1
|
Che X, Oh JH, Kang YJ, Kim DW, Kim SG, Choi JY, Garagiola U. 4-Hexylresorcinol Enhances Glut4 Expression and Glucose Homeostasis via AMPK Activation and Histone H3 Acetylation. Int J Mol Sci 2024; 25:12281. [PMID: 39596347 PMCID: PMC11594624 DOI: 10.3390/ijms252212281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the potential of 4-hexylresorcinol (4HR) as a novel antidiabetic agent by assessing its effects on blood glucose levels, Glut4 expression, AMPK phosphorylation, and Histone H3 acetylation (Ac-H3) in the liver. In vitro experiments utilized Huh7 and HepG2 cells treated with varying concentrations of 4HR. Glut4, p-AMPK, and Ac-H3 expression levels were quantified via Western blotting. Additionally, GAPDH activity and glucose uptake were evaluated. In vivo experiments employed streptozotocin (STZ)-induced diabetic rats, with or without 4HR treatment, monitoring blood glucose, body weight, and hepatic levels of Glut4, p-AMPK, and Ac-H3. In vitro, 4HR treatment increased GAPDH activity and glucose uptake. Elevated Glut4, p-AMPK, and Ac-H3 levels were observed 8 h after 4HR administration. Inhibition of p-AMPK using compound C reduced 4HR-mediated Glut4 expression. In STZ-induced diabetic rats, 4HR significantly upregulated Glut4, p-AMPK, and Ac-H3 expression in the liver. Periodic 4HR injections mitigated weight loss and lowered blood glucose levels in STZ-injected animals. Histological analysis revealed increased glycogen storage in hepatocytes of the 4HR-treated group. Overall, 4HR enhanced Glut4 expression through upregulation of AMPK activity and histone H3 acetylation in vitro and in vivo, improving hepatic glucose homeostasis and suggesting potential as a candidate for diabetes treatment.
Collapse
Affiliation(s)
- Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Ji-Hyeon Oh
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Yei-Jin Kang
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Dae-Won Kim
- Department of Oral Biochemistry, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Umberto Garagiola
- Maxillofacial and Dental Unit, Biomedical, Surgical and Oral Sciences Department, School of Dentistry, University of Milan, 20122 Milan, Italy;
| |
Collapse
|
2
|
Gao L, Ramirez FJ, Cabrera JTO, Varghese MV, Watanabe M, Tsuji-Hosokawa A, Zheng Q, Yang M, Razan MR, Kempf CL, Camp SM, Wang J, Garcia JGN, Makino A. eNAMPT is a novel therapeutic target for mitigation of coronary microvascular disease in type 2 diabetes. Diabetologia 2024; 67:1998-2011. [PMID: 38898303 PMCID: PMC11410976 DOI: 10.1007/s00125-024-06201-9] [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: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
AIMS/HYPOTHESIS Individuals with diabetes are at high risk of cardiovascular complications, which significantly increase morbidity/mortality. Coronary microvascular disease (CMD) is recognised as a critical contributor to the increased cardiac mortality observed in people with diabetes. Therefore, there is an urgent need for treatments that are specific to CMD. eNAMPT (extracellular nicotinamide phosphoribosyltransferase) is a damage-associated molecular pattern and TLR4 ligand, whose plasma levels are elevated in people with diabetes. This study was thus designed to investigate the pathogenic role of intracellular nicotinamide phosphoribosyltransferase (iNAMPT) and eNAMPT in promoting the development of CMD in a preclinical murine model of type 2 diabetes. METHODS An inducible type 2 diabetic mouse model was generated by a single injection of low-dose streptozocin (75 mg/kg, i.p.) combined with a high-fat diet for 16 weeks. The in vivo effects of i/eNAMPT inhibition on cardiac endothelial cell (CEC) function were evaluated by using Nampt+/- heterozygous mice, chronic administration of eNAMPT-neutralising monoclonal antibody (mAb) or use of an NAMPT enzymatic inhibitor (FK866). RESULTS As expected, diabetic wild-type mice exhibited significantly lower coronary flow velocity reserve (CFVR), a determinant of coronary microvascular function, compared with control wild-type mice. eNAMPT plasma levels or expression in CECs were significantly greater in diabetic mice than in control mice. Furthermore, in comparison with diabetic wild-type mice, diabetic Nampt+/- heterozygous mice showed markedly improved CFVR, accompanied by increased left ventricular capillary density and augmented endothelium-dependent relaxation (EDR) in the coronary artery. NAMPT inhibition by FK866 or an eNAMPT-neutralising mAb significantly increased CFVR in diabetic mice. Furthermore, administration of the eNAMPT mAb upregulated expression of angiogenesis- and EDR-related genes in CECs from diabetic mice. Treatment with either eNAMPT or NAD+ significantly decreased CEC migration and reduced EDR in coronary arteries, partly linked to increased production of mitochondrial reactive oxygen species. CONCLUSIONS/INTERPRETATION These data indicate that increased i/eNAMPT expression contributes to the development of diabetic coronary microvascular dysfunction, and provide compelling support for eNAMPT inhibition as a novel and effective therapeutic strategy for CMD in diabetes.
Collapse
Affiliation(s)
- Lei Gao
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Francisco J Ramirez
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Jody Tori O Cabrera
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Makiko Watanabe
- Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | | | - Qiuyu Zheng
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingya Yang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Md Rahatullah Razan
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Carrie L Kempf
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Sara M Camp
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G N Garcia
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA.
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
3
|
Huang R, Chen J, Guo B, Jiang C, Sun W. Diabetes-induced male infertility: potential mechanisms and treatment options. Mol Med 2024; 30:11. [PMID: 38225568 PMCID: PMC10790413 DOI: 10.1186/s10020-023-00771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Male infertility is a physiological phenomenon in which a man is unable to impregnate a fertile woman during a 12-month period of continuous, unprotected sexual intercourse. A growing body of clinical and epidemiological evidence indicates that the increasing incidence of male reproductive problems, especially infertility, shows a very similar trend to the incidence of diabetes within the same age range. In addition, a large number of previous in vivo and in vitro experiments have also suggested that the complex pathophysiological changes caused by diabetes may induce male infertility in multiple aspects, including hypothalamic-pituitary-gonadal axis dysfunction, spermatogenesis and maturation disorders, testicular interstitial cell damage erectile dysfunction. Based on the above related mechanisms, a large number of studies have focused on the potential therapeutic association between diabetes progression and infertility in patients with diabetes and infertility, providing important clues for the treatment of this population. In this paper, we summarized the research results of the effects of diabetes on male reproductive function in recent 5 years, elaborated the potential pathophysiological mechanisms of male infertility induced by diabetes, and reviewed and prospected the therapeutic measures.
Collapse
Affiliation(s)
- Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Jiawang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Buyu Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Weiming Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000.
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
4
|
Ramírez-Hernández D, López-Sánchez P, Lezama-Martínez D, Kuyoc-Arroyo NM, Rodríguez-Rodríguez JE, Fonseca-Coronado S, Valencia-Hernández I, Flores-Monroy J. Timing Matters: Effects of Early and Late Estrogen Replacement Therapy on Glucose Metabolism and Vascular Reactivity in Ovariectomized Aged Wistar Rats. J Renin Angiotensin Aldosterone Syst 2023; 2023:6683989. [PMID: 38025203 PMCID: PMC10665112 DOI: 10.1155/2023/6683989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Cardiovascular disease incidence increases after menopause due to the loss of estrogen cardioprotective effects. However, there are conflicting data regarding the timing of estrogen therapy (ERT) and its effect on vascular dysfunction associated with impaired glucose metabolism. The aim of this work was to evaluate the effect of early and late ERT on blood glucose/insulin balance and vascular reactivity in aged ovariectomized Wistar rats. Eighteen-month-old female Wistar rats were randomized as follows: (1) sham, (2) 10-week postovariectomy (10 w), (3) 10 w postovariectomy+early estradiol therapy (10 w-early E2), (4) 20-week postovariectomy (20 w), and (5) 20-week postovariectomy+late estradiol therapy (20 w-late E2). Early E2 was administered 3 days after ovariectomy and late therapy after 10 weeks, in both groups. 17β-Estradiol (E2) was administered daily for 10 weeks (5 μg/kg/day). Concentration-response curves to angiotensin II, KCl, and acetylcholine (ACh) were performed. Heart rate (HR), diastolic and systolic blood pressure (DBP and SBP), glucose, insulin, HOMA-IR, and nitric oxide (NO) levels were determined. Higher glucose levels were found in all groups compared to the sham group, except the 20 w-late E2 group. Insulin was increased in all ovariectomized groups compared to sham. The HOMA-IR index showed insulin resistance in all ovariectomized groups, except for the 10 w-early E2 group. The 10 w-early E2 group increased NO levels vs. the 10 w group. After 10 w postovariectomy, the vascular response to KCl and Ach increases, despite early E2 administration. Early and late E2 treatment decreased vascular reactivity to Ang II. At 20-week postovariectomy, DBP increased, even with E2 administration, while SBP and HR remained unchanged. The effects of E2 therapy on blood glucose/insulin balance and vascular reactivity depend on the timing of therapy. Early ERT may provide some protective effects on insulin resistance and vascular function, whereas late ERT may not have the same benefits.
Collapse
Affiliation(s)
- Diana Ramírez-Hernández
- Myocardial Pharmacology Laboratory, Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, 54740 State of Mexico, Mexico
| | - Pedro López-Sánchez
- Laboratorio de Farmacología Molecular, Escuela Superior de Medicina, Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico
| | - Diego Lezama-Martínez
- Myocardial Pharmacology Laboratory, Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, 54740 State of Mexico, Mexico
| | - Neidy M. Kuyoc-Arroyo
- Myocardial Pharmacology Laboratory, Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, 54740 State of Mexico, Mexico
| | - Jessica E. Rodríguez-Rodríguez
- Biological Pharmaceutical Chemist Career, Faculty of Higher Education Zaragoza, National Autonomous University of Mexico, Batalla 5 de Mayo S/N, Ejército de Oriente, Iztapalapa, 09230 Mexico City, Mexico
- Laboratory 7, Biomedicine Unit, Faculty of Higher Education Iztacala, National Autonomous University of Mexico, Avenida de los Barrios 1, Los Reyes Ixtacala, 54090 Tlalnepantla de Baz, Mexico
| | - Salvador Fonseca-Coronado
- Immunology Laboratory, Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, 54740 State of Mexico, Mexico
| | - Ignacio Valencia-Hernández
- Laboratorio de Farmacología Cardiovascular, Escuela Superior de Medicina, Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico
| | - Jazmin Flores-Monroy
- Myocardial Pharmacology Laboratory, Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, 54740 State of Mexico, Mexico
| |
Collapse
|
5
|
Alvidrez RIM, Annarapu GK, Srinivasan AJ, Liu Z, Yazdani HO, Nolfi-Donegan D, Simmons RL, Shiva S, Neal MD. High Dose of Metformin Decreases Susceptibility to Occlusive Arterial Thrombosis in Diabetic Mice. JOURNAL OF PHARMACY AND PHARMACOLOGY RESEARCH 2023; 7:192-202. [PMID: 37975061 PMCID: PMC10653203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Introduction Metformin is the most prescribed medication in Diabetes Mellitus(DM). Metformin has shown to decrease mean platelet volume, with promising antiplatelet effects. High doses of Metformin have also been associated with hypercoagulation. We hypothesize that Metformin will protect DM mice from occlusive arterial thrombus formation by altering platelet activation and mitochondrial bioenergetics. Methods DM was developed by low dose of Streptozotocin, non-DM (healthy) mice are controls. Either vehicle or Metformin was administered twice daily via oral gavage for 7-days. Ferric chloride (FeCl3) arterial thrombosis and tail bleeding time were performed. Whole blood aggregometry, platelet activation/adhesion and mitochondrial bioenergetics were evaluated. Results Metformin decreased susceptibility of DM mice to arterial thrombosis. Platelet bioenergetics show DM mice have increased platelet mitochondrial respiration, but no differences were observed with Metformin treatment. In non-DM (healthy) mice, Metformin modulated ADP-dependent increase in platelet adhesion. Non-DM (healthy) mice, Metformin shortens bleeding time with faster thrombotic occlusion. Metformin also increased platelet mitochondrial maximal respiration and spare respiratory capacity uniquely in non-DM (healthy) mice. Conclusion Metformin regulates platelet bioenergetics and ADP-mediated platelet function in DM mice which attenuates susceptibility to arterial thrombosis. Future studies will evaluate clinically relevant doses of Metformin that regulates thrombotic function in diabetic platelets.
Collapse
Affiliation(s)
- Roberto I Mota Alvidrez
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Clinical and Translational Science Center, University of New Mexico, USA 87131
- Department of Pharmaceutical Sciences, University of New Mexico, USA 87131
| | - Gowtham K Annarapu
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Amudan J Srinivasan
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
| | - Zeyu Liu
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- Department of Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Hamza O Yazdani
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Deidre Nolfi-Donegan
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Richard L Simmons
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Matthew D Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| |
Collapse
|
6
|
Mota Alvidrez RI, Annarapu GK, Srinivasan AJ, Liu Z, Yazdani HO, Simmons RL, Shiva S, Neal MD, Nolfi-Donegan D. High Dose of Metformin Decreases Susceptibility to Occlusive Arterial Thrombosis in Diabetic Mice. RESEARCH SQUARE 2023:rs.3.rs-3143156. [PMID: 37503167 PMCID: PMC10371086 DOI: 10.21203/rs.3.rs-3143156/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Introduction Metformin is the most prescribed medication in Diabetes Mellitus(DM). Metformin has shown to decrease mean platelet volume, with promising antiplatelet effects. High doses of Metformin have also been associated with hypercoagulation. We hypothesize that Metformin will protect DM mice from occlusive arterial thrombus formation by altering platelet activation and mitochondrial bioenergetics. Methods DM was developed by low dose of Streptozotocin, healthy (non-DM) mice are controls. Either vehicle or Metformin was administered twice daily via oral gavage for 7-days. Ferric chloride (FeCl3) arterial thrombosis and tail bleeding time were performed. Whole blood aggregometry, platelet activation/adhesion and mitochondrial bioenergetics were evaluated. Results Metformin decreased susceptibility of DM mice to arterial thrombosis. Platelet bioenergetics show DM mice have increased platelet mitochondrial respiration, but no differences were observed with Metformin treatment. In healthy mice, Metformin modulated ADP-dependent increase in platelet adhesion. In healthy mice, Metformin shortens bleeding time with faster thrombotic occlusion. Metformin also increased platelet mitochondrial maximal respiration and spare respiratory capacity uniquely in healthy mice. Conclusion Metformin regulates platelet bioenergetics and ADP-mediated platelet function in DM mice which attenuates susceptibility to arterial thrombosis. Future studies will evaluate clinically relevant doses of Metformin that regulates thrombotic function in diabetic platelets.
Collapse
Affiliation(s)
| | | | | | - Zeyu Liu
- University of Pittsburgh Medical Center
| | | | | | | | | | | |
Collapse
|
7
|
Zhang Y, Weng J, Huan L, Sheng S, Xu F. Mitophagy in atherosclerosis: from mechanism to therapy. Front Immunol 2023; 14:1165507. [PMID: 37261351 PMCID: PMC10228545 DOI: 10.3389/fimmu.2023.1165507] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Mitophagy is a type of autophagy that can selectively eliminate damaged and depolarized mitochondria to maintain mitochondrial activity and cellular homeostasis. Several pathways have been found to participate in different steps of mitophagy. Mitophagy plays a significant role in the homeostasis and physiological function of vascular endothelial cells, vascular smooth muscle cells, and macrophages, and is involved in the development of atherosclerosis (AS). At present, many medications and natural chemicals have been shown to alter mitophagy and slow the progression of AS. This review serves as an introduction to the field of mitophagy for researchers interested in targeting this pathway as part of a potential AS management strategy.
Collapse
Affiliation(s)
- Yanhong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiajun Weng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| | - Luyao Huan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Song Sheng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
8
|
Cabrera JT, Si R, Tsuji-Hosokawa A, Cai H, Yuan JXJ, Dillmann WH, Makino A. Restoration of coronary microvascular function by OGA overexpression in a high-fat diet with low-dose streptozotocin-induced type 2 diabetic mice. Diab Vasc Dis Res 2023; 20:14791641231173630. [PMID: 37186669 PMCID: PMC10196148 DOI: 10.1177/14791641231173630] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Sustained hyperglycemia results in excess protein O-GlcNAcylation, leading to vascular complications in diabetes. This study aims to investigate the role of O-GlcNAcylation in the progression of coronary microvascular disease (CMD) in inducible type 2 diabetic (T2D) mice generated by a high-fat diet with a single injection of low-dose streptozotocin. Inducible T2D mice exhibited an increase in protein O-GlcNAcylation in cardiac endothelial cells (CECs) and decreases in coronary flow velocity reserve (CFVR, an indicator of coronary microvascular function) and capillary density accompanied by increased endothelial apoptosis in the heart. Endothelial-specific O-GlcNAcase (OGA) overexpression significantly lowered protein O-GlcNAcylation in CECs, increased CFVR and capillary density, and decreased endothelial apoptosis in T2D mice. OGA overexpression also improved cardiac contractility in T2D mice. OGA gene transduction augmented angiogenic capacity in high-glucose treated CECs. PCR array analysis revealed that seven out of 92 genes show significant differences among control, T2D, and T2D + OGA mice, and Sp1 might be a great target for future study, the level of which was significantly increased by OGA in T2D mice. Our data suggest that reducing protein O-GlcNAcylation in CECs has a beneficial effect on coronary microvascular function, and OGA is a promising therapeutic target for CMD in diabetic patients.
Collapse
Affiliation(s)
- Jody Tori Cabrera
- Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Rui Si
- Department of Physiology, The University of
Arizona, Tucson, AZ, USA
- Department of Cardiology, Xijing
Hospital, Fourth Military Medical
University, Shaanxi, China
| | | | - Hua Cai
- Department of Anesthesiology, University of California, Los
Angeles, Los Angeles, CA, USA
| | - Jason X-J Yuan
- Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Ayako Makino
- Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
- Department of Physiology, The University of
Arizona, Tucson, AZ, USA
| |
Collapse
|
9
|
Li Y, Yang S, Jin X, Li D, Lu J, Wang X, Wu M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front Pharmacol 2023; 14:1082817. [PMID: 36733506 PMCID: PMC9886688 DOI: 10.3389/fphar.2023.1082817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease (CVD) and is characterized by endothelial damage, lipid deposition, and chronic inflammation. Gut microbiota plays an important role in the occurrence and development of AS by regulating host metabolism and immunity. As human mitochondria evolved from primordial bacteria have homologous characteristics, they are attacked by microbial pathogens as target organelles, thus contributing to energy metabolism disorders, oxidative stress, and apoptosis. Therefore, mitochondria may be a key mediator of intestinal microbiota disorders and AS aggravation. Microbial metabolites, such as short-chain fatty acids, trimethylamine, hydrogen sulfide, and bile acids, also affect mitochondrial function, including mtDNA mutation, oxidative stress, and mitophagy, promoting low-grade inflammation. This further damages cellular homeostasis and the balance of innate immunity, aggravating AS. Herbal medicines and their monomers can effectively ameliorate the intestinal flora and their metabolites, improve mitochondrial function, and inhibit atherosclerotic plaques. This review focuses on the interaction between gut microbiota and mitochondria in AS and explores a therapeutic strategy for restoring mitochondrial function and intestinal microbiota disorders using herbal medicines, aiming to provide new insights for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yujuan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Wang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Min Wu,
| |
Collapse
|
10
|
Qu K, Yan F, Qin X, Zhang K, He W, Dong M, Wu G. Mitochondrial dysfunction in vascular endothelial cells and its role in atherosclerosis. Front Physiol 2022; 13:1084604. [PMID: 36605901 PMCID: PMC9807884 DOI: 10.3389/fphys.2022.1084604] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
The mitochondria are essential organelles that generate large amounts of ATP via the electron transport chain (ECT). Mitochondrial dysfunction causes reactive oxygen species accumulation, energy stress, and cell death. Endothelial mitochondrial dysfunction is an important factor causing abnormal function of the endothelium, which plays a central role during atherosclerosis development. Atherosclerosis-related risk factors, including high glucose levels, hypertension, ischemia, hypoxia, and diabetes, promote mitochondrial dysfunction in endothelial cells. This review summarizes the physiological and pathophysiological roles of endothelial mitochondria in endothelial function and atherosclerosis.
Collapse
Affiliation(s)
- Kai Qu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Fang Yan
- Department of Geriatrics, Geriatric Diseases Institute of Chengdu, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Xian Qin
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Kun Zhang
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,College of Bioengineering Chongqing University, Chongqing, China
| | - Wen He
- Department of Geriatrics, Clinical trial center, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| | - Guicheng Wu
- Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, China,*Correspondence: Mingqing Dong, ; Guicheng Wu,
| |
Collapse
|
11
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
12
|
Coronary Microvascular Dysfunction in Diabetes Mellitus: Pathogenetic Mechanisms and Potential Therapeutic Options. Biomedicines 2022; 10:biomedicines10092274. [PMID: 36140374 PMCID: PMC9496134 DOI: 10.3390/biomedicines10092274] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic patients are frequently affected by coronary microvascular dysfunction (CMD), a condition consisting of a combination of altered vasomotion and long-term structural change to coronary arterioles leading to impaired regulation of blood flow in response to changing cardiomyocyte oxygen requirements. The pathogenesis of this microvascular complication is complex and not completely known, involving several alterations among which hyperglycemia and insulin resistance play particularly central roles leading to oxidative stress, inflammatory activation and altered barrier function of endothelium. CMD significantly contributes to cardiac events such as angina or infarction without obstructive coronary artery disease, as well as heart failure, especially the phenotype associated with preserved ejection fraction, which greatly impact cardiovascular (CV) prognosis. To date, no treatments specifically target this vascular damage, but recent experimental studies and some clinical investigations have produced data in favor of potential beneficial effects on coronary micro vessels caused by two classes of glucose-lowering drugs: glucagon-like peptide 1 (GLP-1)-based therapy and inhibitors of sodium-glucose cotransporter-2 (SGLT2). The purpose of this review is to describe pathophysiological mechanisms, clinical manifestations of CMD with particular reference to diabetes, and to summarize the protective effects of antidiabetic drugs on the myocardial microvascular compartment.
Collapse
|
13
|
Sabe SA, Feng J, Sellke FW, Abid MR. Mechanisms and clinical implications of endothelium-dependent vasomotor dysfunction in coronary microvasculature. Am J Physiol Heart Circ Physiol 2022; 322:H819-H841. [PMID: 35333122 PMCID: PMC9018047 DOI: 10.1152/ajpheart.00603.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
Coronary microvascular disease (CMD), which affects the arterioles and capillary endothelium that regulate myocardial perfusion, is an increasingly recognized source of morbidity and mortality, particularly in the setting of metabolic syndrome. The coronary endothelium plays a pivotal role in maintaining homeostasis, though factors such as diabetes, hypertension, hyperlipidemia, and obesity can contribute to endothelial injury and consequently arteriolar vasomotor dysfunction. These disturbances in the coronary microvasculature clinically manifest as diminished coronary flow reserve, which is a known independent risk factor for cardiac death, even in the absence of macrovascular atherosclerotic disease. Therefore, a growing body of literature has examined the molecular mechanisms by which coronary microvascular injury occurs at the level of the endothelium and the consequences on arteriolar vasomotor responses. This review will begin with an overview of normal coronary microvascular physiology, modalities of measuring coronary microvascular function, and clinical implications of CMD. These introductory topics will be followed by a discussion of recent advances in the understanding of the mechanisms by which inflammation, oxidative stress, insulin resistance, hyperlipidemia, hypertension, shear stress, endothelial cell senescence, and tissue ischemia dysregulate coronary endothelial homeostasis and arteriolar vasomotor function.
Collapse
Affiliation(s)
- Sharif A Sabe
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jun Feng
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
14
|
Metabolic regulation and dysregulation of endothelial small conductance calcium activated potassium channels. Eur J Cell Biol 2022; 101:151208. [DOI: 10.1016/j.ejcb.2022.151208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
|
15
|
Abdelsaid K, Sudhahar V, Harris RA, Das A, Youn SW, Liu Y, McMenamin M, Hou Y, Fulton D, Hamrick MW, Tang Y, Fukai T, Ushio-Fukai M. Exercise improves angiogenic function of circulating exosomes in type 2 diabetes: Role of exosomal SOD3. FASEB J 2022; 36:e22177. [PMID: 35142393 PMCID: PMC8880294 DOI: 10.1096/fj.202101323r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/29/2021] [Accepted: 01/07/2022] [Indexed: 01/31/2023]
Abstract
Exosomes, key mediators of cell-cell communication, derived from type 2 diabetes mellitus (T2DM) exhibit detrimental effects. Exercise improves endothelial function in part via the secretion of exosomes into circulation. Extracellular superoxide dismutase (SOD3) is a major secretory copper (Cu) antioxidant enzyme that catalyzes the dismutation of O2•- to H2 O2 whose activity requires the Cu transporter ATP7A. However, the role of SOD3 in exercise-induced angiogenic effects of circulating plasma exosomes on endothelial cells (ECs) in T2DM remains unknown. Here, we show that both SOD3 and ATP7A proteins were present in plasma exosomes in mice, which was significantly increased after two weeks of volunteer wheel exercise. A single bout of exercise in humans also showed a significant increase in SOD3 and ATP7A protein expression in plasma exosomes. Plasma exosomes from T2DM mice significantly reduced angiogenic responses in human ECs or mouse skin wound healing models, which was associated with a decrease in ATP7A, but not SOD3 expression in exosomes. Exercise training in T2DM mice restored the angiogenic effects of T2DM exosomes in ECs by increasing ATP7A in exosomes, which was not observed in exercised T2DM/SOD3-/- mice. Furthermore, exosomes overexpressing SOD3 significantly enhanced angiogenesis in ECs by increasing local H2 O2 levels in a heparin-binding domain-dependent manner as well as restored defective wound healing and angiogenesis in T2DM or SOD3-/- mice. In conclusion, exercise improves the angiogenic potential of circulating exosomes in T2DM in a SOD3-dependent manner. Exosomal SOD3 may provide an exercise mimetic therapy that supports neovascularization and wound repair in cardiometabolic disease.
Collapse
Affiliation(s)
- Kareem Abdelsaid
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA
| | - Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA
| | | | - Archita Das
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA
| | - Seock-Won Youn
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Physiology and Biophysics, University of Illinois, Chicago, IL
| | - Yutao Liu
- Department of cell biology, Medical College of Georgia at Augusta University, Augusta, GA
| | - Maggie McMenamin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA
| | - Yali Hou
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA
| | - Mark W. Hamrick
- Department of cell biology, Medical College of Georgia at Augusta University, Augusta, GA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA,Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA
| |
Collapse
|
16
|
Akiyoshi K, Boersma GJ, Johnson MD, Velasquez FC, Dunkerly-Eyring B, O’Brien S, Yamaguchi A, Steenbergen C, Tamashiro KLK, Das S. Role of miR-181c in Diet-induced obesity through regulation of lipid synthesis in liver. PLoS One 2021; 16:e0256973. [PMID: 34879063 PMCID: PMC8654194 DOI: 10.1371/journal.pone.0256973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/10/2021] [Indexed: 12/02/2022] Open
Abstract
We recently identified a nuclear-encoded miRNA (miR-181c) in cardiomyocytes that can translocate into mitochondria to regulate mitochondrial gene mt-COX1 and influence obesity-induced cardiac dysfunction through the mitochondrial pathway. Because liver plays a pivotal role during obesity, we hypothesized that miR-181c might contribute to the pathophysiological complications associated with obesity. Therefore, we used miR-181c/d-/- mice to study the role of miR-181c in hepatocyte lipogenesis during diet-induced obesity. The mice were fed a high-fat (HF) diet for 26 weeks, during which indirect calorimetric measurements were made. Quantitative PCR (qPCR) was used to examine the expression of genes involved in lipid synthesis. We found that miR-181c/d-/- mice were not protected against all metabolic consequences of HF exposure. After 26 weeks, the miR-181c/d-/- mice had a significantly higher body fat percentage than did wild-type (WT) mice. Glucose tolerance tests showed hyperinsulinemia and hyperglycemia, indicative of insulin insensitivity in the miR-181c/d-/- mice. miR-181c/d-/- mice fed the HF diet had higher serum and liver triglyceride levels than did WT mice fed the same diet. qPCR data showed that several genes regulated by isocitrate dehydrogenase 1 (IDH1) were more upregulated in miR-181c/d-/- liver than in WT liver. Furthermore, miR-181c delivered in vivo via adeno-associated virus attenuated the lipogenesis by downregulating these same lipid synthesis genes in the liver. In hepatocytes, miR-181c regulates lipid biosynthesis by targeting IDH1. Taken together, the data indicate that overexpression of miR-181c can be beneficial for various lipid metabolism disorders.
Collapse
Affiliation(s)
- Kei Akiyoshi
- Department of Anesthesiology and Critical Care Medicine, Baltimore, MD, United States of America
| | - Gretha J. Boersma
- Department of Psychiatry & Behavioral Sciences, Baltimore, MD, United States of America
| | - Miranda D. Johnson
- Department of Psychiatry & Behavioral Sciences, Baltimore, MD, United States of America
| | | | - Brittany Dunkerly-Eyring
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Shannon O’Brien
- Department of Psychiatry & Behavioral Sciences, Baltimore, MD, United States of America
| | - Atsushi Yamaguchi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Kellie L. K. Tamashiro
- Department of Psychiatry & Behavioral Sciences, Baltimore, MD, United States of America
- * E-mail: (KLKT); (SD)
| | - Samarjit Das
- Department of Anesthesiology and Critical Care Medicine, Baltimore, MD, United States of America
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
- * E-mail: (KLKT); (SD)
| |
Collapse
|
17
|
Si R, Cabrera JTO, Tsuji-Hosokawa A, Guo R, Watanabe M, Gao L, Lee YS, Moon JS, Scott BT, Wang J, Ashton AW, Rao JN, Wang JY, Yuan JXJ, Makino A. HuR/Cx40 downregulation causes coronary microvascular dysfunction in type 2 diabetes. JCI Insight 2021; 6:147982. [PMID: 34747371 PMCID: PMC8663561 DOI: 10.1172/jci.insight.147982] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022] Open
Abstract
Patients with diabetes with coronary microvascular disease (CMD) exhibit higher cardiac mortality than patients without CMD. However, the molecular mechanism by which diabetes promotes CMD is poorly understood. RNA-binding protein human antigen R (HuR) is a key regulator of mRNA stability and translation; therefore, we investigated the role of HuR in the development of CMD in mice with type 2 diabetes. Diabetic mice exhibited decreases in coronary flow velocity reserve (CFVR; a determinant of coronary microvascular function) and capillary density in the left ventricle. HuR levels in cardiac endothelial cells (CECs) were significantly lower in diabetic mice and patients with diabetes than the controls. Endothelial-specific HuR-KO mice also displayed significant reductions in CFVR and capillary density. By examining mRNA levels of 92 genes associated with endothelial function, we found that HuR, Cx40, and Nox4 levels were decreased in CECs from diabetic and HuR-KO mice compared with control mice. Cx40 expression and HuR binding to Cx40 mRNA were downregulated in CECs from diabetic mice. Cx40-KO mice exhibited decreased CFVR and capillary density, whereas endothelium-specific Cx40 overexpression increased capillary density and improved CFVR in diabetic mice. These data suggest that decreased HuR contributes to the development of CMD in diabetes through downregulation of gap junction protein Cx40 in CECs.
Collapse
Affiliation(s)
- Rui Si
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | | | | | - Rui Guo
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA
| | - Makiko Watanabe
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA
| | - Lei Gao
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Yun Sok Lee
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Jae-Su Moon
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Brian T Scott
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Jian Wang
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anthony W Ashton
- Division of Perinatal Research, Kolling Institute of Medical Research, University of Sydney, New South Wales, Australia
| | - Jaladanki N Rao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jian-Ying Wang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Ayako Makino
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
18
|
Rodríguez C, Muñoz M, Contreras C, Prieto D. AMPK, metabolism, and vascular function. FEBS J 2021; 288:3746-3771. [PMID: 33825330 DOI: 10.1111/febs.15863] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/04/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor activated during energy stress that plays a key role in maintaining energy homeostasis. This ubiquitous signaling pathway has been implicated in multiple functions including mitochondrial biogenesis, redox regulation, cell growth and proliferation, cell autophagy and inflammation. The protective role of AMPK in cardiovascular function and the involvement of dysfunctional AMPK in the pathogenesis of cardiovascular disease have been highlighted in recent years. In this review, we summarize and discuss the role of AMPK in the regulation of blood flow in response to metabolic demand and the basis of the AMPK physiological anticontractile, antioxidant, anti-inflammatory, and antiatherogenic actions in the vascular system. Investigations by others and us have demonstrated the key role of vascular AMPK in the regulation of endothelial function, redox homeostasis, and inflammation, in addition to its protective role in the hypoxia and ischemia/reperfusion injury. The pathophysiological implications of AMPK involvement in vascular function with regard to the vascular complications of metabolic disease and the therapeutic potential of AMPK activators are also discussed.
Collapse
Affiliation(s)
- Claudia Rodríguez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| |
Collapse
|
19
|
Moraes RDA, Webb RC, Silva DF. Vascular Dysfunction in Diabetes and Obesity: Focus on TRP Channels. Front Physiol 2021; 12:645109. [PMID: 33716794 PMCID: PMC7952965 DOI: 10.3389/fphys.2021.645109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 01/22/2023] Open
Abstract
Transient receptor potential (TRP) superfamily consists of a diverse group of non-selective cation channels that has a wide tissue distribution and is involved in many physiological processes including sensory perception, secretion of hormones, vasoconstriction/vasorelaxation, and cell cycle modulation. In the blood vessels, TRP channels are present in endothelial cells, vascular smooth muscle cells, perivascular adipose tissue (PVAT) and perivascular sensory nerves, and these channels have been implicated in the regulation of vascular tone, vascular cell proliferation, vascular wall permeability and angiogenesis. Additionally, dysfunction of TRP channels is associated with cardiometabolic diseases, such as diabetes and obesity. Unfortunately, the prevalence of diabetes and obesity is rising worldwide, becoming an important public health problems. These conditions have been associated, highlighting that obesity is a risk factor for type 2 diabetes. As well, both cardiometabolic diseases have been linked to a common disorder, vascular dysfunction. In this review, we briefly consider general aspects of TRP channels, and we focus the attention on TRPC (canonical or classical), TRPV (vanilloid), TRPM (melastatin), and TRPML (mucolipin), which were shown to be involved in vascular alterations of diabetes and obesity or are potentially linked to vascular dysfunction. Therefore, elucidation of the functional and molecular mechanisms underlying the role of TRP channels in vascular dysfunction in diabetes and obesity is important for the prevention of vascular complications and end-organ damage, providing a further therapeutic target in the treatment of these metabolic diseases.
Collapse
Affiliation(s)
- Raiana Dos Anjos Moraes
- Laboratory of Cardiovascular Physiology and Pharmacology, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil.,Postgraduate Course in Biotechnology in Health and Investigative Medicine, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - R Clinton Webb
- Department of Cell Biology and Anatomy and Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Darízy Flávia Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil.,Postgraduate Course in Biotechnology in Health and Investigative Medicine, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| |
Collapse
|
20
|
Tan Y, Xia F, Li L, Peng X, Liu W, Zhang Y, Fang H, Zeng Z, Chen Z. Novel Insights into the Molecular Features and Regulatory Mechanisms of Mitochondrial Dynamic Disorder in the Pathogenesis of Cardiovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6669075. [PMID: 33688392 PMCID: PMC7914101 DOI: 10.1155/2021/6669075] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/20/2022]
Abstract
Mitochondria maintain mitochondrial homeostasis through continuous fusion and fission, that is, mitochondrial dynamics, which is precisely mediated by mitochondrial fission and fusion proteins, including dynamin-related protein 1 (Drp1), mitofusin 1 and 2 (Mfn1/2), and optic atrophy 1 (OPA1). When the mitochondrial fission and fusion of cardiomyocytes are out of balance, they will cause their own morphology and function disorders, which damage the structure and function of the heart, are involved in the occurrence and progression of cardiovascular disease such as ischemia-reperfusion injury (IRI), septic cardiomyopathy, and diabetic cardiomyopathy. In this paper, we focus on the latest findings regarding the molecular features and regulatory mechanisms of mitochondrial dynamic disorder in cardiovascular pathologies. Finally, we will address how these findings can be applied to improve the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fengfan Xia
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528300 Guangdong, China
| | - Lulan Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaojie Peng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenqian Liu
- Department of Critical Care Medicine, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yaoyuan Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haihong Fang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou 510515, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
21
|
Xing H, Zhang Z, Shi G, He Y, Song Y, Liu Y, Harrington EO, Sellke FW, Feng J. Chronic Inhibition of mROS Protects Against Coronary Endothelial Dysfunction in Mice With Diabetes. Front Cell Dev Biol 2021; 9:643810. [PMID: 33681229 PMCID: PMC7930489 DOI: 10.3389/fcell.2021.643810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/29/2021] [Indexed: 11/13/2022] Open
Abstract
Diabetes is associated with coronary endothelial dysfunction. Persistent oxidative stress during diabetes contributes to coronary endothelial dysfunction. The mitochondria are main sources of reactive oxygen species (ROS) in diabetes, and mitochondria-targeted antioxidant mito-Tempo can prevent mitochondrial reactive oxygen species (mROS) generation in a variety of disorders. Inhibition/inactivation of small-conductance Ca2+-activated K+ (SK) channels contribute to diabetic downregulation of coronary endothelial function/relaxation. However, few investigated the role of mROS on endothelial dysfunction/vasodilation and endothelial SK channel downregulation in diabetes. The aim of present study was to investigate the chronic administration of mito-Tempo, on coronary vasodilation, and endothelial SK channel activity of mice with or without diabetes. Mito-Tempo (1 mg/kg/day) was applied to the mice with or without diabetes (n = 10/group) for 4 weeks. In vitro relaxation response of pre-contracted arteries was examined in the presence or absence of the vasodilatory agents. SK channel currents of the isolated mouse heart endothelial cells were measured using whole-cell patch clamp methods. At baseline, coronary endothelium-dependent relaxation responses to ADP and the selective SK channel activator NS309 and endothelial SK channel currents were decreased in diabetic mice compared with that in non-diabetic (ND) mice (p < 0.05). After a 4-week treatment with mito-Tempo, coronary endothelium-dependent relaxation response to ADP or NS309 and endothelial SK channel currents in the diabetic mice was significantly improved when compared with that in untreated diabetic mice (p < 0.05). Interestingly, coronary relaxation responses to ADP and NS309 and endothelial SK channel currents were not significantly changed in ND mice after mito-Tempo treatment, as compared to that of untreated control group. Chronic inhibition of endothelial mROS appears to improve coronary endothelial function/dilation and SK channel activity in diabetes, and mROS inhibitors may be a novel strategy to treat vascular complications in diabetes.
Collapse
Affiliation(s)
- Hang Xing
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Zhiqi Zhang
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Guangbin Shi
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Yixin He
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Yi Song
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Yuhong Liu
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Elizabeth O Harrington
- Vascular Research Laboratory, Providence VA Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, RI, United States
| | - Frank W Sellke
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Jun Feng
- Cardiothoracic Surgery Research Laboratory, Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
22
|
Si R, Zhang Q, Tsuji-Hosokawa A, Watanabe M, Willson C, Lai N, Wang J, Dai A, Scott BT, Dillmann WH, Yuan JXJ, Makino A. Overexpression of p53 due to excess protein O-GlcNAcylation is associated with coronary microvascular disease in type 2 diabetes. Cardiovasc Res 2021; 116:1186-1198. [PMID: 31504245 DOI: 10.1093/cvr/cvz216] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/27/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
AIMS We previously reported that increased protein O-GlcNAcylation in diabetic mice led to vascular rarefaction in the heart. In this study, we aimed to investigate whether and how coronary endothelial cell (EC) apoptosis is enhanced by protein O-GlcNAcylation and thus induces coronary microvascular disease (CMD) and subsequent cardiac dysfunction in diabetes. We hypothesize that excessive protein O-GlcNAcylation increases p53 that leads to CMD and reduced cardiac contractility. METHODS AND RESULTS We conducted in vivo functional experiments in control mice, TALLYHO/Jng (TH) mice, a polygenic type 2 diabetic (T2D) model, and EC-specific O-GlcNAcase (OGA, an enzyme that catalyzes the removal of O-GlcNAc from proteins)-overexpressing TH mice, as well as in vitro experiments in isolated ECs from these mice. TH mice exhibited a significant increase in coronary EC apoptosis and reduction of coronary flow velocity reserve (CFVR), an assessment of coronary microvascular function, in comparison to wild-type mice. The decreased CFVR, due at least partially to EC apoptosis, was associated with decreased cardiac contractility in TH mice. Western blot experiments showed that p53 protein level was significantly higher in coronary ECs from TH mice and T2D patients than in control ECs. High glucose treatment also increased p53 protein level in control ECs. Furthermore, overexpression of OGA decreased protein O-GlcNAcylation and down-regulated p53 in coronary ECs, and conferred a protective effect on cardiac function in TH mice. Inhibition of p53 with pifithrin-α attenuated coronary EC apoptosis and restored CFVR and cardiac contractility in TH mice. CONCLUSIONS The data from this study indicate that inhibition of p53 or down-regulation of p53 by OGA overexpression attenuates coronary EC apoptosis and improves CFVR and cardiac function in diabetes. Lowering coronary endothelial p53 levels via OGA overexpression could be a potential therapeutic approach for CMD in diabetes.
Collapse
Affiliation(s)
- Rui Si
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Rd., Shaanxi 710032, China
| | - Qian Zhang
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA.,Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, 195 W Dongfeng Rd., Guangzhou 510182, China
| | - Atsumi Tsuji-Hosokawa
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA
| | - Makiko Watanabe
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA
| | - Conor Willson
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA
| | - Ning Lai
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, 195 W Dongfeng Rd., Guangzhou 510182, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, 195 W Dongfeng Rd., Guangzhou 510182, China.,Department of Medicine, The University of Arizona, 1501 N. Campbell Ave. Tucson, AZ 85724, USA
| | - Anzhi Dai
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.,Department of Medicine, The University of Arizona, 1501 N. Campbell Ave. Tucson, AZ 85724, USA
| | - Ayako Makino
- Department of Physiology, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724, USA.,Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.,Department of Medicine, The University of Arizona, 1501 N. Campbell Ave. Tucson, AZ 85724, USA
| |
Collapse
|
23
|
Li JC, Velagic A, Qin CX, Li M, Leo CH, Kemp-Harper BK, Ritchie RH, Woodman OL. Diabetes Attenuates the Contribution of Endogenous Nitric Oxide but Not Nitroxyl to Endothelium Dependent Relaxation of Rat Carotid Arteries. Front Pharmacol 2021; 11:585740. [PMID: 33716721 PMCID: PMC7944142 DOI: 10.3389/fphar.2020.585740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction is a major risk factor for several of the vascular complications of diabetes, including ischemic stroke. Nitroxyl (HNO), the one electron reduced and protonated form of nitric oxide (NO•), is resistant to scavenging by superoxide, but the role of HNO in diabetes mellitus associated endothelial dysfunction in the carotid artery remains unknown. Aim: To assess how diabetes affects the role of endogenous NO• and HNO in endothelium-dependent relaxation in rat isolated carotid arteries. Methods: Male Sprague Dawley rats were fed a high-fat-diet (HFD) for 2 weeks prior to administration of low dose streptozotocin (STZ; 35 mg/kg i. p./day) for 2 days. The HFD was continued for a further 12 weeks. Sham rats were fed standard chow and administered with citrate vehicle. After 14 weeks total, rats were anesthetized and carotid arteries collected to assess responses to the endothelium-dependent vasodilator, acetylcholine (ACh) by myography. The combination of calcium-activated potassium channel blockers, TRAM-34 (1 μmol/L) and apamin (1 μmol/L) was used to assess the contribution of endothelium-dependent hyperpolarization to relaxation. The corresponding contribution of NOS-derived nitrogen oxide species to relaxation was assessed using the combination of the NO• synthase inhibitor, L-NAME (200 μmol/L) and the soluble guanylate cyclase inhibitor ODQ (10 μmol/L). Lastly, L-cysteine (3 mmol/L), a selective HNO scavenger, and hydroxocobalamin (HXC; 100 μmol/L), a NO• scavenger, were used to distinguish between NO• and HNO-mediated relaxation. Results: At study end, diabetic rats exhibited significantly retarded body weight gain and elevated blood glucose levels compared to sham rats. The sensitivity and the maximal relaxation response to ACh was significantly impaired in carotid arteries from diabetic rats, indicating endothelial dysfunction. The vasorelaxation evoked by ACh was abolished by L-NAME plus ODQ, but not affected by the apamin plus TRAM-34 combination, indicating that NOS-derived nitrogen oxide species are the predominant endothelium-derived vasodilators in sham and diabetic rat carotid arteries. The maximum relaxation to ACh was significantly decreased by L-cysteine in both sham and diabetic rats, whereas HXC attenuated ACh-induced relaxation only in sham rats, suggesting that diabetes impaired the contribution of NO•, whereas HNO-mediated vasorelaxation remained intact. Conclusion: Both NO• and HNO contribute to endothelium-dependent relaxation in carotid arteries. In diabetes, NO•-mediated relaxation is impaired, whereas HNO-mediated relaxation was preserved. The potential for preserved HNO activity under pathological conditions that are associated with oxidative stress indicates that HNO donors may represent a viable therapeutic approach to the treatment of vascular dysfunction.
Collapse
Affiliation(s)
- Jasmin Chendi Li
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Anida Velagic
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Cheng Xue Qin
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Mandy Li
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chen Huei Leo
- Science, Maths and Technology Cluster, Singapore University of Technology & Design, Singapore, Singapore
| | - Barbara K. Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Rebecca H. Ritchie
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Owen L. Woodman
- Drug, Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Russell JS, Griffith TA, Naghipour S, Vider J, Du Toit EF, Patel HH, Peart JN, Headrick JP. Dietary α-Linolenic Acid Counters Cardioprotective Dysfunction in Diabetic Mice: Unconventional PUFA Protection. Nutrients 2020; 12:nu12092679. [PMID: 32887376 PMCID: PMC7551050 DOI: 10.3390/nu12092679] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Whether dietary omega-3 (n-3) polyunsaturated fatty acid (PUFA) confers cardiac benefit in cardiometabolic disorders is unclear. We test whether dietary -linolenic acid (ALA) enhances myocardial resistance to ischemia-reperfusion (I-R) and responses to ischemic preconditioning (IPC) in type 2 diabetes (T2D); and involvement of conventional PUFA-dependent mechanisms (caveolins/cavins, kinase signaling, mitochondrial function, and inflammation). Eight-week male C57Bl/6 mice received streptozotocin (75 mg/kg) and 21 weeks high-fat/high-carbohydrate feeding. Half received ALA over six weeks. Responses to I-R/IPC were assessed in perfused hearts. Localization and expression of caveolins/cavins, protein kinase B (AKT), and glycogen synthase kinase-3 β (GSK3β); mitochondrial function; and inflammatory mediators were assessed. ALA reduced circulating leptin, without affecting body weight, glycemic dysfunction, or cholesterol. While I-R tolerance was unaltered, paradoxical injury with IPC was reversed to cardioprotection with ALA. However, post-ischemic apoptosis (nucleosome content) appeared unchanged. Benefit was not associated with shifts in localization or expression of caveolins/cavins, p-AKT, p-GSK3β, or mitochondrial function. Despite mixed inflammatory mediator changes, tumor necrosis factor-a (TNF-a) was markedly reduced. Data collectively reveal a novel impact of ALA on cardioprotective dysfunction in T2D mice, unrelated to caveolins/cavins, mitochondrial, or stress kinase modulation. Although evidence suggests inflammatory involvement, the basis of this "un-conventional" protection remains to be identified.
Collapse
Affiliation(s)
- Jake S. Russell
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Tia A. Griffith
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Saba Naghipour
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Jelena Vider
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Eugene F. Du Toit
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Hemal H. Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California, San Diego, CA 92093, USA;
| | - Jason N. Peart
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - John P. Headrick
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
- Correspondence: ; Tel.: +61-7-5552-8292
| |
Collapse
|
25
|
Nitrogen Dioxide Inhalation Exposures Induce Cardiac Mitochondrial Reactive Oxygen Species Production, Impair Mitochondrial Function and Promote Coronary Endothelial Dysfunction. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17155526. [PMID: 32751709 PMCID: PMC7432061 DOI: 10.3390/ijerph17155526] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 01/01/2023]
Abstract
Traffic air pollution is a major health problem and is recognized as an important risk factor for cardiovascular (CV) diseases. In a previous experimental study, we showed that diesel exhaust (DE) exposures induced cardiac mitochondrial and CV dysfunctions associated with the gaseous phase. Here, we hypothesized that NO2 exposures to levels close to those found in DE induce a mitochondrial reactive oxygen species (ROS) production, which contribute to an endothelial dysfunction, an early indicator for numerous CV diseases. For this, we studied the effects of NO2 on ROS production and its impacts on the mitochondrial, coronary endothelial and cardiac functions, after acute (one single exposure) and repeated (three h/day, five days/week for three weeks) exposures in Wistar rats. Acute NO2 exposure induced an early but reversible mitochondrial ROS production. This event was isolated since neither mitochondrial function nor endothelial function were impaired, whereas cardiac function assessment showed a reversible left ventricular dysfunction. Conversely, after three weeks of exposure this alteration was accompanied by a cardiac mitochondrial dysfunction highlighted by an alteration of adenosine triphosphate (ATP) synthesis and oxidative phosphorylation and an increase in mitochondrial ROS production. Moreover, repeated NO2 exposures promoted endothelial dysfunction of the coronary arteries, as shown by reduced acetylcholine-induced vasodilatation, which was due, at least partially, to a superoxide-dependent decrease of nitric oxide (NO) bioavailability. This study shows that NO2 exposures impair cardiac mitochondrial function, which, in conjunction with coronary endothelial dysfunction, contributes to cardiac dysfunction. Together, these results clearly identify NO2 as a probable risk factor in ischemic heart diseases.
Collapse
|
26
|
AMPK, Mitochondrial Function, and Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21144987. [PMID: 32679729 PMCID: PMC7404275 DOI: 10.3390/ijms21144987] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is in charge of numerous catabolic and anabolic signaling pathways to sustain appropriate intracellular adenosine triphosphate levels in response to energetic and/or cellular stress. In addition to its conventional roles as an intracellular energy switch or fuel gauge, emerging research has shown that AMPK is also a redox sensor and modulator, playing pivotal roles in maintaining cardiovascular processes and inhibiting disease progression. Pharmacological reagents, including statins, metformin, berberine, polyphenol, and resveratrol, all of which are widely used therapeutics for cardiovascular disorders, appear to deliver their protective/therapeutic effects partially via AMPK signaling modulation. The functions of AMPK during health and disease are far from clear. Accumulating studies have demonstrated crosstalk between AMPK and mitochondria, such as AMPK regulation of mitochondrial homeostasis and mitochondrial dysfunction causing abnormal AMPK activity. In this review, we begin with the description of AMPK structure and regulation, and then focus on the recent advances toward understanding how mitochondrial dysfunction controls AMPK and how AMPK, as a central mediator of the cellular response to energetic stress, maintains mitochondrial homeostasis. Finally, we systemically review how dysfunctional AMPK contributes to the initiation and progression of cardiovascular diseases via the impact on mitochondrial function.
Collapse
|
27
|
Roman B, Kaur P, Ashok D, Kohr M, Biswas R, O'Rourke B, Steenbergen C, Das S. Nuclear-mitochondrial communication involving miR-181c plays an important role in cardiac dysfunction during obesity. J Mol Cell Cardiol 2020; 144:87-96. [PMID: 32442661 DOI: 10.1016/j.yjmcc.2020.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/09/2020] [Accepted: 05/16/2020] [Indexed: 12/26/2022]
Abstract
AIMS In cardiomyocytes, there is microRNA (miR) in the mitochondria that originates from the nuclear genome and matures in the cytoplasm before translocating into the mitochondria. Overexpression of one such miR, miR-181c, can lead to heart failure by stimulating reactive oxygen species (ROS) production and increasing mitochondrial calcium level ([Ca2+]m). Mitochondrial calcium uptake 1 protein (MICU1), a regulatory protein in the mitochondrial calcium uniporter complex, plays an important role in regulating [Ca2+]m. Obesity results in miR-181c overexpression and a decrease in MICU1. We hypothesize that lowering miR-181c would protect against obesity-induced cardiac dysfunction. METHODS AND RESULTS We used an in vivo mouse model of high-fat diet (HFD) for 18 weeks and induced high lipid load in H9c2 cells with oleate-conjugated bovine serum albumin in vitro. We tested the cardioprotective role of lowering miR-181c by using miR-181c/d-/- mice (in vivo) and AntagomiR against miR-181c (in vitro). HFD significantly upregulated heart levels of miR-181c and led to cardiac hypertrophy in wild-type mice, but not in miR-181c/d-/- mice. HFD also increased ROS production and pyruvate dehydrogenase activity (a surrogate for [Ca2+]m), but the increases were alleviated in miR-181c/d-/- mice. Moreover, miR-181c/d-/- mice fed a HFD had higher levels of MICU1 than did wild-type mice fed a HFD, attenuating the rise in [Ca2+]m. Overexpression of miR-181c in neonatal ventricular cardiomyocytes (NMVM) caused increased ROS production, which oxidized transcription factor Sp1 and led to a loss of Sp1, thereby slowing MICU1 transcription. Hence, miR-181c increases [Ca2+]m through Sp1 oxidation and downregulation of MICU1, suggesting that the cardioprotective effect of miR-181c/d-/- results from inhibition of Sp1 oxidation. CONCLUSION This study has identified a unique nuclear-mitochondrial communication mechanism in the heart orchestrated by miR-181c. Obesity-induced overexpression of miR-181c increases [Ca2+]m via downregulation of MICU1 and leads to cardiac injury. A strategy to inhibit miR-181c in cardiomyocytes can preserve cardiac function during obesity by improving mitochondrial function. Altering miR-181c expression may provide a pharmacologic approach to improve cardiomyopathy in individuals with obesity/type 2 diabetes.
Collapse
Affiliation(s)
- Barbara Roman
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Pawandeep Kaur
- Department of Anesthesiology & Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Deepthi Ashok
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Mark Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America.
| | - Samarjit Das
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America; Department of Anesthesiology & Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States of America.
| |
Collapse
|
28
|
Murugan DD, Md Zain Z, Choy KW, Zamakshshari NH, Choong MJ, Lim YM, Mustafa MR. Edible Bird's Nest Protects Against Hyperglycemia-Induced Oxidative Stress and Endothelial Dysfunction. Front Pharmacol 2020; 10:1624. [PMID: 32116666 PMCID: PMC7010601 DOI: 10.3389/fphar.2019.01624] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022] Open
Abstract
Increased oxidative stress by hyperglycemia is a major cause of vascular complications in diabetes. Bird's nest, which is made from the saliva of swiftlets has both medicinal and nutritional values dated back to ancient China. However, its role in improving endothelial dysfunction due to diabetes is yet to be elucidated. The present study examined the protective effect and mechanism of action of the aqueous extract of hydrolyzed edible bird nest (HBN) on endothelium in models of diabetes, in vitro and in vivo. Male db/m+ and db/db mice were orally administered with or without HBN and glibenclamide for 28 days, followed by vascular reactivity studies in mouse aortas. Human umbilical vein endothelial cells (HUVECs) and isolated mouse aorta from C57BL/6J were treated with high glucose (HG), HBN, sialic acid (SA), glibenclamide, and apocynin, respectively. The effects of HBN on reactive oxygen species (ROS) production and nitric oxide (NO) bioavailability were assessed by Western blot, 2',7'-dichlorofluorescin-diacetate (DCF-DA), and 4-amino-5-methylamino-2',7' difluorofluorescein (DAF-FM DA) in HUVECs, isolated mouse aorta, and db/db diabetic mice. HBN significantly reversed the endothelial dysfunction in diabetic mice and isolated mouse aorta. HBN normalized ROS over-production of NOX2 and nitrotyrosine, reversed the reduction of anti-oxidant marker, SOD-1 as well as restored NO bioavailability in both HUVECs challenged with HG and in db/db diabetic mice. Similarly, HG-induced elevation of oxidative stress in HUVECs were reversed by SA, glibenclamide, and apocynin. This attests that HBN restores endothelial function and protects endothelial cells against oxidative damage induced by HG in HUVECs, isolated mouse aorta, and db/db diabetic mice via modulating ROS mechanism, which subsequently increases NO bioavailability. This result demonstrates the potential role of HBN in preserving endothelial function and management of micro- or macrovascular complications in diabetes.
Collapse
Affiliation(s)
- Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Zuhaida Md Zain
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ker Woon Choy
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Nor Hisam Zamakshshari
- Centre for Natural Product Research and Drug Discovery (CENAR), Wellness Research Cluster, University of Malaya, Kuala Lumpur, Malaysia
| | - Mel June Choong
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Selangor, Malaysia
| | - Yang Mooi Lim
- Department of Pre-clinical Sciences, Centre for Cancer Research, Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Selangor, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Zhang Q, Tsuji-Hosokawa A, Willson C, Watanabe M, Si R, Lai N, Wang Z, Yuan JXJ, Wang J, Makino A. Chloroquine differentially modulates coronary vasodilation in control and diabetic mice. Br J Pharmacol 2020; 177:314-327. [PMID: 31503328 DOI: 10.1111/bph.14864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Chloroquine is a traditional medicine to treat malaria. There is increasing evidence that chloroquine not only induces phagocytosis but regulates vascular tone. Few reports investigating the effect of chloroquine on vascular responsiveness of coronary arteries have been made. In this study, we examined how chloroquine affected endothelium-dependent relaxation in coronary arteries under normal and diabetic conditions. EXPERIMENTAL APPROACH We isolated coronary arteries from mice and examined endothelium-dependent relaxation (EDR). Human coronary endothelial cells and mouse coronary endothelial cells isolated from control and diabetic mouse (TALLYHO/Jng [TH] mice, a spontaneous type 2 diabetic mouse model) were used for the molecular biological or cytosolic NO and Ca2+ measurements. KEY RESULTS Chloroquine inhibited endothelium-derived NO-dependent relaxation but had negligible effect on endothelium-derived hyperpolarization (EDH)-dependent relaxation in coronary arteries of control mice. Chloroquine significantly decreased NO production in control human coronary endothelial cells partly by phosphorylating eNOSThr495 (an inhibitory phosphorylation site of eNOS) and attenuating the rise of cytosolic Ca2+ concentration after stimulation. EDR was significantly inhibited in diabetic mice in comparison to control mice. Interestingly, chloroquine enhanced EDR in diabetic coronary arteries by, specifically, increasing EDH-dependent relaxation due partly to its augmenting effect on gap junction activity in diabetic mouse coronary endothelial cells. CONCLUSIONS AND IMPLICATIONS These data indicate that chloroquine affects vascular relaxation differently under normal and diabetic conditions. Therefore, the patients' health condition such as coronary macrovascular or microvascular disease, with or without diabetes, must be taken account into the consideration when selecting chloroquine for the treatment of malaria.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Physiology, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Conor Willson
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Makiko Watanabe
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Rui Si
- Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Ning Lai
- Department of Medicine, University of California, San Diego, La Jolla, California.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ziyi Wang
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Medicine, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Jian Wang
- Department of Medicine, The University of Arizona, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla, California.,Department of Physiology, The University of Arizona, Tucson, Arizona.,Department of Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
30
|
Vanadium compounds induced damage of human umbilical vein endothelial cells and the protective effect of berberine. Biometals 2019; 32:785-794. [DOI: 10.1007/s10534-019-00211-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/29/2019] [Indexed: 10/26/2022]
|
31
|
The reduction of NDUFC2 expression is associated with mitochondrial impairment in circulating mononuclear cells of patients with acute coronary syndrome. Int J Cardiol 2019; 286:127-133. [PMID: 30808603 DOI: 10.1016/j.ijcard.2019.02.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/10/2019] [Accepted: 02/13/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Deficiency of NADH dehydrogenase [ubiquinone], the mitochondrial complex I, represents an emerging mechanism of cardiovascular diseases. Ndufc2, a subunit of mitochondrial complex I, is involved in stroke development. We aimed to gain some insights on the role of Ndufc2 into acute coronary syndrome (ACS) through the assessment of its gene expression, along with that of anti-oxidant proteins and of mitochondrial function parameters, in circulating mononuclear cells (PBMCs) of ACS versus stable angina (SA) patients. The impact of NDUFC2 silencing in human endothelial and vascular smooth muscle cells was assessed in vitro. METHODS AND RESULTS One hundred twenty-three patients presenting with SA (n = 41) or ACS (n = 82) were enrolled. PBMCs were used to assess the gene expression level of: NDUFC2, uncoupling protein 2 (UCP2), superoxide dysmutases 1 and 2 (SOD1, SOD2), levels of ROS and ATP. The mitochondrial dysfunction was assessed by cytofluorimetry; the structural damage by transmission electron microscopy. Cell viability, angiogenesis, markers of atherogenesis were evaluated in NDUFC2-silenced vascular cells. NDUFC2 mRNA level was significantly downregulated, along with UCP2, SOD1, SOD2 expression, in ACS patients. We found significant increases of ROS levels, reduced ATP levels, higher degree of mitochondrial structural damage and dysfunction in ACS patients. In vitro, NDUFC2 silencing favored mechanisms involved in atherogenesis and plaque vulnerability. CONCLUSIONS A significant reduction of NDUFC2 expression is detected in ACS. In vitro, NDUFC2 silencing affects vascular cell viability and angiogenesis while stimulating the expression of markers of plaque rupture. Our observations suggest that these mechanisms may contribute to ACS development.
Collapse
|
32
|
Pinti MV, Fink GK, Hathaway QA, Durr AJ, Kunovac A, Hollander JM. Mitochondrial dysfunction in type 2 diabetes mellitus: an organ-based analysis. Am J Physiol Endocrinol Metab 2019; 316:E268-E285. [PMID: 30601700 PMCID: PMC6397358 DOI: 10.1152/ajpendo.00314.2018] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/27/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a systemic disease characterized by hyperglycemia, hyperlipidemia, and organismic insulin resistance. This pathological shift in both circulating fuel levels and energy substrate utilization by central and peripheral tissues contributes to mitochondrial dysfunction across organ systems. The mitochondrion lies at the intersection of critical cellular pathways such as energy substrate metabolism, reactive oxygen species (ROS) generation, and apoptosis. It is the disequilibrium of these processes in T2DM that results in downstream deficits in vital functions, including hepatocyte metabolism, cardiac output, skeletal muscle contraction, β-cell insulin production, and neuronal health. Although mitochondria are known to be susceptible to a variety of genetic and environmental insults, the accumulation of mitochondrial DNA (mtDNA) mutations and mtDNA copy number depletion is helping to explain the prevalence of mitochondrial-related diseases such as T2DM. Recent work has uncovered novel mitochondrial biology implicated in disease progressions such as mtDNA heteroplasmy, noncoding RNA (ncRNA), epigenetic modification of the mitochondrial genome, and epitranscriptomic regulation of the mtDNA-encoded mitochondrial transcriptome. The goal of this review is to highlight mitochondrial dysfunction observed throughout major organ systems in the context of T2DM and to present new ideas for future research directions based on novel experimental and technological innovations in mitochondrial biology. Finally, the field of mitochondria-targeted therapeutics is discussed, with an emphasis on novel therapeutic strategies to restore mitochondrial homeostasis in the setting of T2DM.
Collapse
Affiliation(s)
- Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- West Virginia University School of Pharmacy , Morgantown, West Virginia
| | - Garrett K Fink
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- Toxicology Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Andrya J Durr
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| |
Collapse
|
33
|
Vascular endothelial dysfunction, a major mediator in diabetic cardiomyopathy. Acta Pharmacol Sin 2019; 40:1-8. [PMID: 29867137 DOI: 10.1038/s41401-018-0042-6] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is currently a major public health problem. A common complication of diabetes is cardiac dysfunction, which is recognized as a microvascular disease that leads to morbidity and mortality in diabetic patients. While ischemic events are commonly observed in diabetic patients, the risk for developing heart failure is also increased, independent of the severity of coronary artery disease and hypertension. This diabetes-associated clinical entity is considered a distinct disease process referred to as "diabetic cardiomyopathy". However, it is not clear how diabetes promotes cardiac dysfunction. Vascular endothelial dysfunction is thought to be one of the key risk factors. The impact of diabetes on the endothelium involves several alterations, including hyperglycemia, fatty acid oxidation, reduced nitric oxide (NO), oxidative stress, inflammatory activation, and altered barrier function. The current review provides an update on mechanisms that specifically target endothelial dysfunction, which may lead to diabetic cardiomyopathy.
Collapse
|
34
|
Bubb KJ, Ritchie RH, Figtree GA. Modified redox signaling in vasculature after chronic infusion of the insulin receptor antagonist, S961. Microcirculation 2018; 26:e12501. [PMID: 30178465 DOI: 10.1111/micc.12501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/02/2018] [Accepted: 08/30/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Type 2 diabetes and associated vascular complications cause substantial morbidity and mortality. It is important to investigate mechanisms and test therapies in relevant physiological models, yet few animal models adequately recapitulate all aspects of the human condition. OBJECTIVE We sought to determine the potential of using an insulin receptor antagonist, S961, in mice for investigating vascular pathophysiology. METHODS S961 was infused into mice for 4 weeks. Blood glucose was monitored, and insulin was measured at the end of the protocol. Blood pressure and pressor responses to vasodilators were measured in cannulated mice, and vascular reactive oxygen and nitrogen species were measured in isolated tissue. RESULTS S961 infusion-induced hyperglycemia and hyperinsulinemia. There was evidence of increased vascular reactive oxygen and nitrogen species and modification of NO-mediated signaling. Pressor responses to a NO donor were attenuated, but responses to bradykinin were preserved. CONCLUSIONS Infusion of S961, an insulin receptor antagonist, results in the production of a mouse model of type 2 diabetes that may be useful for investigating redox signaling in the vasculature of insulin-resistant mice over the short term. It is limited by both the transient nature of the hyperglycemia and incomplete functional analogy to the human condition.
Collapse
Affiliation(s)
- Kristen J Bubb
- Cardiovascular and Thoracic Health, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gemma A Figtree
- Cardiovascular and Thoracic Health, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Willson C, Watanabe M, Tsuji-Hosokawa A, Makino A. Pulmonary vascular dysfunction in metabolic syndrome. J Physiol 2018; 597:1121-1141. [PMID: 30125956 DOI: 10.1113/jp275856] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome is a critically important precursor to the onset of many diseases, such as cardiovascular disease, and cardiovascular disease is the leading cause of death worldwide. The primary risk factors of metabolic syndrome include hyperglycaemia, abdominal obesity, dyslipidaemia, and high blood pressure. It has been well documented that metabolic syndrome alters vascular endothelial and smooth muscle cell functions in the heart, brain, kidney and peripheral vessels. However, there is less information available regarding how metabolic syndrome can affect pulmonary vascular function and ultimately increase an individual's risk of developing various pulmonary vascular diseases, such as pulmonary hypertension. Here, we review in detail how metabolic syndrome affects pulmonary vascular function.
Collapse
Affiliation(s)
- Conor Willson
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | | | - Ayako Makino
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
36
|
Pan M, Han Y, Basu A, Dai A, Si R, Willson C, Balistrieri A, Scott BT, Makino A. Overexpression of hexokinase 2 reduces mitochondrial calcium overload in coronary endothelial cells of type 2 diabetic mice. Am J Physiol Cell Physiol 2018. [PMID: 29513568 DOI: 10.1152/ajpcell.00350.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Coronary microvascular rarefaction, due to endothelial cell (EC) dysfunction, is one of the causes of increased morbidity and mortality in diabetes. Coronary ECs in diabetes are more apoptotic due partly to mitochondrial calcium overload. This study was designed to investigate the role of hexokinase 2 (HK2, an endogenous inhibitor of voltage-dependent anion channel) in coronary endothelial dysfunction in type 2 diabetes. We used mouse coronary ECs (MCECs) isolated from type 2 diabetic mice and human coronary ECs (HCECs) from type 2 diabetic patients to examine protein levels and mitochondrial function. ECs were more apoptotic and capillary density was lower in the left ventricle of diabetic mice than the control. MCECs from diabetic mice exhibited significant increase in mitochondrial Ca2+ concentration ([Ca2+]mito) compared with the control. Among several regulatory proteins for [Ca2+]mito, hexokinase 1 (HK1) and HK2 were significantly lower in MCECs from diabetic mice than control MCECs. We also found that the level of HK2 ubiquitination was higher in MCECs from diabetic mice than in control MCECs. In line with the data from MCECs, HCECs from diabetic patients showed lower HK2 protein levels than HCECs from nondiabetic patients. High-glucose treatment, but not high-fat treatment, significantly decreased HK2 protein levels in MCECs. HK2 overexpression in MCECs of diabetic mice not only lowered the level of [Ca2+]mito, but also reduced mitochondrial reactive oxygen species production toward the level seen in control MCECs. These data suggest that HK2 is a potential therapeutic target for coronary microvascular disease in diabetes by restoring mitochondrial function in coronary ECs.
Collapse
Affiliation(s)
- Minglin Pan
- Department of Medicine, University of Illinois at Chicago , Chicago, Illinois.,The Second Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Ying Han
- Department of Physiology, University of Arizona , Tucson, Arizona.,Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Aninda Basu
- Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Anzhi Dai
- Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Rui Si
- Department of Physiology, University of Arizona , Tucson, Arizona
| | - Conor Willson
- Department of Physiology, University of Arizona , Tucson, Arizona
| | - Angela Balistrieri
- Department of Physiology, University of Arizona , Tucson, Arizona.,Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ayako Makino
- Department of Physiology, University of Arizona , Tucson, Arizona.,Department of Medicine, University of Arizona , Tucson, Arizona.,Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| |
Collapse
|
37
|
Tang H, Babicheva A, McDermott KM, Gu Y, Ayon RJ, Song S, Wang Z, Gupta A, Zhou T, Sun X, Dash S, Wang Z, Balistrieri A, Zheng Q, Cordery AG, Desai AA, Rischard F, Khalpey Z, Wang J, Black SM, Garcia JGN, Makino A, Yuan JXJ. Endothelial HIF-2α contributes to severe pulmonary hypertension due to endothelial-to-mesenchymal transition. Am J Physiol Lung Cell Mol Physiol 2018; 314:L256-L275. [PMID: 29074488 PMCID: PMC5866501 DOI: 10.1152/ajplung.00096.2017] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 12/26/2022] Open
Abstract
Pulmonary vascular remodeling characterized by concentric wall thickening and intraluminal obliteration is a major contributor to the elevated pulmonary vascular resistance in patients with idiopathic pulmonary arterial hypertension (IPAH). Here we report that increased hypoxia-inducible factor 2α (HIF-2α) in lung vascular endothelial cells (LVECs) under normoxic conditions is involved in the development of pulmonary hypertension (PH) by inducing endothelial-to-mesenchymal transition (EndMT), which subsequently results in vascular remodeling and occlusive lesions. We observed significant EndMT and markedly increased expression of SNAI, an inducer of EndMT, in LVECs from patients with IPAH and animals with experimental PH compared with normal controls. LVECs isolated from IPAH patients had a higher level of HIF-2α than that from normal subjects, whereas HIF-1α was upregulated in pulmonary arterial smooth muscle cells (PASMCs) from IPAH patients. The increased HIF-2α level, due to downregulated prolyl hydroxylase domain protein 2 (PHD2), a prolyl hydroxylase that promotes HIF-2α degradation, was involved in enhanced EndMT and upregulated SNAI1/2 in LVECs from patients with IPAH. Moreover, knockdown of HIF-2α (but not HIF-1α) with siRNA decreases both SNAI1 and SNAI2 expression in IPAH-LVECs. Mice with endothelial cell (EC)-specific knockout (KO) of the PHD2 gene, egln1 (egln1EC-/-), developed severe PH under normoxic conditions, whereas Snai1/2 and EndMT were increased in LVECs of egln1EC-/- mice. EC-specific KO of the HIF-2α gene, hif2a, prevented mice from developing hypoxia-induced PH, whereas EC-specific deletion of the HIF-1α gene, hif1a, or smooth muscle cell (SMC)-specific deletion of hif2a, negligibly affected the development of PH. Also, exposure to hypoxia for 48-72 h increased protein level of HIF-1α in normal human PASMCs and HIF-2α in normal human LVECs. These data indicate that increased HIF-2α in LVECs plays a pathogenic role in the development of severe PH by upregulating SNAI1/2, inducing EndMT, and causing obliterative pulmonary vascular lesions and vascular remodeling.
Collapse
Affiliation(s)
- Haiyang Tang
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Aleksandra Babicheva
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Kimberly M McDermott
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Yali Gu
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ziyi Wang
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Akash Gupta
- Division of Cardiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Tong Zhou
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Xutong Sun
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Swetaleena Dash
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Zilu Wang
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Angela Balistrieri
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Qiuyu Zheng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medicine University , Guangzhou , China
| | - Arlette G Cordery
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ankit A Desai
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Division of Cardiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Franz Rischard
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Zain Khalpey
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Department of Surgery, College of Medicine, University of Arizona , Tucson, Arizona
| | - Jian Wang
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medicine University , Guangzhou , China
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Joe G N Garcia
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Arizona , Tucson, Arizona
- Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
38
|
Leão VF, Ferreira LLDM, Melo CM, Bonfleur ML, da Silva LL, Carneiro EM, Raimundo JM, Ribeiro RA. Taurine supplementation prevents endothelial dysfunction and attenuates structural changes in aortas from hypothalamic obese rats. Eur J Nutr 2018; 58:551-563. [DOI: 10.1007/s00394-018-1616-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/14/2018] [Indexed: 02/07/2023]
|
39
|
Sudhahar V, Okur MN, Bagi Z, O'Bryan JP, Hay N, Makino A, Patel VS, Phillips SA, Stepp D, Ushio-Fukai M, Fukai T. Akt2 (Protein Kinase B Beta) Stabilizes ATP7A, a Copper Transporter for Extracellular Superoxide Dismutase, in Vascular Smooth Muscle: Novel Mechanism to Limit Endothelial Dysfunction in Type 2 Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2018; 38:529-541. [PMID: 29301787 DOI: 10.1161/atvbaha.117.309819] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Copper transporter ATP7A (copper-transporting/ATPase) is required for full activation of SOD3 (extracellular superoxide dismutase), which is secreted from vascular smooth muscle cells (VSMCs) and anchors to endothelial cell surface to preserve endothelial function by scavenging extracellular superoxide. We reported that ATP7A protein expression and SOD3 activity are decreased in insulin-deficient type 1 diabetes mellitus vessels, thereby, inducing superoxide-mediated endothelial dysfunction, which are rescued by insulin treatment. However, it is unknown regarding the mechanism by which insulin increases ATP7A expression in VSMCs and whether ATP7A downregulation is observed in T2DM (type2 diabetes mellitus) mice and human in which insulin-Akt (protein kinase B) pathway is selectively impaired. APPROACH AND RESULTS Here we show that ATP7A protein is markedly downregulated in vessels isolated from T2DM patients, as well as those from high-fat diet-induced or db/db T2DM mice. Akt2 (protein kinase B beta) activated by insulin promotes ATP7A stabilization via preventing ubiquitination/degradation as well as translocation to plasma membrane in VSMCs, which contributes to activation of SOD3 that protects against T2DM-induced endothelial dysfunction. Downregulation of ATP7A in T2DM vessels is restored by constitutive active Akt or PTP1B-/- (protein-tyrosine phosphatase 1B-deficient) T2DM mice, which enhance insulin-Akt signaling. Immunoprecipitation, in vitro kinase assay, and mass spectrometry analysis reveal that insulin stimulates Akt2 binding to ATP7A to induce phosphorylation at Ser1424/1463/1466. Furthermore, SOD3 activity is reduced in Akt2-/- vessels or VSMCs, which is rescued by ATP7A overexpression. CONCLUSION Akt2 plays a critical role in ATP7A protein stabilization and translocation to plasma membrane in VSMCs, which contributes to full activation of vascular SOD3 that protects against endothelial dysfunction in T2DM.
Collapse
Affiliation(s)
- Varadarajan Sudhahar
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Mustafa Nazir Okur
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Zsolt Bagi
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - John P O'Bryan
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Nissim Hay
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Ayako Makino
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Vijay S Patel
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Shane A Phillips
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - David Stepp
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Masuko Ushio-Fukai
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.)
| | - Tohru Fukai
- From the Vascular Biology Center (V.S., Z.B., D.S., M.U.-F., T.F.), Department of Pharmacology and Toxicology (V.S., T.F.), Department of Medicine (Cardiology) (Z.B., M.U.-F.), and Department of Surgery (V.S.P.), Medical College of Georgia at Augusta University; Departments of Medicine (Cardiology) and Pharmacology (V.S., T.F.), Department of Pharmacology (M.N.O., J.P.O., M.U.-F.), Center for Cardiovascular Research (V.S., J.P.O., M.U.-F., T.F.), Department of Physical Therapy (S.A.P.), and Department of Biochemistry and Molecular Genetics (N.H.), University of Illinois at Chicago; Department of Medicine and Physiology, University of Arizona, Tucson (A.M.), Jesse Brown Veterans Affairs Medical Center, Chicago, IL (V.S., T.F.); and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA (V.S., T.F.).
| |
Collapse
|
40
|
Insights into the Dichotomous Regulation of SOD2 in Cancer. Antioxidants (Basel) 2017; 6:antiox6040086. [PMID: 29099803 PMCID: PMC5745496 DOI: 10.3390/antiox6040086] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
While loss of antioxidant expression and the resultant oxidant-dependent damage to cellular macromolecules is key to tumorigenesis, it has become evident that effective oxidant scavenging is conversely necessary for successful metastatic spread. This dichotomous role of antioxidant enzymes in cancer highlights their context-dependent regulation during different stages of tumor development. A prominent example of an antioxidant enzyme with such a dichotomous role and regulation is the mitochondria-localized manganese superoxide dismutase SOD2 (MnSOD). SOD2 has both tumor suppressive and promoting functions, which are primarily related to its role as a mitochondrial superoxide scavenger and H₂O₂ regulator. However, unlike true tumor suppressor- or onco-genes, the SOD2 gene is not frequently lost, or rarely mutated or amplified in cancer. This allows SOD2 to be either repressed or activated contingent on context-dependent stimuli, leading to its dichotomous function in cancer. Here, we describe some of the mechanisms that underlie SOD2 regulation in tumor cells. While much is known about the transcriptional regulation of the SOD2 gene, including downregulation by epigenetics and activation by stress response transcription factors, further research is required to understand the post-translational modifications that regulate SOD2 activity in cancer cells. Moreover, future work examining the spatio-temporal nature of SOD2 regulation in the context of changing tumor microenvironments is necessary to allows us to better design oxidant- or antioxidant-based therapeutic strategies that target the adaptable antioxidant repertoire of tumor cells.
Collapse
|
41
|
Labazi H, Trask AJ. Coronary microvascular disease as an early culprit in the pathophysiology of diabetes and metabolic syndrome. Pharmacol Res 2017; 123:114-121. [PMID: 28700893 DOI: 10.1016/j.phrs.2017.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/19/2017] [Accepted: 07/04/2017] [Indexed: 01/09/2023]
Abstract
Metabolic syndrome (MetS) is a group of cardio-metabolic risk factors that includes obesity, insulin resistance, hypertension, and dyslipidemia; these are also a combination of independent coronary artery disease (CAD) risk factors. Alarmingly, the prevalence of MetS risk factors are increasing and a leading cause for mortality. In the vasculature, complications from MetS and type 2 diabetes (T2D) can be divided into microvascular (retinopathy and nephropathy) and macrovascular (cardiovascular diseases and erectile dysfunction). In addition to vascular and endothelial dysfunction, vascular remodeling and stiffness are also hallmarks of cardiovascular disease (CVD), and well-characterized vascular changes that are observed in the early stages of hypertension, T2D, and obesity [1-3]. In the heart, the link between obstructive atherosclerosis of coronary macrovessels and myocardial ischemia (MI) is well established. However, recent studies show that abnormalities in the coronary microcirculation are associated with functional and structural changes in coronary microvessels (classically defined as being ≤150-200μm internal diameter), which may cause or contribute to MI even in the absence of obstractive CAD. This suggests a prognostic value of an abnormal coronary microcirculation as an early sub-clinical culprit in the pathogenesis and progression of heart disease in T2D and MetS. The aim of this review is to summarize recent studies investigating the coronary microvascular remodeling in an early pre-atherosclerotic phase of MetS and T2D, and to explore potential mechanisms associated with the timing of coronary microvascular remodeling relative to that of the macrovasculature.
Collapse
Affiliation(s)
- Hicham Labazi
- Center for Cardiovascular Research and The Heart Center, The Research Institute at Nationwide Children's Hospital Columbus, OH, United States
| | - Aaron J Trask
- Center for Cardiovascular Research and The Heart Center, The Research Institute at Nationwide Children's Hospital Columbus, OH, United States; Department of Pediatrics, The Ohio State University Columbus, OH, United States.
| |
Collapse
|
42
|
Guo R, Si R, Scott BT, Makino A. Mitochondrial connexin40 regulates mitochondrial calcium uptake in coronary endothelial cells. Am J Physiol Cell Physiol 2017; 312:C398-C406. [PMID: 28122731 PMCID: PMC5407023 DOI: 10.1152/ajpcell.00283.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/30/2023]
Abstract
Connexins (Cxs) are a group of integral membrane proteins that can form gap junctions between adjacent cells. Recently, it was reported that Cx43 is expressed not only in the plasma membrane but also in the inner mitochondrial membrane and that it regulates mitochondrial functions. Cx40 is predominantly expressed in vascular endothelial cells (ECs) and plays an important role in the electrical propagation between ECs and endothelial/smooth muscle cells. However, it is unknown whether Cx40 is expressed in the mitochondria and what the role of mitochondrial Cx40 is in endothelial functions. We observed in coronary ECs that Cx40 protein was expressed in the mitochondria, as determined by Western blot and immunofluorescence studies. We found that mouse coronary ECs (MCECs) isolated from Cx40 knockout (Cx40 KO) mice exhibited significantly lower resting mitochondrial calcium concentration ([Ca2+]mito) than MCECs from wild-type (WT) mice. After increase in cytosolic Ca2+ concentration ([Ca2+]cyto) with cyclopiazonic acid, calcium uptake into the mitochondria was significantly attenuated in MCECs from Cx40 KO mice compared with WT MCECs. There was no difference in resting [Ca2+]cyto and store-operated calcium entry in MCECs from WT and Cx40 KO mice. We also detected a significant decrease in the concentration of mitochondrial reactive oxygen species (ROS) in Cx40 KO MCECs. Cx40 overexpression in ECs significantly increased resting [Ca2+]mito level and calcium uptake by mitochondria in response to increased [Ca2+]cyto and augmented mitochondrial ROS production. These data suggest that mitochondrial Cx40 contributes to the regulation of mitochondrial calcium homeostasis.
Collapse
Affiliation(s)
- Rui Guo
- Department of Physiology, The University of Arizona, Tucson, Arizona; and
| | - Rui Si
- Department of Physiology, The University of Arizona, Tucson, Arizona; and
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ayako Makino
- Department of Physiology, The University of Arizona, Tucson, Arizona; and
| |
Collapse
|
43
|
Pan M, Han Y, Si R, Guo R, Desai A, Makino A. Hypoxia-induced pulmonary hypertension in type 2 diabetic mice. Pulm Circ 2017; 7:175-185. [PMID: 28680577 PMCID: PMC5448524 DOI: 10.1086/690206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/16/2016] [Indexed: 12/20/2022] Open
Abstract
Hypoxia-induced pulmonary hypertension (HPH) is a progressive disease that is mainly caused by chronic exposure to high altitude, chronic obstructive lung disease, and obstructive sleep apnea. The increased pulmonary vascular resistance and increased pulmonary arterial pressure result in increased right ventricular afterload, leading to right heart failure and increased morbidity. There are several clinical reports suggesting a link between PH and diabetes, insulin resistance, or obesity; however, it is unclear whether HPH is associated with diabetes as a progressive complication in diabetes. The major goal of this study is to examine the effect of diabetic "preconditioning" or priming effect on the progression of HPH and define the molecular mechanisms that explain the link between diabetes and HPH. Our data show that HPH is significantly enhanced in diabetic mice, while endothelium-dependent relaxation in pulmonary arteries is significantly attenuated in chronically hypoxic diabetic mice (DH). In addition, we demonstrate that mouse pulmonary endothelial cells (MPECs) isolated from DH mice exhibit a significant increase in mitochondrial reactive oxygen species (ROS) concentration and decreased SOD2 protein expression. Finally, scavenging mitochondrial ROS by mitoTempol restores endothelium-dependent relaxation in pulmonary arteries that is attenuated in DH mice. These data suggest that excessive mitochondrial ROS production in diabetic MPECs leads to the development of severe HPH in diabetic mice exposed to hypoxia.
Collapse
Affiliation(s)
- Minglin Pan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Han
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Rui Si
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Rui Guo
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Ankit Desai
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Ayako Makino
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
44
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 597] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
45
|
Eirin A, Ebrahimi B, Kwon SH, Fiala JA, Williams BJ, Woollard JR, He Q, Gupta RC, Sabbah HN, Prakash YS, Textor SC, Lerman A, Lerman LO. Restoration of Mitochondrial Cardiolipin Attenuates Cardiac Damage in Swine Renovascular Hypertension. J Am Heart Assoc 2016; 5:JAHA.115.003118. [PMID: 27247333 PMCID: PMC4937260 DOI: 10.1161/jaha.115.003118] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Renovascular hypertension (RVH) impairs cardiac structure and left ventricular (LV) function, but whether mitochondrial injury is implicated in RVH‐induced myocardial damage and dysfunction has not been defined. We hypothesized that cardiac remodeling in swine RVH is partly attributable to cardiac mitochondrial injury. Methods and Results After 12 weeks of hypercholesterolemic (HC)‐RVH or control (n=14 each), pigs were treated for another 4 weeks with vehicle or with the mitochondrial‐targeted peptide (MTP), Bendavia (0.1 mg/kg subcutaneously, 5 days/week), which stabilizes mitochondrial inner‐membrane cardiolipin (n=7 each). Cardiac function was subsequently assessed by multidetector‐computed tomography and oxygenation by blood‐oxygen‐level–dependent magnetic resonance imaging. Cardiolipin content, mitochondrial biogenesis, as well as sarcoplasmic‐reticulum calcium cycling, myocardial tissue injury, and coronary endothelial function were assessed ex vivo. Additionally, mitochondrial cardiolipin content, oxidative stress, and bioenergetics were assessed in rat cardiomyocytes incubated with tert‐butyl hydroperoxide (tBHP) untreated or treated with MTP. Chronic mitoprotection in vivo restored cardiolipin content and mitochondrial biogenesis. Thapsigargin‐sensitive sarcoplasmic reticulum Ca2+‐ATPase activity that declined in HC‐RVH normalized in MTP‐treated pigs. Mitoprotection also improved LV relaxation (E/A ratio) and ameliorated cardiac hypertrophy, without affecting blood pressure or systolic function. Myocardial remodeling and coronary endothelial function improved only in MTP‐treated pigs. In tBHP‐treated cardiomyocytes, mitochondrial targeting attenuated a fall in cardiolipin content and bioenergetics. Conclusions Chronic mitoprotection blunted myocardial hypertrophy, improved LV relaxation, and attenuated myocardial cellular and microvascular remodeling, despite sustained HC‐RVH, suggesting that mitochondrial injury partly contributes to hypertensive cardiomyopathy.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Behzad Ebrahimi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Soon Hyo Kwon
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Justin A Fiala
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | | | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Quan He
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Ramech C Gupta
- Division of Cardiovascular Medicine, Henry Ford Health System, Detroit, MI
| | - Hani N Sabbah
- Division of Cardiovascular Medicine, Henry Ford Health System, Detroit, MI
| | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| |
Collapse
|
46
|
Karnewar S, Vasamsetti SB, Gopoju R, Kanugula AK, Ganji SK, Prabhakar S, Rangaraj N, Tupperwar N, Kumar JM, Kotamraju S. Mitochondria-targeted esculetin alleviates mitochondrial dysfunction by AMPK-mediated nitric oxide and SIRT3 regulation in endothelial cells: potential implications in atherosclerosis. Sci Rep 2016; 6:24108. [PMID: 27063143 PMCID: PMC4827087 DOI: 10.1038/srep24108] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/21/2016] [Indexed: 01/10/2023] Open
Abstract
Mitochondria-targeted compounds are emerging as a new class of drugs that can potentially alter the pathophysiology of those diseases where mitochondrial dysfunction plays a critical role. We have synthesized a novel mitochondria-targeted esculetin (Mito-Esc) with an aim to investigate its effect during oxidative stress-induced endothelial cell death and angiotensin (Ang)-II-induced atherosclerosis in ApoE−/− mice. Mito-Esc but not natural esculetin treatment significantly inhibited H2O2- and Ang-II-induced cell death in human aortic endothelial cells by enhancing NO production via AMPK-mediated eNOS phosphorylation. While L-NAME (NOS inhibitor) significantly abrogated Mito-Esc-mediated protective effects, Compound c (inhibitor of AMPK) significantly decreased Mito-Esc-mediated increase in NO production. Notably, Mito-Esc promoted mitochondrial biogenesis by enhancing SIRT3 expression through AMPK activation; and restored H2O2-induced inhibition of mitochondrial respiration. siSIRT3 treatment not only completely reversed Mito-Esc-mediated mitochondrial biogenetic marker expressions but also caused endothelial cell death. Furthermore, Mito-Esc administration to ApoE−/− mice greatly alleviated Ang-II-induced atheromatous plaque formation, monocyte infiltration and serum pro-inflammatory cytokines levels. We conclude that Mito-Esc is preferentially taken up by the mitochondria and preserves endothelial cell survival during oxidative stress by modulating NO generation via AMPK. Also, Mito-Esc-induced SIRT3 plays a pivotal role in mediating mitochondrial biogenesis and perhaps contributes to its anti-atherogenic effects.
Collapse
Affiliation(s)
- Santosh Karnewar
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | - Sathish Babu Vasamsetti
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | - Raja Gopoju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | | | - Sai Krishna Ganji
- National Centre for Mass Spectrometry, Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Sripadi Prabhakar
- National Centre for Mass Spectrometry, Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Nandini Rangaraj
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Nitin Tupperwar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Jerald Mahesh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| |
Collapse
|
47
|
Yang L, Zhang J, Xing W, Zhang X, Xu J, Zhang H, Chen L, Ning X, Ji G, Li J, Zhao Q, Gao F. SIRT3 Deficiency Induces Endothelial Insulin Resistance and Blunts Endothelial-Dependent Vasorelaxation in Mice and Human with Obesity. Sci Rep 2016; 6:23366. [PMID: 27000941 PMCID: PMC4802313 DOI: 10.1038/srep23366] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/03/2016] [Indexed: 02/07/2023] Open
Abstract
Recent evidence implicates the critical role of Sirtuin 3 (SIRT3) in the development of many metabolic diseases, but the contribution of SIRT3 to vascular homeostasis remains largely unknown. The aim of this study was to investigate the role of SIRT3 in endothelial insulin resistance and vascular dysfunction in obesity. We found an impaired insulin-induced mesenteric vasorelaxation and concomitant reduced vascular SIRT3 expression in morbid obese human subjects compared with the non-obese subjects. Downregulation of SIRT3 in cultured human endothelial cells increased mitochondrial reactive oxygen species (mtROS) and impaired insulin signaling as evidenced by decreased phosphorylation of Akt and endothelial nitric oxide synthase and subsequent reduced nitric oxide (NO) release. In addition, obese mice induced by 24-week high-fat diet (HFD) displayed an impaired endothelium-dependent vasorelaxation to both insulin and acetylcholine, which was further exacerbated by the gene deletion of Sirt3. Scavenging of mtROS not only restored insulin-stimulated NO production in SIRT3 knockdown cells, but also improved insulin-induced vasorelaxation in SIRT3 knockout mice fed with HFD. Taken together, our findings suggest that SIRT3 positively regulates endothelial insulin sensitivity and show that SIRT3 deficiency and resultant increased mtROS contribute to vascular dysfunction in obesity.
Collapse
Affiliation(s)
- Lu Yang
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China.,Department of Physiology, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Julei Zhang
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Wenjuan Xing
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Xing Zhang
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Jie Xu
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Haifeng Zhang
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Li Chen
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Xiaona Ning
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Gang Ji
- Department of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Jia Li
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Qingchuan Zhao
- Department of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Feng Gao
- Department of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| |
Collapse
|
48
|
Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA, Lavandero S. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol 2016; 594:509-25. [PMID: 26537557 DOI: 10.1113/jp271301] [Citation(s) in RCA: 421] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/30/2015] [Indexed: 12/14/2022] Open
Abstract
Cardiac hypertrophy is often initiated as an adaptive response to haemodynamic stress or myocardial injury, and allows the heart to meet an increased demand for oxygen. Although initially beneficial, hypertrophy can ultimately contribute to the progression of cardiac disease, leading to an increase in interstitial fibrosis and a decrease in ventricular function. Metabolic changes have emerged as key mechanisms involved in the development and progression of pathological remodelling. As the myocardium is a highly oxidative tissue, mitochondria play a central role in maintaining optimal performance of the heart. 'Mitochondrial dynamics', the processes of mitochondrial fusion, fission, biogenesis and mitophagy that determine mitochondrial morphology, quality and abundance have recently been implicated in cardiovascular disease. Studies link mitochondrial dynamics to the balance between energy demand and nutrient supply, suggesting that changes in mitochondrial morphology may act as a mechanism for bioenergetic adaptation during cardiac pathological remodelling. Another critical function of mitochondrial dynamics is the removal of damaged and dysfunctional mitochondria through mitophagy, which is dependent on the fission/fusion cycle. In this article, we discuss the latest findings regarding the impact of mitochondrial dynamics and mitophagy on the development and progression of cardiovascular pathologies, including diabetic cardiomyopathy, atherosclerosis, damage from ischaemia-reperfusion, cardiac hypertrophy and decompensated heart failure. We will address the ability of mitochondrial fusion and fission to impact all cell types within the myocardium, including cardiac myocytes, cardiac fibroblasts and vascular smooth muscle cells. Finally, we will discuss how these findings can be applied to improve the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- César Vásquez-Trincado
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Ivonne García-Carvajal
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Pennanen
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Sergio Lavandero
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| |
Collapse
|
49
|
Differential Mitochondrial Adaptation in Primary Vascular Smooth Muscle Cells from a Diabetic Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8524267. [PMID: 27034743 PMCID: PMC4737048 DOI: 10.1155/2016/8524267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/10/2015] [Accepted: 11/19/2015] [Indexed: 02/07/2023]
Abstract
Diabetes affects more than 330 million people worldwide and causes elevated cardiovascular disease risk. Mitochondria are critical for vascular function, generate cellular reactive oxygen species (ROS), and are perturbed by diabetes, representing a novel target for therapeutics. We hypothesized that adaptive mitochondrial plasticity in response to nutrient stress would be impaired in diabetes cellular physiology via a nitric oxide synthase- (NOS-) mediated decrease in mitochondrial function. Primary smooth muscle cells (SMCs) from aorta of the nonobese, insulin resistant rat diabetes model Goto-Kakizaki (GK) and the Wistar control rat were exposed to high glucose (25 mM). At baseline, significantly greater nitric oxide evolution, ROS production, and respiratory control ratio (RCR) were observed in GK SMCs. Upon exposure to high glucose, expression of phosphorylated eNOS, uncoupled respiration, and expression of mitochondrial complexes I, II, III, and V were significantly decreased in GK SMCs (p < 0.05). Mitochondrial superoxide increased with high glucose in Wistar SMCs (p < 0.05) with no change in the GK beyond elevated baseline concentrations. Baseline comparisons show persistent metabolic perturbations in a diabetes phenotype. Overall, nutrient stress in GK SMCs caused a persistent decline in eNOS and mitochondrial function and disrupted mitochondrial plasticity, illustrating eNOS and mitochondria as potential therapeutic targets.
Collapse
|
50
|
Xiong S, Wang P, Ma L, Gao P, Gong L, Li L, Li Q, Sun F, Zhou X, He H, Chen J, Yan Z, Liu D, Zhu Z. Ameliorating Endothelial Mitochondrial Dysfunction Restores Coronary Function via Transient Receptor Potential Vanilloid 1-Mediated Protein Kinase A/Uncoupling Protein 2 Pathway. Hypertension 2015; 67:451-60. [PMID: 26667415 DOI: 10.1161/hypertensionaha.115.06223] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023]
Abstract
Coronary heart disease arising from atherosclerosis is a leading cause of cardiogenic death worldwide. Mitochondria are the principal source of reactive oxygen species (ROS), and defective oxidative phosphorylation by the mitochondrial respiratory chain contributes to ROS generation. Uncoupling protein 2 (UCP2), an adaptive antioxidant defense factor, protects against mitochondrial ROS-induced endothelial dysfunction in atherosclerosis. The activation of transient receptor potential vanilloid 1 (TRPV1) attenuates vascular dysfunction. Therefore, whether TRPV1 activation antagonizes coronary lesions by alleviating endothelial mitochondrial dysfunction and enhancing the activity of the protein kinase A/UCP2 pathway warrants examination. ApoE(-/-), ApoE(-/-)/TRPV1(-/-), and ApoE(-/-)/UCP2(-/-) mice were fed standard chow, a high-fat diet (HFD), or the HFD plus 0.01% capsaicin. HFD intake profoundly impaired coronary vasodilatation and myocardial perfusion and shortened the survival duration of ApoE(-/-) mice. TRPV1 or UCP2 deficiency exacerbated HFD-induced coronary dysfunction and was associated with increased ROS generation and reduced nitric oxide production in the endothelium. The activation of TRPV1 by capsaicin upregulated UCP2 expression via protein kinase A phosphorylation, thereby alleviating endothelial mitochondrial dysfunction and inhibiting mitochondrial ROS generation. In vivo, dietary capsaicin supplementation enhanced coronary relaxation and prolonged the survival duration of HFD-fed ApoE(-/-) mice. These effects were not observed in ApoE(-/-) mice lacking the TRPV1 or UCP2 gene. The upregulation of protein kinase A /UCP2 via TRPV1 activation ameliorates coronary dysfunction and prolongs the lifespan of atherosclerotic mice by ameliorating endothelial mitochondrial dysfunction. Dietary capsaicin supplementation may represent a promising intervention for the primary prevention of coronary heart disease.
Collapse
Affiliation(s)
- Shiqiang Xiong
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Peijian Wang
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Liqun Ma
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Peng Gao
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Liuping Gong
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Li Li
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Qiang Li
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Fang Sun
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Xunmei Zhou
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hongbo He
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Jing Chen
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhencheng Yan
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Daoyan Liu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China.
| | - Zhiming Zhu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China.
| |
Collapse
|