1
|
Ahmed MI, Andrikopoulou E, Zheng J, Ulasova E, Pat B, Kelley EE, Powell PC, Denney TS, Lewis C, Davies JE, Darley-Usmar V, Dell’Italia LJ. Interstitial Collagen Loss, Myocardial Remodeling, and Function in Primary Mitral Regurgitation. JACC Basic Transl Sci 2022; 7:973-981. [PMID: 36337921 PMCID: PMC9626893 DOI: 10.1016/j.jacbts.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 11/12/2022]
Abstract
Interstitial collagen loss and cardiomyocyte ultrastructural damage accounts for left ventricular (LV) sphericity and decrease in LV twist and circumferential strain. Normal LV diastolic function belies significantly abnormal left atrial (LA) function and early LV diastolic untwist rate. This underscores the complex interplay of LV and LA myocardial remodeling and function in the pathophysiology of primary mitral regurgitation. In this study, we connect LA function with LV systolic and diastolic myocardial remodeling and function using cardiac magnetic resonance tissue tagging in primary mitral regurgitation.
Collapse
Key Words
- BNP, brain natriuretic peptide
- CMR, cardiac magnetic resonance
- ED, end diastole
- ES, end systole
- ICTP, carboxy-terminal telopeptide of collagen type I
- LA, left atrial
- LV, left ventricle
- LVEF, LV ejection fraction
- PICP, carboxy-terminal propeptide of procollagen type I
- PMR, primary mitral regurgitation
- RV, right ventricle
- SV, stroke volume
- XO, xanthine oxidase
- cardiac magnetic resonance
- collagen loss
- left ventricular remodeling
- primary mitral regurgitation
Collapse
Affiliation(s)
- Mustafa I. Ahmed
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Jingyi Zheng
- Department of Mathematics and Statistics, Auburn University, Auburn, Alabama, USA
| | - Elena Ulasova
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Betty Pat
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| | - Eric E. Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia, USA
| | - Pamela Cox Powell
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| | - Thomas S. Denney
- Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama, USA
| | - Clifton Lewis
- Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James E. Davies
- Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Louis J. Dell’Italia
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| |
Collapse
|
2
|
Miotto MC, Weninger G, Dridi H, Yuan Q, Liu Y, Wronska A, Melville Z, Sittenfeld L, Reiken S, Marks AR. Structural analyses of human ryanodine receptor type 2 channels reveal the mechanisms for sudden cardiac death and treatment. SCIENCE ADVANCES 2022; 8:eabo1272. [PMID: 35857850 PMCID: PMC9299551 DOI: 10.1126/sciadv.abo1272] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/03/2022] [Indexed: 05/29/2023]
Abstract
Ryanodine receptor type 2 (RyR2) mutations have been linked to an inherited form of exercise-induced sudden cardiac death called catecholaminergic polymorphic ventricular tachycardia (CPVT). CPVT results from stress-induced sarcoplasmic reticular Ca2+ leak via the mutant RyR2 channels during diastole. We present atomic models of human wild-type (WT) RyR2 and the CPVT mutant RyR2-R2474S determined by cryo-electron microscopy with overall resolutions in the range of 2.6 to 3.6 Å, and reaching local resolutions of 2.25 Å, unprecedented for RyR2 channels. Under nonactivating conditions, the RyR2-R2474S channel is in a "primed" state between the closed and open states of WT RyR2, rendering it more sensitive to activation that results in stress-induced Ca2+ leak. The Rycal drug ARM210 binds to RyR2-R2474S, reverting the primed state toward the closed state. Together, these studies provide a mechanism for CPVT and for the therapeutic actions of ARM210.
Collapse
Affiliation(s)
- Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Yang Liu
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Leah Sittenfeld
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Cho JS, Han YS, Jensen C, Sieck G. Effects of arginase inhibition on myocardial Ca 2+ and contractile responses. Physiol Rep 2022; 10:e15396. [PMID: 35866269 PMCID: PMC9305075 DOI: 10.14814/phy2.15396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 04/18/2023] Open
Abstract
Nitric oxide (NO) is thought to increase cardiac contractility by increasing cytosolic Ca2+ concentration ([Ca2+ ]cyt ) during excitation. Alternatively, NO could increase the sensitivity of the contractile response to [Ca2+ ]cyt (Ca2+ sensitivity). Arginase regulates NO production by competing with NO synthase (NOS), and thus, arginase inhibition should increase cardiac contractility by increasing NO production. We hypothesized that arginase inhibition increases cardiac contractility by increasing both [Ca2+ ]cyt and Ca2+ sensitivity. [Ca2+ ]cyt and contractile (sarcomere length [SL] shortening) responses to electrical stimulation were measured simultaneously in isolated rat cardiomyocytes using an IonOptix system. In the same cardiomyocytes, measurements were obtained at baseline, following 3-min exposure to an arginase inhibitor (S-[2-boronoethyl]-l-cysteine; BEC) and following 3-min exposure to BEC plus a NOS inhibitor (NG -nitro-l-arginine-methyl ester; l-NAME). These responses were compared to time-matched control cardiomyocytes that were untreated. Compared to baseline, BEC increased the amplitude and the total amount of evoked [Ca2+ ]cyt , and the extent and velocity of SL shortening in cardiomyocytes, whereas addition of l-NAME mitigated these effects. The [Ca2+ ]cyt at 50% contraction and relaxation were not different across treatment groups indicating no effect of BEC on Ca2+ sensitivity. The [Ca2+ ]cyt and SL shortening responses in time-matched controls did not vary with time. Arginase inhibition by BEC significantly increased the amplitude and the total amount of evoked [Ca2+ ]cyt , and the extent and velocity of SL shortening in cardiomyocytes, but did not affect Ca2+ sensitivity. These effects of BEC were mitigated by l-NAME. Together, these results indicate an effect of NO on [Ca2+ ]cyt responses that then increase the contractile response of cardiomyocytes.
Collapse
Affiliation(s)
- Jin Sun Cho
- Department of Anesthesiology and Pain MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Young Soo Han
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Cole Jensen
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary Sieck
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
4
|
Hao T, Qian M, Zhang Y, Liu Q, Midgley AC, Liu Y, Che Y, Hou J, Zhao Q. An Injectable Dual-Function Hydrogel Protects Against Myocardial Ischemia/Reperfusion Injury by Modulating ROS/NO Disequilibrium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105408. [PMID: 35319828 PMCID: PMC9130918 DOI: 10.1002/advs.202105408] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/25/2022] [Indexed: 06/14/2023]
Abstract
Acute myocardial infarction (MI) is the leading cause of death worldwide. Exogenous delivery of nitric oxide (NO) to the infarcted myocardium has proven to be an effective strategy for treating MI due to the multiple physiological functions of NO. However, reperfusion of blood flow to the ischemic tissues is accompanied by the overproduction of toxic reactive oxygen species (ROS), which can further exacerbate tissue damage and compromise the therapeutic efficacy. Here, an injectable hydrogel is synthesized from the chitosan modified by boronate-protected diazeniumdiolate (CS-B-NO) that can release NO in response to ROS stimulation and thereby modulate ROS/NO disequilibrium after ischemia/reperfusion (I/R) injury. Furthermore, administration of CS-B-NO efficiently attenuated cardiac damage and adverse cardiac remodeling, promoted repair of the heart, and ameliorated cardiac function, unlike a hydrogel that only released NO, in a mouse model of myocardial I/R injury. Mechanistically, regulation of the ROS/NO balance activated the antioxidant defense system and protected against oxidative stress induced by I/R injury via adaptive regulation of the Nrf2-Keap1 pathway. Inflammation is then reduced by inhibition of the activation of NF-κB signaling. Collectively, these results show that this dual-function hydrogel may be a promising candidate for the protection of tissues and organs after I/R injury.
Collapse
Affiliation(s)
- Tian Hao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yating Zhang
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Qi Liu
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Yongzhe Che
- School of MedicineNankai UniversityTianjin300071China
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Sustainable Chemical TransformationsKey Laboratory of Bioactive Materials (Ministry of Education)Frontiers Science Center for Cell ResponsesCollege of Life SciencesNankai UniversityTianjin300071China
| |
Collapse
|
5
|
Butts B, Brown JA, Denney TS, Ballinger S, Lloyd SG, Oparil S, Sanders P, Merriman TR, Gaffo A, Singh J, Kelley EE, Calhoun DA, Dell'Italia LJ. Racial Differences in XO (Xanthine Oxidase) and Mitochondrial DNA Damage-Associated Molecular Patterns in Resistant Hypertension. Hypertension 2022; 79:775-784. [PMID: 35164526 PMCID: PMC10652275 DOI: 10.1161/hypertensionaha.121.18298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/22/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously reported increased plasma XO (xanthine oxidase) activity in patients with resistant hypertension. Increased XO can cause mitochondrial DNA damage and promote release of fragments called mitochondrial DNA damage-associated molecular patterns (mtDNA DAMPs). Here, we report racial differences in XO activity and mtDNA DAMPs in Black and White adults with resistant hypertension. METHODS This retrospective study includes 91 resistant hypertension patients (44% Black, 47% female) with blood pressure >140/90 mm Hg on ≥4 medications and 37 normotensive controls (30% Black, 54% female) with plasma XO activity, mtDNA DAMPs, and magnetic resonance imaging of left ventricular morphology and function. RESULTS Black-resistant hypertension patients were younger (mean age 52±10 versus 59±10 years; P=0.001), with higher XO activity and left ventricular wall thickness, and worse diastolic dysfunction than White resistant hypertension patients. Urinary sodium excretion (mg/24 hour per kg) was positively related to left ventricular end-diastolic volume (r=0.527, P=0.001) and left ventricular mass (r=0.394, P=0.02) among Black but not White resistant hypertension patients. Patients with resistant hypertension had increased mtDNA DAMPs versus controls (P<0.001), with Black mtDNA DAMPS greater than Whites (P<0.001). Transmission electron microscopy of skeletal muscle biopsies in resistant hypertension patients demonstrates mitochondria cristae lysis, myofibrillar loss, large lipid droplets, and glycogen accumulation. CONCLUSIONS These data warrant a large study to examine the role of XO and mitochondrial mtDNA DAMPs in cardiac remodeling and heart failure in Black adults with resistant hypertension.
Collapse
Affiliation(s)
- Brittany Butts
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Jamelle A Brown
- Center for Free Radical Biology and Department of Pathology, UAB SOM (J.A.B., S.B.)
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University (T.S.D.)
| | - Scott Ballinger
- Center for Free Radical Biology and Department of Pathology, UAB SOM (J.A.B., S.B.)
| | - Steven G Lloyd
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Suzanne Oparil
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Paul Sanders
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Nephrology Research and Training Center and Division of Nephrology UAB SOM (P.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
| | - Angelo Gaffo
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Jasvinder Singh
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University (E.E.K.)
| | - David A Calhoun
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| |
Collapse
|
6
|
The Role of Oxidative Stress in the Aging Heart. Antioxidants (Basel) 2022; 11:antiox11020336. [PMID: 35204217 PMCID: PMC8868312 DOI: 10.3390/antiox11020336] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Medical advances and the availability of diagnostic tools have considerably increased life expectancy and, consequently, the elderly segment of the world population. As age is a major risk factor in cardiovascular disease (CVD), it is critical to understand the changes in cardiac structure and function during the aging process. The phenotypes and molecular mechanisms of cardiac aging include several factors. An increase in oxidative stress is a major player in cardiac aging. Reactive oxygen species (ROS) production is an important mechanism for maintaining physiological processes; its generation is regulated by a system of antioxidant enzymes. Oxidative stress occurs from an imbalance between ROS production and antioxidant defenses resulting in the accumulation of free radicals. In the heart, ROS activate signaling pathways involved in myocyte hypertrophy, interstitial fibrosis, contractile dysfunction, and inflammation thereby affecting cell structure and function, and contributing to cardiac damage and remodeling. In this manuscript, we review recent published research on cardiac aging. We summarize the aging heart biology, highlighting key molecular pathways and cellular processes that underlie the redox signaling changes during aging. Main ROS sources, antioxidant defenses, and the role of dysfunctional mitochondria in the aging heart are addressed. As metabolism changes contribute to cardiac aging, we also comment on the most prevalent metabolic alterations. This review will help us to understand the mechanisms involved in the heart aging process and will provide a background for attractive molecular targets to prevent age-driven pathology of the heart. A greater understanding of the processes involved in cardiac aging may facilitate our ability to mitigate the escalating burden of CVD in older individuals and promote healthy cardiac aging.
Collapse
|
7
|
Solanki K, Rajpoot S, Bezsonov EE, Orekhov AN, Saluja R, Wary A, Axen C, Wary K, Baig MS. The expanding roles of neuronal nitric oxide synthase (NOS1). PeerJ 2022; 10:e13651. [PMID: 35821897 PMCID: PMC9271274 DOI: 10.7717/peerj.13651] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 01/17/2023] Open
Abstract
The nitric oxide synthases (NOS; EC 1.14.13.39) use L-arginine as a substrate to produce nitric oxide (NO) as a by-product in the tissue microenvironment. NOS1 represents the predominant NO-producing enzyme highly enriched in the brain and known to mediate multiple functions, ranging from learning and memory development to maintaining synaptic plasticity and neuronal development, Alzheimer's disease (AD), psychiatric disorders and behavioral deficits. However, accumulating evidence indicate both canonical and non-canonical roles of NOS1-derived NO in several other tissues and chronic diseases. A better understanding of NOS1-derived NO signaling, and identification and characterization of NO-metabolites in non-neuronal tissues could become useful in diagnosis and prognosis of diseases associated with NOS1 expression. Continued investigation on the roles of NOS1, therefore, will synthesize new knowledge and aid in the discovery of small molecules which could be used to titrate the activities of NOS1-derived NO signaling and NO-metabolites. Here, we address the significance of NOS1 and its byproduct NO in modifying pathophysiological events, which could be beneficial in understanding both the disease mechanisms and therapeutics.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Sajjan Rajpoot
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny E Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Anita Wary
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Cassondra Axen
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Kishore Wary
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| |
Collapse
|
8
|
Xu D, Murakoshi N, Tajiri K, Duo F, Okabe Y, Murakata Y, Yuan Z, Li S, Aonuma K, Song Z, Shimoda Y, Mori H, Sato A, Nogami A, Aonuma K, Ieda M. Xanthine oxidase inhibitor febuxostat reduces atrial fibrillation susceptibility by inhibition of oxidized CaMKII in Dahl salt-sensitive rats. Clin Sci (Lond) 2021; 135:2409-2422. [PMID: 34386810 DOI: 10.1042/cs20210405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.
Collapse
Affiliation(s)
- DongZhu Xu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Feng Duo
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuta Okabe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zixun Yuan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zonghu Song
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuzuno Shimoda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
9
|
Kolodziej F, O’Halloran KD. Re-Evaluating the Oxidative Phenotype: Can Endurance Exercise Save the Western World? Antioxidants (Basel) 2021; 10:609. [PMID: 33921022 PMCID: PMC8071436 DOI: 10.3390/antiox10040609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 01/16/2023] Open
Abstract
Mitochondria are popularly called the "powerhouses" of the cell. They promote energy metabolism through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation, which in contrast to cytosolic glycolysis are oxygen-dependent and significantly more substrate efficient. That is, mitochondrial metabolism provides substantially more cellular energy currency (ATP) per macronutrient metabolised. Enhancement of mitochondrial density and metabolism are associated with endurance training, which allows for the attainment of high relative VO2 max values. However, the sedentary lifestyle and diet currently predominant in the Western world lead to mitochondrial dysfunction. Underdeveloped mitochondrial metabolism leads to nutrient-induced reducing pressure caused by energy surplus, as reduced nicotinamide adenine dinucleotide (NADH)-mediated high electron flow at rest leads to "electron leak" and a chronic generation of superoxide radicals (O2-). Chronic overload of these reactive oxygen species (ROS) damages cell components such as DNA, cell membranes, and proteins. Counterintuitively, transiently generated ROS during exercise contributes to adaptive reduction-oxidation (REDOX) signalling through the process of cellular hormesis or "oxidative eustress" defined by Helmut Sies. However, the unaccustomed, chronic oxidative stress is central to the leading causes of mortality in the 21st century-metabolic syndrome and the associated cardiovascular comorbidities. The endurance exercise training that improves mitochondrial capacity and the protective antioxidant cellular system emerges as a universal intervention for mitochondrial dysfunction and resultant comorbidities. Furthermore, exercise might also be a solution to prevent ageing-related degenerative diseases, which are caused by impaired mitochondrial recycling. This review aims to break down the metabolic components of exercise and how they translate to athletic versus metabolically diseased phenotypes. We outline a reciprocal relationship between oxidative metabolism and inflammation, as well as hypoxia. We highlight the importance of oxidative stress for metabolic and antioxidant adaptation. We discuss the relevance of lactate as an indicator of critical exercise intensity, and inferring from its relationship with hypoxia, we suggest the most appropriate mode of exercise for the case of a lost oxidative identity in metabolically inflexible patients. Finally, we propose a reciprocal signalling model that establishes a healthy balance between the glycolytic/proliferative and oxidative/prolonged-ageing phenotypes. This model is malleable to adaptation with oxidative stress in exercise but is also susceptible to maladaptation associated with chronic oxidative stress in disease. Furthermore, mutations of components involved in the transcriptional regulatory mechanisms of mitochondrial metabolism may lead to the development of a cancerous phenotype, which progressively presents as one of the main causes of death, alongside the metabolic syndrome.
Collapse
Affiliation(s)
- Filip Kolodziej
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, T12 XF62 Cork, Ireland;
| | | |
Collapse
|
10
|
Mosqueira M, Konietzny R, Andresen C, Wang C, H A Fink R. Cardiomyocyte depolarization triggers NOS-dependent NO transient after calcium release, reducing the subsequent calcium transient. Basic Res Cardiol 2021; 116:18. [PMID: 33728868 PMCID: PMC7966140 DOI: 10.1007/s00395-021-00860-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/09/2021] [Indexed: 12/18/2022]
Abstract
Cardiac excitation-contraction coupling and metabolic and signaling activities are centrally modulated by nitric oxide (NO), which is produced by one of three NO synthases (NOSs). Despite the significant role of NO in cardiac Ca2+ homeostasis regulation under different pathophysiological conditions, such as Duchenne muscular dystrophy (DMD), no precise method describes the production, source or effect of NO through two NO signaling pathways: soluble guanylate cyclase-protein kinase G (NO-sGC-PKG) and S-nitrosylation (SNO). Using a novel strategy involving isolated murine cardiomyocytes loaded with a copper-based dye highly specific for NO, we observed a single transient NO production signal after each electrical stimulation event. The NO transient signal started 67.5 ms after the beginning of Rhod-2 Ca2+ transient signal and lasted for approximately 430 ms. Specific NOS isoform blockers or NO scavengers significantly inhibited the NO transient, suggesting that wild-type (WT) cardiomyocytes produce nNOS-dependent NO transients. Conversely, NO transient in mdx cardiomyocyte, a mouse model of DMD, was dependent on inducible NOS (iNOS) and endothelial (eNOS). In a consecutive stimulation protocol, the nNOS-dependent NO transient in WT cardiomyocytes significantly reduced the next Ca2+ transient via NO-sGC-PKG. In mdx cardiomyocytes, this inhibitory effect was iNOS- and eNOS-dependent and occurred through the SNO pathway. Basal NO production was nNOS- and iNOS-dependent in WT cardiomyocytes and eNOS- and iNOS-dependent in mdx cardiomyocytes. These results showed cardiomyocyte produces NO isoform-dependent transients upon membrane depolarization at the millisecond time scale activating a specific signaling pathway to negatively modulate the subsequent Ca2+ transient.
Collapse
Affiliation(s)
- Matias Mosqueira
- Cardio-Ventilatory Muscle Physiology Laboratory, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, Im Neuenheimer Feld 326, R. 305, 69120, Heidelberg, Germany.
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| | - Roland Konietzny
- Cardio-Ventilatory Muscle Physiology Laboratory, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, Im Neuenheimer Feld 326, R. 305, 69120, Heidelberg, Germany
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Carolin Andresen
- Cardio-Ventilatory Muscle Physiology Laboratory, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, Im Neuenheimer Feld 326, R. 305, 69120, Heidelberg, Germany
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chao Wang
- Cardio-Ventilatory Muscle Physiology Laboratory, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, Im Neuenheimer Feld 326, R. 305, 69120, Heidelberg, Germany
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Cardiovascular Department, Wuhan No. 1 Hospital, Hubei, China
| | - Rainer H A Fink
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| |
Collapse
|
11
|
Michel LYM, Farah C, Balligand JL. The Beta3 Adrenergic Receptor in Healthy and Pathological Cardiovascular Tissues. Cells 2020; 9:cells9122584. [PMID: 33276630 PMCID: PMC7761574 DOI: 10.3390/cells9122584] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
The third isotype of beta-adrenoreceptors (β3-AR) has recently come (back) into focus after the observation of its expression in white and beige human adipocytes and its implication in metabolic regulation. This coincides with the recent development and marketing of agonists at the human receptor with superior specificity. Twenty years ago, however, we and others described the expression of β3-AR in human myocardium and its regulation of contractility and cardiac remodeling. Subsequent work from many laboratories has since expanded the characterization of β3-AR involvement in many aspects of cardiovascular physio(patho)logy, justifying the present effort to update current paradigms under the light of the most recent evidence.
Collapse
Affiliation(s)
- Lauriane Y. M. Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
| | - Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
- Department of Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
- Correspondence: ; Tel.: +32-27645262
| |
Collapse
|
12
|
Gerdes HJ, Yang M, Heisner JS, Camara AKS, Stowe DF. Modulation of peroxynitrite produced via mitochondrial nitric oxide synthesis during Ca 2+ and succinate-induced oxidative stress in cardiac isolated mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1861:148290. [PMID: 32828729 DOI: 10.1016/j.bbabio.2020.148290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 01/09/2023]
Abstract
We hypothesized that NO• is generated in isolated cardiac mitochondria as the source for ONOO- production during oxidative stress. We monitored generation of ONOO- from guinea pig isolated cardiac mitochondria subjected to excess Ca2+ uptake before adding succinate and determined if ONOO- production was dependent on a nitric oxide synthase (NOS) located in cardiac mitochondria (mtNOS). Mitochondria were suspended in experimental buffer at pH 7.15, and treated with CaCl2 and then the complex II substrate Na-succinate, followed by menadione, a quinone redox cycler, to generate O2•-. L-tyrosine was added to the mitochondrial suspension where it is oxidized by ONOO- to form dityrosine (diTyr) in proportion to the ONOO- present. We found that exposing mitochondria to excess CaCl2 before succinate resulted in an increase in diTyr and amplex red fluorescence (H2O2) signals, indicating that mitochondrial oxidant stress, induced by elevated mtCa2+ and succinate, increased mitochondrial ONOO- production via NO• and O2•-. Changes in mitochondrial ONOO- production dependent on NOS were evidenced by using NOS inhibitors L-NAME/L-NNA, TEMPOL, a superoxide dismutase (SOD) mimetic, and PTIO, a potent global NO• scavenger. L-NAME and L-NNA decreased succinate and menadione-mediated ONOO- production, PTIO decreased production of ONOO-, and TEMPOL decreased ONOO- levels by converting more O2•- to H2O2. Electron microscopy showed immuno-gold labeled iNOS and nNOS in mitochondria isolated from cardiomyocytes and heart tissue. Western blots demonstrated iNOS and nNOS bands in total heart tissue, bands for both iNOS and nNOS in β-tubulin-free non-purified (crude) mitochondrial preparations, and a prominent iNOS band, but no nNOS band, in purified (Golgi and ER-free) mitochondria. Prior treatment of guinea pigs with lipopolysacharride (LPS) enhanced expression of iNOS in liver mitochondria but not in heart mitochondria. Our results indicate that release of ONOO- into the buffer is dependent both on O2•- released from mitochondria and NO• derived from a mtCa2+-inducible nNOS isoform, possibly attached to mitochondria, and a mtNOS isoform like iNOS that is non-inducible.
Collapse
Affiliation(s)
- Harrison J Gerdes
- Anesthesiology Research Division, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Meiying Yang
- Anesthesiology Research Division, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - James S Heisner
- Anesthesiology Research Division, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Anesthesiology Research Division, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David F Stowe
- Anesthesiology Research Division, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, USA; Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA.
| |
Collapse
|
13
|
Abstract
Xanthine oxidase inhibitors are primarily used in the clinical prevention and treatment of gout associated with hyperuricemia. The archetypal xanthine oxidase inhibitor, Allopurinol has been shown to have other beneficial effects such as a reduction in vascular reactive oxygen species and mechano-energetic uncoupling. This chapter discusses these properties and their relevance to human pathophysiology with a focus on Allopurinol as well as newer xanthine oxidase inhibitors such as Febuxostat and Topiroxostat. Xanthine oxidase (XO) and xanthine dehydrogenase (XDH) are collectively referred to as xanthine oxidoreductase (XOR). XDH is initially synthesised as a 150-kDa protein from which XO is derived, e.g. under conditions of ischemia/hypoxia either reversibly by conformational changes (calcium or SH oxidation) or irreversibly by proteolysis, the latter leading to formation of a 130-kDa form of XO. Both, XO and XDH, catalyse the conversion of hypoxanthine via xanthine to uric acid, the former by using oxygen forming superoxide and hydrogen peroxide and the latter NAD+. However, XDH is in principle also able to generate ROS.
Collapse
|
14
|
The Role of Oxidative Stress in Cardiac Disease: From Physiological Response to Injury Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5732956. [PMID: 32509147 PMCID: PMC7244977 DOI: 10.1155/2020/5732956] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/11/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive chemical species containing oxygen, controlled by both enzymatic and nonenzymatic antioxidant defense systems. In the heart, ROS play an important role in cell homeostasis, by modulating cell proliferation, differentiation, and excitation-contraction coupling. Oxidative stress occurs when ROS production exceeds the buffering capacity of the antioxidant defense systems, leading to cellular and molecular abnormalities, ultimately resulting in cardiac dysfunction. In this review, we will discuss the physiological sources of ROS in the heart, the mechanisms of oxidative stress-related myocardial injury, and the implications of experimental studies and clinical trials with antioxidant therapies in cardiovascular diseases.
Collapse
|
15
|
S-Nitrosylation: An Emerging Paradigm of Redox Signaling. Antioxidants (Basel) 2019; 8:antiox8090404. [PMID: 31533268 PMCID: PMC6769533 DOI: 10.3390/antiox8090404] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a highly reactive molecule, generated through metabolism of L-arginine by NO synthase (NOS). Abnormal NO levels in mammalian cells are associated with multiple human diseases, including cancer. Recent studies have uncovered that the NO signaling is compartmentalized, owing to the localization of NOS and the nature of biochemical reactions of NO, including S-nitrosylation. S-nitrosylation is a selective covalent post-translational modification adding a nitrosyl group to the reactive thiol group of a cysteine to form S-nitrosothiol (SNO), which is a key mechanism in transferring NO-mediated signals. While S-nitrosylation occurs only at select cysteine thiols, such a spatial constraint is partially resolved by transnitrosylation, where the nitrosyl moiety is transferred between two interacting proteins to successively transfer the NO signal to a distant location. As NOS is present in various subcellular locales, a stress could trigger concerted S-nitrosylation and transnitrosylation of a large number of proteins involved in divergent signaling cascades. S-nitrosylation is an emerging paradigm of redox signaling by which cells confer protection against oxidative stress.
Collapse
|
16
|
Zhao J, Yang HT, Wasala L, Zhang K, Yue Y, Duan D, Lai Y. Dystrophin R16/17 protein therapy restores sarcolemmal nNOS in trans and improves muscle perfusion and function. Mol Med 2019; 25:31. [PMID: 31266455 PMCID: PMC6607532 DOI: 10.1186/s10020-019-0101-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Background Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. Methods Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. Results Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. Conclusions Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton. Electronic supplementary material The online version of this article (10.1186/s10020-019-0101-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Hsiao Tung Yang
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
17
|
Avula UMR, Hernandez JJ, Yamazaki M, Valdivia CR, Chu A, Rojas-Pena A, Kaur K, Ramos-Mondragón R, Anumonwo JM, Nattel S, Valdivia HH, Kalifa J. Atrial Infarction-Induced Spontaneous Focal Discharges and Atrial Fibrillation in Sheep: Role of Dantrolene-Sensitive Aberrant Ryanodine Receptor Calcium Release. Circ Arrhythm Electrophysiol 2019. [PMID: 29540372 DOI: 10.1161/circep.117.005659] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms underlying spontaneous atrial fibrillation (AF) associated with atrial ischemia/infarction are incompletely elucidated. Here, we investigate the mechanisms underlying spontaneous AF in an ovine model of left atrial myocardial infarction (LAMI). METHODS AND RESULTS LAMI was created by ligating the atrial branch of the left anterior descending coronary artery. ECG loop recorders were implanted to monitor AF episodes. In 7 sheep, dantrolene-a ryanodine receptor blocker-was administered in vivo during the 8-day observation period (LAMI-D, 2.5 mg/kg, IV, BID). LAMI animals experienced numerous spontaneous AF episodes during the 8-day monitoring period that were suppressed by dantrolene (LAMI, 26.1±5.1; sham, 4.3±1.1; LAMI-D, 2.8±0.8; mean±SEM episodes per sheep, P<0.01). Optical mapping showed spontaneous focal discharges (SFDs) originating from the ischemic/normal-zone border. SFDs were calcium driven, rate dependent, and enhanced by isoproterenol (0.03 µmol/L, from 210±87 to 3816±1450, SFDs per sheep) but suppressed by dantrolene (to 55.8±32.8, SFDs per sheep, mean±SEM). SFDs initiated AF-maintaining reentrant rotors anchored by marked conduction delays at the ischemic/normal-zone border. NOS1 (NO synthase-1) protein expression decreased in ischemic zone myocytes, whereas NADPH (nicotinamide adenine dinucleotide phosphate, reduced form) oxidase and xanthine oxidase enzyme activities and reactive oxygen species (DCF [6-carboxy-2',7'-dichlorodihydrofluorescein diacetate]-fluorescence) increased. CaM (calmodulin) aberrantly increased [3H]ryanodine binding to cardiac RyR2 (ryanodine receptors) in the ischemic zone. Dantrolene restored the physiological binding of CaM to RyR2. CONCLUSIONS Atrial ischemia causes spontaneous AF episodes in sheep, caused by SFDs that initiate reentry. Nitroso-redox imbalance in the ischemic zone is associated with intense reactive oxygen species production and altered RyR2 responses to CaM. Dantrolene administration normalizes the CaM response, prevents LAMI-related SFDs, and AF initiation. These findings provide novel insights into the mechanisms underlying ischemia-related atrial arrhythmias.
Collapse
Affiliation(s)
- Uma Mahesh R Avula
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Jonathan J Hernandez
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Masatoshi Yamazaki
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Carmen R Valdivia
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Antony Chu
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Alvaro Rojas-Pena
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Kuljeet Kaur
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Roberto Ramos-Mondragón
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Justus M Anumonwo
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Stanley Nattel
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Héctor H Valdivia
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Jérôme Kalifa
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.).
| |
Collapse
|
18
|
Butts B, Calhoun DA, Denney TS, Lloyd SG, Gupta H, Gaddam KK, Aban I, Oparil S, Sanders PW, Patel R, Collawn JF, Dell'Italia LJ. Plasma xanthine oxidase activity is related to increased sodium and left ventricular hypertrophy in resistant hypertension. Free Radic Biol Med 2019; 134:343-349. [PMID: 30695690 PMCID: PMC6588431 DOI: 10.1016/j.freeradbiomed.2019.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/30/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The extra-renal effects of aldosterone on left ventricular (LV) structure and function are exacerbated by increased dietary sodium in persons with hypertension. Previous studies demonstrated endothelial dysfunction and increased oxidative stress with high salt diet in normotensive salt-resistant subjects. We hypothesized that increased xanthine oxidase (XO), a product of endothelial cells, is related to 24-h urinary sodium and to LV hypertrophy and function in patients with resistant hypertension (RHTN). METHODS The study group included persons with RHTN (n = 91), defined as a blood pressure > 140/90 mmHg on ≥ 3 medications at pharmacologically effective doses. Plasma XO activity and 24-h urine were collected, and cardiac magnetic resonance imaging (MRI) was performed to assess LV function and morphology. Sixty-seven normotensive persons on no cardiovascular medications served as controls. A subset of RHTN (n = 19) received spironolactone without salt restriction for six months with follow-up XO activity measurements and MRI analyses. RESULTS XO activity was increased two-fold in RHTN vs. normal and was positively correlated with LV mass, LV diastolic function, and 24-h urinary sodium. In RHTN patients receiving spironolactone without salt restriction, LV mass decreased, but LV diastolic function and XO activity did not improve. Baseline urinary sodium was positively associated with rate of change of LV mass to volume ratio and the LV E/A ratio. CONCLUSIONS These results demonstrate a potential role of endothelium-derived oxidative stress and excess dietary salt in the pathophysiology of LV hypertrophy and diastolic dysfunction in persons with RHTN unaffected by the addition of spironolactone.
Collapse
Affiliation(s)
- Brittany Butts
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Nell Hodgson Woodruff School of Nursing, Emory University, USA
| | - David A Calhoun
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University, USA
| | - Steven G Lloyd
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Himanshu Gupta
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Birmingham Department of Veterans Affairs Medical Center, USA
| | - Krishna K Gaddam
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Inmaculada Aban
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | - Suzanne Oparil
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Paul W Sanders
- Division of Nephrology, University of Alabama at Birmingham School of Medicine, USA
| | - Rakesh Patel
- Center for Free Radical Biology and Department of Pathology, University of Alabama at Birmingham, USA
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, USA
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Birmingham Department of Veterans Affairs Medical Center, USA.
| |
Collapse
|
19
|
Crotti L, Ghidoni A, Dagradi F. Genetics of Adult and Fetal Forms of Long QT Syndrome. GENETIC CAUSES OF CARDIAC DISEASE 2019. [DOI: 10.1007/978-3-030-27371-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Patel A, Zhao J, Yue Y, Zhang K, Duan D, Lai Y. Dystrophin R16/17-syntrophin PDZ fusion protein restores sarcolemmal nNOSμ. Skelet Muscle 2018; 8:36. [PMID: 30466494 PMCID: PMC6251231 DOI: 10.1186/s13395-018-0182-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background Loss of sarcolemmal nNOSμ is a common manifestation in a wide variety of muscle diseases and contributes to the dysregulation of multiple muscle activities. Given the critical role sarcolemmal nNOSμ plays in muscle, restoration of sarcolemmal nNOSμ should be considered as an important therapeutic goal. Methods nNOSμ is anchored to the sarcolemma by dystrophin spectrin-like repeats 16 and 17 (R16/17) and the syntrophin PDZ domain (Syn PDZ). To develop a strategy that can independently restore sarcolemmal nNOSμ, we engineered an R16/17-Syn PDZ fusion construct and tested whether this construct alone is sufficient to anchor nNOSμ to the sarcolemma in three different mouse models of Duchenne muscular dystrophy (DMD). Results Membrane-associated nNOSμ is completely lost in DMD. Adeno-associated virus (AAV)-mediated delivery of the R16/17-Syn PDZ fusion construct successfully restored sarcolemmal nNOSμ in all three models. Further, nNOS restoration was independent of the dystrophin-associated protein complex. Conclusions Our results suggest that the R16/17-Syn PDZ fusion construct is sufficient to restore sarcolemmal nNOSμ in the dystrophin-null muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0182-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
21
|
Poles MZ, Bódi N, Bagyánszki M, Fekete É, Mészáros AT, Varga G, Szűcs S, Nászai A, Kiss L, Kozlov AV, Boros M, Kaszaki J. Reduction of nitrosative stress by methane: Neuroprotection through xanthine oxidoreductase inhibition in a rat model of mesenteric ischemia-reperfusion. Free Radic Biol Med 2018; 120:160-169. [PMID: 29550332 DOI: 10.1016/j.freeradbiomed.2018.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
Abstract
Our aim was to characterize the main components of the nitrosative response with quantitative changes of the nitrergic myenteric neurons in adjacent intestinal segments after transient superior mesenteric artery occlusion. We also tested the hypothesis that exogenous methane may modulate the evolution of nitroxidation by influencing xanthine oxidoreductase (XOR) activity. The microcirculatory consequences of a 50 min ischemia or ischemia-reperfusion were investigated in anesthetized rats (n = 124) inhaling normoxic air with or without 2.2% methane. XOR activities, nitrogen monoxide (NO), nitrite/nitrate (NOx), and nitrotyrosine levels were measured, together with relative nitrergic neuron ratios from duodenum, ileum and colon samples. The effects of methane on XOR were also examined in vitro. The intramural flow stopped only in the ileum during ischemia. The highest baseline XOR activity was found in the duodenum, which increased further during ischemia. NO and nitrotyrosine levels rose, and the nNOS-immunopositive neuron ratio and NOx level both dropped. Reperfusion uniformly elevated XOR activity and nitrotyrosine formation, with the highest level attained in the duodenum, where the nitrergic neuron ratio remained depressed. These alterations were eliminated in methane-treated animals, XOR activity and nitrotyrosine formation decreased in all sites, and the duodenal nitrergic neuron ratio was re-established. The inhibitory effect of methane on XOR-linked nitrate reductase activity was also demonstrated in vitro. With segment-specific microcirculatory alterations, the risk for nitrosative stress is highest in transiently hypoxic tissues with high endogenous XOR activities. The XOR-inhibitory effect of methane can reduce nitroxidation and protects the nitrergic neuron population in such conditions.
Collapse
Affiliation(s)
- Marietta Zita Poles
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - Éva Fekete
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726, Szeged, Hungary.
| | - András Tamás Mészáros
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Gabriella Varga
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Szilárd Szűcs
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Anna Nászai
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Liliána Kiss
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, Vienna 1200, Austria.
| | - Mihály Boros
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| | - József Kaszaki
- Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szokefalvi-Nagy Bela u. 6., H-6720 Szeged, Hungary.
| |
Collapse
|
22
|
Sun M, Izumi H, Shinoda Y, Fukunaga K. Neuroprotective effects of protein tyrosine phosphatase 1B inhibitor on cerebral ischemia/reperfusion in mice. Brain Res 2018; 1694:1-12. [PMID: 29705606 DOI: 10.1016/j.brainres.2018.04.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/10/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022]
Abstract
Akt (Protein kinase B, PKB), a serine/threonine kinase, plays a critical role in cell development, growth, and survival. Akt phosphorylation mediates a neuroprotective effect against ischemic injury. Recently, a protein-tyrosine phosphatase-1B (PTP1B) inhibitor (KY-226) was developed to elicit anti-diabetic and anti-obesity effects via enhancement of insulin signaling. Previously, we reported that the nonselective PTP1B inhibitor, sodium orthovanadate, rescued neurons from delayed neuronal death during brain ischemia. In this study, we confirmed the ameliorative effects of KY-226 on ischemia/reperfusion (I/R) injury using a murine model of middle cerebral artery occlusion (MCAO). ICR mice were subjected to MCAO for 2 h followed by reperfusion. Although KY-226 permeability was poor through the blood-brain barrier (BBB) of normal mice, it could penetrate through the BBB of mice after I/R insult. Intraperitoneal KY-226 administration elicited dose-dependent reductions in infarcted brain areas and improved neurological deficits. The neuroprotective effects of KY-266 were obtained when administered within 0.5 h after reperfusion. KY-226 (10 mg/kg) also restored reduced Akt phosphorylation and eNOS phosphorylation (Ser-1177) levels following I/R insult. Moreover, 10 mg/kg of KY-226 improved I/R-induced decreased extracellular signal-regulated kinase (ERK) phosphorylation. Furthermore, KY-226 attenuated the generation of reactive oxygen species (ROS) in mouse cortex. These results suggest that KY-226 may act as a novel therapeutic candidate for ischemic stroke. Activation of Akt and ERK possibly underlie the neuroprotective mechanism of KY-226.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan.
| |
Collapse
|
23
|
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
|
24
|
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]
|
25
|
Joint statement of the European Association for the Study of Obesity and the European Society of Hypertension: obesity and heart failure. J Hypertens 2017; 34:1678-88. [PMID: 27488547 DOI: 10.1097/hjh.0000000000001013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obese individuals are more likely to develop heart failure. Yet, once heart failure is established, the impact of overweight and obesity on prognosis and survival is unclear. The purpose of this joint scientific statement of the European Association for the Study of Obesity and the European Society of Hypertension is to provide an overview on the current scientific literature on obesity and heart failure in terms of prognosis, mechanisms, and clinical management implications. Moreover, the document identifies open questions that ought to be addressed. The need for more tailored weight management recommendations in heart failure will be emphasized and, in line with the emerging evidence, aims to distinguish between primary disease and secondary outcome prevention. In the primary prevention of heart failure, it appears prudent advising obese individuals to lose or achieve a healthy body weight, especially in those with risk factors such as hypertension or type 2 diabetes. However, there is no evidence from clinical trials to guide weight management in overweight or obese patients with established heart failure. Prospective clinical trials are strongly encouraged.
Collapse
|
26
|
Sagy I, Salman AA, Kezerle L, Erez O, Yoel I, Barski L. Peripartum cardiomyopathy is associated with increased uric acid concentrations: A population based study. Heart Lung 2017; 46:369-374. [DOI: 10.1016/j.hrtlng.2017.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/17/2017] [Accepted: 06/20/2017] [Indexed: 10/19/2022]
|
27
|
Hermida N, Michel L, Esfahani H, Dubois-Deruy E, Hammond J, Bouzin C, Markl A, Colin H, Steenbergen AV, De Meester C, Beauloye C, Horman S, Yin X, Mayr M, Balligand JL. Cardiac myocyte β3-adrenergic receptors prevent myocardial fibrosis by modulating oxidant stress-dependent paracrine signaling. Eur Heart J 2017; 39:888-898. [DOI: 10.1093/eurheartj/ehx366] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 06/08/2017] [Indexed: 01/08/2023] Open
Affiliation(s)
- Nerea Hermida
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Lauriane Michel
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Hrag Esfahani
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Emilie Dubois-Deruy
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Joanna Hammond
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Caroline Bouzin
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Andreas Markl
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Henri Colin
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| | - Anne Van Steenbergen
- Division of Cardiology, Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Christophe De Meester
- Division of Cardiology, Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Christophe Beauloye
- Division of Cardiology, Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Sandrine Horman
- Division of Cardiology, Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Xiaoke Yin
- King’s British Heart Foundation Center, King’s College, 125 Coldharbour Lane, SE5 9NU, London, UK
| | - Manuel Mayr
- King’s British Heart Foundation Center, King’s College, 125 Coldharbour Lane, SE5 9NU, London, UK
| | - Jean-Luc Balligand
- Department of Medicine, Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université catholique de Louvain, 52 avenue Mounier, 1200 Brussels, Belgium
| |
Collapse
|
28
|
Carnicer R, Suffredini S, Liu X, Reilly S, Simon JN, Surdo NC, Zhang YH, Lygate CA, Channon KM, Casadei B. The Subcellular Localisation of Neuronal Nitric Oxide Synthase Determines the Downstream Effects of NO on Myocardial Function. Cardiovasc Res 2017; 113:321-331. [PMID: 28158509 PMCID: PMC5408949 DOI: 10.1093/cvr/cvx002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 10/14/2016] [Accepted: 11/26/2016] [Indexed: 01/12/2023] Open
Abstract
Aims In healthy hearts, the neuronal nitric oxide synthase (nNOS) is predominantly localized to the sarcoplasmic reticulum (SR), where it regulates the ryanodine receptor Ca2+ release channel (RyR2) and phospholamban (PLB) phosphorylation, and to a lesser extent to the sarcolemmal membrane where it inhibits the L-type Ca2+ current (I Ca). However, in failing hearts, impaired relaxation and depressed inotropy are associated with a larger proportion of nNOS being localized to the sarcolemmal membrane. Whether there is a causal relationship between altered myocardial function and subcellular localization of nNOS remains to be assessed. Methods and results Adenoviruses (AdV) encoding for a human nNOS.eGFP fusion protein or eGFP were injected into the left ventricle (LV) of nNOS−/− mice. nNOS.eGFP localized to the sarcolemmal and t-tubular membrane and immunoprecipitated with syntrophin and caveolin-3 but not with RyR2. Myocardial transduction of nNOS.eGFP resulted in a significantly increased NOS activity (10-fold, P < 0.01), a 20% increase in myocardial tetrahydrobiopterin (BH4) (P < 0.05), and a 30% reduction in superoxide production (P < 0.001). LV myocytes transduced with nNOS.eGFP showed a significantly lower basal and β-adrenergic stimulated I Ca, [Ca2+]i transient amplitude and cell shortening (vs. eGFP). All differences between groups were abolished after NOS inhibition. In contrast, nNOS.eGFP had no effect on RyR nitrosylation, PLB phosphorylation or the rate of myocardial relaxation and [Ca2+]i decay. Conclusion Our findings indicate that nNOS-mediated regulation of myocardial excitation–contraction (E–C) coupling is exquisitely dependent on nNOS subcellular localization and suggests a partially adaptive role for sarcolemmal nNOS in the human failing myocardium.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6 West Wing, John Radcliffe Hospital, Headley Way, Headington, Oxford. OX3 9DU, UK
| |
Collapse
|
29
|
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]
|
30
|
Singh D, Kumar V, Singh C. IFN-γ regulates xanthine oxidase-mediated iNOS-independent oxidative stress in maneb- and paraquat-treated rat polymorphonuclear leukocytes. Mol Cell Biochem 2016; 427:133-143. [DOI: 10.1007/s11010-016-2905-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/03/2016] [Indexed: 01/25/2023]
|
31
|
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
|
32
|
Pinheiro LC, Oliveira-Paula GH, Portella RL, Guimarães DA, de Angelis CD, Tanus-Santos JE. Omeprazole impairs vascular redox biology and causes xanthine oxidoreductase-mediated endothelial dysfunction. Redox Biol 2016; 9:134-143. [PMID: 27521759 PMCID: PMC4983109 DOI: 10.1016/j.redox.2016.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 07/29/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
Proton pump inhibitors (PPIs) are widely used drugs that may increase the cardiovascular risk by mechanisms not entirely known. While PPIs increase asymmetric dimethylarginine (ADMA) levels and inhibit nitric oxide production, it is unknown whether impaired vascular redox biology resulting of increased xanthine oxidoreductase (XOR) activity mediates PPIs-induced endothelial dysfunction (ED). We examined whether increased XOR activity impairs vascular redox biology and causes ED in rats treated with omeprazole. We also examined whether omeprazole aggravates the ED found in hypertension. Treatment with omeprazole reduced endothelium-dependent aortic responses to acetylcholine without causing hypertension. However, omeprazole did not aggravate two-kidney, one-clip (2K1C) hypertension, nor hypertension-induced ED. Omeprazole and 2K1C increased vascular oxidative stress as assessed with dihydroethidium (DHE), which reacts with superoxide, and by the lucigenin chemiluminescence assay. The selective XOR inhibitor febuxostat blunted both effects induced by omeprazole. Treatment with omeprazole increased plasma ADMA concentrations, XOR activity and systemic markers of oxidative stress. Incubation of aortic rings with ADMA increased XOR activity, DHE fluorescence and lucigenin chemiluminescence signals, and febuxostat blunted these effects. Providing functional evidence that omeprazole causes ED by XOR-mediated mechanisms, we found that febuxostat blunted the ED caused by omeprazole treatment. This study shows that treatment with omeprazole impairs the vascular redox biology by XOR-mediated mechanisms leading to ED. While omeprazole did not further impair hypertension-induced ED, further studies in less severe animal models are warranted. Our findings may have major relevance, particularly to patients with cardiovascular diseases taking PPIs. Proton pump inhibitors are widely used and increase the cardiovascular risk by unknown mechanisms. Omeprazole increased vascular oxidative stress and caused endothelial dysfunction (ED). Those effects were dependent on the prooxidant enzyme xanthine oxidoreductase (XOR). XOR inhibition by the selective XOR inhibitor febuxostat blunted both effects induced by omeprazole. Omeprazole impairs the vascular redox biology by XOR-mediated mechanisms leading to ED.
Collapse
Affiliation(s)
- Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Rafael L Portella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Danielle A Guimarães
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Celio D de Angelis
- Department of Pharmacology, State University of Campinas, Campinas, SP 13081-970, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil.
| |
Collapse
|
33
|
Prognostic Significance of Hyperuricemia in Patients With Acute Heart Failure. Am J Cardiol 2016; 117:1616-1621. [PMID: 27040576 DOI: 10.1016/j.amjcard.2016.02.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 02/07/2023]
Abstract
Serum uric acid (UA) is associated with death and hospitalization in chronic heart failure (HF). However, UA in acute HF has not been well studied with respect to its relation to renal dysfunction and vascular congestion. We measured admission serum UA along with baseline variables in 281 patients with acute HF screened from the Loop Diuretics Administration and Acute Heart Failure (Diur-HF) trial. Hyperuricemia was defined as serum UA >7 mg/dl in men and >6 mg/dl in women. Chronic kidney disease (CKD) was defined as an estimated glomerular filtration rate <60 ml/min/1.73 m(2) before hospital admission. Death or HF hospitalization at 6 months was the primary outcome. The mean UA concentration was 6.4 ± 2.5 mg/dl, and 121 patients (43.1%) were classified as hyperuricemic. UA values were significantly increased in patients with CKD compared to patients without CKD (6.8 ± 2.7 vs 6.1 ± 2.1 mg/dl; p = 0.02); however, UA was not associated with the development of acute kidney injury. Patients with hyperuricemia had greater degrees of pulmonary and systemic congestion than normouricemic patients (congestion score 3.5 vs 2.1, p <0.01). Hyperuricemia was associated with higher risk of death or HF rehospitalization (univariate hazard ratio 1.46 [1.02 to 2.10]; p = 0.04, multivariate hazard ratio 1.69 [1.16 to 2.45]; p = 0.005). In conclusion, hospitalized patients with acute HF, elevated UA levels were associated with both CKD and pulmonary congestion. After controlling for potential confounders, hyperuricemia was associated with rehospitalization and death at 6 months.
Collapse
|
34
|
Human Ischemic Cardiomyopathy Shows Cardiac Nos1 Translocation and its Increased Levels are Related to Left Ventricular Performance. Sci Rep 2016; 6:24060. [PMID: 27041589 PMCID: PMC4819187 DOI: 10.1038/srep24060] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/18/2016] [Indexed: 01/09/2023] Open
Abstract
The role of nitric oxide synthase 1 (NOS1) as a major modulator of cardiac function has been extensively studied in experimental models; however, its role in human ischemic cardiomyopathy (ICM) has never been analysed. Thus, the objectives of this work are to study NOS1 and NOS-related counterparts involved in regulating physiological function of myocyte, to analyze NOS1 localisation, activity, dimerisation, and its relationship with systolic function in ICM. The study has been carried out on left ventricular tissue obtained from explanted human hearts. Here we demonstrate that the upregulation of cardiac NOS1 is not accompanied by an increase in NOS activity, due in part to the alterations found in molecules involved in the regulation of its activity. We observed partial translocation of NOS1 to the sarcolemma in ischemic hearts, and a direct relationship between its protein levels and systolic ventricular function. Our findings indicate that NOS1 may be significant in the pathophysiology of human ischemic heart disease with a preservative role in maintaining myocardial homeostasis.
Collapse
|
35
|
Singh BK, Kumar V, Chauhan AK, Dwivedi A, Singh S, Kumar A, Singh D, Patel DK, Ray RS, Jain SK, Singh C. Neuronal Nitric Oxide Synthase Negatively Regulates Zinc-Induced Nigrostriatal Dopaminergic Neurodegeneration. Mol Neurobiol 2016; 54:2685-2696. [PMID: 26995406 DOI: 10.1007/s12035-016-9857-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/11/2016] [Indexed: 12/21/2022]
Abstract
The study aimed to investigate the role of NO and neuronal NO synthase (nNOS) in Zn-induced neurodegeneration. Animals were treated with zinc sulfate (20 mg/kg), twice a week, for 2-12 weeks along with control. In a few sets, animals were also treated with/without a NO donor, sodium nitroprusside (SNP), or S-nitroso-N-acetyl penicillamine (SNAP) for 12 weeks. Moreover, human neuroblastoma (SH-SY-5Y) cells were also employed to investigate the role of nNOS in Zn-induced toxicity in in vitro in the presence/absence of nNOS inhibitor, 7-nitroindazole (7-NI). Zn caused time-dependent reduction in nitrite content and total/nNOS activity/expression. SNP/SNAP discernibly alleviated Zn-induced neurobehavioral impairments, dopaminergic neurodegeneration, tyrosine hydroxylase (TH) expression, and striatal dopamine depletion. NO donors also salvage from Zn-induced increase in lipid peroxidation (LPO), mitochondrial cytochrome c release, and caspase-3 activation. While Zn elevated LPO content, it attenuated nitrite content, nNOS activity, and glutathione level along with the expression of TH and nNOS in SH-SY-5Y cells. 7-NI further augmented Zn-induced changes in the cell viability, oxidative stress, and expression of TH and nNOS. The results obtained thus demonstrate that Zn inhibits nNOS that partially contributes to an increase in oxidative stress, which subsequently leads to the nigrostriatal dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Brajesh Kumar Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Vinod Kumar
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, 226 001, Uttar Pradesh, India
| | - Amit Kumar Chauhan
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, 226 001, Uttar Pradesh, India
| | - Ashish Dwivedi
- Phototoxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-IITR, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Shweta Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Ashutosh Kumar
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Deepali Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, 226 001, Uttar Pradesh, India
| | - Devendra Kumar Patel
- Analytical Chemistry Laboratory, Regulatory Toxicology Group, CSIR-IITR, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Ratan Singh Ray
- Phototoxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-IITR, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Swatantra Kumar Jain
- Department of Biotechnology, Jamia Hamdard Deemed University, New Delhi, 110 062, Delhi, India
| | - Chetna Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, 226 001, Uttar Pradesh, India.
| |
Collapse
|
36
|
Önen Bayram FE, Sipahi H, Acar ET, Kahveci Ulugöl R, Buran K, Akgün H. The cysteine releasing pattern of some antioxidant thiazolidine-4-carboxylic acids. Eur J Med Chem 2016; 114:337-44. [PMID: 27017266 DOI: 10.1016/j.ejmech.2016.03.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/26/2016] [Accepted: 03/07/2016] [Indexed: 12/26/2022]
Abstract
Oxidative stress that corresponds to a significant increase in free radical concentration in cells can cause considerable damage to crucial biological macromolecules if not prevented by cellular defense mechanisms. The low-molecular-weight thiol glutathione (GSH) constitutes one of the main intracellular antioxidants. It is synthesized via cysteine, an amino acid found only in limited amounts in cells because of its neurotoxicity. Thus, to ensure an efficient GSH synthesis in case of an oxidative stress, cysteine should be provided extracellularly. Yet, given its nucleophilic properties and its rapid conversion into cystine, its corresponding disulfide, cysteine presents some toxicity and therefore is usually supplemented in a prodrug approach. Here, some thiazolidine-4-carboxylic acids were synthesized and evaluated for their antioxidant properties via the DDPH and CUPRAC assays. Then, the cysteine releasing capacity of the obtained compounds was investigated in aqueous and organic medium in order to correlate the relevant antioxidant properties of the molecules with their cysteine releasing pattern. As a result, the structures' antioxidative properties were not only attributed to cysteine release but also to the thiazolidine cycle itself.
Collapse
Affiliation(s)
- F Esra Önen Bayram
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey.
| | - Hande Sipahi
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Ebru Türköz Acar
- Department of Analytical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Reyhan Kahveci Ulugöl
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Kerem Buran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Hülya Akgün
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
37
|
Simon JN, Ziberna K, Casadei B. Compromised redox homeostasis, altered nitroso-redox balance, and therapeutic possibilities in atrial fibrillation. Cardiovasc Res 2016; 109:510-8. [PMID: 26786158 PMCID: PMC4777914 DOI: 10.1093/cvr/cvw012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/15/2016] [Indexed: 12/31/2022] Open
Abstract
Although the initiation, development, and maintenance of atrial fibrillation (AF) have been linked to alterations in myocyte redox state, the field lacks a complete understanding of the impact these changes may have on cellular signalling, atrial electrophysiology, and disease progression. Recent studies demonstrate spatiotemporal changes in reactive oxygen species production shortly after the induction of AF in animal models with an uncoupling of nitric oxide synthase activity ensuing in the presence of long-standing persistent AF, ultimately leading to a major shift in nitroso–redox balance. However, it remains unclear which radical or non-radical species are primarily involved in the underlying mechanisms of AF or which proteins are targeted for redox modification. In most instances, only free radical oxygen species have been assessed; yet evidence from the redox signalling field suggests that non-radical species are more likely to regulate cellular processes. A wider appreciation for the distinction of these species and how both species may be involved in the development and maintenance of AF could impact treatment strategies. In this review, we summarize how redox second-messenger systems are regulated and discuss the recent evidence for alterations in redox regulation in the atrial myocardium in the presence of AF, while identifying some critical missing links. We also examine studies looking at antioxidants for the prevention and treatment of AF and propose alternative redox targets that may serve as superior therapeutic options for the treatment of AF.
Collapse
Affiliation(s)
- Jillian N Simon
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Klemen Ziberna
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Jang JH, Kang MJ, Ko GP, Kim SJ, Yi EC, Zhang YH. Identification of a novel splice variant of neuronal nitric oxide synthase, nNOSβ, in myofilament fraction of murine cardiomyocytes. Nitric Oxide 2015; 50:20-27. [PMID: 26271450 DOI: 10.1016/j.niox.2015.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/07/2015] [Accepted: 07/31/2015] [Indexed: 12/30/2022]
Abstract
Splice variant forms of neuronal nitric oxide synthase (nNOS or NOS1), nNOSα and nNOSμ, are well established to be functionally expressed in discrete compartments in cardiomyocytes (e.g. sarcoplasmic reticulum, SR, caveolae in plasma membrane or mitochondria). So far, whether nNOS is expressed in myofilament fraction of cardiomyocytes and the splice variant form of nNOS are unknown. Immunoblotting results using two nNOS specific antibodies (BD Transduction Laboratories aa 1095-1289 and Santa Cruz Biotechnology aa 2-300) clearly demonstrated that nNOS was abundantly expressed in myofilament-enriched fraction of cardiomyocytes. Whilst the molecular weight of nNOS in membrane/cytosol fractions was ∼165 kDa, nNOS in myofilament was below 140 kDa, suggesting that the predominant splice variant of nNOS in myofilament is nNOSβ. RT-PCR results confirmed the expressions of both nNOSα and nNOSβ mRNAs in rat cardiomyocytes. Similarly, immunoprecipitation experiments using myofilament lysates of cardiomyocytes identified nNOS with low molecular weight (M.W. ∼140 kDa), confirming nNOSβ. Intriguingly, all three splice variants of nNOS were undetectable in the lysates of cardiomyocytes (including myofilament fractions) from nNOS-/- mice (which lacks nNOSα/μ). Furthermore, nNOSβ expression in myofilament of cardiomyocytes was not different in hypertensive rats compared to the level expressed in sham. iTRAQ-based quantitative proteomics analysis revealed that nNOS regulates phosphorylations of ∼20 proteins in cardiac myofilaments. Collectively, we provide direct evidence that different splice variants of nNOS are expressed in myofilament and membrane/cytosol fractions of cardiomyocytes. Discrete expressions of various splice variants in different compartments of cardiomyocytes suggest diverse roles nNOS play in healthy and diseased heart.
Collapse
Affiliation(s)
- Ji Hyun Jang
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Min Jueng Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine and College of Pharmacy, Seoul National University, Republic of Korea
| | - Gwang Pyo Ko
- Department of Environmental Health, Graduate School of Public Health, Seoul National University, Kwanak-ro 1, Kwanak-gu, Seoul, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine and College of Pharmacy, Seoul National University, Republic of Korea.
| | - Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, Republic of Korea; Yanbian University Hospital, Yanji, Jilin Province, China; Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
39
|
Nash KM, Ahmed S. Nanomedicine in the ROS-mediated pathophysiology: Applications and clinical advances. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:2033-40. [PMID: 26255114 DOI: 10.1016/j.nano.2015.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/23/2015] [Accepted: 07/02/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Reactive oxygen species (ROS) are important in regulating normal cell physiological functions, but when produced in excess lead to the augmented pathogenesis of various diseases. Among these, ischemia reperfusion injury, Alzheimer's disease and rheumatoid arthritis are particularly important. Since ROS can be counteracted by a variety of antioxidants, natural and synthetic antioxidants have been developed. However, due to the ubiquitous production of ROS in living systems, poor in vivo efficiency of these agents and lack of target specificity, the current clinical modalities to treat oxidative stress damage are limited. Advances in the developing field of nanomedicine have yielded nanoparticles that can prolong antioxidant activity, and target specificity of these agents. This article reviews recent advances in antioxidant nanoparticles and their applications to manage oxidative stress-mediated diseases. FROM THE CLINICAL EDITOR Production of reactive oxygen species (ROS) is a purely physiological process in many disease conditions. However, excessive and uncontrolled production will lead to oxidative stress and further tissue damage. Advances in nanomedicine have provided many novel strategies to try to combat and counteract ROS. In this review article, the authors comprehensively highlighted the current status and future developments in using nanotechnology for providing novel therapeutic options in this field.
Collapse
Affiliation(s)
- Kevin M Nash
- Department of Pharmacology, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Salahuddin Ahmed
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA, USA.
| |
Collapse
|
40
|
NOS1 induces NADPH oxidases and impairs contraction kinetics in aged murine ventricular myocytes. Basic Res Cardiol 2015; 110:506. [PMID: 26173391 DOI: 10.1007/s00395-015-0506-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/20/2015] [Accepted: 07/07/2015] [Indexed: 01/08/2023]
Abstract
Nitric oxide (NO) modulates calcium transients and contraction of cardiomyocytes. However, it is largely unknown whether NO contributes also to alterations in the contractile function of cardiomyocytes during aging. Therefore, we analyzed the putative role of nitric oxide synthases and NO for the age-related alterations of cardiomyocyte contraction. We used C57BL/6 mice, nitric oxide synthase 1 (NOS1)-deficient mice (NOS1(-/-)) and mice with cardiomyocyte-specific NOS1-overexpression to analyze contractions, calcium transients (Indo-1 fluorescence), acto-myosin ATPase activity (malachite green assay), NADPH oxidase activity (lucigenin chemiluminescence) of isolated ventricular myocytes and cardiac gene expression (Western blots, qPCR). In C57BL/6 mice, cardiac expression of NOS1 was upregulated by aging. Since we found a negative regulation of NOS1 expression by cAMP in isolated cardiomyocytes, we suggest that reduced efficacy of β-adrenergic signaling that is evident in aged hearts promotes upregulation of NOS1. Shortening and relengthening of cardiomyocytes from aged C57BL/6 mice were decelerated, but were normalized by pharmacological inhibition of NOS1/NO. Cardiomyocytes from NOS1(-/-) mice displayed no age-related changes in contraction, calcium transients or acto-myosin ATPase activity. Aging increased cardiac expression of NADPH oxidase subunits NOX2 and NOX4 in C57BL/6 mice, but not in NOS1(-/-) mice. Similarly, cardiac expression of NOX2 and NOX4 was upregulated in a murine model with cardiomyocyte-specific overexpression of NOS1. We conclude that age-dependently upregulated NOS1, putatively via reduced efficacy of β-adrenergic signaling, induces NADPH oxidases. By increasing nitrosative and oxidative stress, both enzyme systems act synergistically to decelerate contraction of aged cardiomyocytes.
Collapse
|
41
|
Kotsuruba AV, Kopjak BS, Sagach VF, Spivak NJ. [Nanocerium restores the erythrocytes stability to acid hemolysis by inhibition of oxygen and nitrogen reactive species in old rats]. ACTA ACUST UNITED AC 2015; 61:3-9. [PMID: 26040029 DOI: 10.15407/fz61.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In experiments in vivo the effect of nanocerium (cerium oxide nanoparticles) on the stability of red blood cells to acid hemolysis, levels of both ROS and RNS generation and H2S pools in plasma and erythrocytes of old rats were investigated. In red blood cells of old rats the proton penetration into the matrix of erythrocytes showed a significant raising and the fate of labile "aging" erythrocytes in old animals compared with adult were up- regulated. These phenomena paralleled with significant up-regulation of ROS and RNS generation. Introduction for 14 days per os to old rats 0.1 mg/kg of nanocerium fully restored resistance of erythrocytes to acid hemolysis by ROS and RNS in both plasma and erythrocytes reduction. Nanocerium decreased the erythrocytes and, conversely, significantly increased the plasma's pools of H2S.
Collapse
|
42
|
Kar R, Kellogg DL, Roman LJ. Oxidative stress induces phosphorylation of neuronal NOS in cardiomyocytes through AMP-activated protein kinase (AMPK). Biochem Biophys Res Commun 2015; 459:393-7. [PMID: 25732085 DOI: 10.1016/j.bbrc.2015.02.113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 02/19/2015] [Indexed: 02/05/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) plays a critical role in regulating cardiomyocyte function. nNOS was reported to decrease superoxide production in the myocardium by inhibiting the function of xanthine oxidoreductase. However, the effect of oxidative stress on nNOS in cardiomyocytes has not been determined. We report here that brief exposure of HL-1 cardiomyocytes to hydrogen peroxide (H2O2) induces phosphorylation of nNOS at serine 1412. This increase in phosphorylation was concomitant with increased nitric oxide (NO) production. Prolonged exposure to the oxidant, however, resulted in decreased expression of the protein. H2O2 treatment for short periods also stimulated phosphorylation of AKT and AMPK. H2O2-induced phosphorylation of nNOS was reduced when AMPK activity was inhibited by compound C, suggesting that AMPK is a mediator of oxidative stress-induced phosphorylation of nNOS. However, inhibition of AKT activity by the pan AKT inhibitor, AKTi, had no effect on nNOS phosphorylation caused by H2O2. These data demonstrate the novel regulation of nNOS phosphorylation and expression by oxidative stress.
Collapse
Affiliation(s)
- Rekha Kar
- Department of Biochemistry, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA.
| | - Dean L Kellogg
- Department of Biochemistry, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA.
| | - Linda J Roman
- Department of Biochemistry, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA.
| |
Collapse
|
43
|
Roof SR, Ho HT, Little SC, Ostler JE, Brundage EA, Periasamy M, Villamena FA, Györke S, Biesiadecki BJ, Heymes C, Houser SR, Davis JP, Ziolo MT. Obligatory role of neuronal nitric oxide synthase in the heart's antioxidant adaptation with exercise. J Mol Cell Cardiol 2015; 81:54-61. [PMID: 25595735 DOI: 10.1016/j.yjmcc.2015.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 12/18/2014] [Accepted: 01/06/2015] [Indexed: 02/07/2023]
Abstract
Excessive oxidative stress in the heart results in contractile dysfunction. While antioxidant therapies have been a disappointment clinically, exercise has shown beneficial results, in part by reducing oxidative stress. We have previously shown that neuronal nitric oxide synthase (nNOS) is essential for cardioprotective adaptations caused by exercise. We hypothesize that part of the cardioprotective role of nNOS is via the augmentation of the antioxidant defense with exercise by positively shifting the nitroso-redox balance. Our results show that nNOS is indispensable for the augmented anti-oxidant defense with exercise. Furthermore, exercise training of nNOS knockout mice resulted in a negative shift in the nitroso-redox balance resulting in contractile dysfunction. Remarkably, overexpressing nNOS (conditional cardiac-specific nNOS overexpression) was able to mimic exercise by increasing VO2max. This study demonstrates that exercise results in a positive shift in the nitroso-redox balance that is nNOS-dependent. Thus, targeting nNOS signaling may mimic the beneficial effects of exercise by combating oxidative stress and may be a viable treatment strategy for heart disease.
Collapse
Affiliation(s)
- Steve R Roof
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Hsiang-Ting Ho
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sean C Little
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Joseph E Ostler
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Elizabeth A Brundage
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Muthu Periasamy
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Frederick A Villamena
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sandor Györke
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Brandon J Biesiadecki
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, Toulouse, France
| | - Steven R Houser
- Department of Physiology, Cardiovascular Research Center, Temple University, Philadelphia, PA, USA
| | - Jonathan P Davis
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mark T Ziolo
- Department of Physiology & Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
44
|
Dulce RA, Mayo V, Rangel EB, Balkan W, Hare JM. Interaction between neuronal nitric oxide synthase signaling and temperature influences sarcoplasmic reticulum calcium leak: role of nitroso-redox balance. Circ Res 2015; 116:46-55. [PMID: 25326127 PMCID: PMC4282621 DOI: 10.1161/circresaha.116.305172] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 10/16/2014] [Indexed: 11/16/2022]
Abstract
RATIONALE Although nitric oxide (NO) signaling modulates cardiac function and excitation-contraction coupling, opposing results because of inconsistent experimental conditions, particularly with respect to temperature, confound the ability to elucidate NO signaling pathways. Here, we show that temperature significantly modulates NO effects. OBJECTIVE To test the hypothesis that temperature profoundly affects nitroso-redox equilibrium, thereby affecting sarcoplasmic reticulum (SR) calcium (Ca(2+)) leak. METHODS AND RESULTS We measured SR Ca(2+) leak in cardiomyocytes from wild-type (WT), NO/redox imbalance (neuronal nitric oxide synthase-deficient mice-1 [NOS1(-/-)]), and hyper S-nitrosoglutathione reductase-deficient (GSNOR(-/-)) mice. In WT cardiomyocytes, SR Ca(2+) leak increased because temperature decreased from 37°C to 23°C, whereas in NOS1(-/-) cells, the leak suddenly increased when the temperature surpassed 30°C. GSNOR(-/-) cardiomyocytes exhibited low leak throughout the temperature range. Exogenously added NO had a biphasic effect on NOS1(-/-) cardiomyocytes; reducing leak at 37°C but increasing it at subphysiological temperatures. Oxypurinol and Tempol diminished the leak in NOS1(-/-) cardiomyocytes. Cooling from 37°C to 23°C increased reactive oxygen species generation in WT but decreased it in NOS1(-/-) cardiomyocytes. Oxypurinol further reduced reactive oxygen species generation. At 23°C in WT cells, leak was decreased by tetrahydrobiopterin, an essential NOS cofactor. Cooling significantly increased SR Ca(2+) content in NOS1(-/-) cells but had no effect in WT or GSNOR(-/-). CONCLUSIONS Ca(2+) leak and temperature are normally inversely proportional, whereas NOS1 deficiency reverses this effect, increasing leak and elevating reactive oxygen species production because temperature increases. Reduced denitrosylation (GSNOR deficiency) eliminates the temperature dependence of leak. Thus, temperature regulates the balance between NO and reactive oxygen species which in turn has a major effect on SR Ca(2+).
Collapse
Affiliation(s)
- Raul A Dulce
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Vera Mayo
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Erika B Rangel
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Wayne Balkan
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL.
| |
Collapse
|
45
|
Plummer BN, Liu H, Wan X, Deschênes I, Laurita KR. Targeted antioxidant treatment decreases cardiac alternans associated with chronic myocardial infarction. Circ Arrhythm Electrophysiol 2014; 8:165-73. [PMID: 25491741 DOI: 10.1161/circep.114.001789] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND In myocardial infarction (MI), repolarization alternans is a potent arrhythmia substrate that has been linked to Ca2+ cycling proteins, such as sarcoplasmic reticulum Ca2+ ATPase (SERCA2a), located in the sarcoplasmic reticulum. MI is also associated with oxidative stress and increased xanthine oxidase (XO) activity, an important source of reactive oxygen species (ROS) in the sarcoplasmic reticulum that may reduce SERCA2a function. We hypothesize that in chronic MI, XO-mediated oxidation of SERCA2a is a mechanism of cardiac alternans. METHODS AND RESULTS Male Lewis rats underwent ligation of the left anterior descending coronary artery (n=54) or sham procedure (n=24). At 4 weeks, optical mapping of intracellular Ca2+ and ROS was performed. ECG T-wave alternans (ECG ALT) and Ca2+ transient alternans (Ca2+ALT) were induced by rapid pacing (300-120 ms) before and after the XO inhibitor allopurinol (ALLO, 50 µmol/L). In MI, ECG ALT (2.32±0.41%) and Ca2+ ALT (22.3±4.5%) were significantly greater compared with sham (0.18±0.08%, P<0.001; 0.79±0.32%, P<0.01). Additionally, ROS was increased by 137% (P<0.01) and oxidation of SERCA2a by 30% (P<0.05) in MI compared with sham. Treatment with ALLO significantly decreased ECG ALT (-77±9%, P<0.05) and Ca2+ ALT (-56±7%, P<0.05) and, importantly, reduced ROS (-65%, P<0.01) and oxidation of SERCA2a (-38%, P<0.05). CaMKII inhibition and general antioxidant treatment had no effect on ECG ALT and Ca2+ ALT. CONCLUSIONS These results demonstrate, for the first time, that in MI, increased ROS from XO is a significant cause of repolarization alternans. This suggests that targeting XO ROS production may be effective at preventing arrhythmia substrates in chronic MI.
Collapse
Affiliation(s)
- Bradley N Plummer
- From The Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus (B.N.P., H.L., X.W., I.D., K.R.L.), and Department of Biomedical Engineering (B.N.P., I.D., K.R.L.), Case Western Reserve University, Cleveland, OH
| | - Haiyan Liu
- From The Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus (B.N.P., H.L., X.W., I.D., K.R.L.), and Department of Biomedical Engineering (B.N.P., I.D., K.R.L.), Case Western Reserve University, Cleveland, OH
| | - Xiaoping Wan
- From The Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus (B.N.P., H.L., X.W., I.D., K.R.L.), and Department of Biomedical Engineering (B.N.P., I.D., K.R.L.), Case Western Reserve University, Cleveland, OH
| | - Isabelle Deschênes
- From The Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus (B.N.P., H.L., X.W., I.D., K.R.L.), and Department of Biomedical Engineering (B.N.P., I.D., K.R.L.), Case Western Reserve University, Cleveland, OH
| | - Kenneth R Laurita
- From The Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus (B.N.P., H.L., X.W., I.D., K.R.L.), and Department of Biomedical Engineering (B.N.P., I.D., K.R.L.), Case Western Reserve University, Cleveland, OH.
| |
Collapse
|
46
|
Treuer AV, Gonzalez DR. Nitric oxide synthases, S-nitrosylation and cardiovascular health: from molecular mechanisms to therapeutic opportunities (review). Mol Med Rep 2014; 11:1555-65. [PMID: 25405382 PMCID: PMC4270315 DOI: 10.3892/mmr.2014.2968] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 08/05/2014] [Indexed: 12/13/2022] Open
Abstract
The understanding of nitric oxide (NO) signaling has grown substantially since the identification of endothelial derived relaxing factor (EDRF). NO has emerged as a ubiquitous signaling molecule involved in diverse physiological and pathological processes. Perhaps the most significant function, independent of EDRF, is that of NO signaling mediated locally in signaling modules rather than relying upon diffusion. In this context, NO modulates protein function via direct post-translational modification of cysteine residues. This review explores NO signaling and related reactive nitrogen species involved in the regulation of the cardiovascular system. A critical concept in the understanding of NO signaling is that of the nitroso-redox balance. Reactive nitrogen species bioactivity is fundamentally linked to the production of reactive oxygen species. This interaction occurs at the chemical, enzymatic and signaling effector levels. Furthermore, the nitroso-redox equilibrium is in a delicate balance, involving the cross-talk between NO and oxygen-derived species signaling systems, including NADPH oxidases and xanthine oxidase.
Collapse
Affiliation(s)
- Adriana V Treuer
- Laboratory of Organic Synthesis, Institute of Chemistry of Natural Resources, University of Talca, Talca 3460000, Chile
| | - Daniel R Gonzalez
- Department of Biomedical Basic Sciences, School of Health Sciences, University of Talca, Talca 3460000, Chile
| |
Collapse
|
47
|
El-Mas MM, Abdel-Rahman AA. Endothelial and neuronal nitric oxide synthases variably modulate the oestrogen-mediated control of blood pressure and cardiovascular autonomic control. Clin Exp Pharmacol Physiol 2014; 41:246-54. [PMID: 24471817 DOI: 10.1111/1440-1681.12207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/09/2013] [Accepted: 12/18/2013] [Indexed: 11/27/2022]
Abstract
1. We have shown previously that long-term oestrogen (E2) replacement lowers blood pressure (BP) and improves cardiovascular autonomic control in ovariectomized (OVX) rats. In the present study, we investigated whether constitutive and/or inducible (i) nitric oxide synthase (NOS) modulate these E2 effects. 2. We evaluated changes in BP, myocardial contractility index (dP/dtmax ) and power spectral indices of haemodynamic variability following selective inhibition of endothelial (e) NOS with N(5)-(1-iminoethyl)-L-ornithine (L-NIO), neuronal (n) NOS with N(ω)-propyl-L-arginine (NPLA) or iNOS with 1400W in telemetered OVX rats treated for 16 weeks with (OVXE2) or without (control; OVXC) E2. 3. The OVXE2 rats exhibited: (i) reduced BP and increased dP/dtmax ; (ii) cardiac parasympathetic dominance, as reflected by the reduced low-frequency (LF; 0.25-0.75 Hz)/high-frequency (HF; 0.75-3 Hz) ratio of interbeat intervals (IBI(LF/HF)); and (iii) reduced LF oscillations of systolic BP, suggesting a reduced vasomotor sympathetic tone. Inhibition of eNOS (L-NIO; 20 mg/kg, i.p.) elicited a shorter-lived pressor response in OVXE2 than OVXC, rats along with reductions in dP/dtmax and increases in the spectral index of spontaneous baroreflex sensitivity (index α). Treatment with 1 mg/kg, i.p., NPLA reduced BP and increased the IBI(LF/HF) ratio in OVXE2 but not OVXC rats. The iNOS inhibitor 1400W (5 mg/kg, i.p.) caused no haemodynamic changes in OVXC or OVXE2 rats. 4. Overall, constitutive NOS isoforms exert restraining tonic modulatory BP effects that encompass eNOS-mediated reductions and nNOS-mediated elevations in BP in OVXE2 rats. Baroreflex facilitation and dP/dtmax reductions may account for the shorter pressor action of L-NIO in E2-treated, compared with untreated, OVX rats.
Collapse
Affiliation(s)
- Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, School of Medicine, East Carolina University, Greenville, NC, USA
| | | |
Collapse
|
48
|
Ziolo MT, Houser SR. Abnormal Ca(2+) cycling in failing ventricular myocytes: role of NOS1-mediated nitroso-redox balance. Antioxid Redox Signal 2014; 21:2044-59. [PMID: 24801117 PMCID: PMC4208612 DOI: 10.1089/ars.2014.5873] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Heart failure (HF) results from poor heart function and is the leading cause of death in Western society. Abnormalities of Ca(2+) handling at the level of the ventricular myocyte are largely responsible for much of the poor heart function. RECENT ADVANCES Although studies have unraveled numerous mechanisms for the abnormal Ca(2+) handling, investigations over the past decade have indicated that much of the contractile dysfunction and adverse remodeling that occurs in HF involves oxidative stress. CRITICAL ISSUES Regrettably, antioxidant therapy has been an immense disappointment in clinical trials. Thus, redox signaling is being reassessed to elucidate why antioxidants failed to treat HF. FUTURE DIRECTIONS A recently identified aspect of redox signaling (specifically the superoxide anion radical) is its interaction with nitric oxide, known as the nitroso-redox balance. There is a large nitroso-redox imbalance with HF, and we suggest that correcting this imbalance may be able to restore myocyte contraction and improve heart function.
Collapse
Affiliation(s)
- Mark T Ziolo
- 1 Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University , Columbus, Ohio
| | | |
Collapse
|
49
|
Simon JN, Duglan D, Casadei B, Carnicer R. Nitric oxide synthase regulation of cardiac excitation-contraction coupling in health and disease. J Mol Cell Cardiol 2014; 73:80-91. [PMID: 24631761 DOI: 10.1016/j.yjmcc.2014.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 02/07/2023]
Abstract
Significant advances in our understanding of the ability of nitric oxide synthases (NOS) to modulate cardiac function have provided key insights into the role NOS play in the regulation of excitation-contraction (EC) coupling in health and disease. Through both cGMP-dependent and cGMP-independent (e.g. S-nitrosylation) mechanisms, NOS have the ability to alter intracellular Ca(2+) handling and the myofilament response to Ca(2+), thereby impacting the systolic and diastolic performance of the myocardium. Findings from experiments using nitric oxide (NO) donors and NOS inhibition or gene deletion clearly implicate dysfunctional NOS as a critical contributor to many cardiovascular disease states. However, studies to date have only partially addressed NOS isoform-specific effects and, more importantly, how subcellular localization of NOS influences ion channels involved in myocardial EC coupling and excitability. In this review, we focus on the contribution of each NOS isoform to cardiac dysfunction and on the role of uncoupled NOS activity in common cardiac disease states, including heart failure, diabetic cardiomyopathy, ischemia/reperfusion injury and atrial fibrillation. We also review evidence that clearly indicates the importance of NO in cardioprotection. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Jillian N Simon
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Drew Duglan
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Ricardo Carnicer
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
50
|
Lai Y, Zhao J, Yue Y, Wasala NB, Duan D. Partial restoration of cardiac function with ΔPDZ nNOS in aged mdx model of Duchenne cardiomyopathy. Hum Mol Genet 2014; 23:3189-99. [PMID: 24463882 DOI: 10.1093/hmg/ddu029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transgenic gene deletion/over-expression studies have established the cardioprotective role of neuronal nitric oxide synthase (nNOS). However, it remains unclear whether nNOS-mediated heart protection can be translated to gene therapy. In this study, we generated an adeno-associated virus (AAV) nNOS vector and tested its therapeutic efficacy in the aged mdx model of Duchenne cardiomyopathy. A PDZ domain-deleted nNOS gene (ΔPDZ nNOS) was packaged into tyrosine mutant AAV-9 and delivered to the heart of ~14-month-old female mdx mice, a phenotypic model of Duchenne cardiomyopathy. Seven months later, we observed robust nNOS expression in the myocardium. Supra-physiological ΔPDZ nNOS expression significantly reduced myocardial fibrosis, inflammation and apoptosis. Importantly, electrocardiography and left ventricular hemodynamics were significantly improved in treated mice. Additional studies revealed increased phosphorylation of phospholamban and p70S6K. Collectively, we have demonstrated the therapeutic efficacy of the AAV ΔPDZ nNOS vector in a symptomatic Duchenne cardiomyopathy model. Our results suggest that the cardioprotective role of ΔPDZ nNOS is likely through reduced apoptosis, enhanced phospholamban phosphorylation and improved Akt/mTOR/p70S6K signaling. Our study has opened the door to treat Duchenne cardiomyopathy with ΔPDZ nNOS gene transfer.
Collapse
Affiliation(s)
- Yi Lai
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| |
Collapse
|