Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chen Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Raffort J, Lareyre F, Clément M, Hassen-Khodja R, Chinetti G, Mallat Z. Diabetes and aortic aneurysm: current state of the art. Cardiovasc Res 2019; 114:1702-1713. [PMID: 30052821 PMCID: PMC6198737 DOI: 10.1093/cvr/cvy174] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/19/2018] [Indexed: 12/17/2022] Open
Abstract
Aortic aneurysm is a life-threatening disease due to the risk of aortic rupture. The only curative treatment available relies on surgical approaches; drug-based therapies are lacking, highlighting an unmet need for clinical practice. Abdominal aortic aneurysm (AAA) is frequently associated with atherosclerosis and cardiovascular risk factors including male sex, age, smoking, hypertension, and dyslipidaemia. Thoracic aortic aneurysm (TAA) is more often linked to genetic disorders of the extracellular matrix and the contractile apparatus but also share similar cardiovascular risk factors. Intriguingly, a large body of evidence points to an inverse association between diabetes and both AAA and TAA. A better understanding of the mechanisms underlying the negative association between diabetes and aortic aneurysm could help the development of innovative diagnostic and therapeutic approaches to tackle the disease. Here, we summarize current knowledge on the relationship between glycaemic parameters, diabetes, and the development of aortic aneurysm. Cellular and molecular pathways that underlie the protective effect of diabetes itself and its treatment are reviewed and discussed, along with their potential implications for clinical translation.
Collapse
Affiliation(s)
- Juliette Raffort
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, Robinson Way, UK.,Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Research Center, Team 5, Hôpital Européen Georges Pompidou, 56 rue Leblanc, Paris, France.,Department of Clinical Biochemistry, University Hospital of Nice, 30 avenue de la Voie Romaine, Nice Cedex 1, France.,Université Côte d'Azur, CHU, Inserm U1065, C3M, 151 Route de Ginestière, Nice Cedex 3, France
| | - Fabien Lareyre
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, Robinson Way, UK.,Université Côte d'Azur, CHU, Inserm U1065, C3M, 151 Route de Ginestière, Nice Cedex 3, France.,Department of Vascular Surgery, University Hospital of Nice, 30 avenue de la Voie Romaine, Nice Cedex 1, France
| | - Marc Clément
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, Robinson Way, UK
| | - Réda Hassen-Khodja
- Université Côte d'Azur, CHU, Inserm U1065, C3M, 151 Route de Ginestière, Nice Cedex 3, France.,Department of Vascular Surgery, University Hospital of Nice, 30 avenue de la Voie Romaine, Nice Cedex 1, France
| | - Giulia Chinetti
- Department of Clinical Biochemistry, University Hospital of Nice, 30 avenue de la Voie Romaine, Nice Cedex 1, France.,Université Côte d'Azur, CHU, Inserm U1065, C3M, 151 Route de Ginestière, Nice Cedex 3, France
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, Robinson Way, UK.,Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Research Center, Team 5, Hôpital Européen Georges Pompidou, 56 rue Leblanc, Paris, France
| |
Collapse
|
30
|
Song KM, Chung DY, Choi MJ, Ghatak K, Minh NN, Limanjaya A, Kwon MH, Ock J, Yin GN, Kim DK, Ryu JK, Suh JK. Vactosertib, a Novel, Orally Bioavailable Activin Receptor-Like Kinase 5 Inhibitor, Promotes Regression of Fibrotic Plaques in a Rat Model of Peyronie's Disease. World J Mens Health 2019; 38:552-563. [PMID: 31496148 PMCID: PMC7502315 DOI: 10.5534/wjmh.190071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose To examine the therapeutic effect of Vactosertib, a small molecule inhibitor of transforming growth factor-β (TGF-β) type I receptor (activin receptor-like kinase-5, ALK5), in an experimental model of Peyronie's disease (PD) and determining anti-fibrotic mechanisms of Vactosertib in primary fibroblasts derived from human PD plaques. Materials and Methods Male rats were randomly divided into three groups (n=6 per group); control rats without treatment; PD rats receiving vehicle; and PD rats receiving Vactosertib (10 mg/kg). PD-like plaques were induced by administering 100 µL of each of human fibrin and thrombin solutions into the tunica albuginea on days 0 and 5. Vactosertib was given orally five times a week for 2 weeks. On day 30, we performed electrical stimulation of the cavernous nerve to measure erectile function, and the penis was obtained for histological examination. Fibroblasts isolated from human PD plaques were used to determine the anti-fibrotic effects of Vactosertib in vitro. Results Vactosertib induced significant regression of fibrotic plaques in PD rats in vivo through reduced infiltration of inflammatory cells and reduced expression of phospho-Smad2, which recovered erectile function. Vactosertib also abrogated TGF-β1-induced enhancement of extracellular matrix protein production and hydroxyproline content in PD fibroblasts in vitro by hindering the TGF-β1-induced Smad2/3 phosphorylation and nuclear translocation, and fibroblast-to-myofibroblast transdifferentiation. Conclusions In view of the critical role of TGF-β and the Smad pathway in the pathogenesis of PD, inhibition of this pathway with an ALK5 inhibitor may represent a novel, targeted therapy for PD.
Collapse
Affiliation(s)
- Kang Moon Song
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Doo Yong Chung
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Min Ji Choi
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Kalyan Ghatak
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Nguyen Nhat Minh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Anita Limanjaya
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Mi Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Dae Kee Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewah Womans University, Seoul, Korea
| | - Ji Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea.
| | - Jun Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea.
| |
Collapse
|
31
|
Kamato D, Ta H, Afroz R, Xu S, Osman N, Little PJ. Mechanisms of PAR-1 mediated kinase receptor transactivation: Smad linker region phosphorylation. J Cell Commun Signal 2019; 13:539-548. [PMID: 31290007 DOI: 10.1007/s12079-019-00527-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/02/2019] [Indexed: 01/08/2023] Open
Abstract
Protease activated receptors (PARs) transactivate both epidermal growth factor receptors (EGFR) and transforming growth factor (TGF)-β receptors (TGFBR1) in vascular smooth muscle leading to the increased expression of genes (CHST11 and CHSY1) which are rate limiting for the enzymes that mediate hyperelongation of glycosaminoglycan (GAG) chains on the lipid-binding proteoglycan, biglycan. This is an excellent model to investigate mechanisms of transactivation as the processes are biochemically distinct. EGFR transactivation is dependent on the classical matrix metalloprotease (MMP) based triple membrane bypass mechanism and TGFBR1 transactivation is dependent on Rho/ROCK signalling and integrins. We have shown that all kinase receptor signalling is targeted towards phosphorylation of the linker region of the transcription factor, Smad2. We investigated the mechanisms of thrombin mediated kinase receptor transactivation signalling using anti-phospho antibodies and Western blotting and gene expression by RT-PCR. Thrombin stimulation of phospho-Smad2 (Ser 245/250/255) and of phospho-Smad2(Thr220) via EGFR transactivation commences quickly and extends out to at least 4 h whereas transactivation via TGFBR1 is delayed for 120 min but also persists for at least 4 h. Signalling of thrombin stimulated Smad linker region phosphorylation is approximately equally inhibited by the MMP inhibitor, GM6001 and the ROCK inhibitor, Y27632, and similarly expression of CHST11 and CHSY1 is approximately equally inhibited by GM6001 and Y27632. The data establishes Smad linker region phosphorylation as a central target of all transactivation signalling of GAG gene expression and thus an upstream kinase may be a target to prevent all transactivation signalling and its pathophysiological consequences.
Collapse
Affiliation(s)
- Danielle Kamato
- School of Pharmacy, University of Queensland, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia. .,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, China.
| | - Hang Ta
- School of Pharmacy, University of Queensland, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia
| | - Rizwana Afroz
- School of Pharmacy, University of Queensland, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Narin Osman
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, 3004, Australia
| | - Peter J Little
- School of Pharmacy, University of Queensland, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, China
| |
Collapse
|
32
|
Barth M, Selig JI, Klose S, Schomakers A, Kiene LS, Raschke S, Boeken U, Akhyari P, Fischer JW, Lichtenberg A. Degenerative aortic valve disease and diabetes: Implications for a link between proteoglycans and diabetic disorders in the aortic valve. Diab Vasc Dis Res 2019; 16:254-269. [PMID: 30563371 DOI: 10.1177/1479164118817922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Degenerative aortic valve disease in combination with diabetes is an increasing burden worldwide. There is growing evidence that particularly small leucine-rich proteoglycans are involved in the development of degenerative aortic valve disease. Nevertheless, the role of these molecules in this disease in the course of diabetes has not been elucidated in detail and previous studies remain controversial. Therefore, the aim of this study is to broaden the knowledge about small leucine-rich proteoglycans in degenerative aortic valve disease and the influence of diabetes and hyperglycaemia on aortic valves and valvular interstitial cells is examined. Analyses were performed using reverse-transcription polymerase chain reaction, Western blot, enzyme-linked immunosorbent assay, (immuno)histology and colorimetric assays. We could show that biglycan, but not decorin and lumican, is upregulated in degenerated human aortic valve cusps. Subgroup analysis reveals that upregulation of biglycan is stage-dependent. In vivo, loss of biglycan leads to stage-dependent calcification and also to migratory effects on interstitial cells within the extracellular matrix. In late stages of degenerative aortic valve disease, diabetes increases the expression of biglycan in aortic valves. In vitro, the combinations of hyperglycaemic with pro-degenerative conditions lead to an upregulation of biglycan. In conclusion, biglycan represents a potential link between degenerative aortic valve disease and diabetes.
Collapse
Affiliation(s)
- Mareike Barth
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jessica I Selig
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Svenja Klose
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Antje Schomakers
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Lena S Kiene
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Silja Raschke
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Udo Boeken
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Payam Akhyari
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jens W Fischer
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Artur Lichtenberg
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
33
|
Jia L, Sun P, Gao H, Shen J, Gao Y, Meng C, Fu S, Yao H, Zhang G. Mangiferin attenuates bleomycin-induced pulmonary fibrosis in mice through inhibiting TLR4/p65 and TGF-β1/Smad2/3 pathway. J Pharm Pharmacol 2019; 71:1017-1028. [PMID: 30847938 DOI: 10.1111/jphp.13077] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/12/2019] [Indexed: 12/30/2022]
Abstract
Abstract
Objectives
Investigating the antipulmonary fibrosis effect of mangiferin from Mangifera indica and the possible molecular mechanism.
Methods
In vivo, bleomycin (BLM)-induced pulmonary fibrosis experimental model was used for evaluating antipulmonary fibrosis effect of mangiferin. Histopathologic examination and collagen deposition were investigated by HE and Masson staining as well as detecting the content of hydroxyproline. The expression of transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (α-SMA), TLR4 and p-P65 in lung tissue was analysed through immunofluorescence. Leucocytes and inflammatory cytokines including IL-1β, IL-6, TNF-α and MCP-1 in bronchoalveolar lavage fluid were detected by cell counting and enzyme-linked immunosorbent assay. In vitro, TGF-β1-induced A549 epithelial–mesenchymal transition (EMT) cell model was used for investigating the possible molecular mechanism. Reactive oxygen species (ROS) generation was detected by DCFH-DA assay. Expression of all proteins was examined by Western blot.
Key findings
Oral administration of mangiferin could attenuate the severity of BLM-induced pulmonary fibrosis through increasing the survival rate, improving histopathological lesion and body weight loss as well as decreasing pulmonary index visibly. Pulmonary hydroxyproline content, TGF-β1, and α-SMA levels were reduced significantly. The molecular mechanism of mangiferin for inhibiting pulmonary fibrosis is that it could obviously inhibit the occurrence of inflammation and the secretion of inflammatory cytokine through inhibiting activation of TLR4 and phosphorylation of p65. Meanwhile, EMT process was suppressed obviously by mangiferin through blocking the phosphorylation of Smad2/3 and reducing MMP-9 expression. Besides, mangiferin could significantly inhibit the process of oxidant stress through downregulating the intracellular ROS generation.
Conclusions
Mangiferin attenuates BLM-induced pulmonary fibrosis in mice through inhibiting TLR4/p65 and TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Li Jia
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Ping Sun
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Hui Gao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Jie Shen
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Yuan Gao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Cheng Meng
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Shidong Fu
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Huijuan Yao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Gong Zhang
- Yanan University Affiliated Hospital, Yanan, Shanxi, China
| |
Collapse
|
34
|
Transforming growth factor-β1 up-regulates connexin43 expression in osteocytes via canonical Smad-dependent signaling pathway. Biosci Rep 2018; 38:BSR20181678. [PMID: 30482881 PMCID: PMC6294634 DOI: 10.1042/bsr20181678] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 02/05/2023] Open
Abstract
Connexin 43 (Cx43)-mediated gap junctional intercellular communication (GJIC) has been shown to be important in regulating multiple functions of bone cells. Transforming growth factor-β1 (TGF-β1) exhibited controversial effects on the expression of Cx43 in different cell types. To date, the effect of TGF-β1 on the Cx43 expression of osteocytes is still unknown. In the present study, we detected the expression of TGF-β1 in osteocytes and bone tissue, and then used recombinant mouse TGF-β1 to elucidate its effect on gap junctions (GJs) of osteocytes. Our data indicated that TGF-β1 up-regulated both mRNA and protein expression of Cx43 in osteocytes. Together with down-regulation of Cx43 expression after being treated with TGF-β type I receptor inhibitor Repsox, we deduced that TGF-β1 can positively regulate Cx43 expression in osteocytes. Thus we next focussed on the downstream signals of TGF-β and found that TGF-β1-mediated smads, Smad3 and Smad4, to translocate into nucleus. These translocated signal proteins bind to the promoter of Gja1 which was responsible for the changed expression of Cx43. The present study provides evidence that TGF-β1 can enhance GJIC between osteocytes through up-regulating Cx43 expression and the underlying mechanism involved in the activation of Smad-dependent pathway.
Collapse
|
35
|
Janssen JN, Batschkus S, Schimmel S, Bode C, Schminke B, Miosge N. The Influence of TGF-β3, EGF, and BGN on SOX9 and RUNX2 Expression in Human Chondrogenic Progenitor Cells. J Histochem Cytochem 2018; 67:117-127. [PMID: 30431382 DOI: 10.1369/0022155418811645] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease and leads to the degradation of the extracellular matrix by an imbalance between anabolic and catabolic processes. TGF-β3 (transforming growth factor beta-3) and epidermal growth factor (EGF) influence the osteochondrogenic potential of chondrocytes. In this study, we compared the expression of mediators and receptors in the TGF-β3 and EGF pathways, as well as biglycan (BGN), in healthy and diseased chondrocytes. Furthermore, we used chondrogenic progenitor cells (CPCs) for in vitro stimulation and knockdown experiments to elucidate the effects of TGF-β3 and EGF on the chondrogenic potential. Our results demonstrate that the expression of TGF-beta receptor type-1 (TGFBRI) and epidermal growth factor receptor (EGFR) is altered in diseased chondrocytes as well as in CPCs. Moreover, TGF-β3 and EGF stimulation influenced the expression levels of BGN, SRY (sex determining region Y)-box 9 (SOX9), and Runt-related transcription factor 2 (RUNX2) in CPCs. Therefore, changes in TGFBRI and EGFR expression likely contribute to the degenerative and regenerative effects seen in late stages of OA.
Collapse
Affiliation(s)
| | | | - Stefan Schimmel
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Christa Bode
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medical Center, Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics
| |
Collapse
|
36
|
Mohamed R, Dayati P, Mehr RN, Kamato D, Seif F, Babaahmadi-Rezaei H, Little PJ. Transforming growth factor-β1 mediated CHST11 and CHSY1 mRNA expression is ROS dependent in vascular smooth muscle cells. J Cell Commun Signal 2018; 13:225-233. [PMID: 30417274 DOI: 10.1007/s12079-018-0495-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/01/2018] [Indexed: 01/08/2023] Open
Abstract
Transforming growth factor (TGF)-β1 mediates glycosaminoglycan (GAG) chain hyperelongation on secreted proteoglycans and these modifications are associated with increased lipid binding in the vessel wall and the development of atherosclerosis. In vascular smooth muscle cells (VSMCs), TGF-β1 regulated GAG elongation via extracellular signal-regulated kinase (ERK) and p38 as well as Smad2 linker region phosphorylation. In this study, our aim was to identify the TGF-β1 mediated signalling pathway involving reactive oxygen species (ROS) and Smad2 linker region phosphorylation that regulate the mRNA expression of GAG synthesizing enzymes, chondroitin 4-O-sulfotransferase 1 (CHST11) and chondroitin sulfate synthase 1 (CHSY1) which are the rate limiting enzymes involved in GAG chain elongation. Signalling molecules were assessed by western blotting, quantitative real-time PCR was used for analysis of gene expression and intracellular ROS level was measured by a fluorescence based assay. TGF-β1 induced ROS production in VSMCs. Nicotinamide adenine dinucleotide phosphate oxidase (Nox) inhibitors, diphenyleneiodonium (DPI) and apocynin blocked TGF-β1 mediated Smad2 linker region phosphorylation. TGF-β1 treatment increased the mRNA levels of CHST11 and CHSY1. Pharmacological inhibition of Nox blocked TGF-β1 mediated mitogen activated protein kinases (MAPKs) phosphorylation and TGF-β1 stimulated CHST11 and CHSY1 mRNA expression. These findings demonstrated that TGF-β1 mediated expression of CHST11 and CHSY1 can occur via Nox-dependent pathways and Smad2 linker region phosphorylation.
Collapse
Affiliation(s)
- Raafat Mohamed
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall St, Woolloongabba, QLD, 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Parisa Dayati
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reyhaneh Niayesh Mehr
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Danielle Kamato
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall St, Woolloongabba, QLD, 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, Guangdong Pr., China
| | - Faezeh Seif
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Peter J Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, 20 Cornwall St, Woolloongabba, QLD, 4102, Australia. .,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, Guangdong Pr., China.
| |
Collapse
|
37
|
Kamato D, Burch M, Zhou Y, Mohamed R, Stow JL, Osman N, Zheng W, Little PJ. Individual Smad2 linker region phosphorylation sites determine the expression of proteoglycan and glycosaminoglycan synthesizing genes. Cell Signal 2018; 53:365-373. [PMID: 30423352 DOI: 10.1016/j.cellsig.2018.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/09/2018] [Accepted: 11/09/2018] [Indexed: 01/04/2023]
Abstract
Growth factors such as thrombin and transforming growth factor (TGF)-β facilitate glycosaminoglycan (GAG) chain hyperelongation on proteoglycans, a phenomenon that increases lipoprotein binding in the vessel wall and the development of atherosclerosis. TGF-β signals via canonical carboxy terminal phosphorylation of R-Smads and also non-canonical linker region phosphorylation of R-Smads. The G protein coupled receptor agonist, thrombin, can transactivate the TGF-β receptor leading to both canonical and non-canonical Smad signalling. Linker region phosphorylation drives the expression of genes for the synthesis of the proteoglycan, biglycan. Proteoglycan synthesis involves core protein synthesis, the initiation of GAG chains and the subsequent elongation of GAG chains. We have explored the relationship between the thrombin stimulated phosphorylation of individual serine and threonine sites in the linker region of Smad2 and the expression of GAG initiation xylosyltransferase-1 (XT-1) and GAG elongation chondroitin 4-sulfotransferase-1 (C4ST-1) and chondroitin synthase-1 (CHSY-1) genes. Thrombin stimulated the phosphorylation of all four target residues (Thr220, Ser245, Ser250 and Ser255 residues) with a similar temporal pattern - phosphorylation was maximal at 15 min (the earliest time point studied) and the level of the phospho-proteins declined thereafter over the following 4 h. Jnk, p38 and PI3K, selectively mediated the phosphorylation of the Thr220 residue whereas the serine residues were variously phosphorylated by multiple kinases. Thrombin stimulated the expression of all three genes - XT-1, C4ST-1 and CHSY-1. The three pathways mediating Thr220 phosphorylation were also involved in the expression of XT-1. The target pathways (excluding Jnk) were involved in the expression of the GAG elongation genes (C4ST-1 and CHSY-1). These findings support the contention that individual Smad linker region phosphorylation sites are linked to the expression of genes for the initiation and elongation of GAG chains on proteoglycans. The context of this work is that a specific inhibitor of GAG elongation represents a potential therapeutic agent for preventing GAG elongation and lipid binding and the results indicate that the specificity of the pathways is such that it might be therapeutically feasible to specifically target GAG elongation without interfering with other physiological processes with which proteoglycans are involved.
Collapse
Affiliation(s)
- Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China.
| | - Micah Burch
- Department of Cardiovascular Medicine, Brigham and Harvard Medical School, Boston, MA 02115, USA
| | - Ying Zhou
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia
| | - Raafat Mohamed
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, 4067, Australia
| | - Narin Osman
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia; School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia
| | - Wenhua Zheng
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia; Faculty of Health Sciences, University of Macau, Taipa, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| |
Collapse
|
38
|
Afroz R, Cao Y, Rostam MA, Ta H, Xu S, Zheng W, Osman N, Kamato D, Little PJ. Signalling pathways regulating galactosaminoglycan synthesis and structure in vascular smooth muscle: Implications for lipoprotein binding and atherosclerosis. Pharmacol Ther 2018; 187:88-97. [DOI: 10.1016/j.pharmthera.2018.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
39
|
Mehr RN, Kheirollah A, Seif F, Dayati P, Babaahmadi-Rezaei H. Reactive Oxygen Species and p38MAPK Have a Role in the Smad2 Linker Region Phosphorylation Induced by TGF-β. IRANIAN JOURNAL OF MEDICAL SCIENCES 2018; 43:401-408. [PMID: 30046209 PMCID: PMC6055211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) in addition to the C-terminal region can phosphorylate receptor-regulated Smads (R-Smads) in their linker region. The aim of the present study was to evaluate the role of signaling mediators such as NAD(P)H oxidases (reactive oxygen species [ROS] generators), ROS, and ROS-sensitive p38 mitogen-activated protein kinase (p38MAPK) in this signaling pathway in cultured human vascular smooth muscle cells (VSMCs). METHODS The present in vitro study was performed on human VSMCs. Proteins were detected by western blotting utilizing an anti-phospho-Smad2 (Ser245/250/255) rabbit polyclonal antibody and a horseradish peroxidase-labeled secondary antibody. Glyceraldehyde-3-phosphate dehydrogenase was used as a loading control. The phospho-Smad2 linker region (pSmad2L) was detected in all the experimental groups: a control group (untreated group), a group treated with TGF-β (2 ng/mL), and a group treated with TGF-β plus different inhibitors. The data were normalized and presented as mean±SEM. The statistical analyses were performed using SPSS, version 16.0, and the nonparametric Kruskal-Wallis test. A P value smaller than 0.05 was considered statistically significant. RESULTS The VSMCs treated with TGF-β (2 ng/mL) showed a time-dependent increase in the pSmad2L level. The highest level was observed at 15 minutes (P=0.03). The inhibitors of NAD(P)H oxidases (diphenyleneiodonium and apocynin) (P=0.04), ROS scavenger (N-acetylcysteine) (P=0.04), and p38MAPK inhibitor (SB-202190) (P=0.04) were able to reduce the increased level of the pSmad2L by TGF-β. CONCLUSION Our results suggested that NAD(P)H oxidases played an important role in the Smad2L phosphorylation in the human VSMCs. Furthermore, our results confirmed that ROS and p38MAPK were involved in this signaling pathway. Thus, TGF-β via a ROS-dependent mechanism can transmit its signals to the pSmad2L.
Collapse
Affiliation(s)
- Reyhaneh Niayesh Mehr
- Atherosclerosis Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Cellular and Molecular Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Faezeh Seif
- Atherosclerosis Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parisa Dayati
- Atherosclerosis Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Atherosclerosis Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
40
|
G protein coupled receptors can transduce signals through carboxy terminal and linker region phosphorylation of Smad transcription factors. Life Sci 2018; 199:10-15. [DOI: 10.1016/j.lfs.2018.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/23/2018] [Accepted: 03/02/2018] [Indexed: 11/22/2022]
|
41
|
Rostam MA, Shajimoon A, Kamato D, Mitra P, Piva TJ, Getachew R, Cao Y, Zheng W, Osman N, Little PJ. Flavopiridol Inhibits TGF- β-Stimulated Biglycan Synthesis by Blocking Linker Region Phosphorylation and Nuclear Translocation of Smad2. J Pharmacol Exp Ther 2018; 365:156-164. [PMID: 29438988 DOI: 10.1124/jpet.117.244483] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/03/2018] [Indexed: 03/08/2025] Open
Abstract
Transforming growth factor-β (TGF-β) is a pleiotropic growth factor implicated in the development of atherosclerosis for its role in mediating glycosaminoglycan (GAG) chain hyperelongation on the proteoglycan biglycan, a phenomenon that increases the binding of atherogenic lipoproteins in the vessel wall. Phosphorylation of the transcription factor Smad has emerged as a critical step in the signaling pathways that control the synthesis of biglycan, both the core protein and the GAG chains. We have used flavopiridol, a well-known cyclin-dependent kinase inhibitor, to study the role of linker region phosphorylation in the TGF-β-stimulated synthesis of biglycan. We used radiosulfate incorporation and SDS-PAGE to assess proteoglycan synthesis, real-time polymerase chain reaction to assess gene expression, and chromatin immunoprecipitation to assess the binding of Smads to the promoter region of GAG Synthesizing genes. Flavopiridol blocked TGF-β-stimulated synthesis of mRNA for the GAG synthesizing enzymes, and chondroitin 4-sulfotransferase (C4ST-1), chondroitin sulfate synthase-1 (ChSy-1) and TGF-β-mediated proteoglycans synthesis as well as GAG hyperelongation. Flavopiridol blocked TGF-β-stimulated Smad2 phosphorylation at both the serine triplet and the isolated threonine residue in the linker region. The binding of Smad to the promoter region of the C4ST-1 and ChSy-1 genes was stimulated by TGF-β, and this response was blocked by flavopiridol, demonstrating that linker region phosphorylated Smad can pass to the nucleus and positively regulate transcription. These results demonstrate the validity of the kinases, which phosphorylate the Smad linker region as potential therapeutic target(s) for the development of an agent to prevent atherosclerosis.
Collapse
Affiliation(s)
- Muhamad A Rostam
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Aravindra Shajimoon
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Danielle Kamato
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Partha Mitra
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Terrence J Piva
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Robel Getachew
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Yingnan Cao
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Wenhua Zheng
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Narin Osman
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| | - Peter J Little
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia (M.A.R.); School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (A.S., T.J.P., R.G., N.O., P.J.L.); School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia (D.K., P.M., P.J.L.); Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe, Guangzhou, China (Y.C., P.J.L.); Faculty of Health Sciences, University of Macau, Taipa, Macau, China (W.Z.); and Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, Victoria, Australia (N.O.)
| |
Collapse
|
42
|
Ponticelli C, Anders HJ. Thrombospondin immune regulation and the kidney. Nephrol Dial Transplant 2018; 32:1084-1089. [PMID: 28088772 DOI: 10.1093/ndt/gfw431] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 12/17/2022] Open
Abstract
Most therapeutic attempts to prevent the progression of kidney diseases have been based on interventions to inhibit the production of transforming growth factor-β (TGF-β). Thrombospondins (TSPs) play an important role in activating TGF-β. In the healthy kidney, two TSPs are expressed, TSP1 and TSP2, which exert contrasting effects. While TSP1 is a major activator of TGF-β in renal cells and exerts pro-inflammatory effects both in vitro and in vivo, TSP2 lacks the ability for TGF-β activation but regulates matrix remodeling and inflammation in experimental kidney disease. The effects of TSPs in the kidney have been mostly investigated by using the murine model of unilateral ureteral obstruction. In this model, TSP1 expression is increased along with the development of interstitial fibrosis and TGF-β. Relief of the obstruction gradually improves renal function and decreases the expression in TSP1 and TGF-β1. Several inhibitors of TSP1 prevented progressive interstitial fibrosis in murine models of ureteral obstruction, suggesting that control of latent TGF-β activation by inhibiting TSP1 might represent a novel potential target for preventing renal interstitial fibrosis. However, further studies are needed to assess whether TSP1-mediated TGF-β activation can be safely used in humans. In fact, TSPs normally act to suppress tumors in vivo. Moreover, TGF-β can exert a pivotal function in the immune system, as it may induce the production of regulatory T cells and suppress B cell responses. Knowledge of the molecular mechanisms involved in TGF-β regulation may help in finding effective treatments of tissue fibrosis, cancer and autoimmune disease.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Renal Unit, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
43
|
Talati N, Kamato D, Piva TJ, Little PJ, Osman N. Thrombin promotes PAI-1 expression and migration in keratinocytes via ERK dependent Smad linker region phosphorylation. Cell Signal 2018; 47:37-43. [PMID: 29577978 DOI: 10.1016/j.cellsig.2018.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 02/19/2018] [Accepted: 03/20/2018] [Indexed: 01/18/2023]
Abstract
Keratinocyte proliferation and migration is essential during re-epithelialisation for the restoration of the epithelial barrier during skin wound healing. Numerous growth factors are involved in the stimulation of keratinocyte proliferation and migration. The signalling pathways that drive these processes during wound healing are not well defined. This study investigated thrombin-mediated signalling in keratinocytes. The thrombin receptor, protease-activated receptor 1 (PAR-1) is a seven transmembrane G-protein coupled receptor that is known to transactivate the epidermal growth factor receptor (EGFR). Immortalized human keratinocytes (HaCaT cells) were treated with thrombin and selective inhibitors to EGFR and MAP kinases. Whole cell lysates were separated on SDS-PAGE and analysed by Western blot using antibodies against transcription factor Smad2. Quantitative real-time polymerase chain reaction was used to measure the mRNA expression of PAI-1 while scratch wound assays were used to measure keratinocyte migration. Western blot data showed that thrombin mediates PAR-1 transactivation of EGFR and the downstream phosphorylation of the transcription factor Smad2 linker (Smad2L) region. ERK1/2 inhibition by UO126 caused a decrease in Smad2L phosphorylation while the p38 inhibitor SB202190 and JNK inhibitor SP600125 did not. Smad2L Ser250 was specifically phosphorylated by this thrombin mediated pathway while Ser245 and Ser255 were not. Thrombin increased PAI-1 mRNA expression and keratinocyte migration and this was reduced when either EGFR or ERK1/2 were blocked. Taken together these results show that thrombin mediated mRNA expression of PAI-1 in keratinocytes and migration occurs via EGFR transactivation and involves signalling intermediates ERK1/2 and Smad2 and may be a key pathway in skin wound healing.
Collapse
Affiliation(s)
- Nirali Talati
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia
| | - Terrence J Piva
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, China
| | - Narin Osman
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Immunology, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
44
|
Yu Y, Wang X, Zhang X, Zhai Y, Lu X, Ma H, Zhu K, Zhao T, Jiao J, Zhao ZA, Li L. ERK inhibition promotes neuroectodermal precursor commitment by blocking self-renewal and primitive streak formation of the epiblast. Stem Cell Res Ther 2018; 9:2. [PMID: 29304842 PMCID: PMC5756365 DOI: 10.1186/s13287-017-0750-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/27/2022] Open
Abstract
Background Pluripotent stem cells hold great promise for regenerative medicine. However, before clinical application, reproducible protocols for pluripotent stem cell differentiation should be established. Extracellular signal-regulated protein kinase (ERK) signaling plays a central role for the self-renewal of epiblast stem cells (EpiSCs), but its role for subsequent germ layer differentiation is still ambiguous. We proposed that ERK could modulate differentiation of the epiblast. Methods PD0325901 was used to inhibit ERK activation during the differentiation of embryonic stem cells and EpiSCs. Immunofluorescence, western blot analysis, real-time PCR and flow cytometry were used to detect germ layer markers and pathway activation. Results We demonstrate that the ERK phosphorylation level is lower in neuroectoderm of mouse E7.5 embryos than that in the primitive streak. ERK inhibition results in neural lineage commitment of epiblast. Mechanistically, PD0325901 abrogates the expression of primitive streak markers by β-catenin retention in the cytoplasm, and inhibits the expression of OCT4 and NANOG during EpiSC differentiation. Thus, EpiSCs differentiate into neuroectodermal lineage efficiently under PD0325901 treatment. These results suggest that neuroectoderm differentiation does not require extrinsic signals, supporting the default differentiation of neural lineage. Conclusions We report that a single ERK inhibitor, PD0325901, can specify epiblasts and EpiSCs into neural-like cells, providing an efficient strategy for neural differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0750-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yang Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Yanhua Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Xukun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haixia Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Zhen-Ao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China. .,Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, 708 Renmin Rd, Suzhou, Jiangsu, 215007, China.
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
45
|
Chen Y, Peng FF, Jin J, Chen HM, Yu H, Zhang BF. Src-mediated ligand release-independent EGFR transactivation involves TGF-β-induced Smad3 activation in mesangial cells. Biochem Biophys Res Commun 2017; 493:914-920. [PMID: 28943431 DOI: 10.1016/j.bbrc.2017.09.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 09/21/2017] [Indexed: 11/28/2022]
Abstract
A great deal of evidence highlighted the pathophysiologic importance of TGF-β1/Smad3 pathway in masangial extracellular matrix (ECM) accumulation, but some alternative signaling pathways are also involved. TGF-β was shown recently to induce rapid and transient epidermal-like growth factor receptor (EGFR) transactivation and subsequent fibronectin expression via heparin-binding epidermal-like growth factors (HB-EGF) release and binding in mesangial cells, which is independent of Smad2 activation. However, whether TGF-β could induce persistent EGFR transactivation remains to be identified. The present study demonstrates that in addition to transient EGFR transactivation, TGF-β1 can also induce continuous EGFR transactivation by a non-ligand-dependent pathway in rat mesangial cells. This sustained EGFR transactivation is mainly due to Src kinase-mediated persistent EGFR tyrosine phosphorylation at Y845 rather than Y1173. TGF-β1-induced early Smad3 phosphorylation is independent of transient EGFR transactivation and ERK1/2 activation initiated by HB-EGF release, whereas Src-mediated chronic EGFR transactivation and ERK1/2 activation participate in Smad3 activation in a relatively modest and delayed manner. Therefore, the present study further clarifies the mechanisms of EGFR transactivation in the TGF-β-initiated ECM upregulation and raises the possibility that targeting EGFR may provide a viable alternative strategy for inhibiting TGF-β in chronic kidney disease.
Collapse
Affiliation(s)
- Yan Chen
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Fang-Fang Peng
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Jing Jin
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Hong-Min Chen
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Hong Yu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Bai-Fang Zhang
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China.
| |
Collapse
|
46
|
Kamato D, Bhaskarala VV, Mantri N, Oh TG, Ling D, Janke R, Zheng W, Little PJ, Osman N. RNA sequencing to determine the contribution of kinase receptor transactivation to G protein coupled receptor signalling in vascular smooth muscle cells. PLoS One 2017; 12:e0180842. [PMID: 28719611 PMCID: PMC5515425 DOI: 10.1371/journal.pone.0180842] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/22/2017] [Indexed: 02/02/2023] Open
Abstract
G protein coupled receptor (GPCR) signalling covers three major mechanisms. GPCR agonist engagement allows for the G proteins to bind to the receptor leading to a classical downstream signalling cascade. The second mechanism is via the utilization of the β-arrestin signalling molecule and thirdly via transactivation dependent signalling. GPCRs can transactivate protein tyrosine kinase receptors (PTKR) to activate respective downstream signalling intermediates. In the past decade GPCR transactivation dependent signalling was expanded to show transactivation of serine/threonine kinase receptors (S/TKR). Kinase receptor transactivation enormously broadens the GPCR signalling paradigm. This work utilizes next generation RNA-sequencing to study the contribution of transactivation dependent signalling to total protease activated receptor (PAR)-1 signalling. Transactivation, assessed as gene expression, accounted for 50 percent of the total genes regulated by thrombin acting through PAR-1 in human coronary artery smooth muscle cells. GPCR transactivation of PTKRs is approximately equally important as the transactivation of the S/TKR with 209 and 177 genes regulated respectively, via either signalling pathway. This work shows that genome wide studies can provide powerful insights into GPCR mediated signalling pathways.
Collapse
Affiliation(s)
- Danielle Kamato
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
- * E-mail:
| | - Venkata Vijayanand Bhaskarala
- Department of Biotechnology and Environmental Biology, School of Applied Sciences, RMIT University, Bundoora, VIC, Australia
| | - Nitin Mantri
- Department of Biotechnology and Environmental Biology, School of Applied Sciences, RMIT University, Bundoora, VIC, Australia
| | - Tae Gyu Oh
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld, Australia
| | - Dora Ling
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Reearna Janke
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Narin Osman
- School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
- Diabetes Complications Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Monash University, Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Cheng H, Yao Q, Song R, Zhai Y, Wang W, Fullerton DA, Meng X. Lysophosphatidylcholine activates the Akt pathway to upregulate extracellular matrix protein production in human aortic valve cells. J Surg Res 2017; 213:243-250. [PMID: 28601321 PMCID: PMC5469546 DOI: 10.1016/j.jss.2017.02.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Overproduction of extracellular matrix (ECM) protein by aortic valve interstitial cells (AVICs) plays an important role in valvular sclerosis (thickening) associated with the early pathobiology of aortic stenosis. Accumulation of oxidized low-density lipoprotein (oxLDL) is observed in sclerotic aortic valve and may have a mechanistic role in valvular disease progression. Lysophosphatidylcholine (LysoPC) is a component of oxLDL and has multiple biological activities. This study was to test the hypothesis that oxLDL and LysoPC upregulate ECM protein production in human AVICs. METHODS AND RESULTS AVICs were isolated from normal human aortic valves. Cells were treated with oxLDL (40 μg/mL) or LysoPC (40 μmol/L). Immunoblotting was applied to analyze ECM proteins (collagens I and III and biglycan) in cell lysate and Picrosirius red staining was used to examine collagen deposition. Both oxLDL and LysoPC upregulated the production of biglycan and collagen I. The upregulation of ECM proteins by LysoPC was preceded by the phosphorylation of Akt and ERK1/2. Inhibition of Akt markedly reduced the effect of LysoPC on ECM protein production and collagen deposition. However, inhibition of ERK1/2 had no effect. CONCLUSIONS LysoPC upregulates the production of biglycan and collagen I in human AVICs and may mediate the effect of oxLDL on ECM protein production. The Akt pathway appears to be critical in mediating the effect of LysoPC. oxLDL accumulation and generation of LysoPC in the aortic valve tissue may contribute to the mechanism of valvular sclerosis associated with the development and progression of aortic stenosis.
Collapse
Affiliation(s)
- Hui Cheng
- Department of Surgery, University of Colorado Denver, Aurora, Colorado; Department of Cardiology, Shantou University Medical College, Shantou, China
| | - Qingzhou Yao
- Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Rui Song
- Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Yufeng Zhai
- Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Wei Wang
- Department of Cardiology, Shantou University Medical College, Shantou, China
| | - David A Fullerton
- Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver, Aurora, Colorado.
| |
Collapse
|
48
|
Kamato D, Mitra P, Davis F, Osman N, Chaplin R, Cabot PJ, Afroz R, Thomas W, Zheng W, Kaur H, Brimble M, Little PJ. Ga q proteins: molecular pharmacology and therapeutic potential. Cell Mol Life Sci 2017; 74:1379-1390. [PMID: 27815595 PMCID: PMC11107756 DOI: 10.1007/s00018-016-2405-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/19/2016] [Accepted: 10/31/2016] [Indexed: 12/15/2022]
Abstract
Seven transmembrane G protein-coupled receptors (GPCRs) have gained much interest in recent years as it is the largest class among cell surface receptors. G proteins lie in the heart of GPCRs signalling and therefore can be therapeutically targeted to overcome complexities in GPCR responses and signalling. G proteins are classified into four families (Gi, Gs, G12/13 and Gq); Gq is further subdivided into four classes. Among them Gαq and Gαq/11 isoforms are most crucial and ubiquitously expressed; these isoforms are almost 88% similar at their amino acid sequence but may exhibit functional divergences. However, uncertainties often arise about Gαq and Gαq/11 inhibitors, these G proteins might also have suitability to the invention of novel-specific inhibitors for each isoforms. YM-254890 and UBO-QIC are discovered as potent inhibitors of Gαq functions and also investigated in thrombin protease-activated receptor (PAR)-1 inhibitors and platelet aggregation inhibition. The most likely G protein involved in PAR-1 stimulates responses is one of the Gαq family isoforms. In this review, we highlight the molecular structures and pharmacological responses of Gαq family which may reflect the biochemical and molecular role of Gαq and Gαq/11. The advanced understanding of Gαq and Gαq/11 role in GPCR signalling may shed light on our understanding on cell biology, cellular physiology and pathophysiology and also lead to the development of novel therapeutic agents for a number of diseases.
Collapse
Affiliation(s)
- Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Partha Mitra
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Felicity Davis
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Narin Osman
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
- School of Medical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
- Department of Immunology, Monash University, Melbounre, VIC, 3004, Australia
| | - Rebecca Chaplin
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Peter J Cabot
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia
| | - Rizwana Afroz
- Department of Biochemistry, Primeasia University, Banani, 1213, Bangladesh
| | - Walter Thomas
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD, 4102, Australia
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Harveen Kaur
- Department of Chemistry, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Margaret Brimble
- Department of Chemistry, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall Street, Woolloongabba, QLD, 4102, Australia.
- School of Medical Sciences, RMIT University, Bundoora, VIC, 3083, Australia.
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, 510520, China.
| |
Collapse
|
49
|
Insights into cellular signalling by G protein coupled receptor transactivation of cell surface protein kinase receptors. J Cell Commun Signal 2017; 11:117-125. [PMID: 28168348 DOI: 10.1007/s12079-017-0375-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/10/2017] [Indexed: 01/22/2023] Open
Abstract
G protein coupled receptor (GPCR) signalling is mediated by transactivation independent and transactivation dependent pathways. GPCRs transactivate protein tyrosine kinase receptors (PTKRs) and protein serine/threonine kinase receptors (PS/TKR). Since the initial observations of transactivation dependent signalling, there has been an effort to understand the mechanisms behind this phenomena. GPCR signalling has evolved to include biased signalling. Biased signalling, whereby selected ligands can activate the same GPCR that can generate multiple signals, but drive only a unique response. To date, there has been no focus on the ability of biased agonists to activate the PTKR and PS/TKR transactivation pathways differentially. As such, this represents a novel direction for future research. This review will discuss the main mechanisms of GPCR mediated receptor transactivation and the pathways involved in intracellular responses.
Collapse
|
50
|
Sharifat N, Mohammad Zadeh G, Ghaffari MA, Dayati P, Kamato D, Little PJ, Babaahmadi-Rezaei H. Endothelin-1 (ET-1) stimulates carboxy terminal Smad2 phosphorylation in vascular endothelial cells by a mechanism dependent on ET receptors and de novo protein synthesis. ACTA ACUST UNITED AC 2016; 69:66-72. [PMID: 27905105 DOI: 10.1111/jphp.12654] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/18/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE G protein-coupled receptor (GPCR) agonists through their receptors can transactivate protein tyrosine kinase receptors such as epidermal growth factor receptor and serine/threonine kinase receptors most notably transforming growth factor (TGF)-β receptor (TβRI). This signalling mechanism represents a major expansion in the cellular outcomes attributable to GPCR signalling. This study addressed the role and mechanisms involved in GPCR agonist, endothelin-1 (ET-1)-mediated transactivation of the TβRI in bovine aortic endothelial cells (BAECs). METHOD The in-vitro model used BAECs. Signalling intermediate phospho-Smad2 in the carboxy terminal was detected and quantified by Western blotting. KEY FINDING ET-1 treatment of BAECs resulted in a time and concentration-dependent increase in pSmad2C. Peak phosphorylation was evident with 100 nm treatment of ET-1 at 4-6 h. TβRI antagonist, SB431542 inhibited ET-1-mediated pSmad2C. In the presence of bosentan, a mixed ETA and ETB receptor antagonist ET-1-mediated pSmad2C levels were inhibited. The ET-mediated pSmad2C was blocked by the protein synthesis inhibitor, cycloheximide. CONCLUSION In BAECs, ET-1 via the ETB receptor is involved in transactivation of the TβRI. The transactivation-dependent response is dependent upon de novo protein synthesis.
Collapse
Affiliation(s)
- Narges Sharifat
- Student Research Committee, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ghorban Mohammad Zadeh
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Ali Ghaffari
- Cellular and Molecular Research Center, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Parisa Dayati
- Student Research Committee, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Danielle Kamato
- Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| | - Peter J Little
- Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| | - Hossein Babaahmadi-Rezaei
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Atherosclerosis Research Center, Ahvaz, Iran
| |
Collapse
|