1
|
Hou YM, Xu BH, Zhang QT, Cheng J, Zhang X, Yang HR, Wang ZY, Wang P, Zhang MX. Deficiency of smooth muscle cell ILF3 alleviates intimal hyperplasia via HMGB1 mRNA degradation-mediated regulation of the STAT3/DUSP16 axis. J Mol Cell Cardiol 2024; 190:62-75. [PMID: 38583797 DOI: 10.1016/j.yjmcc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Intimal hyperplasia is a complicated pathophysiological phenomenon attributable to in-stent restenosis, and the underlying mechanism remains unclear. Interleukin enhancer-binding factor 3 (ILF3), a double-stranded RNA-binding protein involved in regulating mRNA stability, has been recently demonstrated to assume a crucial role in cardiovascular disease; nevertheless, its impact on intimal hyperplasia remains unknown. In current study, we used samples of human restenotic arteries and rodent models of intimal hyperplasia, we found that vascular smooth muscle cell (VSMC) ILF3 expression was markedly elevated in human restenotic arteries and murine ligated carotid arteries. SMC-specific ILF3 knockout mice significantly suppressed injury induced neointimal formation. In vitro, platelet-derived growth factor type BB (PDGF-BB) treatment elevated the level of VSMC ILF3 in a dose- and time-dependent manner. ILF3 silencing markedly inhibited PDGF-BB-induced phenotype switching, proliferation, and migration in VSMCs. Transcriptome sequencing and RNA immunoprecipitation sequencing depicted that ILF3 maintained its stability upon binding to the mRNA of the high-mobility group box 1 protein (HMGB1), thereby exerting an inhibitory effect on the transcription of dual specificity phosphatase 16 (DUSP16) through enhanced phosphorylation of signal transducer and activator of transcription 3 (STAT3). Therefore, the results both in vitro and in vivo indicated that the loss of ILF3 in VSMC ameliorated neointimal hyperplasia by regulating the STAT3/DUSP16 axis through the degradation of HMGB1 mRNA. Our findings revealed that vascular injury activates VSMC ILF3, which in turn promotes intima formation. Consequently, targeting specific VSMC ILF3 may present a potential therapeutic strategy for ameliorating cardiovascular restenosis.
Collapse
Affiliation(s)
- Ya-Min Hou
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Bo-Han Xu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qiu-Ting Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong-Rui Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ze-Ying Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ming-Xiang Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
2
|
Pan H, Lu X, Ye D, Feng Y, Wan J, Ye J. The molecular mechanism of thrombospondin family members in cardiovascular diseases. Front Cardiovasc Med 2024; 11:1337586. [PMID: 38516004 PMCID: PMC10954798 DOI: 10.3389/fcvm.2024.1337586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiovascular diseases have been identified as vital factors in global morbidity and mortality in recent years. The available evidence suggests that various cytokines and pathological proteins participate in these complicated and changeable diseases. The thrombospondin (TSP) family is a series of conserved, multidomain calcium-binding glycoproteins that cause cell-matrix and cell-cell effects via interactions with other extracellular matrix components and cell surface receptors. The TSP family has five members that can be divided into two groups (Group A and Group B) based on their different structures. TSP-1, TSP-2, and TSP-4 are the most studied proteins. Among recent studies and findings, we investigated the functions of several family members, especially TSP-5. We review the basic concepts of TSPs and summarize the relevant molecular mechanisms and cell interactions in the cardiovascular system. Targeting TSPs in CVD and other diseases has a remarkable therapeutic benefit.
Collapse
Affiliation(s)
- Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
3
|
Azman SS, Yazid MD, Abdul Ghani NA, Raja Sabudin RZA, Abdul Rahman MR, Sulaiman N. Generation of a novel ex-vivo model to study re-endothelialization. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:408-416. [PMID: 37584645 DOI: 10.1080/21691401.2023.2245456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Endothelial dysfunction initiates the pathogenesis of a myriad of cardiovascular diseases, yet the precise underlying mechanisms remain unclear. Current model utilises mechanical denudation of arteries resulting in an arterial-injury model with onset of intimal hyperplasia (IH). Our study shows that 5 min enzymatic denudation of human umbilical artery (hUA) lumen at 37 °C efficiently denudes hUA while maintaining vessel integrity without significantly increase intima-media thickness after 7 days in culture. This ex-vivo model will be a valuable tool in understanding the mechanism of re-endothelialization prior to smooth muscle cells (SMC) activation thus placating IH at an early stage.
Collapse
Affiliation(s)
- Siti Sarah Azman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Faculty of Applied Sciences, Universiti Teknologi MARA, Perak Branch, Tapah Campus, Perak, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nur Azurah Abdul Ghani
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Raja Zahratul Azma Raja Sabudin
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| |
Collapse
|
4
|
Pervaiz N, Kathuria I, Aithabathula RV, Singla B. Matricellular proteins in atherosclerosis development. Matrix Biol 2023; 120:1-23. [PMID: 37086928 PMCID: PMC10225360 DOI: 10.1016/j.matbio.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
The extracellular matrix (ECM) is an intricate network composed of various multi-domain macromolecules like collagen, proteoglycans, and fibronectin, etc., that form a structurally stable composite, contributing to the mechanical properties of tissue. However, matricellular proteins are non-structural, secretory extracellular matrix proteins, which modulate various cellular functions via interacting with cell surface receptors, proteases, hormones, and cell-matrix. They play essential roles in maintaining tissue homeostasis by regulating cell differentiation, proliferation, adhesion, migration, and several signal transduction pathways. Matricellular proteins display a broad functionality regulated by their multiple structural domains and their ability to interact with different extracellular substrates and/or cell surface receptors. The expression of these proteins is low in adults, however, gets upregulated following injuries, inflammation, and during tumor growth. The marked elevation in the expression of these proteins during atherosclerosis suggests a positive association between their expression and atherosclerotic lesion formation. The role of matricellular proteins in atherosclerosis development has remained an area of research interest in the last two decades and studies revealed these proteins as important players in governing vascular function, remodeling, and plaque formation. Despite extensive research, many aspects of the matrix protein biology in atherosclerosis are still unknown and future studies are required to investigate whether targeting pathways stimulated by these proteins represent viable therapeutic approaches for patients with atherosclerotic vascular diseases. This review summarizes the characteristics of distinct matricellular proteins, discusses the available literature on the involvement of matrix proteins in the pathogenesis of atherosclerosis and suggests new avenues for future research.
Collapse
Affiliation(s)
- Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, USA.
| |
Collapse
|
5
|
Déglise S, Bechelli C, Allagnat F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front Physiol 2023; 13:1081881. [PMID: 36685215 PMCID: PMC9845604 DOI: 10.3389/fphys.2022.1081881] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Arterial occlusive disease is the leading cause of death in Western countries. Core contemporary therapies for this disease include angioplasties, stents, endarterectomies and bypass surgery. However, these treatments suffer from high failure rates due to re-occlusive vascular wall adaptations and restenosis. Restenosis following vascular surgery is largely due to intimal hyperplasia. Intimal hyperplasia develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel's innermost layer or intima. In this review, we describe the current state of knowledge on the origin and mechanisms underlying the dysregulated proliferation of vascular smooth muscle cells in intimal hyperplasia, and we present the new avenues of research targeting VSMC phenotype and proliferation.
Collapse
Affiliation(s)
| | | | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
6
|
Lin S, Lin R, Zhang H, Xu Q, He Y. Peripheral vascular remodeling during ischemia. Front Pharmacol 2022; 13:1078047. [DOI: 10.3389/fphar.2022.1078047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
About 230 million people worldwide suffer from peripheral arterial disease (PAD), and the prevalence is increasing year by year. Multiple risk factors, including smoking, dyslipidemia, diabetes, and hypertension, can contribute to the development of PAD. PAD is typically characterized by intermittent claudication and resting pain, and there is a risk of severe limb ischemia, leading to major adverse limb events, such as amputation. Currently, a major progress in the research field of the pathogenesis of vascular remodeling, including atherosclerosis and neointima hyperplasia has been made. For example, the molecular mechanisms of endothelial dysfunction and smooth muscle phenotype switching have been described. Interestingly, a series of focused studies on fibroblasts of the vessel wall has demonstrated their impact on smooth muscle proliferation and even endothelial function via cell-cell communications. In this review, we aim to focus on the functional changes of peripheral arterial cells and the mechanisms of the pathogenesis of PAD. At the same time, we summarize the progress of the current clinical treatment and potential therapeutic methods for PAD and shine a light on future perspectives.
Collapse
|
7
|
Macabrey D, Longchamp A, Déglise S, Allagnat F. Clinical Use of Hydrogen Sulfide to Protect Against Intimal Hyperplasia. Front Cardiovasc Med 2022; 9:876639. [PMID: 35479275 PMCID: PMC9035533 DOI: 10.3389/fcvm.2022.876639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/18/2022] [Indexed: 12/27/2022] Open
Abstract
Arterial occlusive disease is the narrowing of the arteries via atherosclerotic plaque buildup. The major risk factors for arterial occlusive disease are age, high levels of cholesterol and triglycerides, diabetes, high blood pressure, and smoking. Arterial occlusive disease is the leading cause of death in Western countries. Patients who suffer from arterial occlusive disease develop peripheral arterial disease (PAD) when the narrowing affects limbs, stroke when the narrowing affects carotid arteries, and heart disease when the narrowing affects coronary arteries. When lifestyle interventions (exercise, diet…) fail, the only solution remains surgical endovascular and open revascularization. Unfortunately, these surgeries still suffer from high failure rates due to re-occlusive vascular wall adaptations, which is largely due to intimal hyperplasia (IH). IH develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel’s innermost layer or intima. Re-occlusive IH lesions result in costly and complex recurrent end-organ ischemia, and often lead to loss of limb, brain function, or life. Despite decades of IH research, limited therapies are currently available. Hydrogen sulfide (H2S) is an endogenous gasotransmitter derived from cysteine metabolism. Although environmental exposure to exogenous high H2S is toxic, endogenous H2S has important vasorelaxant, cytoprotective and anti-inflammatory properties. Its vasculo-protective properties have attracted a remarkable amount of attention, especially its ability to inhibit IH. This review summarizes IH pathophysiology and treatment, and provides an overview of the potential clinical role of H2S to prevent IH and restenosis.
Collapse
Affiliation(s)
- Diane Macabrey
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
Zhang BH, Liu H, Yuan Y, Weng XD, Du Y, Chen H, Chen ZY, Wang L, Liu XH. Knockdown of TRIM8 Protects HK-2 Cells Against Hypoxia/Reoxygenation-Induced Injury by Inhibiting Oxidative Stress-Mediated Apoptosis and Pyroptosis via PI3K/Akt Signal Pathway. Drug Des Devel Ther 2021; 15:4973-4983. [PMID: 34916780 PMCID: PMC8670861 DOI: 10.2147/dddt.s333372] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022] Open
Abstract
Background Acute kidney injury (AKI) emerges as an acute and critical disease. Tripartite motif 8 (TRIM8), one number of the TRIM protein family, is proved to participate in ischemia/reperfusion (I/R) injury. However, whether TRIM8 is involved in renal I/R injury and the associated mechanisms are currently unclear. Purpose This study aimed to investigate the precise role of TRIM8 and relevant mechanisms in renal I/R injury. Materials and Methods In this study, human renal proximal tubular epithelial cells (HK-2 cells) underwent 12 hours of hypoxia and 2 h, 3 h or 4 h of reoxygenation to establish an in vitro hypoxia/reoxygenation (H/R) model. The siRNAs specific to TRIM8 (si-TRIM8) were transfected into HK-2 cells to knockdown TRIM8. The cell H/R model included various groups including Control, H/R, H/R+DMSO, H/R+NAC, si-NC+H/R, si-TRIM8+H/R and si-TRIM8+LY294002+H/R. The cell viability and levels of reactive oxygen species (ROS), hydrogen peroxide (H2O2), mRNA, apoptotic proteins, pyroptosis-related proteins and PI3K/AKT pathway-associated proteins were assessed. Results In vitro, realtime-quantitative PCR and western-blot analysis showed that the mRNA and protein expression of TRIM8 were obviously upregulated after H/R treatment in HK-2 cells. Compared with the H/R model group, knockdown of TRIM8 significantly increased cell viability and reduced the levels of ROS, H2O2, apoptotic proteins (Cleaved caspasebase-3 and BAX) and pyroptosis-related proteins (NLRP3, ASC, Caspase-1, Caspase-11, IL-1β and GSDMD-N). Western-blot analysis also authenticated that PI3K/AKT pathway was activated after TRIM8 inhibition. The application of 5 mM N-acetyl-cysteine, one highly efficient ROS inhibitor, significantly suppressed the expression of apoptotic proteins and pyroptosis-related proteins. Moreover, the combined treatment of TRIM8 knockdown and LY294002 reversed the effects of inhibiting oxidative stress. Conclusion Knockdown of TRIM8 can alleviate H/R-induced oxidative stress by triggering the PI3K/AKT pathway, thus attenuating pyropyosis and apoptosis in vitro.
Collapse
Affiliation(s)
- Bang-Hua Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.,Hubei Key Laboratory of Digestive System Disease, Wuhan, Hubei, People's Republic of China
| | - Hao Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.,Hubei Key Laboratory of Digestive System Disease, Wuhan, Hubei, People's Republic of China
| | - Yan Yuan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.,Hubei Key Laboratory of Digestive System Disease, Wuhan, Hubei, People's Republic of China
| | - Xiao-Dong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yang Du
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhi-Yuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
9
|
Yao Z, Bao B, Qian S, Li Z, Lu Q, Min S, Li M, Wang H. [Correlation of serum ADAMTS13 and TSP1 levels with myocardial injury and prognosis in patients with acute coronary syndrome]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:710-715. [PMID: 34134958 DOI: 10.12122/j.issn.1673-4254.2021.05.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate serum levels of von Willebrand factor lytic protease (ADAMTS13) and thrombospondin-1 (TSP1) in patients with different types of acute coronary syndrome (ACS) and their correlation with the patients' clinical prognosis. OBJECTIVE According to their disease history, results of angiography and clinical biochemical tests, a total of 405 patients undergoing coronary angiography, were divided into unstable angina (UAP) group (n=215), acute myocardial infarction (AMI) group (n=96), and angiographically normal group (n=94). Serum ADAMTS13 and TSP1 levels were detected in all the patients, who were followed up for 15 months to evaluate the occurrence of long-term major cardiac adverse events (MACE). OBJECTIVE Serum ADAMTS13 level was significantly lower and TSP1 level was significantly higher in AMI group and UAP group than in the normal group (P < 0.001). Serum ADAMTS13 and TSP1 levels were negative correlated in ACS patients (R=-0.577, P < 0.001). The patients experiencing MACE had significantly different serum TSP1 level from those without MACE (P < 0.05). Cox proportion regression model analysis showed that TSP1 was a risk factor affecting the occurrence of MACE in ACS patients; Kaplan-Meier survival analysis showed that the patients with high levels of TSP1 had a higher incidence of longterm MACE than those with low TSP1 levels. OBJECTIVE A lowered serum ADAMTS13 level and an elevated TSP1 level can support the diagnosis of ACS. An elevated TSP1 level may serve as an indicator for predicting the risk of MACE in patients with ACS.
Collapse
Affiliation(s)
- Z Yao
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - B Bao
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - S Qian
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Z Li
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Q Lu
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - S Min
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Bengbu 233000, China
| | - M Li
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - H Wang
- Department of Cardiovascular Disease, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| |
Collapse
|
10
|
Buchmann GK, Schürmann C, Spaeth M, Abplanalp W, Tombor L, John D, Warwick T, Rezende F, Weigert A, Shah AM, Hansmann ML, Weissmann N, Dimmeler S, Schröder K, Brandes RP. The hydrogen-peroxide producing NADPH oxidase 4 does not limit neointima development after vascular injury in mice. Redox Biol 2021; 45:102050. [PMID: 34218201 PMCID: PMC8256285 DOI: 10.1016/j.redox.2021.102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 11/26/2022] Open
Abstract
Objective The NADPH oxidase Nox4 is an important source of H2O2. Nox4-derived H2O2 limits vascular inflammation and promotes smooth muscle differentiation. On this basis, the role of Nox4 for restenosis development was determined in the mouse carotid artery injury model. Methods and results Genetic deletion of Nox4 by a tamoxifen-activated Cre-Lox-system did not impact on neointima formation in the carotid artery wire injury model. To understand this unexpected finding, time-resolved single-cell RNA-sequencing (scRNAseq) from injured carotid arteries of control mice and massive-analysis-of-cDNA-ends (MACE)-RNAseq from the neointima harvested by laser capture microdissection of control and Nox4 knockout mice was performed. This revealed that resting smooth muscle cells (SMCs) and fibroblasts exhibit high Nox4 expression, but that the proliferating de-differentiated SMCs, which give rise to the neointima, have low Nox4 expression. In line with this, the first weeks after injury, gene expression was unchanged between the carotid artery neointimas of control and Nox4 knockout mice. Conclusion Upon vascular injury, Nox4 expression is transiently lost in the cells which comprise the neointima. NADPH oxidase 4 therefore does not interfere with restenosis development after wire-induced vascular injury.
Collapse
Affiliation(s)
- Giulia K Buchmann
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Manuela Spaeth
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Wesley Abplanalp
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Lukas Tombor
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - David John
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London, British Heart Foundation Centre, London, UK
| | | | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Stefanie Dimmeler
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany.
| |
Collapse
|
11
|
Que Y, Shu X, Wang L, Wang S, Li S, Hu P, Tong X. Inactivation of SERCA2 Cys 674 accelerates aortic aneurysms by suppressing PPARγ. Br J Pharmacol 2021; 178:2305-2323. [PMID: 33591571 DOI: 10.1111/bph.15411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/24/2020] [Accepted: 02/04/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Inactivation of Cys674 (C674) in the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) causes intracellular Ca2+ accumulation, which activates calcineurin-mediated nuclear factor of activated T-lymphocytes (NFAT)/NF-κB pathways, and results in the phenotypic modulation of smooth muscle cells (SMCs) to accelerate angiotensin II-induced aortic aneurysms. Our goal was to investigate the mechanism involved. EXPERIMENTAL APPROACH We used heterozygous SERCA2 C674S knock-in (SKI) mice, where half of C674 was substituted by serine, to mimic partial irreversible oxidation of C674. The aortas of SKI mice and their littermate wild-type mice were collected for RNA sequencing, cell culture, protein expression, luciferase activity and aortic aneurysm analysis. KEY RESULTS Inactivation of C674 inhibited the promoter activity and protein expression of PPARγ, which could be reversed by inhibitors of calcineurin or NF-κB. In SKI SMCs, inhibition of NF-κB by pyrrolidinedithiocarbamic acid (PDTC) or overexpression of PPARγ2 reversed the protein expression of SMC phenotypic modulation markers and inhibited cell proliferation, migration, and macrophage adhesion to SMCs. Pioglitazone, a PPARγ agonist, blocked the activation of NFAT/NF-κB, reversed the protein expression of SMC phenotypic modulation markers, and inhibited cell proliferation, migration, and macrophage adhesion to SMCs in SKI SMCs. Furthermore, pioglitazone also ameliorated angiotensin II-induced aortic aneurysms in SKI mice. CONCLUSIONS AND IMPLICATIONS The inactivation of SERCA2 C674 promotes the development of aortic aneurysms by disrupting the balance between PPARγ and NFAT/NF-κB. Our study highlights the importance of C674 redox status in regulating PPARγ to maintain aortic homeostasis.
Collapse
Affiliation(s)
- Yumei Que
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xi Shu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Langtao Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Sai Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Siqi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Pingping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaoyong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
12
|
Yu W, Xiao L, Que Y, Li S, Chen L, Hu P, Xiong R, Seta F, Chen H, Tong X. Smooth muscle NADPH oxidase 4 promotes angiotensin II-induced aortic aneurysm and atherosclerosis by regulating osteopontin. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165912. [PMID: 32777344 DOI: 10.1016/j.bbadis.2020.165912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Angiotensin II (Ang II) is commonly used to induce aortic aneurysm and atherosclerosis in animal models. Ang II upregulates NADPH oxidase isoform Nox4 in aortic smooth muscle cells (SMCs) in mice. However, whether smooth muscle Nox4 is directly involved in Ang II-induced aortic aneurysm and atherosclerosis is unclear. METHODS & RESULTS To address this, we used smooth muscle-specific Nox4 dominant-negative (SDN) transgenic mice, in which Nox4 activity is constitutively inhibited. In non-transgenic (NTg) mice, Ang II increased the expression of proteins known to contribute to both aortic aneurysm and atherosclerosis, namely osteopontin (OPN), collagen type I&III (Col I&III), matrix metalloproteinase 2 (MMP2), and vascular cell adhesion molecule 1 (VCAM1), which were all significantly downregulated in SDN mice. The number and size of Ang II-induced aorta collateral aneurysms and atherosclerotic lesions in the renal artery and aortic root of SDN mice were significantly decreased compared to NTg mice, and directly correlated with a decrease in OPN expression. Replenishing OPN in SDN SMCs, increased the expression of Col I&III, MMP2, and VCAM1, and promoted SMC proliferation, migration, and inflammation. CONCLUSIONS Our data demonstrate that smooth muscle Nox4 directly promotes the development of Ang II-induced aortic aneurysm and atherosclerosis, at least in part, through regulating OPN expression.
Collapse
Affiliation(s)
- Weimin Yu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Li Xiao
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Lili Chen
- Wuhan EasyDiagnosis Biomedicine Co., Ltd., Wuhan 430075, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Rui Xiong
- Chongqing General Hospital, University of Chinese Academy of Science, Chongqing 400013, China
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hao Chen
- Chongqing General Hospital, University of Chinese Academy of Science, Chongqing 400013, China
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
13
|
Yang C, Chen Z, Yu H, Liu X. Inhibition of Disruptor of Telomeric Silencing 1-Like Alleviated Renal Ischemia and Reperfusion Injury-Induced Fibrosis by Blocking PI3K/AKT-Mediated Oxidative Stress. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:4375-4387. [PMID: 31920287 PMCID: PMC6939406 DOI: 10.2147/dddt.s224909] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/05/2019] [Indexed: 12/26/2022]
Abstract
Background Ischemia/reperfusion (I/R) injury is a major cause of acute kidney injury, usually occurs during renal surgeries, and may eventually lead to chronic kidney diseases. However, effective therapeutic targets for renal I/R injury remain limited. Purpose In the present study, we investigated whether inhibition of disruptor of telomeric silencing 1-like (Dot1l) could alleviate renal I/R in vivo and in vitro, as well as the potential mechanisms involved in this process. Methods Sprague–Dawley rats were subjected to right renal ischemia for 45 mins and reperfusion for 0, 7, or 14 days with and without the Dot1l inhibitor EPZ004777. In addition, human renal proximal tubular epithelial cell line human kidney-2 cells were subjected to the hypoxia/reoxygenation (H/R) process (ie, 3 hrs hypoxia, 12 hrs and 24 hrs reoxygenation), with or without Dot1l inhibitor or genetic knockdown. Results Inhibition of Dot1l through EPZ004777 or genetic knockdown reduced the expression of alpha-smooth muscle actin, vimentin, and fibronectin in I/R- and H/R-induced injury. Moreover, H/R-induced fibrosis depended on oxidative stress in vitro. In addition, I/R- and H/R-induced generation of reactive oxygen species (ROS) was attenuated by EPZ004777 or small interfering RNA for Dot1l. Furthermore, the elevation of ROS induced by Dot1l was regulated via phosphatidylinositol 3-kinase (PI3K) and serine-threonine protein kinase (AKT) phosphorylation in vivo and in vitro. Conclusion Inhibition of Dot1l alleviated renal fibrosis by preventing the generation of ROS via the PI3K/AKT pathway. These results indicate that inhibitor of Dot1l could be a potential therapeutic target for renal I/R injury.
Collapse
Affiliation(s)
- Chuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Department of Urology, The People's Hospital of Hanchuan City, Hanchuan, People's Republic of China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Hua Yu
- Department of Urology, The People's Hospital of Hanchuan City, Hanchuan, People's Republic of China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
14
|
Inhibition of PRMT5 Attenuates Oxidative Stress-Induced Pyroptosis via Activation of the Nrf2/HO-1 Signal Pathway in a Mouse Model of Renal Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2345658. [PMID: 31885778 PMCID: PMC6899313 DOI: 10.1155/2019/2345658] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/11/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023]
Abstract
Background Extensive evidence has demonstrated that oxidative stress, pyroptosis, and proinflammatory programmed cell death are related to renal ischemia/reperfusion (I/R) injury. However, the underlying mechanism remains to be illustrated. Protein arginine methylation transferase 5 (PRMT5), which mediates arginine methylation involved in the regulation of epigenetics, exhibits a variety of biological functions and essential roles in diseases. The present study investigated the role of PRMT5 in oxidative stress and pyroptosis induced by I/R injury in a mouse model and in a hypoxia/reoxygenation (H/R) model of HK-2 cells. Methods C57 mice were used as an animal model. All mice underwent right nephrectomy, and the left renal pedicles were either clamped or not. Renal I/R injury was induced by ligating the left renal pedicle for 30 min followed by reperfusion for 24 h. HK-2 cells were exposed to normal conditions or stimulation through H/R. EPZ015666(EPZ)—a selective potent chemical inhibitor—and small interfering RNA (siRNA) were administered to suppress the function and expression of PRMT5. The levels of urea nitrogen and creatinine in the serum and renal tissue injury were assessed. Immunohistochemistry, western blotting, and reverse transcription-polymerase chain reaction were used to evaluate pyroptosis-related proteins including nod-like receptor protein-3, ASC, caspase-1, caspase-11, GSDMD-N, and interleukin-1β. Cell apoptosis and cell viability were detected through flow cytometry, and the levels of reactive oxygen species (ROS) and hydrogen peroxide (H2O2) were measured. Ki-67 was used to assess the proliferation of renal tubular epithelium. In addition, the activity of malondialdehyde and superoxide dismutase was determined. Results I/R or H/R induced an increase in the expression of PRMT5. Inhibition of PRMT5 by EPZ alleviated oxidative stress and I/R- or H/R-induced pyroptosis. In renal tissue, the application of EPZ promoted the proliferation of tubular epithelium. In addition, H/R-induced pyroptosis in HK-2 cells was dependent on oxidative stress in vitro. Administration of either EPZ or siRNA led to decreased expression of pyroptosis-related proteins. Inhibition of PRMT5 also attenuated the I/R- or H/R-induced oxidative stress in vivo and in HK-2 cells, respectively. It also resulted in a distinct decrease in the levels of malondialdehyde and H2O2, and an apparent increase in superoxide dismutase activity in mouse renal tissue. Moreover, it led to a significant decrease in the levels of ROS and H2O2 in HK-2 cells. When activated, NF-E2-related factor/heme oxygenase-1 (Nrf2/HO-1)—a key regulator of various cytoprotective proteins that withstand oxidative damage—can decrease the generation of ROS. Nrf2/HO-1 was downregulated during I/R in tissues and H/R in HK-2 cells, and this effect was reversed by the PRMT5 inhibitor. Furthermore, the expressions of Nrf2 and HO-1 proteins were markedly upregulated by EPZ or siRNA against PRMT5. Conclusion PRMT5 is involved in ischemia- and hypoxia-induced oxidative stress and pyroptosis in vitro and in vivo. Inhibition of PRMT5 may ameliorate renal I/R injury by suppressing oxidative stress and pyroptosis via the activation of the Nrf2/HO-1 pathway, as well as promoting the proliferation of tubular epithelium. Therefore, PRMT5 may be a promising therapeutic target.
Collapse
|
15
|
Zhang WX, Tai GJ, Li XX, Xu M. Inhibition of neointima hyperplasia by the combined therapy of linagliptin and metformin via AMPK/Nox4 signaling in diabetic rats. Free Radic Biol Med 2019; 143:153-163. [PMID: 31369842 DOI: 10.1016/j.freeradbiomed.2019.07.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/06/2019] [Accepted: 07/28/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND Neointima hyperplasia is the pathological basis of atherosclerosis and restenosis which have been associated with diabetes mellitus (DM). It is controversial for linagliptin and metformin to protect against vascular neointimal hyperplasia caused by DM. Given the combined therapy of linagliptin and metformin in clinical practice, we investigated whether the combination therapy inhibited neointimal hyperplasia in the carotid artery in diabetic rats. METHODS AND RESULTS Neointima hyperplasia in the carotid artery was induced by balloon-injury in the rats fed with high fat diet (HFD) combined with low dose streptozotocin (STZ) administration. In vitro, vascular smooth muscle cells (VSMCs) were incubated with high glucose (HG, 30 mM) and the proliferation, migration, apoptosis and collagen deposition were analyzed in VSMCs. We found that the combined therapy, not the monotherapy of linagliptin and metformin significantly inhibited the neointima hyperplasia and improved the endothelium-independent contraction in the balloon-injured cardia artery of diabetic rats, which was associated with the inhibition of superoxide (O2-.) production in the cardia artery. In vitro, HG-induced VSMC remodeling was shown as the remarkable upregulation of PCNA, collagan1, MMP-9, Bcl-2 and migration rate as well as the decreased apoptosis rate. Such abnormal changes were dramatically reversed by the combined use of linagliptin and metformin. Moreover, the AMP-activated protein kinase (AMPK)/Nox4 signal pathway was found to mediate VSMC remodeling responding to HG. Linagliptin and metformin were synergistical to target AMPK/Nox4 signal pathway in VSMCs incubated with HG and in the cardia artery of diabetic rats, which was superior to the monotherapy. CONCLUSIONS We demonstrated that the potential protection of the combined use of linagliptin and metformin on VSMC remodeling through AMPK/Nox4 signal pathway, resulting in the improvement of neointima hyperplasia in diabetic rats. This study provided new therapeutic strategies for vascular stenosis associated with diabetes.
Collapse
Affiliation(s)
- Wen-Xu Zhang
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiao-Xue Li
- Department of Pharmacology, Southeast University School of Medicine, Nanjing, 210009, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Burtenshaw D, Kitching M, Redmond EM, Megson IL, Cahill PA. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. Front Cardiovasc Med 2019; 6:89. [PMID: 31428618 PMCID: PMC6688526 DOI: 10.3389/fcvm.2019.00089] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Arteriosclerosis causes significant morbidity and mortality worldwide. Central to this process is the development of subclinical non-atherosclerotic intimal lesions before the appearance of pathologic intimal thickening and advanced atherosclerotic plaques. Intimal thickening is associated with several risk factors, including oxidative stress due to reactive oxygen species (ROS), inflammatory cytokines and lipid. The main ROS producing systems in-vivo are reduced nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX). ROS effects are context specific. Exogenous ROS induces apoptosis and senescence, whereas intracellular ROS promotes stem cell differentiation, proliferation, and migration. Lineage tracing studies using murine models of subclinical atherosclerosis have revealed the contributory role of medial smooth muscle cells (SMCs), resident vascular stem cells, circulating bone-marrow progenitors and endothelial cells that undergo endothelial-mesenchymal-transition (EndMT). This review will address the putative physiological and patho-physiological roles of ROS in controlling vascular cell fate and ROS contribution to vascular regeneration and disease progression.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Ian L Megson
- Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Paul A Cahill
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
17
|
Inhibition of Brd4 alleviates renal ischemia/reperfusion injury-induced apoptosis and endoplasmic reticulum stress by blocking FoxO4-mediated oxidative stress. Redox Biol 2019; 24:101195. [PMID: 31004990 PMCID: PMC6475721 DOI: 10.1016/j.redox.2019.101195] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 01/09/2023] Open
Abstract
Ischemia/reperfusion injury (I/R) is one of the leading causes of acute kidney injury (AKI) that typically occurs in renal surgeries. However, renal I/R still currently lacks effective therapeutic targets. In this study, we proved that inhibition of Brd4 with its selective inhibitor, JQ1, could exert a protective role in renal I/R injury in mice. Inhibiting Brd4 with either JQ1 or genetic knockdown resulted in reduction of endoplasmic reticulum stress (ERS)-associated protein and proapoptotic protein expression both in I/R-induced injury and hypoxia/reoxygenation (H/R) stimulation in HK-2 cells. H/R-induced apoptosis and ERS depended on oxidative stress in vitro. Moreover, FoxO4, which is involved in the generation of hydrogen peroxide, was up-regulated during H/R stimulation-mediated apoptosis and ERS, and this upregulation could be abolished by Brd4 inhibition. Consistently, FoxO4-mediated ROS generation was attenuated upon inhibition of Brd4 with JQ1 or siRNA against Brd4. Further, the transcriptional activity of FoxO4 was suppressed by PI3K and AKT phosphorylation, which are upstream signals of FoxO4 expression, and were enhanced by Brd4 both in vivo and in vitro. In conclusion, our results proved that Brd4 inhibition blocked renal apoptotic and ERS protein expression by preventing FoxO4-dependent ROS generation through the PI3K/AKT pathway, indicating that Brd4 could be a potential therapeutic target for renal I/R injury. Brd4 was up-regulated in renal I/R injury. Brd4 inhibitor JQ1 alleviated renal I/R injury. Brd4 inhibition blocked H/R-induced oxidative stress, apoptosis and ERS through FoxO4. Brd4 regulated FoxO4 through the PI3K/AKT pathway.
Collapse
|
18
|
Wu W, Zhang W, Choi M, Zhao J, Gao P, Xue M, Singer HA, Jourd'heuil D, Long X. Vascular smooth muscle-MAPK14 is required for neointimal hyperplasia by suppressing VSMC differentiation and inducing proliferation and inflammation. Redox Biol 2019; 22:101137. [PMID: 30771750 PMCID: PMC6377391 DOI: 10.1016/j.redox.2019.101137] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
Injury-induced stenosis is a serious vascular complication. We previously reported that p38α (MAPK14), a redox-regulated p38MAPK family member was a negative regulator of the VSMC contractile phenotype in vitro. Here we evaluated the function of VSMC-MAPK14 in vivo in injury-induced neointima hyperplasia and the underlying mechanism using an inducible SMC-MAPK14 knockout mouse line (iSMC-MAPK14-/-). We show that MAPK14 expression and activity were induced in VSMCs after carotid artery ligation injury in mice and ex vivo cultured human saphenous veins. While the vasculature from iSMC-MAPK14-/- mice was indistinguishable from wildtype littermate controls at baseline, these mice exhibited reduced neointima formation following carotid artery ligation injury. Concomitantly, there was an increased VSMC contractile protein expression in the injured vessels and a decrease in proliferating cells. Blockade of MAPK14 through a selective inhibitor suppressed, while activation of MAPK14 by forced expression of an upstream MAPK14 kinase promoted VSMC proliferation in cultured VSMCs. Genome wide RNA array combined with VSMC lineage tracing studies uncovered that vascular injury evoked robust inflammatory responses including the activation of proinflammatory gene expression and accumulation of CD45 positive inflammatory cells, which were attenuated in iSMC-MAPK14-/- mice. Using multiple pharmacological and molecular approaches to manipulate MAPK14 pathway, we further confirmed the critical role of MAPK14 in activating proinflammatory gene expression in cultured VSMCs, which occurs in a p65/NFkB-dependent pathway. Finally, we found that NOX4 contributes to MAPK14 suppression of the VSMC contractile phenotype. Our results revealed that VSMC-MAPK14 is required for injury-induced neointima formation, likely through suppressing VSMC differentiation and promoting VSMC proliferation and inflammation. Our study will provide mechanistic insights into therapeutic strategies for mitigation of vascular stenosis.
Collapse
Affiliation(s)
- Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Wei Zhang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Mihyun Choi
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Min Xue
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States.
| |
Collapse
|
19
|
Liu X, Qin Z, Liu C, Song M, Luo X, Zhao H, Qian D, Chen J, Huang L. Nox4 and soluble epoxide hydrolase synergistically mediate homocysteine-induced inflammation in vascular smooth muscle cells. Vascul Pharmacol 2019; 120:106544. [PMID: 30610956 DOI: 10.1016/j.vph.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/05/2018] [Accepted: 01/01/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Hyperhomocysteinemia leads to a vascular smooth muscle cell (VSMC) inflammatory response. Meanwhile, Nox4 dependent reactive oxygen species (ROS) signaling and soluble epoxide hydrolase (sEH)/epoxyeicosatrienoic acids (EETs) are both involved in vascular inflammation. Herein, we hypothesized that Nox4 and soluble epoxide hydrolase cross regulated during homocysteine-induced VSMC inflammation. METHODS AND RESULTS In cultured VSMCs, the expression of the inflammatory factors VCAM1 and ICAM1 was measured by real-time PCR and Western blotting, while supernatant MCP1 was measured by ELISA. Upon VSMC stimulation with 50 μΜ homocysteine, we observed the VCAM1 and ICAM1 mRNA levels were increased by 1.15 and 1.0 folds, respectively. The MCP1 levels in the supernatant of cultured VSMCs treated with 100 μΜ increased to 1.76 folds. As expected, homocysteine induced Nox4 expression and Nox4-dependent ROS generation. The sEH expression was also upregulated in the presence of homocysteine in a dose-dependent manner. Furthermore, we knocked down Nox4 with siRNA. Knockdown of Nox4 decreased ROS generation and homocysteine-induced sEH expression. Overexpression of Nox4 with an adenovirus stimulated sEH expression. Similarly, knockdown or chemical inhibition of sEH blunted the upregulation of Nox4 by homocysteine. In vivo, in homocysteine-fed mice, concomitant upregulation of Nox4 and sEH was associated with increased VCAM1 and ICAM1 expression in the aortic wall. CONCLUSIONS The inflammatory response induced by homocysteine in VSMCs was accompanied by Nox4 and sEH upregulation. Nox4 and soluble epoxide hydrolase synergistically contribute to homocysteine-induced inflammation.
Collapse
Affiliation(s)
- Xi Liu
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Mingbao Song
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiaolin Luo
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Hongqing Zhao
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Dehui Qian
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jianfei Chen
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| |
Collapse
|
20
|
Durgin BG, Straub AC. Redox control of vascular smooth muscle cell function and plasticity. J Transl Med 2018; 98:1254-1262. [PMID: 29463879 PMCID: PMC6102093 DOI: 10.1038/s41374-018-0032-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (SMC) play a major role in vascular diseases, such as atherosclerosis and hypertension. It has long been established in vitro that contractile SMC can phenotypically switch to function as proliferative and/or migratory cells in response to stimulation by oxidative stress, growth factors, and inflammatory cytokines. Reactive oxygen species (ROS) are oxidative stressors implicated in driving vascular diseases, shifting cell bioenergetics, and increasing SMC proliferation, migration, and apoptosis. In this review, we summarize our current knowledge of how disruptions to redox balance can functionally change SMC and how this may influence vascular disease pathogenesis. Specifically, we focus on our current understanding of the role of vascular nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) 1, 4, and 5 in SMC function. We also review the evidence implicating mitochondrial fission in SMC phenotypic transitions and mitochondrial fusion in maintenance of SMC homeostasis. Finally, we discuss the importance of the redox regulation of the soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway as a potential oxidative and therapeutic target for regulating SMC function.
Collapse
Affiliation(s)
- Brittany G Durgin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Jiménez-Altayó F, Meirelles T, Crosas-Molist E, Sorolla MA, Del Blanco DG, López-Luque J, Mas-Stachurska A, Siegert AM, Bonorino F, Barberà L, García C, Condom E, Sitges M, Rodríguez-Pascual F, Laurindo F, Schröder K, Ros J, Fabregat I, Egea G. Redox stress in Marfan syndrome: Dissecting the role of the NADPH oxidase NOX4 in aortic aneurysm. Free Radic Biol Med 2018; 118:44-58. [PMID: 29471108 DOI: 10.1016/j.freeradbiomed.2018.02.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 11/16/2022]
Abstract
Marfan syndrome (MFS) is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix fibrillin-containing microfibrils and dysfunction of TGF-β signaling. Here we identify the molecular targets of redox stress in aortic aneurysms from MFS patients, and investigate the role of NOX4, whose expression is strongly induced by TGF-β, in aneurysm formation and progression in a murine model of MFS. Working models included aortae and cultured vascular smooth muscle cells (VSMC) from MFS patients, and a NOX4-deficient Marfan mouse model (Fbn1C1039G/+-Nox4-/-). Increased tyrosine nitration and reactive oxygen species levels were found in the tunica media of human aortic aneurysms and in cultured VSMC. Proteomic analysis identified nitrated and carbonylated proteins, which included smooth muscle α-actin (αSMA) and annexin A2. NOX4 immunostaining increased in the tunica media of human Marfan aorta and was transcriptionally overexpressed in VSMC. Fbn1C1039G/+-Nox4-/- mice aortas showed a reduction of fragmented elastic fibers, which was accompanied by an amelioration in the Marfan-associated enlargement of the aortic root. Increase in the contractile phenotype marker calponin in the tunica media of MFS mice aortas was abrogated in Fbn1C1039G/+-Nox4-/- mice. Endothelial dysfunction evaluated by myography in the Marfan ascending aorta was prevented by the absence of Nox4 or catalase-induced H2O2 decomposition. We conclude that redox stress occurs in MFS, whose targets are actin-based cytoskeleton members and regulators of extracellular matrix homeostasis. Likewise, NOX4 have an impact in the progression of the aortic dilation in MFS and in the structural organization of the aortic tunica media, the VSMC phenotypic modulation, and endothelial function.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Eva Crosas-Molist
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Alba Sorolla
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Darya Gorbenko Del Blanco
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Judit López-Luque
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Ana-Maria Siegert
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Fabio Bonorino
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Laura Barberà
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Carolina García
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Enric Condom
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Marta Sitges
- Cardiovascular Institute, Hospital Clinic, IDIBAPS-University of Barcelona, Barcelona, Spain
| | | | - Francisco Laurindo
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Katrin Schröder
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Joaquim Ros
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Gustavo Egea
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain.
| |
Collapse
|
22
|
Roberts DD, Kaur S, Isenberg JS. Regulation of Cellular Redox Signaling by Matricellular Proteins in Vascular Biology, Immunology, and Cancer. Antioxid Redox Signal 2017; 27:874-911. [PMID: 28712304 PMCID: PMC5653149 DOI: 10.1089/ars.2017.7140] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE In contrast to structural elements of the extracellular matrix, matricellular proteins appear transiently during development and injury responses, but their sustained expression can contribute to chronic disease. Through interactions with other matrix components and specific cell surface receptors, matricellular proteins regulate multiple signaling pathways, including those mediated by reactive oxygen and nitrogen species and H2S. Dysregulation of matricellular proteins contributes to the pathogenesis of vascular diseases and cancer. Defining the molecular mechanisms and receptors involved is revealing new therapeutic opportunities. Recent Advances: Thrombospondin-1 (TSP1) regulates NO, H2S, and superoxide production and signaling in several cell types. The TSP1 receptor CD47 plays a central role in inhibition of NO signaling, but other TSP1 receptors also modulate redox signaling. The matricellular protein CCN1 engages some of the same receptors to regulate redox signaling, and ADAMTS1 regulates NO signaling in Marfan syndrome. In addition to mediating matricellular protein signaling, redox signaling is emerging as an important pathway that controls the expression of several matricellular proteins. CRITICAL ISSUES Redox signaling remains unexplored for many matricellular proteins. Their interactions with multiple cellular receptors remains an obstacle to defining signaling mechanisms, but improved transgenic models could overcome this barrier. FUTURE DIRECTIONS Therapeutics targeting the TSP1 receptor CD47 may have beneficial effects for treating cardiovascular disease and cancer and have recently entered clinical trials. Biomarkers are needed to assess their effects on redox signaling in patients and to evaluate how these contribute to their therapeutic efficacy and potential side effects. Antioxid. Redox Signal. 27, 874-911.
Collapse
Affiliation(s)
- David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey S. Isenberg
- Division of Pulmonary, Allergy and Critical Care, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
23
|
Labrousse-Arias D, Martínez-Ruiz A, Calzada MJ. Hypoxia and Redox Signaling on Extracellular Matrix Remodeling: From Mechanisms to Pathological Implications. Antioxid Redox Signal 2017; 27:802-822. [PMID: 28715969 DOI: 10.1089/ars.2017.7275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) is an essential modulator of cell behavior that influences tissue organization. It has a strong relevance in homeostasis and translational implications for human disease. In addition to ECM structural proteins, matricellular proteins are important regulators of the ECM that are involved in a myriad of different pathologies. Recent Advances: Biochemical studies, animal models, and study of human diseases have contributed to the knowledge of molecular mechanisms involved in remodeling of the ECM, both in homeostasis and disease. Some of them might help in the development of new therapeutic strategies. This review aims to review what is known about some of the most studied matricellular proteins and their regulation by hypoxia and redox signaling, as well as the pathological implications of such regulation. CRITICAL ISSUES Matricellular proteins have complex regulatory functions and are modulated by hypoxia and redox signaling through diverse mechanisms, in some cases with controversial effects that can be cell or tissue specific and context dependent. Therefore, a better understanding of these regulatory processes would be of great benefit and will open new avenues of considerable therapeutic potential. FUTURE DIRECTIONS Characterizing the specific molecular mechanisms that modulate matricellular proteins in pathological processes that involve hypoxia and redox signaling warrants additional consideration to harness the potential therapeutic value of these regulatory proteins. Antioxid. Redox Signal. 27, 802-822.
Collapse
Affiliation(s)
- David Labrousse-Arias
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain
| | - Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,2 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) , Madrid, Spain
| | - María J Calzada
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) , Madrid, Spain .,3 Departmento de Medicina, Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
24
|
Tong X, Hou X, Wason C, Kopel T, Cohen RA, Dember LM. Smooth Muscle Nitric Oxide Responsiveness and Clinical Maturation of Hemodialysis Arteriovenous Fistulae. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2095-2101. [PMID: 28822538 DOI: 10.1016/j.ajpath.2017.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/02/2017] [Indexed: 01/08/2023]
Abstract
The arteriovenous fistula is the preferred type of hemodialysis vascular access for patients with end-stage renal disease, but a high proportion of newly created fistulas fail to mature for use. Stenosis caused by neointimal hyperplasia often is present in fistulas with maturation failure, suggesting that local mechanisms controlling vascular smooth muscle cell (SMC) migration and proliferation are important contributors to maturation failure. SMCs cultured from explants of vein tissue obtained at the time of fistula creation from 19 patients with end-stage renal disease were studied to determine whether smooth muscle responsiveness to nitric oxide is associated with fistula maturation outcomes. Nitric oxide-induced inhibition of smooth muscle cell migration, but not proliferation, was greater in cells from patients with subsequent fistula maturation success than from patients with subsequent fistula maturation failure (mean inhibition percentage, 17 versus 5.7, respectively; P = 0.035). Impaired nitric oxide responsiveness was associated with oxidation of the calcium regulatory protein, sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA), and was reversed by overexpressing SERCA (1.8-fold increase in inhibition, P = 0.0128) or down-regulating Nox4-based NADPH oxidase (2.3-fold increase in inhibition; P = 0.005). Our data suggest that the nitric oxide responsiveness of SMC migration is associated with fistula maturation success and raises the possibility that therapeutic restoration of nitric oxide responsiveness through manipulation of local mediators may prevent fistula maturation failure.
Collapse
Affiliation(s)
- Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing, China; Vascular Biology Section, Boston University Medical Center, Boston, Massachusetts.
| | - Xiuyun Hou
- Vascular Biology Section, Boston University Medical Center, Boston, Massachusetts
| | - Christopher Wason
- Vascular Biology Section, Boston University Medical Center, Boston, Massachusetts
| | - Tal Kopel
- Nephrology Division, University of Montreal Hospital Centre, Hopital Saint-Luc, Montreal, Quebec, Canada
| | - Richard A Cohen
- Vascular Biology Section, Boston University Medical Center, Boston, Massachusetts
| | - Laura M Dember
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
25
|
Tanaka LY, Laurindo FRM. Vascular remodeling: A redox-modulated mechanism of vessel caliber regulation. Free Radic Biol Med 2017; 109:11-21. [PMID: 28109889 DOI: 10.1016/j.freeradbiomed.2017.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/13/2017] [Accepted: 01/15/2017] [Indexed: 11/17/2022]
Abstract
Vascular remodeling, i.e. whole-vessel structural reshaping, determines lumen caliber in (patho)physiology. Here we review mechanisms underlying vessel remodeling, with emphasis in redox regulation. First, we discuss confusing terminology and focus on strictu sensu remodeling. Second, we propose a mechanobiological remodeling paradigm based on the concept of tensional homeostasis as a setpoint regulator. We first focus on shear-mediated models as prototypes of remodeling closely dominated by highly redox-sensitive endothelial function. More detailed discussions focus on mechanosensors, integrins, extracellular matrix, cytoskeleton and inflammatory pathways as potential of mechanisms potentially coupling tensional homeostasis to redox regulation. Further discussion of remodeling associated with atherosclerosis and injury repair highlights important aspects of redox vascular responses. While neointima formation has not shown consistent responsiveness to antioxidants, vessel remodeling has been more clearly responsive, indicating that despite the multilevel redox signaling pathways, there is a coordinated response of the whole vessel. Among mechanisms that may orchestrate redox pathways, we discuss roles of superoxide dismutase activity and extracellular protein disulfide isomerase. We then discuss redox modulation of aneurysms, a special case of expansive remodeling. We propose that the redox modulation of vascular remodeling may reflect (1) remodeling pathophysiology is dominated by a particularly redox-sensitive cell type, e.g., endothelial cells (2) redox pathways are temporospatially coordinated at an organ level across distinct cellular and acellular structures or (3) the tensional homeostasis setpoint is closely connected to redox signaling. The mechanobiological/redox model discussed here can be a basis for improved understanding of remodeling and helps clarifying mechanisms underlying prevalent hard-to-treat diseases.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil.
| |
Collapse
|
26
|
Thrombospondins: A Role in Cardiovascular Disease. Int J Mol Sci 2017; 18:ijms18071540. [PMID: 28714932 PMCID: PMC5536028 DOI: 10.3390/ijms18071540] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Thrombospondins (TSPs) represent extracellular matrix (ECM) proteins belonging to the TSP family that comprises five members. All TSPs have a complex multidomain structure that permits the interaction with various partners including other ECM proteins, cytokines, receptors, growth factors, etc. Among TSPs, TSP1, TSP2, and TSP4 are the most studied and functionally tested. TSP1 possesses anti-angiogenic activity and is able to activate transforming growth factor (TGF)-β, a potent profibrotic and anti-inflammatory factor. Both TSP2 and TSP4 are implicated in the control of ECM composition in hypertrophic hearts. TSP1, TSP2, and TSP4 also influence cardiac remodeling by affecting collagen production, activity of matrix metalloproteinases and TGF-β signaling, myofibroblast differentiation, cardiomyocyte apoptosis, and stretch-mediated enhancement of myocardial contraction. The development and evaluation of TSP-deficient animal models provided an option to assess the contribution of TSPs to cardiovascular pathology such as (myocardial infarction) MI, cardiac hypertrophy, heart failure, atherosclerosis, and aortic valve stenosis. Targeting of TSPs has a significant therapeutic value for treatment of cardiovascular disease. The activation of cardiac TSP signaling in stress and pressure overload may be therefore beneficial.
Collapse
|
27
|
Forte M, Palmerio S, Yee D, Frati G, Sciarretta S. Functional Role of Nox4 in Autophagy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:307-326. [DOI: 10.1007/978-3-319-55330-6_16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
Nowak WN, Deng J, Ruan XZ, Xu Q. Reactive Oxygen Species Generation and Atherosclerosis. Arterioscler Thromb Vasc Biol 2017; 37:e41-e52. [DOI: 10.1161/atvbaha.117.309228] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Witold N. Nowak
- From the Cardiovascular Division, King’s BHF Centre, King’s College London, United Kingdom (W.N.N., J.D., Q.X.); Centre for Nephrology and Urology, Health Science Centre, Shenzhen University, China (X.Z.R.); and Centre for Nephrology, University College London, United Kingdom (X.Z.R.)
| | - Jiacheng Deng
- From the Cardiovascular Division, King’s BHF Centre, King’s College London, United Kingdom (W.N.N., J.D., Q.X.); Centre for Nephrology and Urology, Health Science Centre, Shenzhen University, China (X.Z.R.); and Centre for Nephrology, University College London, United Kingdom (X.Z.R.)
| | - Xiong Z. Ruan
- From the Cardiovascular Division, King’s BHF Centre, King’s College London, United Kingdom (W.N.N., J.D., Q.X.); Centre for Nephrology and Urology, Health Science Centre, Shenzhen University, China (X.Z.R.); and Centre for Nephrology, University College London, United Kingdom (X.Z.R.)
| | - Qingbo Xu
- From the Cardiovascular Division, King’s BHF Centre, King’s College London, United Kingdom (W.N.N., J.D., Q.X.); Centre for Nephrology and Urology, Health Science Centre, Shenzhen University, China (X.Z.R.); and Centre for Nephrology, University College London, United Kingdom (X.Z.R.)
| |
Collapse
|
29
|
He YH, Wang XQ, Zhang J, Liu ZH, Pan WQ, Shen Y, Zhu ZB, Wang LJ, Yan XX, Yang K, Zhang RY, Shen WF, Ding FH, Lu L. Association of Serum HMGB2 Levels With In-Stent Restenosis. Arterioscler Thromb Vasc Biol 2017; 37:717-729. [DOI: 10.1161/atvbaha.116.308210] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Objective—
In a previous study, we established diabetic and nondiabetic minipig models with coronary artery in-stent restenosis (ISR). Mass spectrometry showed that high-mobility group box (HMGB) 2 level was higher in ISR than in non-ISR tissue from diabetic minipigs. We here investigated whether serum HMGB2 levels were related to ISR in coronary artery disease patients. The effect of HMGB2 was evaluated in mice with femoral artery wire injury and in human aortic smooth muscle cells.
Approach and Results—
From 2513 patients undergoing coronary artery intervention and follow-up angiography at ≈1 year, 262 patients were diagnosed with ISR, and 298 patients with no ISR were randomly included as controls. Serum HMGB2 levels were significantly higher in patients with ISR than in those without ISR and were associated with ISR severity. Multivariable logistic regression analysis showed that HMGB2 level was independently associated with ISR. In experiments, HMGB2 expression was increased in vascular tissue after injury. Perivascular HMGB2 administration promoted injury-induced neointimal hyperplasia in C57Bl/6 mice compared with in the control, whereas such pathophysiological features were attenuated in
Hmgb2
–/–
mice. Mechanistically, HMGB2 enhanced neointimal hyperplasia in mice and proliferation and migration in human aortic smooth muscle cells by inducing reactive oxygen species through increased p47phox phosphorylation. Knocking down p47phox, however, inhibited HMGB2-induced effects in human aortic smooth muscle cells. Finally, HMGB2-induced effects were significantly declined in receptor of advanced glycation end products knockdown or deficient cells, but not in Toll-like receptor 4 knockdown or deficient cells.
Conclusions—
Serum HMGB2 levels were associated with ISR in patients. HMGB2 promoted neointimal hyperplasia in mice with arterial wire injury through reactive oxygen species activation.
Collapse
Affiliation(s)
- Yu Hu He
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Xiao Qun Wang
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Jian Zhang
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Zhu Hui Liu
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Wen Qi Pan
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Ying Shen
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Zheng Bin Zhu
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Ling Jie Wang
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Xiao Xiang Yan
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Ke Yang
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Rui Yan Zhang
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Wei Feng Shen
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Feng Hua Ding
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| | - Lin Lu
- From the Department of Cardiology, Rui Jin Hospital (Y.H.H., X.Q.W., Z.H.L., W.Q.P., Y.S., Z.B.Z., L.J.W., X.X.Y., R.Y.Z., W.F.S., F.H.D., L.L.) and Institute of Cardiovascular Diseases (Y.H.H., X.Q.W., Z.H.L., L.J.W., X.X.Y., K.Y., W.F.S., L.L.), Shanghai Jiaotong University School of Medicine, China; and Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (J.Z.)
| |
Collapse
|
30
|
Hu P, Wu X, Khandelwal AR, Yu W, Xu Z, Chen L, Yang J, Weisbrod RM, Lee KSS, Seta F, Hammock BD, Cohen RA, Zeng C, Tong X. Endothelial Nox4-based NADPH oxidase regulates atherosclerosis via soluble epoxide hydrolase. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1382-1391. [PMID: 28185955 DOI: 10.1016/j.bbadis.2017.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/05/2017] [Accepted: 02/02/2017] [Indexed: 12/17/2022]
Abstract
Nox4-based NADPH oxidase is a major reactive oxygen species-generating enzyme in the vasculature, but its role in atherosclerosis remains controversial. OBJECTIVE Our goal was to investigate the mechanisms of endothelial Nox4 in regulating atherosclerosis. APPROACH AND RESULTS Atherosclerosis-prone conditions (disturbed blood flow, type I diabetes, and Western diet) downregulated endothelial Nox4 mRNA in arteries. To address whether the downregulated endothelial Nox4 was directly involved in the development of atherosclerosis, we generated mice carrying a human Nox4 P437H dominant negative mutation (Nox4DN), driven by the endothelial specific promoter Tie-2, on atherosclerosis-prone genetic background (ApoE deficient mice) to mimic the effect of decreased endothelial Nox4. Nox4DN significantly increased type I diabetes-induced aortic stiffness and atherosclerotic lesions. Gene analysis indicated that soluble epoxide hydrolase 2 (sEH) was significantly upregulated in Nox4DN endothelial cells (EC). Inhibition of sEH activity in Nox4DN EC suppressed inflammation and macrophage adhesion to EC. On the contrary, overexpression of endothelial wild type Nox4 suppressed sEH, ameliorated Western diet-induced atherosclerosis and decreased aortic stiffness. CONCLUSIONS Atherosclerosis-prone conditions downregulated endothelial Nox4 to accelerate the progress of atherosclerosis, at least in part, by upregulating sEH to enhance inflammation.
Collapse
Affiliation(s)
- Pingping Hu
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Xiaojuan Wu
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Alok R Khandelwal
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Weimin Yu
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lili Chen
- Wuhan EasyDiagnosis Biomedicine Co., Ltd., Wuhan 430075, China
| | - Jian Yang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Robert M Weisbrod
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kin Sing Stephen Lee
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, CA 95616, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bruce D Hammock
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, CA 95616, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
31
|
Zhou B, Mu J, Gong Y, Lu C, Zhao Y, He T, Qin Z. Brd4 inhibition attenuates unilateral ureteral obstruction-induced fibrosis by blocking TGF-β-mediated Nox4 expression. Redox Biol 2016; 11:390-402. [PMID: 28063381 PMCID: PMC5219604 DOI: 10.1016/j.redox.2016.12.031] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/20/2016] [Accepted: 12/29/2016] [Indexed: 01/09/2023] Open
Abstract
Uncovering new therapeutic targets for renal fibrosis holds promise for the treatment of chronic kidney diseases. Bromodomain and extra-terminal (BET) protein inhibitors have been shown to effectively ameliorate pathological fibrotic responses. However, the pharmacological effects and underlying mechanisms of these inhibitors in renal fibrosis remain elusive. In this study, we determined that the inhibition of Brd4, a BET family member, with a selective potent chemical inhibitor, JQ1, could prevent the development of renal fibrosis and block the progression of fibrosis in rats that have undergone unilateral ureteral obstruction (UUO). Inhibiting Brd4 with either JQ1 or genetic knockdown resulted in decreased expression of fibrotic genes such as α-smooth muscle actin, collagen IV and fibronectin both in UUO-induced fibrosis and upon TGF-β1 stimulation in HK-2 cells. Brd4 inhibition also suppressed the oxidative stress induced by UUO in vivo or by TGF-β1 in HK-2 cells. Moreover, Nox4, which is constitutively active in renal cells and is involved in the generation of hydrogen peroxide, was up-regulated during UUO-mediated fibrosis and induced by TGF-β1 in HK-2 cells, and this up-regulation could be blunted by Brd4 inhibition. Consistently, Nox4-mediated ROS generation and fibrotic gene expression were attenuated upon Brd4 inhibition. Further, the transcriptional activity of Nox4 was suppressed by JQ1 or siRNA against Brd4. Additionally, Smad3 and ERK1/2 phosphorylation, which are upstream signals of Nox4 expression, were inhibited both in JQ1-administered UUO rats and Brd4-inhibited HK-2 cells. In conclusion, these results indicated that the inhibition of Brd4 might protect against renal fibrosis by blocking the TGF-β-Nox4-ROS-fibrosis axis, suggesting that Brd4 could be a promising therapeutic target. Brd4 was up-regulated in the progression of renal fibrosis. Brd4 inhibitor JQ1 prevented renal fibrosis and delayed the fibrotic progression. Brd4 inhibition blocked TGF-β1-induced oxidative stress and fibrosis through Nox4. Brd4 regulated Nox4 expression via Smad and ERK pathways.
Collapse
Affiliation(s)
- Baoshang Zhou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jiao Mu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yi Gong
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Caibao Lu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Youguang Zhao
- Department of Urology, Chengdu Military General Hospital, Chengdu 610083, China
| | - Ting He
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
32
|
García-Redondo AB, Aguado A, Briones AM, Salaices M. NADPH oxidases and vascular remodeling in cardiovascular diseases. Pharmacol Res 2016; 114:110-120. [PMID: 27773825 DOI: 10.1016/j.phrs.2016.10.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/10/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are key signaling molecules that regulate vascular function and structure in physiological conditions. A misbalance between the production and detoxification of ROS increases oxidative stress that is involved in the vascular remodeling associated with cardiovascular diseases such as hypertension by affecting inflammation, hypertrophy, migration, growth/apoptosis and extracellular matrix protein turnover. The major and more specific source of ROS in the cardiovascular system is the NADPH oxidase (NOX) family of enzymes composed of seven members (NOX1-5, DUOX 1/2). Vascular cells express several NOXs being NOX-1 and NOX-4 the most abundant NOXs present in vascular smooth muscle cells. This review focuses on specific aspects of NOX-1 and NOX-4 isoforms including information on regulation, function and their role in vascular remodeling. In order to obtain a more integrated view about the role of the different NOX isoforms in different types of vascular remodeling, we discuss the available literature not only on hypertension but also in atherosclerosis, restenosis and aortic dilation.
Collapse
Affiliation(s)
- Ana B García-Redondo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28029, Madrid, Spain
| | - Andrea Aguado
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28029, Madrid, Spain
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28029, Madrid, Spain.
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28029, Madrid, Spain.
| |
Collapse
|
33
|
Liu XH, Zhang QY, Pan LL, Liu SY, Xu P, Luo XL, Zou SL, Xin H, Qu LF, Zhu YZ. NADPH oxidase 4 contributes to connective tissue growth factor expression through Smad3-dependent signaling pathway. Free Radic Biol Med 2016; 94:174-84. [PMID: 26945889 DOI: 10.1016/j.freeradbiomed.2016.02.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/21/2016] [Accepted: 02/28/2016] [Indexed: 01/28/2023]
Abstract
Transforming growth factor-β (TGF-β)/Smad signaling has been implicated in connective tissue growth factor (CTGF) expression in vascular smooth muscle cells (VSMC). Reactive oxygen species (ROS) are involved in activation of TGF-β/Smad signaling. However, detailed mechanisms underlying the process remain unclear. In present study, we demonstrated TGF-β1 strongly induced CTGF expression, Smad3 activation, NADPH oxidase 4 (Nox4) expression and increased ROS production in primary rat VSMC in vitro. NADPH oxidases inhibitor diphenylene iodonium (DPI) eliminated TGF-β1-induced CTGF expression and ROS generation. In addition, small-interfering RNA (siRNA) silencing of Smad3 or Nox4 significantly suppressed TGF-β1-mediated CTGF expression in VSMC. Furthermore, Nox4 silencing or inhibition eliminated TGF-β1-induced Smad3 activation and interaction between Nox4 and Smad3. In vivo studies further identified a positive correlation of Nox4 levels with Smad3 activation and CTGF expression in atherosclerotic arteries of patients and animal models. These data established that a novel mechanistic link of Nox4-dependent activation of Smad3 to increased TGF-β1-induced CTGF in the process of vascular remodeling, which suggested a new potential pathway for therapeutic interventions.
Collapse
Affiliation(s)
- Xin-Hua Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Qiu-Yan Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Li-Long Pan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Yu Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Peng Xu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Xiao-Ling Luo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Li Zou
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Hong Xin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Le-Feng Qu
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Macau University of Science and Technology, Macau.
| |
Collapse
|
34
|
Tong X, Khandelwal AR, Wu X, Xu Z, Yu W, Chen C, Zhao W, Yang J, Qin Z, Weisbrod RM, Seta F, Ago T, Lee KSS, Hammock BD, Sadoshima J, Cohen RA, Zeng C. Pro-atherogenic role of smooth muscle Nox4-based NADPH oxidase. J Mol Cell Cardiol 2016; 92:30-40. [PMID: 26812119 PMCID: PMC5008453 DOI: 10.1016/j.yjmcc.2016.01.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/14/2016] [Accepted: 01/22/2016] [Indexed: 11/17/2022]
Abstract
UNLABELLED Nox4-based NADPH oxidase is a major reactive oxygen species-generating enzyme in the vasculature, but its role in atherosclerosis remains controversial. OBJECTIVE Our goal was to investigate the role of smooth muscle Nox4 in atherosclerosis. APPROACH AND RESULTS Atherosclerosis-prone conditions (disturbed blood flow and Western diet) increased Nox4 mRNA level in smooth muscle of arteries. To address whether upregulated smooth muscle Nox4 under atherosclerosis-prone conditions was directly involved in the development of atherosclerosis, mice carrying a human Nox4 P437H dominant negative mutation (Nox4DN), specifically in smooth muscle, were generated on a FVB/N ApoE deficient genetic background to counter the effect of increased smooth muscle Nox4. Nox4DN significantly decreased aortic stiffness and atherosclerotic lesions, with no effect on blood pressure. Gene analysis indicated that soluble epoxide hydrolase 2 (sEH) was significantly downregulated in Nox4DN smooth muscle cells (SMC), at both mRNA and protein levels. Downregulation of sEH by siRNA decreased SMC proliferation and migration, and suppressed inflammation and macrophage adhesion to SMC. CONCLUSIONS Downregulation of smooth muscle Nox4 inhibits atherosclerosis by suppressing sEH, which, at least in part, accounts for inhibition of SMC proliferation, migration and inflammation.
Collapse
Affiliation(s)
- Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China.
| | - Alok R Khandelwal
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xiaojuan Wu
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Weimin Yu
- Innovative Drug Research Centre, Chongqing University, Chongqing 401331, China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Wanzhou Zhao
- The Nanjing Han & Zaenker Cancer Institute, OG Pharmaceuticals, Nanjing 210019, China
| | - Jian Yang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhexue Qin
- Department of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Robert M Weisbrod
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 812-8581, Japan
| | - Kin Sing Stephen Lee
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, CA 95616, USA
| | - Bruce D Hammock
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, CA 95616, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|