1
|
Shao R, Chen R, Zheng Q, Yao M, Li K, Cao Y, Jiang L. Oxidative stress disrupts vascular microenvironmental homeostasis affecting the development of atherosclerosis. Cell Biol Int 2024. [PMID: 39370593 DOI: 10.1002/cbin.12239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis is primarily an inflammatory reaction of the cardiovascular system caused by endothelial damage, leading to progressive thickening and hardening of the vessel walls, as well as extensive necrosis and fibrosis of the surrounding tissues, the most necessary pathological process causing cardiovascular disease. When the body responds to harmful internal and external stimuli, excess oxygen free radicals are produced causing oxidative stress to occur in cells and tissues. Simultaneously, the activation of inflammatory immunological processes is followed by an elevation in oxygen free radicals, which directly initiates the release of cytokines and chemokines, resulting in a detrimental cycle of vascular homeostasis abnormalities. Oxidative stress contributes to the harm inflicted upon vascular endothelial cells and the decrease in nitric oxide levels. Nitric oxide is crucial for maintaining vascular homeostasis and is implicated in the development of atherosclerosis. This study examines the influence of oxidative stress on the formation of atherosclerosis, which is facilitated by the vascular milieu. It also provides an overview of the pertinent targets and pharmaceutical approaches for treating this condition.
Collapse
Affiliation(s)
- Ruifei Shao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Rui Chen
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Qiang Zheng
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Mengyu Yao
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
| | - Kunlin Li
- Department of General Surgery II, The First People's Hospital of Yunnan Province, Kunming, China
| | - Yu Cao
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lihong Jiang
- Medical School, Center for Translational Research in Clinical Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Innovative Application of Traditional Chinese Medicine, Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
2
|
Sun HJ, Ni ZR, Liu Y, Fu X, Liu SY, Hu JY, Sun QY, Li YC, Hou XH, Zhang JR, Zhu XX, Lu QB. Deficiency of neutral cholesterol ester hydrolase 1 (NCEH1) impairs endothelial function in diet-induced diabetic mice. Cardiovasc Diabetol 2024; 23:138. [PMID: 38664801 PMCID: PMC11046792 DOI: 10.1186/s12933-024-02239-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neutral cholesterol ester hydrolase 1 (NCEH1) plays a critical role in the regulation of cholesterol ester metabolism. Deficiency of NCHE1 accelerated atherosclerotic lesion formation in mice. Nonetheless, the role of NCEH1 in endothelial dysfunction associated with diabetes has not been explored. The present study sought to investigate whether NCEH1 improved endothelial function in diabetes, and the underlying mechanisms were explored. METHODS The expression and activity of NCEH1 were determined in obese mice with high-fat diet (HFD) feeding, high glucose (HG)-induced mouse aortae or primary endothelial cells (ECs). Endothelium-dependent relaxation (EDR) in aortae response to acetylcholine (Ach) was measured. RESULTS Results showed that the expression and activity of NCEH1 were lower in HFD-induced mouse aortae, HG-exposed mouse aortae ex vivo, and HG-incubated primary ECs. HG exposure reduced EDR in mouse aortae, which was exaggerated by endothelial-specific deficiency of NCEH1, whereas NCEH1 overexpression restored the impaired EDR. Similar results were observed in HFD mice. Mechanically, NCEH1 ameliorated the disrupted EDR by dissociating endothelial nitric oxide synthase (eNOS) from caveolin-1 (Cav-1), leading to eNOS activation and nitric oxide (NO) release. Moreover, interaction of NCEH1 with the E3 ubiquitin-protein ligase ZNRF1 led to the degradation of Cav-1 through the ubiquitination pathway. Silencing Cav-1 and upregulating ZNRF1 were sufficient to improve EDR of diabetic aortas, while overexpression of Cav-1 and downregulation of ZNRF1 abolished the effects of NCEH1 on endothelial function in diabetes. Thus, NCEH1 preserves endothelial function through increasing NO bioavailability secondary to the disruption of the Cav-1/eNOS complex in the endothelium of diabetic mice, depending on ZNRF1-induced ubiquitination of Cav-1. CONCLUSIONS NCEH1 may be a promising candidate for the prevention and treatment of vascular complications of diabetes.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Aorta/enzymology
- Aorta/physiopathology
- Aorta/metabolism
- Aorta/drug effects
- Aorta/pathology
- Caveolin 1/metabolism
- Caveolin 1/deficiency
- Caveolin 1/genetics
- Cells, Cultured
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/physiopathology
- Diet, High-Fat
- Endothelial Cells/enzymology
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Obesity/enzymology
- Obesity/physiopathology
- Obesity/metabolism
- Signal Transduction
- Sterol Esterase/metabolism
- Sterol Esterase/genetics
- Ubiquitination
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Zhang-Rong Ni
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yao Liu
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Xiao Fu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Shi-Yi Liu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jin-Yi Hu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Qing-Yi Sun
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yu-Chao Li
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Xiao-Hui Hou
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Ji-Ru Zhang
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Xue-Xue Zhu
- Department of Physiology, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Qing-Bo Lu
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, China.
| |
Collapse
|
3
|
Allen-Gondringer A, Gau D, Dutta P, Roy P. Haplo-insufficiency of Profilin1 in vascular endothelial cells is beneficial but not sufficient to confer protection against experimentally induced atherosclerosis. Cytoskeleton (Hoboken) 2024:10.1002/cm.21859. [PMID: 38623956 PMCID: PMC11480255 DOI: 10.1002/cm.21859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
Actin cytoskeleton plays an important role in various aspects of atherosclerosis, a key driver of ischemic heart disease. Actin-binding protein Profilin1 (Pfn1) is overexpressed in atherosclerotic plaques in human disease, and Pfn1, when partially depleted globally in all cell types, confers atheroprotection in vivo. This study investigates the impact of endothelial cell (EC)-specific partial loss of Pfn1 expression in atherosclerosis development. We utilized mice engineered for conditional heterozygous knockout of the Pfn1 gene in ECs, with atherosclerosis induced by depletion of hepatic LDL receptor by gene delivery of PCSK9 combined with high-cholesterol diet. Our studies show that partial depletion of EC Pfn1 has certain beneficial effects marked by dampening of select pro-atherogenic cytokines (CXCL10 and IL7) with concomitant reduction in cytotoxic T cell abundance but is not sufficient to reduce hyperlipidemia and confer atheroprotection in vivo. In light of these findings, we conclude that atheroprotective phenotype conferred by global Pfn1 haplo-insufficiency requires contributions of additional cell types that are relevant for atherosclerosis progression.
Collapse
Affiliation(s)
| | - David Gau
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Partha Dutta
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Partha Roy
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Pathology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
4
|
Allen-Gondringer A, Gau D, Dutta P, Roy P. Haplo-insufficiency of Profilin1 in vascular endothelial cells is beneficial but not sufficient to confer protection against experimentally induced atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.06.570450. [PMID: 38106044 PMCID: PMC10723386 DOI: 10.1101/2023.12.06.570450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Actin cytoskeleton plays an important role in various aspects of atherosclerosis, a key driver of ischemic heart disease. Actin-binding protein Profilin1 (Pfn1) is overexpressed in atherosclerotic plaques in human disease, and Pfn1, when partially depleted globally in all cell types, confers atheroprotection in vivo . This study investigates the impact of endothelial cell (EC)-specific partial loss of Pfn1 expression in atherosclerosis development. We utilized mice engineered for conditional heterozygous knockout of the Pfn1 gene in ECs, with atherosclerosis induced by depletion of hepatic LDL receptor by gene delivery of PCSK9 combined with high-cholesterol diet. Our studies show that partial depletion of EC Pfn1 has certain beneficial effects marked by dampening of select pro-atherogenic cytokines (CXCL10 and IL7) with concomitant reduction in cytotoxic T cell abundance but is not sufficient to reduce hyperlipidemia and confer atheroprotection in vivo . In light of these findings, we conclude that atheroprotective phenotype conferred by global Pfn1 haplo-insufficiency requires contributions of additional cell types that are relevant for atherosclerosis progression.
Collapse
|
5
|
Sharma A, Choi J, Sim L, Dey A, Mohan M, Kantharidis P, Dietz L, Sandner P, de Haan JB. Ameliorating diabetes-associated atherosclerosis and diabetic nephropathy through modulation of soluble guanylate cyclase. Front Cardiovasc Med 2023; 10:1220095. [PMID: 37502180 PMCID: PMC10368983 DOI: 10.3389/fcvm.2023.1220095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Diabetes mellitus (DM) is an independent risk factor for micro- and macrovascular complications such as nephropathy and atherosclerosis respectively, which are the major causes of premature morbidity and mortality in Type 1 and Type 2 diabetic patients. Endothelial dysfunction is the critical first step of vascular disease and is characterized by reduced bioavailability of the essential endothelial vasodilator, nitric oxide (NO), coupled with an elevation in inflammation and oxidative stress. A novel pathway to bolster NO activity is to upregulate soluble guanylate cyclase (sGC), an enzyme responsible for mediating the protective actions of NO. Two classes of sGC modulators exist, activators and stimulators, with differing sensitivity to oxidative stress. In this study, we investigated the therapeutic effects of the sGC stimulator BAY 41-2272 (Bay 41) and the sGC activator BAY 60-2770 (Bay 60) on endpoints of atherosclerosis and renal disease as well as inflammation and oxidative stress in diabetic Apolipoprotein E knockout (ApoE-/-) mice. We hypothesized that under oxidative conditions known to accompany diabetes, sGC activation might be more efficacious than sGC stimulation in limiting diabetic vascular complications. We demonstrate that Bay 60 not only significantly decreased nitrotyrosine staining (P < 0.01) and F4/80 positive cells by 75% (P < 0.05), but it also significantly reduced total plaque area (P < 0.05) and improved endothelial function (P < 0.01). Our data suggest an important anti-atherogenic role for Bay 60 accompanied by reduced oxidative stress and inflammation under diabetic settings. Treatment with the stimulator Bay 41, on the other hand, had minimal effects or caused no changes with respect to cardiovascular or renal pathology. In the kidneys, treatment with Bay 60 significantly lessened urinary albuminuria, mesangial expansion and nitrotyrosine staining under diabetic conditions. In summary, our head-to-head comparator is the first preclinical study to show that a sGC activator is more efficacious than a sGC stimulator for the treatment of diabetes-associated vascular and renal complications.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Cardiovascular Inflammation and Redox Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Diabetes, Monash University, Central Clinical School, Melbourne, VIC, Australia
| | - Judy Choi
- Cardiovascular Inflammation and Redox Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Lachlan Sim
- Cardiovascular Inflammation and Redox Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Abhiroop Dey
- Cardiovascular Inflammation and Redox Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Monash University, Central Clinical School, Melbourne, VIC, Australia
| | - Phillip Kantharidis
- Department of Diabetes, Monash University, Central Clinical School, Melbourne, VIC, Australia
| | - Lisa Dietz
- Pharmaceuticals Research and Development, Bayer AG, Wuppertal, Germany
| | - Peter Sandner
- Pharmaceuticals Research and Development, Bayer AG, Wuppertal, Germany
- Institute of Pharmacology, Hannover Medical School, Hanover, Germany
| | - Judy B. de Haan
- Cardiovascular Inflammation and Redox Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Peche VS, Pietka TA, Jacome-Sosa M, Samovski D, Palacios H, Chatterjee-Basu G, Dudley AC, Beatty W, Meyer GA, Goldberg IJ, Abumrad NA. Endothelial cell CD36 regulates membrane ceramide formation, exosome fatty acid transfer and circulating fatty acid levels. Nat Commun 2023; 14:4029. [PMID: 37419919 PMCID: PMC10329018 DOI: 10.1038/s41467-023-39752-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/28/2023] [Indexed: 07/09/2023] Open
Abstract
Endothelial cell (EC) CD36 controls tissue fatty acid (FA) uptake. Here we examine how ECs transfer FAs. FA interaction with apical membrane CD36 induces Src phosphorylation of caveolin-1 tyrosine-14 (Cav-1Y14) and ceramide generation in caveolae. Ensuing fission of caveolae yields vesicles containing FAs, CD36 and ceramide that are secreted basolaterally as small (80-100 nm) exosome-like extracellular vesicles (sEVs). We visualize in transwells EC transfer of FAs in sEVs to underlying myotubes. In mice with EC-expression of the exosome marker emeraldGFP-CD63, muscle fibers accumulate circulating FAs in emGFP-labeled puncta. The FA-sEV pathway is mapped through its suppression by CD36 depletion, blocking actin-remodeling, Src inhibition, Cav-1Y14 mutation, and neutral sphingomyelinase 2 inhibition. Suppression of sEV formation in mice reduces muscle FA uptake, raises circulating FAs, which remain in blood vessels, and lowers glucose, mimicking prominent Cd36-/- mice phenotypes. The findings show that FA uptake influences membrane ceramide, endocytosis, and EC communication with parenchymal cells.
Collapse
Affiliation(s)
- V S Peche
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - T A Pietka
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - M Jacome-Sosa
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - D Samovski
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - H Palacios
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - G Chatterjee-Basu
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - A C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - W Beatty
- Department of Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - G A Meyer
- Departments of Physical Therapy, Neurology and Orthopedic Surgery, Washington University School of Medicine, St. Louis, 63110, USA
| | - I J Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - N A Abumrad
- Department of Medicine, Division of Nutritional Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
7
|
Troncoso MF, Díaz-Vesga MC, Sanhueza-Olivares F, Riquelme JA, Müller M, Garrido L, Gabrielli L, Chiong M, Corbalan R, Castro PF, Lavandero S. Targeting VCAM-1: a therapeutic opportunity for vascular damage. Expert Opin Ther Targets 2023; 27:207-223. [PMID: 36880349 DOI: 10.1080/14728222.2023.2187778] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
INTRODUCTION The vascular cell adhesion molecule (VCAM-1) is a transmembrane sialoglycoprotein detected in activated endothelial and vascular smooth muscle cells involved in the adhesion and transmigration of inflammatory cells into damaged tissue. Widely used as a pro-inflammatory marker, its potential role as a targeting molecule has not been thoroughly explored. AREAS COVERED We discuss the current evidence supporting the potential targeting of VCAM-1 in atherosclerosis, diabetes, hypertension and ischemia/reperfusion injury. EXPERT OPINION There is emerging evidence that VCAM-1 is more than a biomarker and may be a promising therapeutic target for vascular diseases. While there are neutralizing antibodies that allow preclinical research, the development of pharmacological tools to activate or inhibit this protein are required to thoroughly assess its therapeutic potential.
Collapse
Affiliation(s)
- Mayarling F Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Magda C Díaz-Vesga
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Pontificia Universidad Javeriana de Cali, Cali, Colombia
| | - Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jaime A Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Garrido
- Division of Cardiovascular Diseases, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luigi Gabrielli
- Division Surgery, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ramon Corbalan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo F Castro
- Division Surgery, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
8
|
Role of Echocardiography in Diabetic Cardiomyopathy: From Mechanisms to Clinical Practice. J Cardiovasc Dev Dis 2023; 10:jcdd10020046. [PMID: 36826542 PMCID: PMC9959745 DOI: 10.3390/jcdd10020046] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/17/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023] Open
Abstract
It has been well established that diabetes mellitus (DM) is considered as a core risk factor for the development of cardiovascular diseases. However, what is less appreciated is the fact that DM may affect cardiac function irrespective of cardiac pathologies to which it contributes, such as coronary artery disease and hypertension. Although echocardiography provides accurate and reproducible diagnostic and prognostic data in patients with DM, its use in these patients is still underappreciated, resulting in progression of DM-related heart failure in many patients. Hence, in the present review, we aimed to discuss the role of echocardiography in the contemporary management of diabetic cardiomyopathy (DCM), as well as the role of emerging echocardiographic techniques, which may contribute to earlier diagnosis and more appropriate management of this complication of DM. In order to improve outcomes, focus must be placed on early diagnosis of this condition using a combination of echocardiography and emerging biomarkers, but perhaps the more important thing is to change perspective when it comes to the clinical importance of DCM.
Collapse
|
9
|
Maassen H, Said MY, Frenay ARS, Koning A, Post A, Riphagen IJ, Heiner-Fokkema MR, Drabert K, Fernandez BO, Gans ROB, van den Berg E, Navis G, Tsikas D, Feelisch M, Bakker SJL, van Goor H. Nitric oxide and long-term outcomes after kidney transplantation: Results of the TransplantLines cohort study. Nitric Oxide 2022; 125-126:1-11. [PMID: 35660109 DOI: 10.1016/j.niox.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022]
Abstract
Impaired endogenous nitric oxide (NO) production may contribute to graft failure and premature mortality in kidney transplant recipients (KTR). We investigated potential associations of 24-h urinary NOx (NO3- + NO2-) excretion (uNOx) with long-term outcomes. uNOx was determined by HPLC and GC-MS in 698 KTR and in 132 kidney donors before and after donation. Additionally, we measured urinary nitroso species (RXNO) by gas-phase chemiluminescence. Median uNOx was lower in KTR compared to kidney donors (688 [393-1076] vs. 1301 [868-1863] before donation and 1312 [982-1853] μmol/24h after donation, P < 0.001). During median follow-up of 5.4 [4.8-6.1] years, 150 KTR died (61 due to cardiovascular disease) and 83 experienced graft failure. uNOx was inversely associated with all-cause mortality (HR per doubling of uNOx: 0.84 [95% CI 0.75-0.93], P < 0.001) and cardiovascular mortality (HR 0.78 [95% CI 0.67-0.92], P = 0.002). The association of uNOx with graft failure was lost when adjusted for renal function (HR per doubling of uNOx: 0.89 [95% CI 0.76-1.05], P = 0.17). There were no significant associations of urinary RXNO with outcomes. Our study suggests that KTR have lower NO production than healthy subjects and that lower uNOx is associated with a higher risk of all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Hanno Maassen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - M Yusof Said
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Anne-Roos S Frenay
- Department of Gynecology and Obstetrics, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Anne Koning
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Adrian Post
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kathrin Drabert
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Bernadette O Fernandez
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Reinold O B Gans
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Else van den Berg
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
10
|
Geiger M, Hayter E, Martin R, Spence D. Red blood cells in type 1 diabetes and multiple sclerosis and technologies to measure their emerging roles. J Transl Autoimmun 2022; 5:100161. [PMID: 36039310 PMCID: PMC9418496 DOI: 10.1016/j.jtauto.2022.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- M. Geiger
- Institute of Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - E. Hayter
- Department of Chemistry, Saint Louis University, St. Louis, MO 63103, USA
| | - R.S. Martin
- Department of Chemistry, Saint Louis University, St. Louis, MO 63103, USA
| | - D. Spence
- Institute of Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
- Corresponding author. 775 Woodlot Drive, East Lansing, MI 48824, USA.
| |
Collapse
|
11
|
Wang C, Chen J, Wang P, Qing S, Li W, Lu J. Endogenous Protective Factors and Potential Therapeutic Agents for Diabetes-Associated Atherosclerosis. Front Endocrinol (Lausanne) 2022; 13:821028. [PMID: 35557850 PMCID: PMC9086429 DOI: 10.3389/fendo.2022.821028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
The complications of macrovascular atherosclerosis are the leading cause of disability and mortality in patients with diabetes. It is generally believed that the pathogenesis of diabetic vascular complications is initiated by the imbalance between injury and endogenous protective factors. Multiple endogenous protective factors secreted by endothelium, liver, skeletal muscle and other tissues are recognized of their importance in combating injury factors and maintaining the homeostasis of vasculatures in diabetes. Among them, glucagon-like peptide-1 based drugs were clinically proven to be effective and recommended as the first-line medicine for the treatment of type 2 diabetic patients with high risks or established arteriosclerotic cardiovascular disease (CVD). Some molecules such as irisin and lipoxins have recently been perceived as new protective factors on diabetic atherosclerosis, while the protective role of HDL has been reinterpreted since the failure of several clinical trials to raise HDL therapy on cardiovascular events. The current review aims to summarize systemic endogenous protective factors for diabetes-associated atherosclerosis and discuss their mechanisms and potential therapeutic strategy or their analogues. In particular, we focus on the existing barriers or obstacles that need to be overcome in developing new therapeutic approaches for macrovascular complications of diabetes.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jin Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Pin Wang
- Department of Pharmacology, Naval Medical University, Shanghai, China
| | - Shengli Qing
- Department of Pharmacology, Naval Medical University, Shanghai, China
| | - Wenwen Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- *Correspondence: Jin Lu, ; Wenwen Li,
| | - Jin Lu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- *Correspondence: Jin Lu, ; Wenwen Li,
| |
Collapse
|
12
|
Cieślik P, Kalinowski L, Wierońska JM. Procognitive activity of nitric oxide inhibitors and donors in animal models. Nitric Oxide 2021; 119:29-40. [PMID: 34896554 DOI: 10.1016/j.niox.2021.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/17/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Nitric oxide is a small gaseous molecule that plays important roles in the majority of biological functions. Impairments of NO-related pathways contribute to the majority of neurological disorders, such as Alzheimer's disease (AD), and mental disorders, such as schizophrenia. Cognitive decline is one of the most serious impairments accompanying both AD and schizophrenia. In the present study, the activities of NO donors, slow (spermine NONOate) or fast (DETANONOate) releasers, and selective inhibitor of neuronal nitric oxide synthase N(ω)-propyl-l-arginine (NPLA) were investigated in pharmacological models of schizophrenia and AD. Cognitive impairments were induced by administration of MK-801 or scopolamine and were measured in novel object recognition (NOR) and Y-maze tests. The compounds were investigated at doses of 0.05-0.5 mg/kg. The dose-dependent effectiveness of all the compounds was observed in the NOR test, while only the highest doses of spermine NONOate and NPLA were active in the Y-maze test. DETANONOate was not active in the Y-maze test. The impact of the investigated compounds on motor coordination was tested at doses of 0.5 and 1 mg/kg. Only NPLA at a dose of 1 mg/kg slightly disturbed motor coordination in animals.
Collapse
Affiliation(s)
- Paulina Cieślik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Neurobiology, 12 Smętna Street, 31-343, Kraków, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, 7 Dębinki Street, 80-211, Gdańsk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Consortium Poland (BBMRI.pl), Poland; BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | - Joanna M Wierońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Neurobiology, 12 Smętna Street, 31-343, Kraków, Poland.
| |
Collapse
|
13
|
Gokani S, Bhatt LK. Caveolin-1: A promising therapeutic target for diverse diseases. Curr Mol Pharmacol 2021; 15:701-715. [PMID: 34847854 DOI: 10.2174/1874467214666211130155902] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/26/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022]
Abstract
The plasma membrane of eukaryotic cells contains small flask-shaped invaginations known as caveolae that are involved in the regulation of cellular signaling. Caveolin-1 is a 21-24kDa protein localized in the caveolar membrane. Caveolin-1 (Cav-1) has been considered as a master regulator among the various signaling molecules. It has been emerging as a chief protein regulating cellular events associated with homeostasis, caveolae formation, and caveolae trafficking. In addition to the physiological role of cav-1, it has a complex role in the progression of various diseases. Caveolin-1 has been identified as a prognosticator in patients with cancer and has a dual role in tumorigenesis. The expression of Cav-1 in hippocampal neurons and synapses is related to neurodegeneration, cognitive decline, and aging. Despite the ubiquitous association of caveolin-1 in various pathological processes, the mechanisms associated with these events are still unclear. Caveolin-1 has a significant role in various events of the viral cycle, such as viral entry. This review will summarize the role of cav-1 in the development of cancer, neurodegeneration, glaucoma, cardiovascular diseases, and infectious diseases. The therapeutic perspectives involving clinical applications of Caveolin-1 have also been discussed. The understanding of the involvement of caveolin-1 in various diseased states provides insights into how it can be explored as a novel therapeutic target.
Collapse
Affiliation(s)
- Shivani Gokani
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai. India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai. India
| |
Collapse
|
14
|
Khosravi Bakhtiari M, Sharifiyazdi H, Nazifi S, Ghaemi M, Hadadipour Zarandi M. Effects of citral on serum antioxidant status and liver genes expressions of paraoxonase 1 and nitric oxide synthase in a rat model of streptozotocin-induced diabetes mellitus. IRANIAN JOURNAL OF VETERINARY RESEARCH 2021; 22:195-202. [PMID: 34777519 PMCID: PMC8573404 DOI: 10.22099/ijvr.2021.38416.5585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 11/12/2022]
Abstract
Background: Citral (C10H16O) is the main ingredient of Cymbopogon citratus (lemongrass oil) and can reduce the side effects of oxidative stress. Diabetes caused by insulin deficiency induces oxidative stress in the liver. Aims: This study aimed to investigate the ameliorative effects of citral on selected oxidative parameters and the gene expression of paraoxonase 1 (PON1) and endothelial nitric oxide synthase (eNOS) in a rat model of streptozotocin (STZ)-induced diabetes mellitus. Methods: Forty rats were divided into four groups at random: control (C), control citral (CC), and two STZ-induced diabetic groups (diabetic (D) and citral diabetic (CD)). After diabetes confirmation (day 7), gavage treatment with citral (300 mg/kg body weight (BW)) was started in the CD and CC groups and continued for two weeks. Results: On day 21 of the study, following treatment with citral for 14 days, the serum levels of total antioxidant capacity (TAC), and PON1 in the CD group were significantly increased compared to those in the D group (P<0.05). While treatment with citral caused a significant decrease in the Malondialdehyde (MDA), and eNOS in the CD group compared to those of the D group (P<0.001). The expression rate of liver PON1 gene was considerably upregulated in the CD group compared to that in the D group (P<0.001); while the opposite was observed for eNOS gene expression. However, there was no significant difference between the CC and C groups in terms of all examined parameters (P>0.05). Conclusion: This study showed positive effects of citral on serum antioxidant status and liver gene expression of PON1 and eNOS in diabetic rats.
Collapse
Affiliation(s)
- M Khosravi Bakhtiari
- Resident of Clinical Pathology, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - H Sharifiyazdi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - S Nazifi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - M Ghaemi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - M Hadadipour Zarandi
- Resident of Clinical Pathology, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
15
|
Liao K, Lv DY, Yu HL, Chen H, Luo SX. iNOS regulates activation of the NLRP3 inflammasome through the sGC/cGMP/PKG/TACE/TNF-α axis in response to cigarette smoke resulting in aortic endothelial pyroptosis and vascular dysfunction. Int Immunopharmacol 2021; 101:108334. [PMID: 34768128 DOI: 10.1016/j.intimp.2021.108334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cigarette smoke (CS) is associated with vascular injury and dysfunction, which may be mediated by iNOS and NLRP3. However, the exact mechanism is unknown. METHODS iNOS-knockout and NLRP3-knockout C57BL/6 mice were exposed to air or CS. The vascular structure was examined by hematoxylin-eosin staining. The vascular tension was measured by a vascular reactivity assay. The expression of iNOS, NLRP3, caspase-1p20, IL-1β and eNOS were measured by western blotting. Human aortic endothelial cells (HAECs) were exposed to L-NIL (iNOS inhibitor), MCC950 (NLRP3 inhibitor), ODQ (sGC inhibitor), KT5823 (PKG inhibitor) or TAPI-1 (TACE/ADAM17 inhibitor) for 1 h prior to cigarette smoke extract (CSE) treatment. The cell viability and lactate dehydrogenase activity were assessed and pyroptosis was determined by scanning electron microscopy. The mRNA expression of TNF-α, and protein expression of iNOS, active-TACE, NLRP3, caspase-1p20, IL-1β, and eNOS were measured. RESULTS CS resulted in shrinkage of endothelial cells, impaired aorta relaxation, reduced eNOS expression, and induced expression of iNOS, NLRP3, caspase-1p20 and IL-1β, which could be prevented by knockdown of iNOS and NLRP3. CSE reduced cell viability, induced LDH release and pyroptosis, and promoted iNOS, NLRP3, caspase-1p20, and IL-1β expression and reduced eNOS reduction, which could be reversed by inhibition of iNOS or NLRP3 in HAECs. Altogether, activation of the NLRP3 inflammasome by iNOS in CS-exposed HAECs may be mediated by the sGC/cGMP/PKG/TACE/TNF- α pathway. CONCLUSION These results link iNOS to NLRP3 in CSE-stimulated HAECs through the sGC/cGMP/PKG/TACE/TNF-α pathway. The findings identify a mechanism through which iNOS and NLRP3 contribute to the pathogenesis of CS-induced pyroptosis and impaired aorta relaxation in HAECs.
Collapse
Affiliation(s)
- Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Ding-Yi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hui-Lin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hong Chen
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| | - Su-Xin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
16
|
Horie K, Maeda H, Nanashima N, Oey I. Potential Vasculoprotective Effects of Blackcurrant ( Ribes nigrum) Extract in Diabetic KK-A y Mice. Molecules 2021; 26:molecules26216459. [PMID: 34770868 PMCID: PMC8587626 DOI: 10.3390/molecules26216459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/19/2023] Open
Abstract
Polyphenols are bioactive compounds found naturally in fruits and vegetables; they are widely used in disease prevention and health maintenance. Polyphenol-rich blackcurrant extract (BCE) exerts beneficial effects on vascular health in menopausal model animals. However, the vasculoprotective effects in diabetes mellitus (DM) and atherosclerotic vascular disease secondary to DM are unknown. Therefore, we investigated whether BCE is effective in preventing atherosclerosis using KK-Ay mice as a diabetes model. The mice were divided into three groups and fed a high-fat diet supplemented with 1% BCE (BCE1), 3% BCE (BCE2), or Control for 9 weeks. The mice in the BCE2 group showed a considerable reduction in the disturbance of elastic lamina, foam cell formation, and vascular remodeling compared to those in the BCE1 and Control groups. Immunohistochemical staining indicated that the score of endothelial nitric oxide synthase staining intensity was significantly higher in both BCE2 (2.9) and BCE1 (1.9) compared to that in the Control (1.1). Furthermore, the score for the percentage of alpha-smooth muscle actin was significantly lower in the BCE2 (2.9%) than in the Control (2.1%). Our results suggest that the intake of anthocyanin-rich BCE could have beneficial effects on the blood vessels of diabetic patients.
Collapse
Affiliation(s)
- Kayo Horie
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki 036-8564, Japan;
- Correspondence: ; Tel.: +81-172-39-5527
| | - Hayato Maeda
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan;
| | - Naoki Nanashima
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki 036-8564, Japan;
| | - Indrawati Oey
- Department of Food Science, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand;
- Riddet Institute, Massey University, Private Bag 11 222, Palmerston North 4442, New Zealand
| |
Collapse
|
17
|
Zhuang A, Yang C, Liu Y, Tan Y, Bond ST, Walker S, Sikora T, Laskowski A, Sharma A, de Haan JB, Meikle PJ, Shimizu T, Coughlan MT, Calkin AC, Drew BG. SOD2 in skeletal muscle: New insights from an inducible deletion model. Redox Biol 2021; 47:102135. [PMID: 34598016 PMCID: PMC8487078 DOI: 10.1016/j.redox.2021.102135] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 01/01/2023] Open
Abstract
Metabolic conditions such as obesity, insulin resistance and glucose intolerance are frequently associated with impairments in skeletal muscle function and metabolism. This is often linked to dysregulation of homeostatic pathways including an increase in reactive oxygen species (ROS) and oxidative stress. One of the main sites of ROS production is the mitochondria, where the flux of substrates through the electron transport chain (ETC) can result in the generation of oxygen free radicals. Fortunately, several mechanisms exist to buffer bursts of intracellular ROS and peroxide production, including the enzymes Catalase, Glutathione Peroxidase and Superoxide Dismutase (SOD). Of the latter, there are two intracellular isoforms; SOD1 which is mostly cytoplasmic, and SOD2 which is found exclusively in the mitochondria. Developmental and chronic loss of these enzymes has been linked to disease in several studies, however the temporal effects of these disturbances remain largely unexplored. Here, we induced a post-developmental (8-week old mice) deletion of SOD2 in skeletal muscle (SOD2-iMKO) and demonstrate that 16 weeks of SOD2 deletion leads to no major impairment in whole body metabolism, despite these mice displaying alterations in aspects of mitochondrial abundance and voluntary ambulatory movement. This is likely partly explained by the suggestive data that a compensatory response may exist from other redox enzymes, including catalase and glutathione peroxidases. Nevertheless, we demonstrated that inducible SOD2 deletion impacts on specific aspects of muscle lipid metabolism, including the abundance of phospholipids and phosphatidic acid (PA), the latter being a key intermediate in several cellular signaling pathways. Thus, our findings suggest that post-developmental deletion of SOD2 induces a more subtle phenotype than previous embryonic models have shown, allowing us to highlight a previously unrecognized link between SOD2, mitochondrial function and bioactive lipid species including PA.
Collapse
Affiliation(s)
- Aowen Zhuang
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Christine Yang
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia
| | - Yingying Liu
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia
| | - Yanie Tan
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Simon T Bond
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Shannen Walker
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Tim Sikora
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia
| | - Adrienne Laskowski
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Arpeeta Sharma
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia
| | - Judy B de Haan
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, 3083, Australia; Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, 3122, Australia
| | - Peter J Meikle
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Takahiko Shimizu
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Melinda T Coughlan
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Anna C Calkin
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Brian G Drew
- Baker Heart & Diabetes Institute, Melbourne, 3004, Australia; Central Clinical School, Monash University, Melbourne, 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
18
|
He J, Cui Z, Zhu Y. The role of caveolae in endothelial dysfunction. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:78-91. [PMID: 37724072 PMCID: PMC10388784 DOI: 10.1515/mr-2021-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Caveolae, the specialized cell-surface plasma membrane invaginations which are abundant in endothelial cells, play critical roles in regulating various cellular processes, including cholesterol homeostasis, nitric oxide production, and signal transduction. Endothelial caveolae serve as a membrane platform for compartmentalization, modulation, and integration of signal events associated with endothelial nitric oxide synthase, ATP synthase β, and integrins, which are involved in the regulation of endothelial dysfunction and related cardiovascular diseases, such as atherosclerosis and hypertension. Furthermore, these dynamic microdomains on cell membrane are modulated by various extracellular stimuli, including cholesterol and flow shear stress. In this brief review, we summarize the critical roles of caveolae in the orchestration of endothelial function based on recent findings as well as our work over the past two decades.
Collapse
Affiliation(s)
- Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| | - Zhen Cui
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| |
Collapse
|
19
|
Das M, Devi KP, Belwal T, Devkota HP, Tewari D, Sahebnasagh A, Nabavi SF, Khayat Kashani HR, Rasekhian M, Xu S, Amirizadeh M, Amini K, Banach M, Xiao J, Aghaabdollahian S, Nabavi SM. Harnessing polyphenol power by targeting eNOS for vascular diseases. Crit Rev Food Sci Nutr 2021; 63:2093-2118. [PMID: 34553653 DOI: 10.1080/10408398.2021.1971153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vascular diseases arise due to vascular endothelium dysfunction in response to several pro-inflammatory stimuli and invading pathogens. Thickening of the vessel wall, formation of atherosclerotic plaques consisting of proliferating smooth muscle cells, macrophages and lymphocytes are the major consequences of impaired endothelium resulting in atherosclerosis, hypercholesterolemia, hypertension, type 2 diabetes mellitus, chronic renal failure and many others. Decreased nitric oxide (NO) bioavailability was found to be associated with anomalous endothelial function because of either its reduced production level by endothelial NO synthase (eNOS) which synthesize this potent endogenous vasodilator from L-arginine or its enhanced breakdown due to severe oxidative stress and eNOS uncoupling. Polyphenols are a group of bioactive compounds having more than 7000 chemical entities present in different cereals, fruits and vegetables. These natural compounds possess many OH groups which are largely responsible for their strong antioxidative, anti-inflammatory antithrombotic and anti-hypersensitive properties. Several flavonoid-derived polyphenols like flavones, isoflavones, flavanones, flavonols and anthocyanidins and non-flavonoid polyphenols like tannins, curcumins and resveratrol have attracted scientific interest for their beneficial effects in preventing endothelial dysfunction. This article will focus on in vitro as well as in vivo and clinical studies evidences of the polyphenols with eNOS modulating activity against vascular disease condition while their molecular mechanism will also be discussed.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, India
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, India
| | - Tarun Belwal
- College of Biosystems Engineering and Food Science, Zhejiang University, China
| | | | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Rasekhian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Suowen Xu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mehran Amirizadeh
- Department of Pharmacotherapy, Faculty of pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Kiumarth Amini
- Student Research Committee, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Poland
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China.,Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense, Spain
| | - Safieh Aghaabdollahian
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Owusu J, Barrett E. Early Microvascular Dysfunction: Is the Vasa Vasorum a "Missing Link" in Insulin Resistance and Atherosclerosis. Int J Mol Sci 2021; 22:ijms22147574. [PMID: 34299190 PMCID: PMC8303323 DOI: 10.3390/ijms22147574] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 11/16/2022] Open
Abstract
The arterial vasa vasorum is a specialized microvasculature that provides critical perfusion required for the health of the arterial wall, and is increasingly recognized to play a central role in atherogenesis. Cardio-metabolic disease (CMD) (including hypertension, metabolic syndrome, obesity, diabetes, and pre-diabetes) is associated with insulin resistance, and characteristically injures the microvasculature in multiple tissues, (e.g., the eye, kidney, muscle, and heart). CMD also increases the risk for atherosclerotic vascular disease. Despite this, the impact of CMD on vasa vasorum structure and function has been little studied. Here we review emerging information on the early impact of CMD on the microvasculature in multiple tissues and consider the potential impact on atherosclerosis development and progression, if vasa vasorum is similarly affected.
Collapse
Affiliation(s)
- Jeanette Owusu
- Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA;
| | - Eugene Barrett
- Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA;
- Department of Pediatrics, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Correspondence: ; Tel.: +1-434-924-1263
| |
Collapse
|
21
|
Choi JSY, de Haan JB, Sharma A. Animal models of diabetes-associated vascular diseases: an update on available models and experimental analysis. Br J Pharmacol 2021; 179:748-769. [PMID: 34131901 DOI: 10.1111/bph.15591] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/08/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a chronic metabolic disorder associated with the accelerated development of macrovascular (atherosclerosis and coronary artery disease) and microvascular complications (nephropathy, retinopathy and neuropathy), which remain the principal cause of mortality and morbidity in this population. Current understanding of cellular and molecular pathways of diabetes-driven vascular complications, as well as therapeutic interventions has arisen from studying disease pathogenesis in animal models. Diabetes-associated vascular complications are multi-faceted, involving the interaction between various cellular and molecular pathways. Thus, the choice of an appropriate animal model to study vascular pathogenesis is important in our quest to identify innovative and mechanism-based targeted therapies to reduce the burden of diabetic complications. Herein, we provide up-to-date information on available mouse models of both Type 1 and Type 2 diabetic vascular complications as well as experimental analysis and research outputs.
Collapse
Affiliation(s)
- Judy S Y Choi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia.,Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Arpeeta Sharma
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Central Clinical School, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Kumric M, Ticinovic Kurir T, Borovac JA, Bozic J. Role of novel biomarkers in diabetic cardiomyopathy. World J Diabetes 2021; 12:685-705. [PMID: 34168722 PMCID: PMC8192249 DOI: 10.4239/wjd.v12.i6.685] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/22/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is commonly defined as cardiomyopathy in patients with diabetes mellitus in the absence of coronary artery disease and hypertension. As DCM is now recognized as a cause of substantial morbidity and mortality among patients with diabetes mellitus and clinical diagnosis is still inappropriate, various expert groups struggled to identify a suitable biomarker that will help in the recognition and management of DCM, with little success so far. Hence, we thought it important to address the role of biomarkers that have shown potential in either human or animal studies and which could eventually result in mitigating the poor outcomes of DCM. Among the array of biomarkers we thoroughly analyzed, long noncoding ribonucleic acids, soluble form of suppression of tumorigenicity 2 and galectin-3 seem to be most beneficial for DCM detection, as their plasma/serum levels accurately correlate with the early stages of DCM. The combination of relatively inexpensive and accurate speckle tracking echocardiography with some of the highlighted biomarkers may be a promising screening method for newly diagnosed diabetes mellitus type 2 patients. The purpose of the screening test would be to direct affected patients to more specific confirmation tests. This perspective is in concordance with current guidelines that accentuate the importance of an interdisciplinary team-based approach.
Collapse
Affiliation(s)
- Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Department of Endocrinology, University Hospital of Split, Split 21000, Croatia
| | - Josip A Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Emergency Medicine, Institute of Emergency Medicine of Split-Dalmatia County, Split 21000, Croatia
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| |
Collapse
|
23
|
Troncoso MF, Ortiz-Quintero J, Garrido-Moreno V, Sanhueza-Olivares F, Guerrero-Moncayo A, Chiong M, Castro PF, García L, Gabrielli L, Corbalán R, Garrido-Olivares L, Lavandero S. VCAM-1 as a predictor biomarker in cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166170. [PMID: 34000374 DOI: 10.1016/j.bbadis.2021.166170] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
The vascular cellular adhesion molecule-1 (VCAM-1) is a protein that canonically participates in the adhesion and transmigration of leukocytes to the interstitium during inflammation. VCAM-1 expression, together with soluble VCAM-1 (sVCAM-1) induced by the shedding of VCAM-1 by metalloproteinases, have been proposed as biomarkers in immunological diseases, cancer, autoimmune myocarditis, and as predictors of mortality and morbidity in patients with chronic heart failure (HF), endothelial injury in patients with coronary artery disease, and arrhythmias. This revision aims to discuss the role of sVCAM-1 as a biomarker to predict the occurrence, development, and preservation of cardiovascular disease.
Collapse
Affiliation(s)
- Mayarling Francisca Troncoso
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jafet Ortiz-Quintero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Bioanalysis & Immunology, Faculty of Sciences, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Valeria Garrido-Moreno
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernanda Sanhueza-Olivares
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandra Guerrero-Moncayo
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo F Castro
- Division of Cardiovascular Diseases, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lorena García
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luigi Gabrielli
- Division of Cardiovascular Diseases, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ramón Corbalán
- Division of Cardiovascular Diseases, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Garrido-Olivares
- Division of Surgery, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Desoye G, Wells JCK. Pregnancies in Diabetes and Obesity: The Capacity-Load Model of Placental Adaptation. Diabetes 2021; 70:823-830. [PMID: 33741605 PMCID: PMC7980199 DOI: 10.2337/db20-1111] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Excess nutritional supply to the growing fetus, resulting from maternal diabetes and obesity, is associated with increased risks of fetal maldevelopment and adverse metabolic conditions in postnatal life. The placenta, interposed between mother and fetus, serves as the gateway between the two circulations and is usually considered to mediate maternal exposures to the fetus through a direct supply line. In this Perspective, however, we argue that the placenta is not an innocent bystander and mounts responses to fetal "signals of distress" to sustain its own adequate function and protect the fetus. We describe several types of protection that the placenta can offer the fetus against maternal metabolic perturbations and offer a theoretical model of how the placenta responds to the intrauterine environment in maternal diabetes and obesity to stabilize the fetal environment. Our approach supports growing calls for early screening and control of pregnancy metabolism to minimize harmful fetal outcomes.
Collapse
|
25
|
Sharma A, Choi JSY, Stefanovic N, Al-Sharea A, Simpson DS, Mukhamedova N, Jandeleit-Dahm K, Murphy AJ, Sviridov D, Vince JE, Ritchie RH, de Haan JB. Specific NLRP3 Inhibition Protects Against Diabetes-Associated Atherosclerosis. Diabetes 2021; 70:772-787. [PMID: 33323396 DOI: 10.2337/db20-0357] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/01/2020] [Indexed: 11/13/2022]
Abstract
Low-grade persistent inflammation is a feature of diabetes-driven vascular complications, in particular activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome to trigger the maturation and release of the inflammatory cytokine interleukin-1β (IL-1β). We investigated whether inhibiting the NLRP3 inflammasome, through the use of the specific small-molecule NLRP3 inhibitor MCC950, could reduce inflammation, improve vascular function, and protect against diabetes-associated atherosclerosis in the streptozotocin-induced diabetic apolipoprotein E-knockout mouse. Diabetes led to an approximately fourfold increase in atherosclerotic lesions throughout the aorta, which were significantly attenuated with MCC950 (P < 0.001). This reduction in lesions was associated with decreased monocyte-macrophage content, reduced necrotic core, attenuated inflammatory gene expression (IL-1β, tumor necrosis factor-α, intracellular adhesion molecule 1, and MCP-1; P < 0.05), and reduced oxidative stress, while maintaining fibrous cap thickness. Additionally, vascular function was improved in diabetic vessels of mice treated with MCC950 (P < 0.05). In a range of cell lines (murine bone marrow-derived macrophages, human monocytic THP-1 cells, phorbol 12-myristate 13-acetate-differentiated human macrophages, and aortic smooth muscle cells from humans with diabetes), MCC950 significantly reduced IL-1β and/or caspase-1 secretion and attenuated leukocyte-smooth muscle cell interactions under high glucose or lipopolysaccharide conditions. In summary, MCC950 reduces plaque development, promotes plaque stability, and improves vascular function, suggesting that targeting NLRP3-mediated inflammation is a novel therapeutic strategy to improve diabetes-associated vascular disease.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Judy S Y Choi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nada Stefanovic
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Annas Al-Sharea
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel S Simpson
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Karin Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - James E Vince
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Abstract
Since the initial reports implicating caveolin-1 (CAV1) in neoplasia, the scientific community has made tremendous strides towards understanding how CAV1-dependent signaling and caveolae assembly modulate solid tumor growth. Once a solid neoplastic tumor reaches a certain size, it will increasingly rely on its stroma to meet the metabolic demands of the rapidly proliferating cancer cells, a limitation typically but not exclusively addressed via the formation of new blood vessels. Landmark studies using xenograft tumor models have highlighted the importance of stromal CAV1 during neoplastic blood vessel growth from preexisting vasculature, a process called angiogenesis, and helped identify endothelium-specific signaling events regulated by CAV1, such as vascular endothelial growth factor (VEGF) receptors as well as the endothelial nitric oxide (NO) synthase (eNOS) systems. This chapter provides a glimpse into the signaling events modulated by CAV1 and its scaffolding domain (CSD) during endothelial-specific aspects of neoplastic growth, such as vascular permeability, angiogenesis, and mechanotransduction.
Collapse
Affiliation(s)
- Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), 2176 Health Sciences mall, room 217, Vancouver, BC, V6T 1Z3, Canada. .,Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada.
| |
Collapse
|
27
|
The Inflammatory Cytokine IL-3 Hampers Cardioprotection Mediated by Endothelial Cell-Derived Extracellular Vesicles Possibly via Their Protein Cargo. Cells 2020; 10:cells10010013. [PMID: 33374685 PMCID: PMC7822476 DOI: 10.3390/cells10010013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/09/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
The biological relevance of extracellular vesicles (EV) released in an ischemia/reperfusion setting is still unclear. We hypothesized that the inflammatory microenvironment prevents cardioprotection mediated by endothelial cell (EC)-derived extracellular vesicles. The effects of naïve EC-derived EV (eEV) or eEV released in response to interleukin-3 (IL-3) (eEV-IL-3) were evaluated in cardiomyoblasts (H9c2) and rat hearts. In transwell assay, eEV protected the H9c2 exposed to hypoxia/reoxygenation (H/R) more efficiently than eEV-IL-3. Conversely, only eEV directly protected H9c2 cells to H/R-induced damage. Consistent with this latter observation, eEV, but not eEV-IL-3, exerted beneficial effects in the whole heart. Protein profiles of eEV and eEV-IL-3, established using label-free mass spectrometry, demonstrated that IL-3 drives changes in eEV-IL-3 protein cargo. Gene ontology analysis revealed that both eEV and eEV-IL-3 were equipped with full cardioprotective machinery, including the Nitric Oxide Signaling in the Cardiovascular System. eEV-IL-3 were also enriched in the endothelial-nitric oxide-synthase (eNOS)-antagonist caveolin-1 and proteins related to the inflammatory response. In vitro and ex vivo experiments demonstrated that a functional Mitogen-Activated Protein Kinase Kinase (MEK1/2)/eNOS/guanylyl-cyclase (GC) pathway is required for eEV-mediated cardioprotection. Consistently, eEV were found enriched in MEK1/2 and able to induce the expression of B-cell-lymphoma-2 (Bcl-2) and the phosphorylation of eNOS in vitro. We conclude that an inflammatory microenvironment containing IL-3 changes the eEV cargo and impairs eEV cardioprotective action.
Collapse
|
28
|
Han WM, Chen XC, Li GR, Wang Y. Acacetin Protects Against High Glucose-Induced Endothelial Cells Injury by Preserving Mitochondrial Function via Activating Sirt1/Sirt3/AMPK Signals. Front Pharmacol 2020; 11:607796. [PMID: 33519472 PMCID: PMC7844858 DOI: 10.3389/fphar.2020.607796] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022] Open
Abstract
The strategy of decreasing atherosclerotic cardiovascular disorder is imperative for reducing premature death and improving quality of life in patients with diabetes mellitus. The aim of this study was to investigate whether the natural flavone acacetin could protect against endothelial injury induced by high glucose and attenuate diabetes-accelerated atherosclerosis in streptozotocin-(STZ) induced diabetic ApoE−/− mice model. It was found that in human umbilical vein endothelial cells (HUVECs) cultured with normal 5.5 mM or high 33 mM glucose, acacetin (0.3–3 μM) exerted strong cytoprotective effects by reversing high glucose-induced viability reduction and reducing apoptosis and excess production of intracellular reactive oxygen species (ROS) and malondialdehyde in a concentration-dependent manner. Acacetin countered high glucose-induced depolarization of mitochondrial membrane potential and reduction of ATP product and mitoBcl-2/mitoBax ratio. Silencing Sirt3 abolished the beneficial effects of acacetin. Further analysis revealed that these effects of acacetin rely on Sirt1 activation by increasing NAD+ followed by increasing Sirt3, pAMPK and PGC-1α. In STZ-diabetic mice, acacetin significantly upregulated the decreased signaling molecules (i.e. SOD, Bcl-2, PGC-1α, pAMPK, Sirt3 and Sirt1) in aorta tissue and attenuated atherosclerosis. These results indicate that vascular endothelial protection of acacetin by activating Sirt1/Sirt3/AMPK signals is likely involved in alleviating diabetes-accelerated atherosclerosis by preserving mitochondrial function, which suggests that acacetin may be a drug candidate for treating cardiovascular disorder in patients with diabetes.
Collapse
Affiliation(s)
- Wei-Min Han
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xu-Chang Chen
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China.,Nanjing Amazigh Pharma Limited, Nanjing, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
29
|
Oe Y, Mitsui S, Sato E, Shibata N, Kisu K, Sekimoto A, Miyazaki M, Sato H, Ito S, Takahashi N. Lack of Endothelial Nitric Oxide Synthase Accelerates Ectopic Calcification in Uremic Mice Fed an Adenine and High Phosphorus Diet. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:283-293. [PMID: 33159888 DOI: 10.1016/j.ajpath.2020.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Ectopic calcification is a risk of cardiovascular disease in chronic kidney disease (CKD) patients, and impaired endothelial nitric oxide synthase (eNOS) is involved in the CKD complications. However, whether eNOS dysfunction is a cause of ectopic calcification in CKD remains to be elucidated. To address this issue, we investigated the role of eNOS in ectopic calcification in mice with renal injury caused by an adenine and high-phosphorus (Ade + HP) diet. DBA/2J mice, a calcification-sensitive strain, were fed Ade + HP for 3 weeks. Expression levels of eNOS-related genes were reduced significantly in their calcified aorta. C57BL/6J is a calcification-resistant strain, and wild-type mice showed mild calcified lesions in the aorta and kidney when given an Ade + HP diet for 4 weeks. In contrast, a lack of eNOS led to the development of severe aortic calcification accompanied by an increase in runt-related transcription factor 2, an osteochondrogenic marker. Increased renal calcium deposition and the tubular injury score were remarkable in mice lacking eNOS-fed Ade + HP. Exacerbation of ectopic calcification by a lack of eNOS is associated with increased oxidative stress markers such as nicotinamide adenine dinucleotide phosphate oxidases. In conclusion, eNOS is critically important in preventing ectopic calcification. Therefore, the maintenance of eNOS is useful to reduce cardiovascular disease events and to improve prognosis in CKD patients.
Collapse
Affiliation(s)
- Yuji Oe
- Department of Community Medical Support, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shohei Mitsui
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Emiko Sato
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Naoko Shibata
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyomi Kisu
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiyo Sekimoto
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Sato
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Takahashi
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan.
| |
Collapse
|
30
|
Ning K, Wang MJ, Lin G, Zhang YL, Li MY, Yang BF, Chen Y, Huang Y, Li ZM, Huang YJ, Zhu L, Liang K, Yu B, Zhu YZ, Zhu YC. eNOS-Nitric Oxide System Contributes to a Novel Antiatherogenic Effect of Leonurine via Inflammation Inhibition and Plaque Stabilization. J Pharmacol Exp Ther 2020; 373:463-475. [PMID: 32238453 DOI: 10.1124/jpet.119.264887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
Leonurine (LEO) is a bioactive small molecular compound that has protective effects on the cardiovascular system and prevents the early progression of atherosclerosis; however, it is not clear whether LEO is effective for plaque stability. A novel mouse atherosclerosis model involving tandem stenosis (TS) of the right carotid artery combined with western diet (WD) feeding was used. Apolipoprotein E gene-deficient mice were fed with a WD and received LEO administration daily for 13 weeks. TS was introduced 6 weeks after the onset of experiments. We found that LEO enhanced plaque stability by increasing fibrous cap thickness and collagen content while decreasing the population of CD68-positive cells. Enhanced plaque stability by LEO was associated with the nitric oxide synthase (NOS)-nitric oxide (NO) system. LEO restored the balance between endothelial NOS(E)- and inducible NOS(iNOS)-derived NO production; suppressed the NF-κB signaling pathway; reduced the level of the inflammatory infiltration in plaque, including cytokine interleukin 6; and downregulated the expression of adhesion molecules. These findings support the distinct role of LEO in plaque stabilization. In vitro studies with oxidized low-density lipoprotein-challenged human umbilical vein endothelial cells revealed that LEO balanced NO production and inhibited NF-κB/P65 nuclear translocation, thus mitigating inflammation. In conclusion, the restored balance of the NOS-NO system and mitigated inflammation contribute to the plaque-stabilizing effect of LEO. SIGNIFICANCE STATEMENT: LEO restored the balance between endothelial NOS and inducible NOS in NO production and inhibited excessive inflammation in atherosclerotic "unstable" and rupture-prone plaques in apolipoprotein E gene-deficient mice. The protective effect of LEO for stabilizing atherosclerotic plaques was due to improved collagen content, increased fibrous cap thickness, and decreased accumulation of macrophages/foam cells. So far, LEO has passed the safety and feasibility test of phase I clinical trial.
Collapse
Affiliation(s)
- Ke Ning
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ming-Jie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ge Lin
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Lin Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Bao-Feng Yang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yong Huang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Zhi-Ming Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Jun Huang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Lei Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Kun Liang
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Bo Yu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, Shanghai Medical College, School of Basic Medical Sciences (K.N., M.-J.W., G.L., Y.-L.Z., M.-Y.L., Y.C., Y.H., Z.-M.L., Y.-C.Z.), Department of Vascular Surgery, Huashan Hospital (Y.-J.H., L.Z., K.L., B.Y.), and Institutes of Science and Technology for Brain-inspired intelligence (B.-F.Y.), Fudan University, Shanghai, China; and State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (Y.-Z.Z.)
| |
Collapse
|
31
|
Role of Caveolin-1 in Diabetes and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9761539. [PMID: 32082483 PMCID: PMC7007939 DOI: 10.1155/2020/9761539] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/10/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
It is estimated that in 2017 there were 451 million people with diabetes worldwide. These figures are expected to increase to 693 million by 2045; thus, innovative preventative programs and treatments are a necessity to fight this escalating pandemic disorder. Caveolin-1 (CAV1), an integral membrane protein, is the principal component of caveolae in membranes and is involved in multiple cellular functions such as endocytosis, cholesterol homeostasis, signal transduction, and mechanoprotection. Previous studies demonstrated that CAV1 is critical for insulin receptor-mediated signaling, insulin secretion, and potentially the development of insulin resistance. Here, we summarize the recent progress on the role of CAV1 in diabetes and diabetic complications.
Collapse
|
32
|
Qin CX, Anthonisz J, Leo CH, Kahlberg N, Velagic A, Li M, Jap E, Woodman OL, Parry LJ, Horowitz JD, Kemp-Harper BK, Ritchie RH. Nitric Oxide Resistance, Induced in the Myocardium by Diabetes, Is Circumvented by the Nitric Oxide Redox Sibling, Nitroxyl. Antioxid Redox Signal 2020; 32:60-77. [PMID: 31680536 DOI: 10.1089/ars.2018.7706] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aim: Impairment of tissue responsiveness to exogenous and endogenous nitric oxide (NO•), known as NO• resistance, occurs in many cardiovascular disease states, prominently in diabetes and especially in the presence of marked hyperglycemia. In this study, we sought to determine in moderate and severe diabetes (i) whether NO• resistance also occurs in the myocardium, and (ii) whether the NO• redox sibling nitroxyl (HNO) circumvents this. Results: The spectrum of acute NO• effects (induced by diethylamine-NONOate), including vasodilation, and enhanced myocardial contraction and relaxation were impaired by moderately diabetic rats ([blood glucose] ∼20 mM). In contrast, acute HNO effects (induced by isopropylamine-NONOate) were preserved even in more severe diabetes ([blood glucose] >28 mM). Intriguingly, the positive inotropic effects of HNO were significantly enhanced in diabetic rat hearts. Further, progressive attenuation of soluble guanylyl cyclase (sGC) contribution to myocardial NO• responses occurred with increasing severity of diabetes. Nevertheless, activation of sGC by HNO remained intact in the myocardium. Innovation: Diabetes is associated with marked attenuation of vascular and myocardial effects of NO and NO donors, and this NO• resistance is circumvented by HNO, suggesting potential therapeutic utility for HNO donors in cardiovascular emergencies in diabetics. Conclusion: These results provide the first evidence that NO• resistance occurs in diabetic hearts, and that HNO largely circumvents this problem. Further, the positive inotropic and lusitropic effects of HNO are enhanced in a severely diabetic myocardium, a finding that warrants further mechanistic interrogation. The results support a potential role for therapeutic HNO administration in acute treatment of ischemia and/or heart failure in diabetics.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Jarryd Anthonisz
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Chen Huei Leo
- School of Biosciences, University of Melbourne, Parkville, Australia.,Science and Maths Cluster, Singapore University of Technology & Design, Singapore Singapore
| | - Nicola Kahlberg
- School of Biosciences, University of Melbourne, Parkville, Australia
| | - Anida Velagic
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Mandy Li
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Edwina Jap
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Owen L Woodman
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Laura J Parry
- School of Biosciences, University of Melbourne, Parkville, Australia
| | - John D Horowitz
- Cardiology Unit, The Queen Elizabeth Hospital, Basil Hetzel Institute, The University of Adelaide, Woodville SA, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| |
Collapse
|
33
|
Potential Impact of MicroRNA Gene Polymorphisms in the Pathogenesis of Diabetes and Atherosclerotic Cardiovascular Disease. J Pers Med 2019; 9:jpm9040051. [PMID: 31775219 PMCID: PMC6963792 DOI: 10.3390/jpm9040051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, small (18–23 nucleotides), non-coding RNA molecules. They regulate the posttranscriptional expression of their target genes. MiRNAs control vital physiological processes such as metabolism, development, differentiation, cell cycle and apoptosis. The control of the gene expression by miRNAs requires efficient binding between the miRNA and their target mRNAs. Genome-wide association studies (GWASs) have suggested the association of single-nucleotide polymorphisms (SNPs) with certain diseases in various populations. Gene polymorphisms of miRNA target sites have been implicated in diseases such as cancers, diabetes, cardiovascular and Parkinson’s disease. Likewise, gene polymorphisms of miRNAs have been reported to be associated with diseases. In this review, we discuss the SNPs in miRNA genes that have been associated with diabetes and atherosclerotic cardiovascular disease in different populations. We also discuss briefly the potential underlining mechanisms through which these SNPs increase the risk of developing these diseases.
Collapse
|
34
|
Xue M, Shi Y, Pang A, Men L, Hu Y, Zhou P, Long G, Tian X, Wang R, Zhao Y, Liao X, Shen Y, Cui Y. Corin plays a protective role via upregulating MAPK and downregulating eNOS in diabetic nephropathy endothelial dysfunction. FASEB J 2019; 34:95-106. [PMID: 31914697 DOI: 10.1096/fj.201900531rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 09/22/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
Diabetic nephropathy (DN) is one of the leading causes of mortality in diabetic patients, but its pathogenesis is unclear. We aimed to study the role of the pro-ANP convertase Corin in the pathogenesis of DN. Corin and ANP expression in DN rat kidneys and high-glucose-treated HK-2 cells was analyzed by real-time PCR, western blotting, and immunohistochemical staining. The effect of Corin-siRNA or ANP-siRNA HK-2 cells on EA.hy926 cell migration was determined by scratch-wound healing assay. The expression of mitogen-activated protein kinase (MAPK) and endothelial NO synthase (eNOS) in EA.hy926 cells treated with conditioned medium from Corin-siRNA- or ANP-siRNA-transfected HK-2 cells was determined by western blotting. We found a significant reduction in Corin and ANP expression in DN rat kidneys. These results were recapitulated in HK-2 cells treated with high glucose. EA.hy926 cells treated with conditioned medium from Corin-deficient HK-2 cells had inhibited migration, increased MAPK activity, and decreased eNOS activity. Similar effects were observed with ANP-siRNA transfection. Finally, adding ANP to the Corin-deficient HK-2 conditioned medium rescued the above defects, indicating that Corin mediates its effects through ANP. In conclusion, Corin plays a renoprotective role through pro-ANP processing, and defects in Corin cause endothelial dysfunction through MAPK and eNOS signaling in DN.
Collapse
Affiliation(s)
- Meiting Xue
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yue Shi
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Li Men
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yahui Hu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Pengfei Zhou
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Guangfeng Long
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xin Tian
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Rong Wang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xudong Liao
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Yanna Shen
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yujie Cui
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| |
Collapse
|
35
|
White Z, Milad N, Tehrani AY, Chen WWH, Donen G, Sellers SL, Bernatchez P. Angiotensin II receptor blocker losartan exacerbates muscle damage and exhibits weak blood pressure-lowering activity in a dysferlin-null model of Limb-Girdle muscular dystrophy type 2B. PLoS One 2019; 14:e0220903. [PMID: 31404091 PMCID: PMC6690544 DOI: 10.1371/journal.pone.0220903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/25/2019] [Indexed: 01/01/2023] Open
Abstract
There is no cure or beneficial management option for Limb-Girdle muscular dystrophy (MD) type 2B (LGMD2B). Losartan, a blood pressure (BP) lowering angiotensin II (AngII) receptor type 1 (ATR1) blocker (ARB) with unique anti-transforming growth factor-β (TGF-β) properties, can protect muscles in various types of MD such as Duchenne MD, suggesting a potential benefit for LGMD2B patients. Herein, we show in a mild, dysferlin-null mouse model of LGMD2B that losartan increased quadriceps muscle fibrosis (142%; P<0.0001). In a severe, atherogenic diet-fed model of LGMD2B recently described by our group, losartan further exacerbated dysferlin-null mouse muscle wasting in quadriceps and triceps brachii, two muscles typically affected by LGMD2B, by 40% and 51%, respectively (P<0.05). Lower TGF-β signalling was not observed with losartan, therefore plasma levels of atherogenic lipids known to aggravate LGMD2B severity were investigated. We report that losartan increased both plasma triglycerides and cholesterol concentrations in dysferlin-null mice. Other protective properties of losartan, such as increased nitric oxide release and BP lowering, were also reduced in the absence of dysferlin expression. Our data suggest that LGMD2B patients may show some resistance to the primary BP-lowering effects of losartan along with accelerated muscle wasting and dyslipidemia. Hence, we urge caution on the use of ARBs in this population as their ATR1 pathway may be dysfunctional.
Collapse
Affiliation(s)
- Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
- * E-mail: (ZW); (PB)
| | - Nadia Milad
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Arash Y. Tehrani
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - William Wei-Han Chen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Graham Donen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Stephanie L. Sellers
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
- * E-mail: (ZW); (PB)
| |
Collapse
|
36
|
White Z, Milad N, Tehrani AY, Lamothe J, Hogg JC, Esfandiarei M, Seidman M, Booth S, Hackett TL, Morissette MC, Bernatchez P. Sildenafil Prevents Marfan-Associated Emphysema and Early Pulmonary Artery Dilation in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1536-1546. [PMID: 31125551 DOI: 10.1016/j.ajpath.2019.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 01/24/2023]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in fibrillin-1 (Fbn1). Although aortic rupture is the major cause of mortality in MFS, patients also experience pulmonary complications, which are poorly understood. Loss of basal nitric oxide (NO) production and vascular integrity has been implicated in MFS aortic root disease, yet their contribution to lung complications remains unknown. Because of its capacity to potentiate the vasodilatory NO/cyclic guanylate monophosphate signaling pathway, we assessed whether the phosphodiesterase-5 inhibitor, sildenafil (SIL), could attenuate aortic root remodeling and emphysema in a mouse model of MFS. Despite increasing NO-dependent vasodilation, SIL unexpectedly elevated mean arterial blood pressure, failed to inhibit MFS aortic root dilation, and exacerbated elastic fiber fragmentation. In the lung, early pulmonary artery dilation observed in untreated MFS mice was delayed by SIL treatment, and the severe emphysema-like alveolar destruction was prevented. In addition, improvements in select parameters of lung function were documented. Subsequent microarray analyses showed changes to gene signatures involved in the inflammatory response in the MFS lung treated with SIL, without significant down-regulation of connective tissue or transforming growth factor-β signaling genes. Because phosphodiesterase-5 inhibition leads to improved lung histopathology and function, the effects of SIL against emphysema warrant further investigation in the settings of MFS despite limited efficacy on aortic root remodeling.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada; Quebec Heart and Lung Institute, Université Laval, Québec City, Quebec, Canada; Department of Medicine, Université Laval, Québec City, Quebec, Canada
| | - Arash Y Tehrani
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Jennifer Lamothe
- Quebec Heart and Lung Institute, Université Laval, Québec City, Quebec, Canada; Department of Medicine, Université Laval, Québec City, Quebec, Canada
| | - James C Hogg
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mitra Esfandiarei
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, Arizona
| | - Michael Seidman
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven Booth
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Mathieu C Morissette
- Quebec Heart and Lung Institute, Université Laval, Québec City, Quebec, Canada; Department of Medicine, Université Laval, Québec City, Quebec, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
37
|
Sellers SL, Milad N, Chan R, Mielnik M, Jermilova U, Huang PL, de Crom R, Hirota JA, Hogg JC, Sandor GG, Van Breemen C, Esfandiarei M, Seidman MA, Bernatchez P. Inhibition of Marfan Syndrome Aortic Root Dilation by Losartan: Role of Angiotensin II Receptor Type 1-Independent Activation of Endothelial Function. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 188:574-585. [PMID: 29433732 DOI: 10.1016/j.ajpath.2017.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/18/2017] [Accepted: 11/16/2017] [Indexed: 01/13/2023]
Abstract
Marfan syndrome (MFS) is a genetic disorder that frequently leads to aortic root dissection and aneurysm. Despite promising preclinical and pilot clinical data, a recent large-scale study using antihypertensive angiotensin II (AngII) receptor type 1 (ATR1) blocker losartan has failed to meet expectations at preventing MFS-associated aortic root dilation, casting doubts about optimal therapy. To study the deleterious role of normal ATR1 signaling in aortic root widening, we generated MFS mice lacking ATR1a expression in an attempt to preserve protective ATR2 signaling. Despite being hypotensive and resistant to AngII vasopressor effects, MFS/ATR1a-null mice showed unabated aortic root enlargement and remained fully responsive to losartan, confirming that blood pressure lowering is of minor therapeutic value in MFS and that losartan's antiremodeling properties may be ATR1 independent. Having shown that MFS causes endothelial dysfunction and that losartan can activate endothelial function in mice and patients, we found that nitric oxide synthase (NOS) inhibition renders losartan therapeutically inactive, whereas multiple transgenic and pharmacologic models of endothelial NOS activation block aortic root dilation by correcting extracellular signal-regulated kinase signaling. In vitro, losartan can increase endothelial NO release in the absence of AngII and correct MFS NO levels in vivo. Our data suggest that increased protective endothelial function, rather than ATR1 inhibition or blood pressure lowering, might be of therapeutic significance in preventing aortic root disease in MFS.
Collapse
Affiliation(s)
- Stephanie L Sellers
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Rayleigh Chan
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Michael Mielnik
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Una Jermilova
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Paul L Huang
- Cardiovascular Research Centre, Massachusetts General Hospital, Harvard University, Charlestown, Massachusetts
| | - Rini de Crom
- Department of Cell Biology and Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeremy A Hirota
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Division of Respiratory Medicine, Department of Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - James C Hogg
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - George G Sandor
- Providence Health Care, and the Child and Family Research Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Casey Van Breemen
- Providence Health Care, and the Child and Family Research Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Mitra Esfandiarei
- Department of Biomedical Sciences, Midwestern University, Glendale, Arizona
| | - Michael A Seidman
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada.
| |
Collapse
|
38
|
Vlacil AK, Schuett J, Schieffer B, Grote K. Variety matters: Diverse functions of monocyte subtypes in vascular inflammation and atherogenesis. Vascul Pharmacol 2018; 113:9-19. [PMID: 30553027 DOI: 10.1016/j.vph.2018.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022]
Abstract
Monocytes are important mediators of the innate immunity by recognizing and attacking especially bacterial pathogens but also play crucial roles in various inflammatory diseases, including vascular inflammation and atherosclerosis. Maturation, differentiation and function of monocytes have been intensively explored for a long time in innumerable experimental and clinical studies. Monocytes do not represent a uniform cell type but could be further subdivided into subpopulations with distinct features and functions. Those subpopulations have been identified in experimental mouse models as well as in humans, albeit distinguished by different cell surface markers. While Ly6C is used for subpopulation differentiation in mice, corresponding human subsets are differentiated by CD14 and CD16. In this review, we specifically focused on new experimental insights from recent years mainly in regard to murine monocyte subpopulations and their roles in vascular inflammation und atherogenesis.
Collapse
Affiliation(s)
| | - Jutta Schuett
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | | | - Karsten Grote
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
39
|
Pickering RJ, Rosado CJ, Sharma A, Buksh S, Tate M, de Haan JB. Recent novel approaches to limit oxidative stress and inflammation in diabetic complications. Clin Transl Immunology 2018; 7:e1016. [PMID: 29713471 PMCID: PMC5905388 DOI: 10.1002/cti2.1016] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 12/25/2022] Open
Abstract
Diabetes is considered a major burden on the healthcare system of Western and non‐Western societies with the disease reaching epidemic proportions globally. Diabetic patients are highly susceptible to developing micro‐ and macrovascular complications, which contribute significantly to morbidity and mortality rates. Over the past decade, a plethora of research has demonstrated that oxidative stress and inflammation are intricately linked and significant drivers of these diabetic complications. Thus, the focus now has been towards specific mechanism‐based strategies that can target both oxidative stress and inflammatory pathways to improve the outcome of disease burden. This review will focus on the mechanisms that drive these diabetic complications and the feasibility of emerging new therapies to combat oxidative stress and inflammation in the diabetic milieu.
Collapse
Affiliation(s)
- Raelene J Pickering
- Department of Diabetes Central Clinical School Monash University Melbourne VIC Australia
| | - Carlos J Rosado
- Department of Diabetes Central Clinical School Monash University Melbourne VIC Australia
| | - Arpeeta Sharma
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Shareefa Buksh
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Mitchel Tate
- Heart Failure Pharmacology Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Judy B de Haan
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| |
Collapse
|
40
|
Oktay AA, Akturk HK, Esenboğa K, Javed F, Polin NM, Jahangir E. Pathophysiology and Prevention of Heart Disease in Diabetes Mellitus. Curr Probl Cardiol 2018; 43:68-110. [DOI: 10.1016/j.cpcardiol.2017.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Yu L, Yin M, Yang X, Lu M, Tang F, Wang H. Calpain inhibitor I attenuates atherosclerosis and inflammation in atherosclerotic rats through eNOS/NO/NF-κB pathway. Can J Physiol Pharmacol 2018; 96:60-67. [DOI: 10.1139/cjpp-2016-0652] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that calpain, the Ca2+-sensitive cysteine protease, gets involved in atherogenesis. This study aimed to investigate the effects of calpain inhibitor I (CAI, 5 mg/kg per day) with or without NG-nitro-l-arginine-methyl ester (l-NAME) (100 mg/kg per day), the inhibitor of nitric oxide synthase (NOS), on atherosclerosis and inflammation in a rat model induced by high-cholesterol diet (HCD). The results demonstrated HCD increased protein expression of calpain-1 but not calpain-2 in aortic tissue. In addition, CAI reduced the thickness of atherosclerotic intima compared with HCD group, which was weakened by the l-NAME combination. CAI with or without l-NAME decreased the activity of calpain in the aorta. Also, CAI decreased the expressions of vascular cell adhesion molecule-1 (VCAM-1), intracellular cell adhesion molecule-1 (ICAM-1), and monocyte chemoattractant protein-1 (MCP-1) in the aorta at the levels of both mRNA and protein. Furthermore, CAI increased the activity and the protein expression of endothelial NOS (eNOS) accompanied by increased content of NO and downregulated the protein expression of nuclear factor κB (NF-κB) of the nucleus in the aorta. However, the abovementioned effects were at least partly cancelled by l-NAME except for the protein expression of eNOS. The results suggested that CAI attenuated atherosclerosis and inflammation through eNOS/NO/NF-κB pathway.
Collapse
Affiliation(s)
- Lan Yu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
- Central Hospital of Yingkou Development Areas, Yingkou 115007, China
| | - Meihui Yin
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Xueyan Yang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
- Internal Medicine-Cardiovascular Departments, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Futian Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
42
|
Sellers SL, Milad N, White Z, Pascoe C, Chan R, Payne GW, Seow C, Rossi F, Seidman MA, Bernatchez P. Increased nonHDL cholesterol levels cause muscle wasting and ambulatory dysfunction in the mouse model of LGMD2B. J Lipid Res 2017; 59:261-272. [PMID: 29175948 DOI: 10.1194/jlr.m079459] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/19/2017] [Indexed: 12/22/2022] Open
Abstract
Progressive limb and girdle muscle atrophy leading to loss of ambulation is a hallmark of dysferlinopathies, which include limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. However, animal models fail to fully reproduce the disease severity observed in humans, with dysferlin-null (Dysf-/-) mice exhibiting minor muscle damage and weakness without dramatic ambulatory dysfunction. As we have previously reported significant Dysf expression in blood vessels, we investigated the role of vascular function in development of muscle pathology by generating a Dysf-deficient mouse model with vascular disease. This was achieved by crossing Dysf-/- mice with ApoE-/- mice, which have high levels of nonHDL-associated cholesterol. Double-knockout Dysf-/-ApoE-/- mice exhibited severe ambulatory dysfunction by 11 months of age. In limb-girdle muscles, histology confirmed dramatic muscle wasting, fibrofatty replacement, and myofiber damage in Dysf-/-ApoE-/- mice without affecting the ratio of centrally nucleated myofibers. Although there were no major changes in ex vivo diaphragm and soleus muscle function, histological analyses revealed these muscles to be untouched by damage and remodelling. In all, these data suggest that cholesterol may be deleterious to dysferlinopathic muscle and lead to ambulatory dysfunction. Moreover, differences in plasma lipid handling between mice and humans could be a key factor affecting dysferlinopathy severity.
Collapse
Affiliation(s)
- Stephanie L Sellers
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Chris Pascoe
- St. Paul's Hospital, University of British Columbia, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Rayleigh Chan
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Geoffrey W Payne
- Providence Health Care, University of Northern British Columbia, Prince George, Canada
| | - Chun Seow
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Fabio Rossi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Biomedical Research Centre, University of British Columbia, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Michael A Seidman
- St. Paul's Hospital, University of British Columbia, Vancouver, Canada.,Department of Pathology, Prince George, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada .,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
43
|
Pyrrolidine dithiocarbamate ameliorates endothelial dysfunction in thoracic aorta of diabetic rats by preserving vascular DDAH activity. PLoS One 2017; 12:e0179908. [PMID: 28715444 PMCID: PMC5513417 DOI: 10.1371/journal.pone.0179908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/07/2017] [Indexed: 01/27/2023] Open
Abstract
Objective Endothelial dysfunction plays a pivotal role in the development of diabetic cardiovascular complications. Accumulation of endogenous nitric oxide synthase (NOS) inhibitor asymmetric dimethylarginine (ADMA) and inhibition of dimethylarginine dimethylaminohydrolase (DDAH) activity have been involved in diabetic endothelial dysfunction. This study was to investigate the effect of pyrrolidine dithiocarbamate (PDTC) on impairment of endothelium-dependent vasodilatation in diabetic rats and its potential mechanism. Methods Diabetic rats were induced by a single intraperitoneal injection of streptozotocin (60mg/kg), and PDTC (10mg/kg) was given in drinking water for 8 weeks. Blood glucose and serum ADMA concentrations were measured in experimental rats. Recombinant adenovirus encoding human DDAH2 gene were constructed and ex vivo transferred to isolated rat aortas. The maximal relaxation (Emax) and half maximal effective concentration (EC50) of aortic rings response to accumulative concentrations of acetylcholine and vascular DDAH activity were examined before and after gene transfection. Results Diabetic rats displayed significant elevations of blood glucose and serum ADMA levels compared to control group (P<0.01). Vascular DDAH activity and endothelium-dependent relaxation of aortas were inhibited, as expressed by the decreased Emax and increased EC50 in diabetic rats compared to control rats (P<0.01). Treatment with PDTC not only decreased blood glucose and serum ADMA concentration (P<0.01) but also restored vascular DDAH activity and endothelium-dependent relaxation, evidenced by the higher Emax and lower EC50 in PDTC-treated diabetic rats compared to untreated diabetic rats (P<0.01). Similar restoration of Emax, EC50 and DDAH activity were observed in diabetic aortas after DDAH2-gene transfection. Conclusions These results indicate that PDTC could ameliorate impairment of endothelium-dependent relaxation in diabetic rats. The underlying mechanisms might be related to preservation of vascular DDAH activity and consequent reduction of endogenous ADMA in endothelium via its antioxidant action. This study highlights the therapeutic potential of PDTC in impaired vasodilation and provides a new strategy for treatment of diabetic cardiovascular complications.
Collapse
|
44
|
Li L, Huang B, Song S, Sohun H, Rao Z, Tao L, Jin Q, Zeng J, Wu R, Ji K, Lin J, Wu L, Chu M. A20 functions as mediator in TNFα-induced injury of human umbilical vein endothelial cells through TAK1-dependent MAPK/eNOS pathway. Oncotarget 2017; 8:65230-65239. [PMID: 29029426 PMCID: PMC5630326 DOI: 10.18632/oncotarget.18191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 12/15/2022] Open
Abstract
A20, a negative regulator of nuclear factor κB signaling, has been shown to attenuate atherosclerotic events. Transforming growth factor beta-activated kinase 1 (TAK1) plays a critical role in TNFα-induced atherosclerosis via endothelial nitric oxide (NO) synthase (eNOS) uncoupling and NO reduction. In the study, we investigated the hypothesis that A20 protected endothelial cell injury induced by TNFα through modulating eNOS activity and TAK1 signalling. Human umbilical vein endothelial cells (HUVECs) were stimulated by TNFα. The impact of A20 on cell apoptosis, eNOS expression and NO production and related TAK1 pathway were detected. Both eNOS and NO production were remarkably reduced. TAK1, p38 MAPK phosphorylation and HUVECs apoptosis were enhanced after TNFα stimulation for 2 hrs. Inhibition of A20 significantly activated TAK1, p38 MAPK phosphorylation, and cell apoptosis, but blocked eNOS expression and NO production. Furthermore, p38 MAPK expression was suppressed by A20 over-expression, but re-enhanced by inhibiting A20 or activation of TAK1. Furtherly, TNFα-induced suppression of eNOS and NO production were largely prevented by silencing p38 MAPK. Collectively, our results suggested that A20-mediated TAK1 inactivation suppresses p38 MAPK and regulated MAPK/eNOS pathway, which contributes to endothelial cell survival and function preservation.
Collapse
Affiliation(s)
- Lei Li
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | | | - Shiyang Song
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hareshwaree Sohun
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhiheng Rao
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Luyuan Tao
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qike Jin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jingjing Zeng
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Rongzhou Wu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Kangting Ji
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiafeng Lin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Lianpin Wu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Maoping Chu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
45
|
Kraehling JR, Sessa WC. Contemporary Approaches to Modulating the Nitric Oxide-cGMP Pathway in Cardiovascular Disease. Circ Res 2017; 120:1174-1182. [PMID: 28360348 DOI: 10.1161/circresaha.117.303776] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endothelial cells lining the vessel wall control important aspects of vascular homeostasis. In particular, the production of endothelium-derived nitric oxide and activation of soluble guanylate cyclase promotes endothelial quiescence and governs vasomotor function and proportional remodeling of blood vessels. Here, we discuss novel approaches to improve endothelial nitric oxide generation and preserve its bioavailability. We also discuss therapeutic opportunities aimed at activation of soluble guanylate cyclase for multiple cardiovascular indications.
Collapse
Affiliation(s)
- Jan R Kraehling
- From the Vascular Biology and Therapeutics Program (J.R.K.) and Department of Pharmacology (W.C.S.), Yale University, School of Medicine, New Haven, CT
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (J.R.K.) and Department of Pharmacology (W.C.S.), Yale University, School of Medicine, New Haven, CT.
| |
Collapse
|
46
|
Camaré C, Pucelle M, Nègre-Salvayre A, Salvayre R. Angiogenesis in the atherosclerotic plaque. Redox Biol 2017; 12:18-34. [PMID: 28212521 PMCID: PMC5312547 DOI: 10.1016/j.redox.2017.01.007] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifocal alteration of the vascular wall of medium and large arteries characterized by a local accumulation of cholesterol and non-resolving inflammation. Atherothrombotic complications are the leading cause of disability and mortality in western countries. Neovascularization in atherosclerotic lesions plays a major role in plaque growth and instability. The angiogenic process is mediated by classical angiogenic factors and by additional factors specific to atherosclerotic angiogenesis. In addition to its role in plaque progression, neovascularization may take part in plaque destabilization and thromboembolic events. Anti-angiogenic agents are effective to reduce atherosclerosis progression in various animal models. However, clinical trials with anti-angiogenic drugs, mainly anti-VEGF/VEGFR, used in anti-cancer therapy show cardiovascular adverse effects, and require additional investigations.
Collapse
Affiliation(s)
- Caroline Camaré
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France
| | - Mélanie Pucelle
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France
| | - Anne Nègre-Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France.
| | - Robert Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France.
| |
Collapse
|
47
|
Wang Q, Zhang M, Torres G, Wu S, Ouyang C, Xie Z, Zou MH. Metformin Suppresses Diabetes-Accelerated Atherosclerosis via the Inhibition of Drp1-Mediated Mitochondrial Fission. Diabetes 2017; 66:193-205. [PMID: 27737949 PMCID: PMC5204316 DOI: 10.2337/db16-0915] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022]
Abstract
Metformin is a widely used antidiabetic drug that exerts cardiovascular protective effects in patients with diabetes. How metformin protects against diabetes-related cardiovascular diseases remains poorly understood. Here, we show that metformin abated the progression of diabetes-accelerated atherosclerosis by inhibiting mitochondrial fission in endothelial cells. Metformin treatments markedly reduced mitochondrial fragmentation, mitigated mitochondrial-derived superoxide release, improved endothelial-dependent vasodilation, inhibited vascular inflammation, and suppressed atherosclerotic lesions in streptozotocin (STZ)-induced diabetic ApoE-/- mice. In high glucose-exposed endothelial cells, metformin treatment and adenoviral overexpression of constitutively active AMPK downregulated mitochondrial superoxide, lowered levels of dynamin-related protein (Drp1) and its translocation into mitochondria, and prevented mitochondrial fragmentation. In contrast, AMPK-α2 deficiency abolished the effects of metformin on Drp1 expression, oxidative stress, and atherosclerosis in diabetic ApoE-/-/AMPK-α2-/- mice, indicating that metformin exerts an antiatherosclerotic action in vivo via the AMPK-mediated blockage of Drp1-mediated mitochondrial fission. Consistently, mitochondrial division inhibitor 1, a potent and selective Drp1 inhibitor, reduced mitochondrial fragmentation, attenuated oxidative stress, ameliorated endothelial dysfunction, inhibited inflammation, and suppressed atherosclerosis in diabetic mice. These findings show that metformin attenuated the development of atherosclerosis by reducing Drp1-mediated mitochondrial fission in an AMPK-dependent manner. Suppression of mitochondrial fission may be a therapeutic approach for treating macrovascular complications in patients with diabetes.
Collapse
Affiliation(s)
- Qilong Wang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Miao Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gloria Torres
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Shengnan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Changhan Ouyang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Zhonglin Xie
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
- Corresponding author: Ming-Hui Zou, , or Zhonglin Xie,
| |
Collapse
|
48
|
Myeloid Cell Prostaglandin E2 Receptor EP4 Modulates Cytokine Production but Not Atherogenesis in a Mouse Model of Type 1 Diabetes. PLoS One 2016; 11:e0158316. [PMID: 27351842 PMCID: PMC4924840 DOI: 10.1371/journal.pone.0158316] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/14/2016] [Indexed: 11/19/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is associated with cardiovascular complications induced by atherosclerosis. Prostaglandin E2 (PGE2) is often raised in states of inflammation, including diabetes, and regulates inflammatory processes. In myeloid cells, a key cell type in atherosclerosis, PGE2 acts predominately through its Prostaglandin E Receptor 4 (EP4; Ptger4) to modulate inflammation. The effect of PGE2-mediated EP4 signaling specifically in myeloid cells on atherosclerosis in the presence and absence of diabetes is unknown. Because diabetes promotes atherosclerosis through increased arterial myeloid cell accumulation, we generated a myeloid cell-targeted EP4-deficient mouse model (EP4M-/-) of T1DM-accelerated atherogenesis to investigate the relationship between myeloid cell EP4, inflammatory phenotypes of myeloid cells, and atherogenesis. Diabetic mice exhibited elevated plasma PGE metabolite levels and elevated Ptger4 mRNA in macrophages, as compared with non-diabetic littermates. PGE2 increased Il6, Il1b, Il23 and Ccr7 mRNA while reducing Tnfa mRNA through EP4 in isolated myeloid cells. Consistently, the stimulatory effect of diabetes on peritoneal macrophage Il6 was mediated by PGE2-EP4, while PGE2-EP4 suppressed the effect of diabetes on Tnfa in these cells. In addition, diabetes exerted effects independent of myeloid cell EP4, including a reduction in macrophage Ccr7 levels and increased early atherogenesis characterized by relative lesional macrophage accumulation. These studies suggest that this mouse model of T1DM is associated with increased myeloid cell PGE2-EP4 signaling, which is required for the stimulatory effect of diabetes on IL-6, markedly blunts the effect of diabetes on TNF-α and does not modulate diabetes-accelerated atherogenesis.
Collapse
|
49
|
κ-Opioid Receptor Stimulation Improves Endothelial Function via Akt-stimulated NO Production in Hyperlipidemic Rats. Sci Rep 2016; 6:26807. [PMID: 27226238 PMCID: PMC4881032 DOI: 10.1038/srep26807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/09/2016] [Indexed: 11/24/2022] Open
Abstract
This study was designed to investigate the effect of U50,488H (a selective κ-opioid receptor agonist) on endothelial function impaired by hyperlipidemia and to determine the role of Akt-stimulated NO production in it. Hyperlipidemic model was established by feeding rats with a high-fat diet for 14 weeks. U50,488H and nor-BNI (a selective κ-opioid receptor antagonist) were administered intraperitoneally. In vitro, the involvement of the PI3K/Akt/eNOS pathway in the effect of U50,488H was studied using cultured endothelial cells subjected to artificial hyperlipidemia. Serum total cholesterol and low-density lipoprotein cholesterol concentrations dramatically increased after high-fat diet feeding. Administration of U50,488H significantly alleviated endothelial ultrastructural destruction and endothelium-dependent vasorelaxation impairment caused by hyperlipidemia. U50,488H also increased Akt/eNOS phosphorylation and serum/medium NO level both in vivo and in vitro. U50,488H increased eNOS activity and suppressed iNOS activity in vivo. The effects of U50,488H were abolished in vitro by siRNAs targeting κ-opioid receptor and Akt or PI3K/Akt/eNOS inhibitors. All effects of U50,488H were blocked by nor-BNI. These results demonstrate that κ-opioid receptor stimulation normalizes endothelial ultrastructure and function under hyperlipidemic condition. Its mechanism is related to the preservation of eNOS phosphorylation through activation of the PI3K/Akt signaling pathway and downregulation of iNOS expression/activity.
Collapse
|
50
|
Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch 2016; 468:1125-1137. [DOI: 10.1007/s00424-016-1839-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
|