1
|
Li H, Cheng Z, Wu D, Hu Q. Nitric oxide and mitochondrial function in cardiovascular diseases. Nitric Oxide 2024; 154:42-50. [PMID: 39577487 DOI: 10.1016/j.niox.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/11/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Nitric oxide (NO) has been highlighted as an important factor in cardiovascular system. As a signaling molecule in the cardiovascular system, NO can relax blood vessels, lower blood pressure, and prevent platelet aggregation. Mitochondria serve as a central hub for cellular metabolism and intracellular signaling, and their dysfunction can lead to a variety of diseases. Accumulating evidence suggests that NO can act as a regulator of mitochondria, affecting mitochondrial function and cellular activity, which in turn mediates the onset and progression of disease. However, there is a lack of comprehensive understanding of how NO regulates mitochondrial function in the cardiovascular system. This review aims to summarize the regulation of mitochondrial function by nitric oxide in cardiovascular related diseases, as well as the multifaceted and complex roles of NO in the cardiovascular system. Understanding the mechanism of NO mediated mitochondrial function can provide new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoqi Li
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zijie Cheng
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
2
|
Cui Q, Jiang T, Xie X, Wang H, Qian L, Cheng Y, Li Q, Lu T, Yao Q, Liu J, Lai B, Chen C, Xiao L, Wang N. S-nitrosylation attenuates pregnane X receptor hyperactivity and acetaminophen-induced liver injury. JCI Insight 2024; 9:e172632. [PMID: 38032737 PMCID: PMC10906221 DOI: 10.1172/jci.insight.172632] [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: 05/31/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Drug-induced liver injury (DILI), especially acetaminophen overdose, is the leading cause of acute liver failure. Pregnane X receptor (PXR) is a nuclear receptor and the master regulator of drug metabolism. Aberrant activation of PXR plays a pathogenic role in the acetaminophen hepatotoxicity. Here, we aimed to examine the S-nitrosylation of PXR (SNO-PXR) in response to acetaminophen. We found that PXR was S-nitrosylated in hepatocytes and the mouse livers after exposure to acetaminophen or S-nitrosoglutathione (GSNO). Mass spectrometry and site-directed mutagenesis identified the cysteine 307 as the primary residue for S-nitrosylation (SNO) modification. In hepatocytes, SNO suppressed both agonist-induced (rifampicin and SR12813) and constitutively active PXR (VP-PXR, a human PXR fused to the minimal transactivator domain of the herpes virus transcription factor VP16) activations. Furthermore, in acetaminophen-overdosed mouse livers, PXR protein was decreased at the centrilobular regions overlapping with increased SNO. In PXR-/- mice, replenishing the livers with the SNO-deficient PXR significantly aggravated hepatic necrosis, increased HMGB1 release, and exacerbated liver injury and inflammation. Particularly, we demonstrated that S-nitrosoglutathione reductase (GSNOR) inhibitor N6022 promoted hepatoprotection by increasing the levels of SNO-PXR. In conclusion, PXR is posttranslationally modified by SNO in hepatocytes in response to acetaminophen. This modification mitigated the acetaminophen-induced PXR hyperactivity. It may serve as a target for therapeutical intervention.
Collapse
Affiliation(s)
- Qi Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Tingting Jiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Xinya Xie
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Haodong Wang
- East China Normal University Health Science Center, Shanghai, China
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yanyan Cheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Qiang Li
- School of Public Health, Xi’an Jiaotong University, Xi’an, China
| | - Tingxu Lu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Qinyu Yao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Jia Liu
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Baochang Lai
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lei Xiao
- School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Nanping Wang
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
3
|
Bai W, Guo T, Wang H, Li B, Sun Q, Wu W, Zhang J, Zhou J, Luo J, Zhu M, Lu J, Li P, Dong B, Han S, Pang X, Zhang G, Bai Y, Wang S. S-nitrosylation of AMPKγ impairs coronary collateral circulation and disrupts VSMC reprogramming. EMBO Rep 2024; 25:128-143. [PMID: 38177907 PMCID: PMC10897329 DOI: 10.1038/s44319-023-00015-3] [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: 07/07/2022] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 01/06/2024] Open
Abstract
Collateral circulation is essential for blood resupply to the ischemic heart, which is dictated by the contractile phenotypic restoration of vascular smooth muscle cells (VSMC). Here we investigate whether S-nitrosylation of AMP-activated protein kinase (AMPK), a key regulator of the VSMC phenotype, impairs collateral circulation. In rats with collateral growth and development, nitroglycerin decreases coronary collateral blood flow (CCBF), inhibits vascular contractile phenotypic restoration, and increases myocardial infarct size, accompanied by reduced AMPK activity in the collateral zone. Nitric oxide (NO) S-nitrosylates human recombinant AMPKγ1 at cysteine 131 and decreases AMP sensitivity of AMPK. In VSMCs, exogenous expression of S-nitrosylation-resistant AMPKγ1 or deficient NO synthase (iNOS) prevents the disruption of VSMC reprogramming. Finally, hyperhomocysteinemia or hyperglycemia increases AMPKγ1 S-nitrosylation, prevents vascular contractile phenotypic restoration, reduces CCBF, and increases the infarct size of the heart in Apoe-/- mice, all of which is rescued in Apoe-/-/iNOSsm-/- mice or Apoe-/- mice with enforced expression of the AMPKγ1-C130A mutant following RI/MI. We conclude that nitrosative stress disrupts coronary collateral circulation during hyperhomocysteinemia or hyperglycemia through AMPK S-nitrosylation.
Collapse
Affiliation(s)
- Wenwu Bai
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Guo
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Han Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Bin Li
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Quan Sun
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wanzhou Wu
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxiong Zhang
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jipeng Zhou
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingmin Luo
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Moli Zhu
- School of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan, China
| | - Junxiu Lu
- School of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan, China
| | - Peng Li
- School of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Shufang Han
- Department of Cardiology, The 960th Hospital of PLA Joint Logistics Support Force, Jinan, China
| | - Xinyan Pang
- Department of Cardiovascular Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Guogang Zhang
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongping Bai
- Department of Geriatric Medicine and Coronary Circulation Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- School of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shuangxi Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
4
|
Al-Owais MM, Hettiarachchi NT, Dallas ML, Scragg JL, Lippiat JD, Holden AV, Steele DS, Peers C. Inhibition of the voltage-gated potassium channel Kv1.5 by hydrogen sulfide attenuates remodeling through S-nitrosylation-mediated signaling. Commun Biol 2023; 6:651. [PMID: 37336943 PMCID: PMC10279668 DOI: 10.1038/s42003-023-05016-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
The voltage-gated K+ channel plays a key role in atrial excitability, conducting the ultra-rapid rectifier K+ current (IKur) and contributing to the repolarization of the atrial action potential. In this study, we examine its regulation by hydrogen sulfide (H2S) in HL-1 cardiomyocytes and in HEK293 cells expressing human Kv1.5. Pacing induced remodeling resulted in shorting action potential duration, enhanced both Kv1.5 channel and H2S producing enzymes protein expression in HL-1 cardiomyocytes. H2S supplementation reduced these remodeling changes and restored action potential duration through inhibition of Kv1.5 channel. H2S also inhibited recombinant hKv1.5, lead to nitric oxide (NO) mediated S-nitrosylation and activated endothelial nitric oxide synthase (eNOS) by increased phosphorylation of Ser1177, prevention of NO formation precluded these effects. Regulation of Ikur by H2S has important cardiovascular implications and represents a novel and potential therapeutic target.
Collapse
Affiliation(s)
- Moza M Al-Owais
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Jonathan D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Arun V Holden
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
5
|
Nagarajan N, Oka SI, Nah J, Wu C, Zhai P, Mukai R, Xu X, Kashyap S, Huang CY, Sung EA, Mizushima W, Titus AS, Takayama K, Mourad Y, Francisco J, Liu T, Chen T, Li H, Sadoshima J. Thioredoxin 1 promotes autophagy through transnitrosylation of Atg7 during myocardial ischemia. J Clin Invest 2023; 133:e162326. [PMID: 36480290 PMCID: PMC9888389 DOI: 10.1172/jci162326] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Modification of cysteine residues by oxidative and nitrosative stress affects structure and function of proteins, thereby contributing to the pathogenesis of cardiovascular disease. Although the major function of thioredoxin 1 (Trx1) is to reduce disulfide bonds, it can also act as either a denitrosylase or transnitrosylase in a context-dependent manner. Here we show that Trx1 transnitrosylates Atg7, an E1-like enzyme, thereby stimulating autophagy. During ischemia, Trx1 was oxidized at Cys32-Cys35 of the oxidoreductase catalytic center and S-nitrosylated at Cys73. Unexpectedly, Atg7 Cys545-Cys548 reduced the disulfide bond in Trx1 at Cys32-Cys35 through thiol-disulfide exchange and this then allowed NO to be released from Cys73 in Trx1 and transferred to Atg7 at Cys402. Experiments conducted with Atg7 C402S-knockin mice showed that S-nitrosylation of Atg7 at Cys402 promotes autophagy by stimulating E1-like activity, thereby protecting the heart against ischemia. These results suggest that the thiol-disulfide exchange and the NO transfer are functionally coupled, allowing oxidized Trx1 to mediate a salutary effect during myocardial ischemia through transnitrosylation of Atg7 and stimulation of autophagy.
Collapse
Affiliation(s)
- Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Changgong Wu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Koichiro Takayama
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Tong Chen
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
6
|
Yin YL, Chen Y, Ren F, Wang L, Zhu ML, Lu JX, Wang QQ, Lu CB, Liu C, Bai YY, Wang SX, Wang JZ, Li P. Nitrosative stress induced by homocysteine thiolactone drives vascular cognitive impairments via GTP cyclohydrolase 1 S-nitrosylation in vivo. Redox Biol 2022; 58:102540. [DOI: 10.1016/j.redox.2022.102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
|
7
|
Cui C, Wu C, Shu P, Liu T, Li H, Beuve A. Soluble guanylyl cyclase mediates noncanonical nitric oxide signaling by nitrosothiol transfer under oxidative stress. Redox Biol 2022; 55:102425. [PMID: 35961098 PMCID: PMC9372771 DOI: 10.1016/j.redox.2022.102425] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
Soluble guanylyl cyclase (GC1) is an α/β heterodimer producing cGMP when stimulated by nitric oxide (NO). The NO-GC1-cGMP pathway is essential for cardiovascular homeostasis but is disrupted by oxidative stress, which causes GC1 desensitization to NO by heme oxidation and S-nitrosation (SNO) of specific cysteines. We discovered that under these conditions, GC1-α subunit increases cellular S-nitrosation via transfer of nitrosothiols to other proteins (transnitrosation) in cardiac and smooth muscle cells. One of the GC1 SNO-targets was the oxidized form of Thioredoxin1 (oTrx1), which is unidirectionally transnitrosated by GC1 with αC610 as a SNO-donor. Because oTrx1 itself drives transnitrosation, we sought and identified SNO-proteins targeted by both GC1 and Trx1. We found that transnitrosation of the small GTPase RhoA by SNO-GC1 requires oTrx1 as a nitrosothiol relay, suggesting a SNO-GC1→oTrx1→RhoA cascade. The RhoA signaling pathway, which is antagonized by the canonical NO-cGMP pathway, was alternatively inhibited by GC1-α-dependent S-nitrosation under oxidative conditions. We propose that SNO-GC1, via transnitrosation, mediates adaptive responses triggered by oxidation of the canonical NO-cGMP pathway.
Collapse
Affiliation(s)
- Chuanlong Cui
- Rutgers School of Graduate Studies, Newark Health Science, Newark, NJ, 07103, USA; Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Changgong Wu
- Thermo Fisher Scientific, Somerset, NJ, 08873, USA
| | - Ping Shu
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA
| | - Annie Beuve
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School at Rutgers, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Huang AW, Janssen PML. The Case for, and Challenges of, Human Cardiac Tissue in Advancing Phosphoprotein Research. Front Physiol 2022; 13:853511. [PMID: 35399265 PMCID: PMC8984461 DOI: 10.3389/fphys.2022.853511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Cardiovascular disease (CVD) and stroke affect over 92 million Americans and account for nearly 1 out of 3 deaths in the US. The use of animal models in cardiovascular research has led to considerable advances in treatment and in our understanding of the pathophysiology of many CVDs. Still, animals may not fully recapitulate human disease states; species differences have long been postulated to be one of the main reasons for a failure of translation between animals and humans in drug discovery and development. Indeed, it has become increasingly clear over the past few decades that to answer certain biomedical questions, like the physiological mechanisms that go awry in many human CVDs, animal tissues may not always be the best option to use. While human cardiac tissue has long been used for laboratory research, published findings often contradict each other, leading to difficulties in interpretation. Current difficulties in utilizing human cardiac tissue include differences in acquisition time, varying tissue procurement protocols, and the struggle to define a human “control” sample. With the tremendous emphasis on translational research that continues to grow, research studies using human tissues are becoming more common. This mini review will discuss advantages, disadvantages, and considerations of using human cardiac tissue in the study of CVDs, paying specific attention to the study of phosphoproteins.
Collapse
|
9
|
Veitch CR, Power AS, Erickson JR. CaMKII Inhibition is a Novel Therapeutic Strategy to Prevent Diabetic Cardiomyopathy. Front Pharmacol 2021; 12:695401. [PMID: 34381362 PMCID: PMC8350113 DOI: 10.3389/fphar.2021.695401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing prevalence of diabetes mellitus worldwide has pushed the complex disease state to the foreground of biomedical research, especially concerning its multifaceted impacts on the cardiovascular system. Current therapies for diabetic cardiomyopathy have had a positive impact, but with diabetic patients still suffering from a significantly greater burden of cardiac pathology compared to the general population, the need for novel therapeutic approaches is great. A new therapeutic target, calcium/calmodulin-dependent kinase II (CaMKII), has emerged as a potential treatment option for preventing cardiac dysfunction in the setting of diabetes. Within the last 10 years, new evidence has emerged describing the pathophysiological consequences of CaMKII activation in the diabetic heart, the mechanisms that underlie persistent CaMKII activation, and the protective effects of CaMKII inhibition to prevent diabetic cardiomyopathy. This review will examine recent evidence tying cardiac dysfunction in diabetes to CaMKII activation. It will then discuss the current understanding of the mechanisms by which CaMKII activity is enhanced during diabetes. Finally, it will examine the benefits of CaMKII inhibition to treat diabetic cardiomyopathy, including contractile dysfunction, heart failure with preserved ejection fraction, and arrhythmogenesis. We intend this review to serve as a critical examination of CaMKII inhibition as a therapeutic strategy, including potential drawbacks of this approach.
Collapse
Affiliation(s)
- Christopher R Veitch
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Amelia S Power
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
10
|
S-nitrosylation-mediated coupling of G-protein alpha-2 with CXCR5 induces Hippo/YAP-dependent diabetes-accelerated atherosclerosis. Nat Commun 2021; 12:4452. [PMID: 34294713 PMCID: PMC8298471 DOI: 10.1038/s41467-021-24736-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/01/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis-associated cardiovascular disease is one of the main causes of death and disability among patients with diabetes mellitus. However, little is known about the impact of S-nitrosylation in diabetes-accelerated atherosclerosis. Here, we show increased levels of S-nitrosylation of guanine nucleotide-binding protein G(i) subunit alpha-2 (SNO-GNAI2) at Cysteine 66 in coronary artery samples from diabetic patients with atherosclerosis, consistently with results from mice. Mechanistically, SNO-GNAI2 acted by coupling with CXCR5 to dephosphorylate the Hippo pathway kinase LATS1, thereby leading to nuclear translocation of YAP and promoting an inflammatory response in endothelial cells. Furthermore, Cys-mutant GNAI2 refractory to S-nitrosylation abrogated GNAI2-CXCR5 coupling, alleviated atherosclerosis in diabetic mice, restored Hippo activity, and reduced endothelial inflammation. In addition, we showed that melatonin treatment restored endothelial function and protected against diabetes-accelerated atherosclerosis by preventing GNAI2 S-nitrosylation. In conclusion, SNO-GNAI2 drives diabetes-accelerated atherosclerosis by coupling with CXCR5 and activating YAP-dependent endothelial inflammation, and reducing SNO-GNAI2 is an efficient strategy for alleviating diabetes-accelerated atherosclerosis.
Collapse
|
11
|
Wang P, Wei M, Zhu X, Liu Y, Yoshimura K, Zheng M, Liu G, Kume S, Morishima M, Kurokawa T, Ono K. Nitric oxide down-regulates voltage-gated Na + channel in cardiomyocytes possibly through S-nitrosylation-mediated signaling. Sci Rep 2021; 11:11273. [PMID: 34050231 PMCID: PMC8163867 DOI: 10.1038/s41598-021-90840-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
Nitric oxide (NO) is produced from endothelial cells and cardiomyocytes composing the myocardium and benefits cardiac function through both vascular-dependent and—independent effects. This study was purposed to investigate the possible adverse effect of NO focusing on the voltage-gated Na+ channel in cardiomyocytes. We carried out patch-clamp experiments on rat neonatal cardiomyocytes demonstrating that NOC-18, an NO donor, significantly reduced Na+ channel current in a dose-dependent manner by a long-term application for 24 h, accompanied by a reduction of Nav1.5-mRNA and the protein, and an increase of a transcription factor forkhead box protein O1 (FOXO1) in the nucleus. The effect of NOC-18 on the Na+ channel was blocked by an inhibitor of thiol oxidation N-ethylmaleimide, a disulfide reducing agent disulfide 1,4-Dithioerythritol, or a FOXO1 activator paclitaxel, suggesting that NO is a negative regulator of the voltage-gated Na+ channel through thiols in regulatory protein(s) for the channel transcription.
Collapse
Affiliation(s)
- Pu Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Mengyan Wei
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Xiufang Zhu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Yangong Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Kenshi Yoshimura
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China
| | - Shinichiro Kume
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Masaki Morishima
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Tatsuki Kurokawa
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Katsushige Ono
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan.
| |
Collapse
|
12
|
Rocca C, Grande F, Granieri MC, Colombo B, De Bartolo A, Giordano F, Rago V, Amodio N, Tota B, Cerra MC, Rizzuti B, Corti A, Angelone T, Pasqua T. The chromogranin A 1-373 fragment reveals how a single change in the protein sequence exerts strong cardioregulatory effects by engaging neuropilin-1. Acta Physiol (Oxf) 2021; 231:e13570. [PMID: 33073482 DOI: 10.1111/apha.13570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/16/2022]
Abstract
AIM Chromogranin A (CgA), a 439-residue long protein, is an important cardiovascular regulator and a precursor of various bioactive fragments. Under stressful/pathological conditions, CgA cleavage generates the CgA1-373 proangiogenic fragment. The present work investigated the possibility that human CgA1-373 influences the mammalian cardiac performance, evaluating the role of its C-terminal sequence. METHODS Haemodynamic assessment was performed on an ex vivo Langendorff rat heart model, while mechanistic studies were performed using perfused hearts, H9c2 cardiomyocytes and in silico. RESULTS On the ex vivo heart, CgA1-373 elicited direct dose-dependent negative inotropism and vasodilation, while CgA1-372 , a fragment lacking the C-terminal R373 residue, was ineffective. Antibodies against the PGPQLR373 C-terminal sequence abrogated the CgA1-373 -dependent cardiac and coronary modulation. Ex vivo studies showed that CgA1-373 -dependent effects were mediated by endothelium, neuropilin-1 (NRP1) receptor, Akt/NO/Erk1,2 pathways, nitric oxide (NO) production and S-nitrosylation. In vitro experiments on H9c2 cardiomyocytes indicated that CgA1-373 also induced eNOS activation directly on the cardiomyocyte component by NRP1 targeting and NO involvement and provided beneficial action against isoproterenol-induced hypertrophy, by reducing the increase in cell surface area and brain natriuretic peptide (BNP) release. Molecular docking and all-atom molecular dynamics simulations strongly supported the hypothesis that the C-terminal R373 residue of CgA1-373 directly interacts with NRP1. CONCLUSION These results suggest that CgA1-373 is a new cardioregulatory hormone and that the removal of R373 represents a critical switch for turning "off" its cardioregulatory activity.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
| | - Fedora Grande
- Laboratory of Medicinal and Analytical Chemistry Department of Pharmacy, Health and Nutritional Sciences University of Calabria Rende Italy
| | - Maria Concetta Granieri
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
| | - Barbara Colombo
- Division of Experimental Oncology Vita‐Salute San Raffaele University–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute Milan Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria Rende Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria Rende Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria Rende Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine Magna Graecia University of Catanzaro Catanzaro Italy
| | - Bruno Tota
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
- Laboratory of Organ and System Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
| | - Bruno Rizzuti
- CNR‐NANOTEC Licryl‐UOS Cosenza and CEMIF.Cal Department of Physics University of Calabria Rende Italy
| | - Angelo Corti
- Division of Experimental Oncology Vita‐Salute San Raffaele University–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute Milan Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
- National Institute of Cardiovascular Research (INRC) Bologna Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Patho‐Physiology Department of Biology, E. and E.S. University of Calabria Rende Italy
- "Fondazione Umberto Veronesi" Milan Italy
| |
Collapse
|
13
|
Nitric Oxide and S-Nitrosylation in Cardiac Regulation: G Protein-Coupled Receptor Kinase-2 and β-Arrestins as Targets. Int J Mol Sci 2021; 22:ijms22020521. [PMID: 33430208 PMCID: PMC7825736 DOI: 10.3390/ijms22020521] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiac diseases including heart failure (HF), are the leading cause of morbidity and mortality globally. Among the prominent characteristics of HF is the loss of β-adrenoceptor (AR)-mediated inotropic reserve. This is primarily due to the derangements in myocardial regulatory signaling proteins, G protein-coupled receptor (GPCR) kinases (GRKs) and β-arrestins (β-Arr) that modulate β-AR signal termination via receptor desensitization and downregulation. GRK2 and β-Arr2 activities are elevated in the heart after injury/stress and participate in HF through receptor inactivation. These GPCR regulators are modulated profoundly by nitric oxide (NO) produced by NO synthase (NOS) enzymes through S-nitrosylation due to receptor-coupled NO generation. S-nitrosylation, which is NO-mediated modification of protein cysteine residues to generate an S-nitrosothiol (SNO), mediates many effects of NO independently from its canonical guanylyl cyclase/cGMP/protein kinase G signaling. Herein, we review the knowledge on the NO system in the heart and S-nitrosylation-dependent modifications of myocardial GPCR signaling components GRKs and β-Arrs.
Collapse
|
14
|
Li XY, Zhang HM, An GP, Liu MY, Han SF, Jin Q, Song Y, Lin YM, Dong B, Wang SX, Meng LB. S-Nitrosylation of Akt by organic nitrate delays revascularization and the recovery of cardiac function in mice following myocardial infarction. J Cell Mol Med 2020; 25:27-36. [PMID: 33128338 PMCID: PMC7810919 DOI: 10.1111/jcmm.15263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/29/2020] [Accepted: 03/20/2020] [Indexed: 12/26/2022] Open
Abstract
The effects of long‐term nitrate therapy are compromised due to protein S‐Nitrosylation, which is mediated by nitric oxide (NO). This study is to determine the role of Akt S‐Nitrosylation in the recovery of heart functions after ischaemia. In recombinant Akt protein and in HEK293 cells, NO donor decreased Akt activity and induced Akt S‐Nitrosylation, but was abolished if Akt protein was mutated by replacing cysteine 296/344 with alanine (Akt‐C296/344A). In endothelial cells, NO induced Akt S‐Nitrosylation, reduced Akt activity and damaged multiple cellular functions including proliferation, migration and tube formation. These alterations were ablated if cells expressed Akt‐C296/344A mutant. In Apoe−/− mice, nitroglycerine infusion increased both Akt S‐Nitrosylation and infarct size, reduced Akt activity and capillary density, and delayed the recovery of cardiac function in ischaemic hearts, compared with mice infused with vehicle. Importantly, these in vivo effects of nitroglycerine in Apoe−/− mice were remarkably prevented by adenovirus‐mediated enforced expression of Akt‐C296/344A mutant. In conclusion, long‐term usage of organic nitrate may inactivate Akt to delay ischaemia‐induced revascularization and the recovery of cardiac function through NO‐mediated S‐Nitrosylation.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Hong-Ming Zhang
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Gui-Peng An
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mo-Yan Liu
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Shu-Fang Han
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Qun Jin
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Ying Song
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Yi-Meng Lin
- Department of Cardiology, the 960thHospital of Chinese People's Liberation Army, Jinan, China
| | - Bo Dong
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.,Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Shuang-Xi Wang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ling-Bo Meng
- Department of Cardiology, The Second Hospital affiliated to Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Boardman NT, Migally B, Pileggi C, Parmar GS, Xuan JY, Menzies K, Harper ME. Glutaredoxin-2 and Sirtuin-3 deficiencies impair cardiac mitochondrial energetics but their effects are not additive. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165982. [PMID: 33002579 DOI: 10.1016/j.bbadis.2020.165982] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/06/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Altered redox biology and oxidative stress have been implicated in the progression of heart failure. Glutaredoxin-2 (GRX2) is a glutathione-dependent oxidoreductase and catalyzes the reversible deglutathionylation of mitochondrial proteins. Sirtuin-3 (SIRT3) is a class III histone deacetylase and regulates lysine acetylation in mitochondria. Both GRX2 and SIRT3 are considered as key in the protection against oxidative damage in the myocardium. Knockout of either contributes to adverse heart pathologies including hypertrophy, hypertension, and cardiac dysfunction. Here, we created and characterized a GRX2 and SIRT3 double-knockout mouse model, hypothesizing that their deletions would have an additive effect on oxidative stress, and exacerbate mitochondrial function and myocardial structural remodeling. Wildtype, single-gene knockout (Sirt3-/-, Grx2-/-), and double-knockout mice (Grx2-/-/Sirt3-/-) were compared in heart weight, histology, mitochondrial respiration and H2O2 production. Overall, the hearts from Grx2-/-/Sirt3-/- mice displayed increased fibrosis and hypertrophy versus wildtype. In the Grx2-/- and the Sirt3-/- we observed changes in mitochondrial oxidative capacity, however this was associated with elevated H2O2 emission only in the Sirt3-/-. Similar changes were observed but not worsened in hearts from Grx2-/-/Sirt3-/- mice, suggesting that these changes were not additive. In human myocardium, using genetic and histopathological data from the human Genotype-Tissue Expression consortium, we confirmed that SIRT3 expression correlates inversely with heart pathology. Altogether, GRX2 and SIRT3 are important in the control of cardiac mitochondrial redox and oxidative processes, but their combined absence does not exacerbate effects, consistent with the overall conclusion that they function together in the complex redox and antioxidant systems in the heart.
Collapse
Affiliation(s)
- Neoma T Boardman
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Department of Medical Biology, Faculty of Health Sciences, UiT-Arctic University of Norway, Tromsø, Norway
| | - Baher Migally
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Chantal Pileggi
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Gaganvir S Parmar
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jian Ying Xuan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Keir Menzies
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
16
|
Molecular mechanisms by which iNOS uncoupling can induce cardiovascular dysfunction during sepsis: Role of posttranslational modifications (PTMs). Life Sci 2020; 255:117821. [PMID: 32445759 DOI: 10.1016/j.lfs.2020.117821] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023]
Abstract
Human sepsis is the result of a multifaceted pathological process causing marked dysregulation of cardiovascular responses. A more sophisticated understanding of the pathogenesis of sepsis is certainly prerequisite. Evidence from studies provide further insight into the role of inducible nitric oxide synthase (iNOS) isoform. Results on inhibition of iNOS in sepsis models remain inconclusive. Concern has been devoted to improving our knowledge and understanding of the role of iNOS. The aim of this review is to define the role of iNOS in redox homeostasis disturbance, the detailed mechanisms linking iNOS and posttranslational modifications (PTMs) to cardiovascular dysfunctions, and their future implications in sepsis settings. Many questions related to the iNOS and PTMs still remain open, and much more work is needed on this.
Collapse
|
17
|
Sun J, Hao W, Fillmore N, Ma H, Springer D, Yu ZX, Sadowska A, Garcia A, Chen R, Muniz-Medina V, Rosenthal K, Lin J, Kuruvilla D, Osbourn J, Karathanasis SK, Walker J, Murphy E. Human Relaxin-2 Fusion Protein Treatment Prevents and Reverses Isoproterenol-Induced Hypertrophy and Fibrosis in Mouse Heart. J Am Heart Assoc 2019; 8:e013465. [PMID: 31818212 PMCID: PMC6951077 DOI: 10.1161/jaha.119.013465] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Heart failure is one of the leading causes of death in Western countries, and there is a need for new therapeutic approaches. Relaxin‐2 is a peptide hormone that mediates pleiotropic cardiovascular effects, including antifibrotic, angiogenic, vasodilatory, antiapoptotic, and anti‐inflammatory effects in vitro and in vivo. Methods and Results We developed RELAX10, a fusion protein composed of human relaxin‐2 hormone and the Fc of a human antibody, to test the hypothesis that extended exposure of the relaxin‐2 peptide could reduce cardiac hypertrophy and fibrosis. RELAX10 demonstrated the same specificity and similar in vitro activity as the relaxin‐2 peptide. The terminal half‐life of RELAX10 was 7 days in mouse and 3.75 days in rat after subcutaneous administration. We evaluated whether treatment with RELAX10 could prevent and reverse isoproterenol‐induced cardiac hypertrophy and fibrosis in mice. Isoproterenol administration in mice resulted in increased cardiac hypertrophy and fibrosis compared with vehicle. Coadministration with RELAX10 significantly attenuated the cardiac hypertrophy and fibrosis compared with untreated animals. Isoproterenol administration significantly increased transforming growth factor β1 (TGF‐β1)–induced fibrotic signaling, which was attenuated by RELAX10. We found that RELAX10 also significantly increased protein kinase B/endothelial NO synthase signaling and protein S‐nitrosylation. In the reversal study, RELAX10‐treated animals showed significantly reduced cardiac hypertrophy and collagen levels. Conclusions These findings support a potential role for RELAX10 in the treatment of heart failure.
Collapse
Affiliation(s)
- Junhui Sun
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | | | - Natasha Fillmore
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Hanley Ma
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Danielle Springer
- Murine Phenotyping Core National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | - Zu-Xi Yu
- Pathology Core National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| | | | | | | | | | | | | | | | | | | | | | - Elizabeth Murphy
- Cardiac Physiology Section/Cardiovascular Branch National Heart, Lung, and Blood Institute/National Institutes of Health Bethesda MD
| |
Collapse
|
18
|
Richards MA, Simon JN, Ma R, Loonat AA, Crabtree MJ, Paterson DJ, Fahlman RP, Casadei B, Fliegel L, Swietach P. Nitric oxide modulates cardiomyocyte pH control through a biphasic effect on sodium/hydrogen exchanger-1. Cardiovasc Res 2019; 116:1958-1971. [PMID: 31742355 PMCID: PMC7567331 DOI: 10.1093/cvr/cvz311] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/31/2019] [Accepted: 11/16/2019] [Indexed: 11/14/2022] Open
Abstract
AIMS When activated, Na+/H+ exchanger-1 (NHE1) produces some of the largest ionic fluxes in the heart. NHE1-dependent H+ extrusion and Na+ entry strongly modulate cardiac physiology through the direct effects of pH on proteins and by influencing intracellular Ca2+ handling. To attain an appropriate level of activation, cardiac NHE1 must respond to myocyte-derived cues. Among physiologically important cues is nitric oxide (NO), which regulates a myriad of cardiac functions, but its actions on NHE1 are unclear. METHODS AND RESULTS NHE1 activity was measured using pH-sensitive cSNARF1 fluorescence after acid-loading adult ventricular myocytes by an ammonium prepulse solution manoeuvre. NO signalling was manipulated by knockout of its major constitutive synthase nNOS, adenoviral nNOS gene delivery, nNOS inhibition, and application of NO-donors. NHE1 flux was found to be activated by low [NO], but inhibited at high [NO]. These responses involved cGMP-dependent signalling, rather than S-nitros(yl)ation. Stronger cGMP signals, that can inhibit phosphodiesterase enzymes, allowed [cAMP] to rise, as demonstrated by a FRET-based sensor. Inferring from the actions of membrane-permeant analogues, cGMP was determined to activate NHE1, whereas cAMP was inhibitory, which explains the biphasic regulation by NO. Activation of NHE1-dependent Na+ influx by low [NO] also increased the frequency of spontaneous Ca2+ waves, whereas high [NO] suppressed these aberrant forms of Ca2+ signalling. CONCLUSIONS Physiological levels of NO stimulation increase NHE1 activity, which boosts pH control during acid-disturbances and results in Na+-driven cellular Ca2+ loading. These responses are positively inotropic but also increase the likelihood of aberrant Ca2+ signals, and hence arrhythmia. Stronger NO signals inhibit NHE1, leading to a reversal of the aforementioned effects, ostensibly as a potential cardioprotective intervention to curtail NHE1 overdrive.
Collapse
Affiliation(s)
- Mark A Richards
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Ruichong Ma
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Aminah A Loonat
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Mark J Crabtree
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - David J Paterson
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Richard P Fahlman
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
19
|
Dynnik VV, Grishina EV, Fedotcheva NI. The mitochondrial NO-synthase/guanylate cyclase/protein kinase G signaling system underpins the dual effects of nitric oxide on mitochondrial respiration and opening of the permeability transition pore. FEBS J 2019; 287:1525-1536. [PMID: 31602795 DOI: 10.1111/febs.15090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/07/2019] [Accepted: 10/09/2019] [Indexed: 12/30/2022]
Abstract
The available data on the involvement of nitric oxide (NO) and mitochondrial calcium-dependent NO synthase (mtNOS) in the control of mitochondrial respiration and the permeability transition pore (mPTP) are contradictory. We have proposed that the mitochondrial mtNOS/guanylate cyclase/protein kinase G signaling system (mtNOS-SS) is also implicated in the control of respiration and mPTP, providing the interplay between NO and mtNOS-SS, which, in turn, may result in inconsistent effects of NO. Therefore, using rat liver mitochondria, we applied specific inhibitors of the enzymes of this signaling system to evaluate its role in the control of respiration and mPTP opening. Steady-state respiration was supported by pyruvate, glutamate, or succinate in the presence of hexokinase, glucose, and ADP. When applied at low concentrations, l-arginine (to 500 µm) and NO donors (to 50 µm) activated the respiration and increased the threshold concentrations of calcium and d,l-palmitoylcarnitine required for the dissipation of the mitochondrial membrane potential and pore opening. Both effects were eliminated by the inhibitors of NO synthase, guanylate cyclase, and kinase G, which denotes the involvement of mtNOS-SS in the activation of respiration and deceleration of mPTP opening. At high concentrations, l-arginine and NO donors inhibited the respiration and promoted pore opening, indicating that adverse effects induced by an NO excess dominate over the protection provided by mtNOS-SS. Thus, these results demonstrate the opposite impact of NO and mtNOS-SS on the respiration and mPTP control, which can explain the dual effects of NO.
Collapse
Affiliation(s)
- Vladimir V Dynnik
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| | - Elena V Grishina
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| | - Nadezhda I Fedotcheva
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| |
Collapse
|
20
|
Maes M, Simeonova D, Stoyanov D, Leunis J. Upregulation of the nitrosylome in bipolar disorder type 1 (BP1) and major depression, but not BP2: Increased IgM antibodies to nitrosylated conjugates are associated with indicants of leaky gut. Nitric Oxide 2019; 91:67-76. [PMID: 31323278 DOI: 10.1016/j.niox.2019.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/01/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022]
|
21
|
Fert-Bober J, Murray CI, Parker SJ, Van Eyk JE. Precision Profiling of the Cardiovascular Post-Translationally Modified Proteome: Where There Is a Will, There Is a Way. Circ Res 2019; 122:1221-1237. [PMID: 29700069 DOI: 10.1161/circresaha.118.310966] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is an exponential increase in biological complexity as initial gene transcripts are spliced, translated into amino acid sequence, and post-translationally modified. Each protein can exist as multiple chemical or sequence-specific proteoforms, and each has the potential to be a critical mediator of a physiological or pathophysiological signaling cascade. Here, we provide an overview of how different proteoforms come about in biological systems and how they are most commonly measured using mass spectrometry-based proteomics and bioinformatics. Our goal is to present this information at a level accessible to every scientist interested in mass spectrometry and its application to proteome profiling. We will specifically discuss recent data linking various protein post-translational modifications to cardiovascular disease and conclude with a discussion for enablement and democratization of proteomics across the cardiovascular and scientific community. The aim is to inform and inspire the readership to explore a larger breadth of proteoform, particularity post-translational modifications, related to their particular areas of expertise in cardiovascular physiology.
Collapse
Affiliation(s)
- Justyna Fert-Bober
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Christopher I Murray
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Sarah J Parker
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA.
| | - Jennifer E Van Eyk
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
22
|
e Silva KSF, Lima RM, Baeza LC, Lima PDS, Cordeiro TDM, Charneau S, da Silva RA, Soares CMDA, Pereira M. Interactome of Glyceraldehyde-3-Phosphate Dehydrogenase Points to the Existence of Metabolons in Paracoccidioides lutzii. Front Microbiol 2019; 10:1537. [PMID: 31338083 PMCID: PMC6629890 DOI: 10.3389/fmicb.2019.01537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/20/2019] [Indexed: 11/13/2022] Open
Abstract
Paracoccidioides is a dimorphic fungus, the causative agent of paracoccidioidomycosis. The disease is endemic within Latin America and prevalent in Brazil. The treatment is based on azoles, sulfonamides and amphotericin B. The seeking for new treatment approaches is a real necessity for neglected infections. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is an essential glycolytic enzyme, well known for its multitude of functions within cells, therefore categorized as a moonlight protein. To our knowledge, this is the first approach performed on the Paracoccidioides genus regarding the description of PPIs having GAPDH as a target. Here, we show an overview of experimental GAPDH interactome in different phases of Paracoccidioides lutzii and an in silico analysis of 18 proteins partners. GAPDH interacted with 207 proteins in P. lutzii. Several proteins bound to GAPDH in mycelium, transition and yeast phases are common to important pathways such as glycolysis and TCA. We performed a co-immunoprecipitation assay to validate the complex formed by GAPDH with triose phosphate isomerase, enolase, isocitrate lyase and 2-methylcitrate synthase. We found GAPDH participating in complexes with proteins of specific pathways, indicating the existence of a glycolytic and a TCA metabolon in P. lutzii. GAPDH interacted with several proteins that undergoes regulation by nitrosylation. In addition, we modeled the GAPDH 3-D structure, performed molecular dynamics and molecular docking in order to identify the interacting interface between GAPDH and the interacting proteins. Despite the large number of interacting proteins, GAPDH has only four main regions of contact with interacting proteins, reflecting its ancestrality and conservation over evolution.
Collapse
Affiliation(s)
| | - Raisa Melo Lima
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Lilian Cristiane Baeza
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Patrícia de Sousa Lima
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Thuany de Moura Cordeiro
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brasília, Brazil
| | - Sébastien Charneau
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brasília, Brazil
| | - Roosevelt Alves da Silva
- Núcleo Colaborativo de Biossistemas, Instituto de Ciências Exatas, Universidade Federal de Jataí, Goiás, Brazil
| | | | - Maristela Pereira
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|
23
|
Suprun EV. Protein post-translational modifications – A challenge for bioelectrochemistry. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.04.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Mnatsakanyan R, Markoutsa S, Walbrunn K, Roos A, Verhelst SHL, Zahedi RP. Proteome-wide detection of S-nitrosylation targets and motifs using bioorthogonal cleavable-linker-based enrichment and switch technique. Nat Commun 2019; 10:2195. [PMID: 31097712 PMCID: PMC6522481 DOI: 10.1038/s41467-019-10182-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/18/2019] [Indexed: 01/03/2023] Open
Abstract
Cysteine modifications emerge as important players in cellular signaling and homeostasis. Here, we present a chemical proteomics strategy for quantitative analysis of reversibly modified Cysteines using bioorthogonal cleavable-linker and switch technique (Cys-BOOST). Compared to iodoTMT for total Cysteine analysis, Cys-BOOST shows a threefold higher sensitivity and considerably higher specificity and precision. Analyzing S-nitrosylation (SNO) in S-nitrosoglutathione (GSNO)-treated and non-treated HeLa extracts Cys-BOOST identifies 8,304 SNO sites on 3,632 proteins covering a wide dynamic range of the proteome. Consensus motifs of SNO sites with differential GSNO reactivity confirm the relevance of both acid-base catalysis and local hydrophobicity for NO targeting to particular Cysteines. Applying Cys-BOOST to SH-SY5Y cells, we identify 2,151 SNO sites under basal conditions and reveal significantly changed SNO levels as response to early nitrosative stress, involving neuro(axono)genesis, glutamatergic synaptic transmission, protein folding/translation, and DNA replication. Our work suggests SNO as a global regulator of protein function akin to phosphorylation and ubiquitination. Reversible cysteine modifications play important roles in cellular redox signaling. Here, the authors develop a chemical proteomics strategy that enables the quantitative analysis of endogenous cysteine nitrosylation sites and their dynamic regulation under nitrosative stress conditions.
Collapse
Affiliation(s)
- Ruzanna Mnatsakanyan
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany
| | - Stavroula Markoutsa
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany
| | - Kim Walbrunn
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany.,Department of Neuropediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Steven H L Verhelst
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany.,Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49, Box 802, 3000, Leuven, Belgium
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Str. 6b, 44227, Dortmund, Germany. .,Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, 5100 de Maisonneuve Blvd. West, Montreal, Quebec, H4A 3T2, Canada. .,Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, 3755 Côte Ste-Catherine Road, Montreal, Quebec, H3T 1E2, Canada.
| |
Collapse
|
25
|
Mnatsakanyan R, Shema G, Basik M, Batist G, Borchers CH, Sickmann A, Zahedi RP. Detecting post-translational modification signatures as potential biomarkers in clinical mass spectrometry. Expert Rev Proteomics 2019; 15:515-535. [PMID: 29893147 DOI: 10.1080/14789450.2018.1483340] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Numerous diseases are caused by changes in post-translational modifications (PTMs). Therefore, the number of clinical proteomics studies that include the analysis of PTMs is increasing. Combining complementary information-for example changes in protein abundance, PTM levels, with the genome and transcriptome (proteogenomics)-holds great promise for discovering important drivers and markers of disease, as variations in copy number, expression levels, or mutations without spatial/functional/isoform information is often insufficient or even misleading. Areas covered: We discuss general considerations, requirements, pitfalls, and future perspectives in applying PTM-centric proteomics to clinical samples. This includes samples obtained from a human subject, for instance (i) bodily fluids such as plasma, urine, or cerebrospinal fluid, (ii) primary cells such as reproductive cells, blood cells, and (iii) tissue samples/biopsies. Expert commentary: PTM-centric discovery proteomics can substantially contribute to the understanding of disease mechanisms by identifying signatures with potential diagnostic or even therapeutic relevance but may require coordinated efforts of interdisciplinary and eventually multi-national consortia, such as initiated in the cancer moonshot program. Additionally, robust and standardized mass spectrometry (MS) assays-particularly targeted MS, MALDI imaging, and immuno-MALDI-may be transferred to the clinic to improve patient stratification for precision medicine, and guide therapies.
Collapse
Affiliation(s)
- Ruzanna Mnatsakanyan
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany
| | - Gerta Shema
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany
| | - Mark Basik
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada
| | - Gerald Batist
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada
| | - Christoph H Borchers
- b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada.,c University of Victoria-Genome British Columbia Proteomics Centre, University of Victoria , Victoria , British Columbia V8Z 7X8 , Canada.,d Department of Biochemistry and Microbiology , University of Victoria , Victoria , British Columbia , V8P 5C2 , Canada.,e Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University , Montreal , Quebec H3T 1E2 , Canada
| | - Albert Sickmann
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany.,f Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum , 44801 Bochum , Germany.,g Department of Chemistry , College of Physical Sciences, University of Aberdeen , Aberdeen AB24 3FX , Scotland , United Kingdom
| | - René P Zahedi
- a Protein Dynamics , Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V , Dortmund , 44227 , Germany.,b Gerald Bronfman Department of Oncology , Jewish General Hospital, McGill University , Montreal , Quebec H4A 3T2 , Canada.,e Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University , Montreal , Quebec H3T 1E2 , Canada
| |
Collapse
|
26
|
Xu X, Qiu H, Shi F, Wang Z, Wang X, Jin L, Chi L, Zhang Q. The protein S-nitrosylation of splicing and translational machinery in vascular endothelial cells is susceptible to oxidative stress induced by oxidized low-density lipoprotein. J Proteomics 2019; 195:11-22. [PMID: 30630120 DOI: 10.1016/j.jprot.2019.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/18/2018] [Accepted: 01/05/2019] [Indexed: 12/24/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL) can impair endothelial function and lead to the atherosclerosis development. Protein S-nitrosylation is sensitive to cellular redox state and acts as a crucial regulator and executor of nitric oxide (NO) signaling pathways. Aberrant S-nitrosylation contributes to the pathogenesis of cardiovascular and cerebrovascular diseases. However, the effect of ox-LDL on S-nitrosylation and its significance for endothelial dysfunction have not been studied at proteome level. Herein, the combined quantitative analysis of proteome and S-nitrosoproteome was performed using an integrated biotin switch and iTRAQ labeling approach in EA.hy926 cell line derived from human umbilical vein endothelial cell (HUVEC) treated with ox-LDL. A total of 2204 S-nitrosylated (SNO) peptides of 1318 SNO-proteins were quantified. Notably, 352 SNO-peptides of 262 SNO-proteins were significantly regulated after excluding S-nitrosylation changes caused by protein expression alterations. Many of them belonged to mRNA splicing, ribosomal structure and translational regulatory proteins, covering the entire translation process. The results indicated that S-nitrosylation of the splicing and translational machinery in vascular endothelial cells was susceptible to ox-LDL. Abnormal protein S-nitrosylation may be one pivotal mechanism underlying endothelial dysfunction induced by ox-LDL. This study potentially enriches the present understanding of pro-atherogenic effect of ox-LDL from the perspective of S-nitrosylation. SIGNIFICANCE: The role of ox-LDL in endothelial dysfunction and atherosclerosis development has been recognized from the aspect of impaired NO production. However, its effect on S-nitrosylation, which is directly related to NO signaling pathway, still remains largely unexplored. Our work initially provided a systematic characterization of S-nitrosoproteome in ox-LDL-treated endothelial cells after ruling out the changes of S-nitrosylation modification caused by protein expression alone. MS-based approach coupled with iTRAQ technique indicated 262 SNO-proteins were significantly regulated. Functional enrichment and interaction network analysis revealed that proteins involved in mRNA splicing and translational machinery were susceptible to abnormal S-nitrosylation under ox-LDL treatment. This achievement suggested one potential mechanism underlying endothelial dysfunction induced by ox-LDL from the perspective of S-nitrosoproteome.
Collapse
Affiliation(s)
- Xiaohui Xu
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Hongyan Qiu
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Feng Shi
- Scientific Research Division, Shandong Institute for Food and Drug Control, Jinan, Shandong 250101, China
| | - Zhe Wang
- Division of Endocrinology and Metabolism, Provincial Hospital affiliated with Shandong University, Jinan, Shandong 250021, China
| | - Xiaowei Wang
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Lan Jin
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Lianli Chi
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China.
| | - Qunye Zhang
- National Glycoengineering Research Center, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250021, China.
| |
Collapse
|
27
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 305] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Zhou SN, Lu JX, Wang XQ, Shan MR, Miao Z, Pan GP, Jian X, Li P, Ping S, Pang XY, Bai YP, Liu C, Wang SX. S-Nitrosylation of Prostacyclin Synthase Instigates Nitrate Cross-Tolerance In Vivo. Clin Pharmacol Ther 2018; 105:201-209. [PMID: 29672839 DOI: 10.1002/cpt.1094] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
Development of nitrate tolerance is a major drawback to nitrate therapy. Prostacyclin (PGI2) is a powerful vasodilator produced from prostaglandin (PGH2) by prostacyclin synthase (PGIS) in endothelial cells. This study aimed to determine the role of PGIS S-nitrosylation in nitrate tolerance induced by nitroglycerin (GTN). In endothelial cells, GTN increased PGIS S-nitrosylation and disturbed PGH2 metabolism, which were normalized by mutants of PGIS cysteine 231/441 to alanine (C231/441A). Clearance of nitric oxide by carboxy-PTIO or inhibition of S-nitrosylation by N-acetyl-cysteine decreased GTN-induced PGIS S-nitrosylation. Enforced expression of mutated PGIS with C231/441A markedly abolished GTN-induced PGIS S-nitrosylation and nitrate cross-tolerance in Apoe-/- mice. Inhibition of cyclooxygenase 1 by aspirin, supplementation of PGI2 by beraprost, and inhibition of PGIS S-nitrosylation by N-acetyl-cysteine improved GTN-induced nitrate cross-tolerance in rats. In patients, increased PGIS S-nitrosylation was associated with nitrate tolerance. In conclusion, GTN induces nitrate cross-tolerance through PGIS S-nitrosylation at cysteine 231/441.
Collapse
Affiliation(s)
- Sheng-Nan Zhou
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Jun-Xiu Lu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xue-Qing Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Mei-Rong Shan
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Zhang Miao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Guo-Pin Pan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xu Jian
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Song Ping
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xin-Yan Pang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Yong-Ping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Liu
- Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Shuang-Xi Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
29
|
eNOS S-nitrosylation mediated OxLDL-induced endothelial dysfunction via increasing the interaction of eNOS with β‑catenin. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1793-1801. [PMID: 29471036 DOI: 10.1016/j.bbadis.2018.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/19/2018] [Accepted: 02/13/2018] [Indexed: 01/03/2023]
Abstract
Protein S-nitrosylation plays an important role in the progression of cardiovascular diseases. eNOS can be S-nitrosylated in endothelial cells, and this modification reversibly attenuates enzyme activity. Under physiological conditions, eNOS directly interacts with β‑catenin. However, whether and how eNOS S-nitrosylation regulates the β‑catenin signal pathway and participates in endothelial dysfunction remains unknown. Here, we show that OxLDL induces the S-nitrosylation of eNOS, which enhances the interaction between eNOS and β‑catenin, transcriptional activity of β‑catenin, cell migration and adhesion molecule expression in endothelial cells. In addition, these effects are partially abolished after eNOS is mutated at Cys94 and Cys99, but not Cys441, in endothelial cells. Furthermore, OxLDL increases iNOS expression. The specific iNOS inhibitor 1400 W decreases eNOS S-nitrosylation and the association of eNOS and β‑catenin, thereby blocking the β‑catenin signal pathway to alleviate OxLDL-induced endothelial dysfunction. Taken together, OxLDL induces eNOS S-nitrosylation at Cys94 and Cys99 via an iNOS-dependent manner, which may increase β‑catenin activation and trigger endothelial injury. This study describes a novel mechanism of endothelial dysfunction.
Collapse
|
30
|
Abstract
Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.
Collapse
Affiliation(s)
- Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| |
Collapse
|
31
|
NO Signaling in the Cardiovascular System and Exercise. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:211-245. [DOI: 10.1007/978-981-10-4304-8_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
|
33
|
Morris G, Walder K, Carvalho AF, Tye SJ, Lucas K, Berk M, Maes M. The role of hypernitrosylation in the pathogenesis and pathophysiology of neuroprogressive diseases. Neurosci Biobehav Rev 2017; 84:453-469. [PMID: 28789902 DOI: 10.1016/j.neubiorev.2017.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/02/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022]
Abstract
There is a wealth of data indicating that de novo protein S-nitrosylation in general and protein transnitrosylation in particular mediates the bulk of nitric oxide signalling. These processes enable redox sensing and facilitate homeostatic regulation of redox dependent protein signalling, function, stability and trafficking. Increased S-nitrosylation in an environment of increasing oxidative and nitrosative stress (O&NS) is initially a protective mechanism aimed at maintaining protein structure and function. When O&NS becomes severe, mechanisms governing denitrosylation and transnitrosylation break down leading to the pathological state referred to as hypernitrosylation (HN). Such a state has been implicated in the pathogenesis and pathophysiology of several neuropsychiatric and neurodegenerative diseases and we investigate its potential role in the development and maintenance of neuroprogressive disorders. In this paper, we propose a model whereby the hypernitrosylation of a range of functional proteins and enzymes lead to changes in activity which conspire to produce at least some of the core abnormalities contributing to the development and maintenance of pathology in these illnesses.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW, Wales, United Kingdom
| | - Ken Walder
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil
| | - Susannah J Tye
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia; Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil; Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, 55128 Mainz, Germany
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia.
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
34
|
Chouchani ET, James AM, Methner C, Pell VR, Prime TA, Erickson BK, Forkink M, Lau GY, Bright TP, Menger KE, Fearnley IM, Krieg T, Murphy MP. Identification and quantification of protein S-nitrosation by nitrite in the mouse heart during ischemia. J Biol Chem 2017; 292:14486-14495. [PMID: 28710281 PMCID: PMC5582841 DOI: 10.1074/jbc.m117.798744] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/12/2017] [Indexed: 12/22/2022] Open
Abstract
Nitrate (NO3-) and nitrite (NO2-) are known to be cardioprotective and to alter energy metabolism in vivo NO3- action results from its conversion to NO2- by salivary bacteria, but the mechanism(s) by which NO2- affects metabolism remains obscure. NO2- may act by S-nitrosating protein thiols, thereby altering protein activity. But how this occurs, and the functional importance of S-nitrosation sites across the mammalian proteome, remain largely uncharacterized. Here we analyzed protein thiols within mouse hearts in vivo using quantitative proteomics to determine S-nitrosation site occupancy. We extended the thiol-redox proteomic technique, isotope-coded affinity tag labeling, to quantify the extent of NO2--dependent S-nitrosation of proteins thiols in vivo Using this approach, called SNOxICAT (S-nitrosothiol redox isotope-coded affinity tag), we found that exposure to NO2- under normoxic conditions or exposure to ischemia alone results in minimal S-nitrosation of protein thiols. However, exposure to NO2- in conjunction with ischemia led to extensive S-nitrosation of protein thiols across all cellular compartments. Several mitochondrial protein thiols exposed to the mitochondrial matrix were selectively S-nitrosated under these conditions, potentially contributing to the beneficial effects of NO2- on mitochondrial metabolism. The permeability of the mitochondrial inner membrane to HNO2, but not to NO2-, combined with the lack of S-nitrosation during anoxia alone or by NO2- during normoxia places constraints on how S-nitrosation occurs in vivo and on its mechanisms of cardioprotection and modulation of energy metabolism. Quantifying S-nitrosated protein thiols now allows determination of modified cysteines across the proteome and identification of those most likely responsible for the functional consequences of NO2- exposure.
Collapse
Affiliation(s)
- Edward T Chouchani
- From the Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02284-9168, .,the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Andrew M James
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Carmen Methner
- the Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| | - Victoria R Pell
- the Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| | - Tracy A Prime
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Brian K Erickson
- From the Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02284-9168.,the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Marleen Forkink
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Gigi Y Lau
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Thomas P Bright
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Katja E Menger
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Ian M Fearnley
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| | - Thomas Krieg
- the Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, United Kingdom
| | - Michael P Murphy
- the Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, United Kingdom, and
| |
Collapse
|
35
|
Badreddin A, Fady Y, Attia H, Hafez M, Khairallah A, Johar D, Bernstein L. What role does the stress response have in congestive heart failure? J Cell Physiol 2017; 233:2863-2870. [PMID: 28493471 DOI: 10.1002/jcp.26003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/10/2017] [Indexed: 01/10/2023]
Abstract
This review is concerned with cardiac malfunction as a result of an imbalance in protein proteostasis, the homeostatic balance between protein removal and regeneration in a long remodeling process involving the endoplasmic reticulum (ER) and the unfolded protein response (UPR). The importance of this is of special significance with regard to cardiac function as a high energy requiring muscular organ that has a high oxygen requirement and is highly dependent on mitochondria. The importance of mitochondria is not only concerned with high energy dependence on mitochondrial electron transport, but it also has a role in the signaling between the mitochondria and the ER under stress. Proteins made in the ER are folded as a result of sulfhydryl groups (-SH) and attractive and repulsive reactions in the tertiary structure. We discuss how this matters with respect to an imbalance between muscle breakdown and repair in a stressful environment, especially as a result of oxidative and nitrosative byproducts of mitochondrial activity. The normal repair is a remodeling, but under this circumstance, the cell undergoes or even lysosomal "self eating" autophagy, or even necrosis instead of apoptosis. We shall discuss the relationship of the UPR pathway to chronic congestive heart failure (CHF).
Collapse
Affiliation(s)
- Ahmed Badreddin
- Department of Cardiothoracic Surgery, Beni-Suef University Faculty of Medicine, Beni-Suef, Egypt
| | - Youssef Fady
- Department of Cardiac Surgery, Cardiac Surgery Center Sultan Qaboos Hospital, Salalah, Dhofar, Sultanate of Oman, Salalah, Oman
| | - Hamdy Attia
- Kasr Al'Ainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Hafez
- Kasr Al'Ainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Khairallah
- Medical Research Division, Department of Pharmacology, National Research Centre, Dokki, Cairo, Egypt
| | - Dina Johar
- Faculty of Women for Arts, Sciences, and Education, Department of Biochemistry and Nutrition, Ain Shams University, Heliopolis, Cairo, Egypt.,Max Rady Faculty of Health Sciences, Department of Physiology and Pathophysiology, Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
36
|
Lopez JR, Kolster J, Zhang R, Adams J. Increased constitutive nitric oxide production by whole body periodic acceleration ameliorates alterations in cardiomyocytes associated with utrophin/dystrophin deficiency. J Mol Cell Cardiol 2017. [PMID: 28623080 DOI: 10.1016/j.yjmcc.2017.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) cardiomyopathy is a progressive lethal disease caused by the lack of the dystrophin protein in the heart. The most widely used animal model of DMD is the dystrophin-deficient mdx mouse; however, these mice exhibit a mild dystrophic phenotype with heart failure only late in life. In contrast, mice deficient for both dystrophin and utrophin (mdx/utrn-/-, or dKO) can be used to model severe DMD cardiomyopathy where pathophysiological indicators of heart failure are detectable by 8-10weeks of age. Nitric oxide (NO) is an important signaling molecule involved in vital functions of regulating rhythm, contractility, and microcirculation of the heart, and constitutive NO production affects the function of proteins involved in excitation-contraction coupling. In this study, we explored the efficacy of enhancing NO production as a therapeutic strategy for treating DMD cardiomyopathy using the dKO mouse model of DMD. Specifically, NO production was induced via whole body periodic acceleration (pGz), a novel non-pharmacologic intervention which enhances NO synthase (NOS) activity through sinusoidal motion of the body in a headward-footward direction, introducing pulsatile shear stress to the vascular endothelium and cardiomyocyte plasma membrane. Male dKO mice were randomized at 8weeks of age to receive daily pGz (480cpm, Gz±3.0m/s2, 1h/d) for 4weeks or no treatment, and a separate age-matched group of WT animals (pGz-treated and untreated) served as non-diseased controls. At the conclusion of the protocol, cardiomyocytes from untreated dKO animals had, respectively, 4.3-fold and 3.5-fold higher diastolic resting concentration of Ca2+ ([Ca2+]d) and Na+ ([Na+]d) compared to WT, while pGz treatment significantly reduced these levels. For dKO cardiomyocytes, pGz treatment also improved the depressed contractile function, decreased oxidative stress, blunted the elevation in calpain activity, and mitigated the abnormal increase in [Ca2+]d upon mechanical stress. These improvements culminated in a significant reduction in circulating cardiac troponin T (cTnT) and an extension of the median lifespan of dKO mice from 16 to 31weeks. Treatment with L-NAME (NOS inhibitor) significantly decreased overall lifespan and abolished the cardioprotective properties elicited by pGz. Our results provide evidence that enhancement of NO synthesis by pGz can ameliorate cellular dysfunction in dKO cardiomyocytes and may represent a novel therapeutic intervention in DMD cardiomyopathy patients.
Collapse
Affiliation(s)
- Jose R Lopez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, United States; Division of Neonatology, Mount Sinai Medical Center, Miami, FL 33140, United States.
| | - Juan Kolster
- Centro de Investigaciones Biomédicas, México, D.F., Mexico
| | - Rui Zhang
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, United States
| | - Jose Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami, FL 33140, United States
| |
Collapse
|
37
|
Abstract
Nitric oxide (NO) is an imperative regulator of the cardiovascular system and is a critical mechanism in preventing the pathogenesis and progression of the diseased heart. The scenario of bioavailable NO in the myocardium is complex: 1) NO is derived from both endogenous NO synthases (endothelial, neuronal, and/or inducible NOSs [eNOS, nNOS, and/or iNOS]) and exogenous sources (entero-salivary NO pathway) and the amount of NO from exogenous sources varies significantly; 2) NOSs are located at discrete compartments of cardiac myocytes and are regulated by distinctive mechanisms under stress; 3) NO regulates diverse target proteins through different modes of post-transcriptional modification (soluble guanylate cyclase [sGC]/cyclic guanosine monophosphate [cGMP]/protein kinase G [PKG]-dependent phosphorylation,
S-nitrosylation, and transnitrosylation); 4) the downstream effectors of NO are multidimensional and vary from ion channels in the plasma membrane to signalling proteins and enzymes in the mitochondria, cytosol, nucleus, and myofilament; 5) NOS produces several radicals in addition to NO (e.g. superoxide, hydrogen peroxide, peroxynitrite, and different NO-related derivatives) and triggers redox-dependent responses. However, nNOS inhibits cardiac oxidases to reduce the sources of oxidative stress in diseased hearts. Recent consensus indicates the importance of nNOS protein in cardiac protection under pathological stress. In addition, a dietary regime with high nitrate intake from fruit and vegetables together with unsaturated fatty acids is strongly associated with reduced cardiovascular events. Collectively, NO-dependent mechanisms in healthy and diseased hearts are better understood and shed light on the therapeutic prospects for NO and NOSs in clinical applications for fatal human heart diseases.
Collapse
Affiliation(s)
- Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, College of Medicine, Seoul National University, 103 Dae Hak Ro, Chong No Gu, 110-799 Seoul, Korea, South.,Yanbian University Hospital, Yanji, Jilin Province, 133000, China.,Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
38
|
Tullio F, Penna C, Cabiale K, Femminò S, Galloni M, Pagliaro P. Cardioprotective effects of calcitonin gene-related peptide in isolated rat heart and in H9c2 cells via redox signaling. Biomed Pharmacother 2017; 90:194-202. [PMID: 28364596 DOI: 10.1016/j.biopha.2017.03.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 02/02/2023] Open
Abstract
The calcitonin-gene-related-peptide (CGRP) release is coupled to the signaling of Angeli's salt in determining vasodilator effects. However, it is unknown whether CGRP is involved in Angeli's salt cardioprotective effects and which are the mechanisms of protection. We aimed to determine whether CGRP is involved in myocardial protection induced by Angeli's salt. We also analyzed the intracellular signaling pathway activated by CGRP. Isolated rat hearts were pre-treated with Angeli's salt or Angeli's salt plus CGRP8-37, a specific CGRP-receptor antagonist, and subjected to ischemia (30-min) and reperfusion (120-min). Moreover, we studied CGRP-induced protection during oxidative stress (H2O2) and hypoxia/reoxygenation protocols in H9c2 cardiomyocytes. Cell vitality and mitochondrial membrane potential (ΔYm, MMP) were measured using MTT and JC-1 dyes. Angeli's salt reduced infarct size and ameliorated post-ischemic cardiac function via a CGRP-receptor-dependent mechanism. Pre-treatment with CGRP increased H9c2 survival upon challenging with either H2O2 (redox stress) or hypoxia/reoxygenation (H/R stress). Under these stress conditions, reduction in MMP and cell death were partly prevented by CGRP. These CGRP beneficial effects were blocked by CGRP8-37. During H/R stress, pre-treatment with either CGRP-receptor, protein kinase C (PKC) or mitochondrial KATP channel antagonists, and pre-treatment with an antioxidant (2-mercaptopropionylglycine) blocked the protection mediated by CGRP. In conclusion, CGRP is involved in the cardioprotective effects of Angeli's salt. In H9c2 cardiomyocytes, CGRP elicits PKC-dependent and mitochondrial-KATP-redox-dependent mechanisms. Hence, CGRP is an important factor in the redox-sensible cardioprotective effects of Angeli's salt.
Collapse
Affiliation(s)
- Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| | - Karine Cabiale
- Department of Clinical and Biological Sciences, University of Turin, Italy; Department of Veterinary Science, University of Torino, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Marco Galloni
- Department of Veterinary Science, University of Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| |
Collapse
|
39
|
Hrstka SCL, Li X, Nelson TJ. NOTCH1-Dependent Nitric Oxide Signaling Deficiency in Hypoplastic Left Heart Syndrome Revealed Through Patient-Specific Phenotypes Detected in Bioengineered Cardiogenesis. Stem Cells 2017; 35:1106-1119. [PMID: 28142228 DOI: 10.1002/stem.2582] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 11/12/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart defect (CHD) attributable to multifactorial molecular underpinnings. Multiple genetic loci have been implicated to increase the risk of disease, yet genotype-phenotype relationships remain poorly defined. Whole genome sequencing complemented by cardiac phenotype from five individuals in an HLHS-affected family enabled the identification of NOTCH1 as a prioritized candidate gene linked to CHD in three individuals with mutant allele burden significantly impairing Notch signaling in the HLHS-affected proband. To better understand a mechanistic basis through which NOTCH1 contributes to heart development, human induced pluripotent stem cells (hiPSCs) were created from the HLHS-affected parent-proband triad and differentiated into cardiovascular cell lineages for molecular characterization. HLHS-affected hiPSCs exhibited a deficiency in Notch signaling pathway components and a diminished capacity to generate hiPSC-cardiomyocytes. Optimization of conditions to procure HLHS-hiPSC-cardiomyocytes led to an approach that compensated for dysregulated nitric oxide (NO)-dependent Notch signaling in the earliest specification stages. Augmentation of HLHS-hiPSCs with small molecules stimulating NO signaling in the first 4 days of differentiation provided a cardiomyocyte yield equivalent to the parental hiPSCs. No discernable differences in calcium dynamics were observed between the bioengineered cardiomyocytes derived from the proband and the parents. We conclude that in vitro modeling with HLHS-hiPSCs bearing NOTCH1 mutations facilitated the discovery of a NO-dependent signaling component essential for cardiovascular cell lineage specification. Potentiation of NO signaling with small therapeutic molecules restored cardiogenesis in vitro and may identify a potential therapeutic target for patients affected by functionally compromised NOTCH1 variants. Stem Cells 2017;35:1106-1119.
Collapse
Affiliation(s)
- Sybil C L Hrstka
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA.,Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy J Nelson
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,General Internal Medicine and Transplant Center, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
40
|
Dulce RA, Kulandavelu S, Schulman IH, Fritsch J, Hare JM. Nitric Oxide Regulation of Cardiovascular Physiology and Pathophysiology. Nitric Oxide 2017. [DOI: 10.1016/b978-0-12-804273-1.00024-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
41
|
Zhang YH. Neuronal nitric oxide synthase in hypertension - an update. Clin Hypertens 2016; 22:20. [PMID: 27822383 PMCID: PMC5093926 DOI: 10.1186/s40885-016-0055-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023] Open
Abstract
Hypertension is a prevalent condition worldwide and is the key risk factor for fatal cardiovascular complications, such as stroke, sudden cardiac death and heart failure. Reduced bioavailability of nitric oxide (NO) in the endothelium is an important precursor for impaired vasodilation and hypertension. In the heart, NO deficiency deteriorates the adverse consequences of pressure-overload and causes cardiac hypertrophy, fibrosis and myocardial infarction which lead to fatal heart failure and sudden cardiac death. Recent consensus is that both endothelial and neuronal nitric oxide synthases (eNOS or NOS3 and nNOS or NOS1) are the constitutive sources of NO in the myocardium. Between the two, nNOS is the predominant isoform of NOS that controls intracellular Ca2+ homeostasis, myocyte contraction, relaxation and signaling pathways including nitroso-redox balance. Notably, our recent research indicates that cardiac eNOS protein is reduced but nNOS protein expression and activity are increased in hypertension. Furthermore, nNOS is induced by the interplay between angiotensin II (Ang II) type 1 receptor (AT1R) and Ang II type 2 receptor (AT2R), mediated by NADPH oxidase and reactive oxygen species (ROS)-dependent eNOS activity in cardiac myocytes. nNOS, in turn, protects the heart from pathogenesis via positive lusitropy in hypertension. Soluble guanylate cyclase (sGC)-cGMP/PKG-dependent phosphorylation of myofilament proteins are novel targets of nNOS in hypertensive myocardium. In this short review, we will endeavor to overview new findings of the up-stream and downstream regulation of cardiac nNOS in hypertension, shed light on the underlying mechanisms which may be of therapeutic value in hypertensive cardiomyopathy.
Collapse
Affiliation(s)
- Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, 103 Dae Hak Ro, Chong No Gu, 110-799 Seoul Korea ; Yanbian University Hospital, Yanji, Jilin Province 133000 China ; Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
42
|
Stram AR, Payne RM. Post-translational modifications in mitochondria: protein signaling in the powerhouse. Cell Mol Life Sci 2016; 73:4063-73. [PMID: 27233499 PMCID: PMC5045789 DOI: 10.1007/s00018-016-2280-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 02/03/2023]
Abstract
There is an intimate interplay between cellular metabolism and the pathophysiology of disease. Mitochondria are essential to maintaining and regulating metabolic function of cells and organs. Mitochondrial dysfunction is implicated in diverse diseases, such as cardiovascular disease, diabetes and metabolic syndrome, neurodegeneration, cancer, and aging. Multiple reversible post-translational protein modifications are located in the mitochondria that are responsive to nutrient availability and redox conditions, and which can act in protein-protein interactions to modify diverse mitochondrial functions. Included in this are physiologic redox signaling via reactive oxygen and nitrogen species, phosphorylation, O-GlcNAcylation, acetylation, and succinylation, among others. With the advent of mass proteomic screening techniques, there has been a vast increase in the array of known mitochondrial post-translational modifications and their protein targets. The functional significance of these processes in disease etiology, and the pathologic response to their disruption, are still under investigation. However, many of these reversible modifications act as regulatory mechanisms in mitochondria and show promise for mitochondrial-targeted therapeutic strategies. This review addresses the current knowledge of post-translational processing and signaling mechanisms in mitochondria, and their implications in health and disease.
Collapse
Affiliation(s)
- Amanda R Stram
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA
| | - R Mark Payne
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA.
| |
Collapse
|
43
|
Lee CF, Chavez JD, Garcia-Menendez L, Choi Y, Roe ND, Chiao YA, Edgar JS, Goo YA, Goodlett DR, Bruce JE, Tian R. Normalization of NAD+ Redox Balance as a Therapy for Heart Failure. Circulation 2016; 134:883-94. [PMID: 27489254 DOI: 10.1161/circulationaha.116.022495] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/08/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND Impairments of mitochondrial function in the heart are linked intricately to the development of heart failure, but there is no therapy for mitochondrial dysfunction. METHODS We assessed the reduced/oxidized ratio of nicotinamide adenine dinucleotide (NADH/NAD(+) ratio) and protein acetylation in the failing heart. Proteome and acetylome analyses were followed by docking calculation, mutagenesis, and mitochondrial calcium uptake assays to determine the functional role of specific acetylation sites. The therapeutic effects of normalizing mitochondrial protein acetylation by expanding the NAD(+) pool also were tested. RESULTS Increased NADH/NAD(+) and protein hyperacetylation, previously observed in genetic models of defective mitochondrial function, also are present in human failing hearts as well as in mouse hearts with pathologic hypertrophy. Elevation of NAD(+) levels by stimulating the NAD(+) salvage pathway suppressed mitochondrial protein hyperacetylation and cardiac hypertrophy, and improved cardiac function in responses to stresses. Acetylome analysis identified a subpopulation of mitochondrial proteins that was sensitive to changes in the NADH/NAD(+) ratio. Hyperacetylation of mitochondrial malate-aspartate shuttle proteins impaired the transport and oxidation of cytosolic NADH in the mitochondria, resulting in altered cytosolic redox state and energy deficiency. Furthermore, acetylation of oligomycin-sensitive conferring protein at lysine-70 in adenosine triphosphate synthase complex promoted its interaction with cyclophilin D, and sensitized the opening of mitochondrial permeability transition pore. Both could be alleviated by normalizing the NAD(+) redox balance either genetically or pharmacologically. CONCLUSIONS We show that mitochondrial protein hyperacetylation due to NAD(+) redox imbalance contributes to the pathologic remodeling of the heart via 2 distinct mechanisms. Our preclinical data demonstrate a clear benefit of normalizing NADH/NAD(+) imbalance in the failing hearts. These findings have a high translational potential as the pharmacologic strategy of increasing NAD(+) precursors are feasible in humans.
Collapse
Affiliation(s)
- Chi Fung Lee
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Juan D Chavez
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Lorena Garcia-Menendez
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Yongseon Choi
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Nathan D Roe
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Ying Ann Chiao
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - John S Edgar
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Young Ah Goo
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - David R Goodlett
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - James E Bruce
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA
| | - Rong Tian
- From Mitochondria and Metabolism Center (C.F.L., L.G.-M., Y.C., N.D.R., R.T.), Department of Anesthesiology and Pain Medicine (C.F.L., L.G.-M, Y.C., N.D.R., R.T.), Department of Genome Sciences (J.D.C., J.E.B.), Department of Pathology (Y.A.C.), and Department of Medicinal Chemistry (J.S.E., Y.A.G., D.R.G.), University of Washington, Seattle, WA.
| |
Collapse
|
44
|
Wu B, Yu H, Wang Y, Pan Z, Zhang Y, Li T, Li L, Zhang W, Ge L, Chen Y, Ho CK, Zhu D, Huang X, Lou Y. Peroxiredoxin-2 nitrosylation facilitates cardiomyogenesis of mouse embryonic stem cells via XBP-1s/PI3K pathway. Free Radic Biol Med 2016; 97:179-191. [PMID: 27261193 DOI: 10.1016/j.freeradbiomed.2016.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/10/2016] [Accepted: 05/28/2016] [Indexed: 11/24/2022]
Abstract
Protein nitrosylation is a ubiquitous post-translational modification in almost all biological systems. However, its function on stem cell biology is so far incompletely understood. Here, we demonstrated that peroxiredoxin 2 (Prdx-2) nitrosylation was involved in cardiomyocyte differentiation of mouse embryonic stem (ES) cells induced by S-nitrosoglutathione (GSNO). We found that temporary GSNO exposure could promote ES cell-derived cardiomyogenesis. Using a stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics approach, coupled with biotin switch technique, a total of 104 nitrosylated proteins were identified. Specifically, one of the antioxidant enzymes, Prdx-2, was abundantly nitrosylated and temporarily reduced in antioxidant activity, causing transient endogenous hydrogen peroxide (H2O2) accumulation and subsequent X-box binding protein-1s/phosphatidylinositol 3-kinase pathway activation. The present study reveals the mechanism in which GSNO favors cardiomyocyte differentiation. Prdx-2 nitrosylation could be a potent strategy to affect the pluripotent stem cell-derived cardiomyogenesis.
Collapse
Affiliation(s)
- Bowen Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Key Science and Technology Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Yu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China
| | - Yifan Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China
| | - Zongfu Pan
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihan Zhang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tong Li
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lu Li
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Key Science and Technology Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weichen Zhang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China
| | - Lijun Ge
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Chen
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Chu Kochen Honors College, Zhejiang University, Hangzhou 310058, China
| | - Choe Kyong Ho
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; College of International Education, Zhejiang University, Hangzhou 310058, China; Haeju Medical University, Haeju, Democratic People's Republic of Korea
| | - Danyan Zhu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Key Science and Technology Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xin Huang
- Key Science and Technology Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Cardiovascular Key Laboratory of Zhejiang Province, The 2nd Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Yijia Lou
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Key Science and Technology Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
45
|
Murphy E, Ardehali H, Balaban RS, DiLisa F, Dorn GW, Kitsis RN, Otsu K, Ping P, Rizzuto R, Sack MN, Wallace D, Youle RJ. Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ Res 2016; 118:1960-91. [PMID: 27126807 PMCID: PMC6398603 DOI: 10.1161/res.0000000000000104] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease is a major leading cause of morbidity and mortality in the United States and elsewhere. Alterations in mitochondrial function are increasingly being recognized as a contributing factor in myocardial infarction and in patients presenting with cardiomyopathy. Recent understanding of the complex interaction of the mitochondria in regulating metabolism and cell death can provide novel insight and therapeutic targets. The purpose of this statement is to better define the potential role of mitochondria in the genesis of cardiovascular disease such as ischemia and heart failure. To accomplish this, we will define the key mitochondrial processes that play a role in cardiovascular disease that are potential targets for novel therapeutic interventions. This is an exciting time in mitochondrial research. The past decade has provided novel insight into the role of mitochondria function and their importance in complex diseases. This statement will define the key roles that mitochondria play in cardiovascular physiology and disease and provide insight into how mitochondrial defects can contribute to cardiovascular disease; it will also discuss potential biomarkers of mitochondrial disease and suggest potential novel therapeutic approaches.
Collapse
|
46
|
Margaritelis NV, Cobley JN, Paschalis V, Veskoukis AS, Theodorou AA, Kyparos A, Nikolaidis MG. Principles for integrating reactive species into in vivo biological processes: Examples from exercise physiology. Cell Signal 2016; 28:256-71. [DOI: 10.1016/j.cellsig.2015.12.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/07/2015] [Accepted: 12/20/2015] [Indexed: 12/14/2022]
|
47
|
Wojdyla K, Wrzesinski K, Williamson J, Fey SJ, Rogowska-Wrzesinska A. Acetaminophen-induced S-nitrosylation and S-sulfenylation signalling in 3D cultured hepatocarcinoma cell spheroids. Toxicol Res (Camb) 2016; 5:905-920. [PMID: 30090399 PMCID: PMC6072433 DOI: 10.1039/c5tx00469a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/26/2016] [Indexed: 12/12/2022] Open
Abstract
Acetaminophen (APAP) is possibly the most widely used medication globally and yet little is known of its molecular effects at therapeutic doses. Using a novel approach, we have analysed the redox proteome of the hepatocellular cell line HepG2/C3A treated with therapeutic doses of APAP and quantitated both individual protein abundance and their reversible S-nitrosylation (SNO) and S-sulfenylation (SOH) modifications by mass spectrometry. APAP treatment results in a late, transient increase in ATP production and a multiplicity of alterations in protein abundance and modifications. The majority of the differentially SNO or SOH modified proteins are found in the endoplasmic reticulum and cytosol, suggesting that the source of reactive species is there. The cellular response indicates: constraint of fatty acid metabolism; reduction in ribosome construction and protein synthesis (to conserve ATP); maintenance of glutathione levels (by increased synthetic capacity); and an increased NADPH production (via the pentose phosphate pathway). This response appears to be coordinated, directly or indirectly, by the canonical Wnt and Nrf2 signalling pathways. Combined with the known role of NAPQI, these studies suggest that the physiological and toxicological responses form a continuum: therapeutic doses of APAP produce reactive species and NAPQI in the cytoplasm but result in little permanent damage. The cell mounts a multifaceted response which minimises disruption and repairs are effected within a day or two. Higher doses of APAP lead to intensified reactive species production, which increasingly disturbs mitochondrial function and eventually leads to cell death.
Collapse
Affiliation(s)
- Katarzyna Wojdyla
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| | - Krzysztof Wrzesinski
- Tissue Culture Engineering Laboratory , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark
| | - James Williamson
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| | - Stephen J Fey
- Tissue Culture Engineering Laboratory , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark
| | - Adelina Rogowska-Wrzesinska
- Protein Research Group , Department of Biochemistry and Molecular Biology , University of Southern Denmark , Campusvej 55 , 5230 Odense M , Denmark .
| |
Collapse
|
48
|
Affiliation(s)
- Divya Seth
- From the Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University (D.S., J.S.S.) and Harrington Discovery Institute (J.S.S.), University Hospitals Case Medical Center, Cleveland, OH
| | - Jonathan S Stamler
- From the Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University (D.S., J.S.S.) and Harrington Discovery Institute (J.S.S.), University Hospitals Case Medical Center, Cleveland, OH.
| |
Collapse
|
49
|
Cardioprotective effects of inorganic nitrate/nitrite in chronic anthracycline cardiotoxicity: Comparison with dexrazoxane. J Mol Cell Cardiol 2015; 91:92-103. [PMID: 26724189 DOI: 10.1016/j.yjmcc.2015.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 12/29/2022]
Abstract
Dexrazoxane (DEX) is a clinically available cardioprotectant that reduces the toxicity induced by anthracycline (ANT) anticancer drugs; however, DEX is seldom used and its action is poorly understood. Inorganic nitrate/nitrite has shown promising results in myocardial ischemia-reperfusion injury and recently in acute high-dose ANT cardiotoxicity. However, the utility of this approach for overcoming clinically more relevant chronic forms of cardiotoxicity remains elusive. Hence, in this study, the protective potential of inorganic nitrate and nitrite against chronic ANT cardiotoxicity was investigated, and the results were compared to those using DEX. Chronic cardiotoxicity was induced in rabbits with daunorubicin (DAU). Sodium nitrate (1g/L) was administered daily in drinking water, while sodium nitrite (0.15 or 5mg/kg) or DEX (60mg/kg) was administered parenterally before each DAU dose. Although oral nitrate induced a marked increase in plasma NOx, it showed no improvement in DAU-induced mortality, myocardial damage or heart failure. Instead, the higher nitrite dose reduced the incidence of end-stage cardiotoxicity, prevented related premature deaths and significantly ameliorated several molecular and cellular perturbations induced by DAU, particularly those concerning mitochondria. The latter result was also confirmed in vitro. Nevertheless, inorganic nitrite failed to prevent DAU-induced cardiac dysfunction and molecular remodeling in vivo and failed to overcome the cytotoxicity of DAU to cardiomyocytes in vitro. In contrast, DEX completely prevented all of the investigated molecular, cellular and functional perturbations that were induced by DAU. Our data suggest that the difference in cardioprotective efficacy between DEX and inorganic nitrite may be related to their different abilities to address a recently proposed upstream target for ANT in the heart - topoisomerase IIβ.
Collapse
|
50
|
Bell JR, Raaijmakers AJA, Janssens JV, Delbridge LMD. CaMKIIδand cardiomyocyte Ca2+signalling new perspectives on splice variant targeting. Clin Exp Pharmacol Physiol 2015; 42:1327-32. [DOI: 10.1111/1440-1681.12489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 12/29/2022]
Affiliation(s)
- James R Bell
- Department of Physiology; University of Melbourne; Victoria Australia
| | | | | | - Lea MD Delbridge
- Department of Physiology; University of Melbourne; Victoria Australia
| |
Collapse
|