1
|
Leinhos L, Robinson P, Poloni G, Broadway-Stringer S, Beglov J, Lokman AB, Douglas G, Nuthay S, Fonseka O, Schmid M, Singer E, Hooper C, Thomson K, Bagnall RD, Ingles J, Semsarian C, Ormondroyd E, Toepfer CN, Davies B, Redwood C, Watkins H, Gehmlich K. An ALPK3 truncation variant causing autosomal dominant hypertrophic cardiomyopathy is partially rescued by mavacamten. Sci Rep 2025; 15:10090. [PMID: 40128237 PMCID: PMC11933305 DOI: 10.1038/s41598-025-94371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
The ALPK3 gene encodes alpha-protein kinase 3, a cardiac pseudo-kinase of unknown function. Heterozygous truncating variants (ALPK3tv) can cause dominant adult-onset hypertrophic cardiomyopathy (HCM). Here we confirm an excess of ALPK3tv in sarcomere-gene negative HCM patients. Moreover, we generated a novel knock-in mouse model carrying an ALPK3tv (K201X). Homozygous animals displayed hypertrophy and systolic dysfunction. Heterozygous animals demonstrated no obvious baseline; however, they had an aggravated hypertrophic response upon chronic adrenergic challenge. Isolated, unloaded cardiomyocytes from heterozygous and homozygous mice showed reduced basal sarcomere length with prolonged relaxation, whilst calcium transients showed increased diastolic calcium levels. Protein kinase A-mediated phosphorylation, including that of cardiac troponin I, was significantly decreased. In agreement with the cellular HCM phenotype, reduced ratios of myosin heads in the super-relaxed state were measured. Contractile and calcium handling defects were partly corrected by treatment with mavacamten, a novel myosin inhibitor. For the first time with a non-sarcomere HCM variant, we have demonstrated hallmark changes in cardiac contractility and calcium handling. Mavacamten is able to partially rescue the cellular phenotype, hence could be beneficial to HCM patients with ALPK3tv. Moreover, our data points at a potential role of ALPK3 as a modulator of protein kinase A signalling.
Collapse
Affiliation(s)
- Lisa Leinhos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Giulia Poloni
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Sophie Broadway-Stringer
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, Institute of Biomedical Research (IBR) room 229, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Julia Beglov
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Adam B Lokman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Sajjad Nuthay
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, Institute of Biomedical Research (IBR) room 229, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Oveena Fonseka
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Manuel Schmid
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Evie Singer
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, Institute of Biomedical Research (IBR) room 229, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Charlotte Hooper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Kate Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Richard D Bagnall
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
- Faculty of Medicine and Heath, The University of Sydney, Sydney, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research and University of New South Wales, Sydney, Australia
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
- Faculty of Medicine and Heath, The University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Elizabeth Ormondroyd
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Christopher N Toepfer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, Transgenic Core, University of Oxford, Oxford, UK
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK.
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, Institute of Biomedical Research (IBR) room 229, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
2
|
Liang Y, Ornatowski W, Lu Q, Sun X, Yegambaram M, Feng A, Dong Y, Aggarwal S, Unwalla HJ, Fineman JR, Black SM, Wang T. Chloroquine Restores eNOS Signaling in Shunt Endothelial Cells via Inhibiting eNOS Uncoupling. Int J Mol Sci 2025; 26:1352. [PMID: 39941119 PMCID: PMC11818845 DOI: 10.3390/ijms26031352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by increased lung vascular stiffness and impaired vessel relaxation, primarily due to reduced nitric oxide (NO) production in endothelial cells. Recent studies indicate that chloroquine, an autophagy inhibitor, may help lower pulmonary arterial pressure and enhance lung vascular function. This study investigates the mechanisms underlying the chloroquine-mediated restoration of NO bioavailability in endothelial cells derived from aortopulmonary shunt lambs, a relevant model for congenital heart defect (CHD)-associated PAH. We found that NO production was significantly reduced in shunt pulmonary artery endothelial cells (PAECs), attributable to decreased levels of tetrahydrobiopterin (BH4) and diminished expression of GTP cyclohydrolase 1 (GCH1), despite a slight increase in endothelial nitric oxide synthase (eNOS) levels. Chloroquine robustly restored endothelial NO production, which correlated with increased BH4 levels and restored GCH1 expression. The mechanistically upregulated carboxyl terminus of Hsp70-interacting protein (CHIP) in shunt PAECs is responsible for heightened GCH1 degradation, and chloroquine disrupted the assembly of the GCH1-HSP70-CHIP complex to preserve cellular GCH1. Similarly, another autophagy inhibitor, bafilomycin A1, demonstrated comparable effects. These findings suggest that autophagy inhibition can effectively enhance NO synthesis in endothelial cells experiencing depleted NO bioavailability, presenting a potential therapeutic strategy for managing PAH.
Collapse
Affiliation(s)
- Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Wojciech Ornatowski
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
3
|
Xia W, Zhang M, Liu C, Wang S, Xu A, Xia Z, Pang L, Cai Y. Exploring the therapeutic potential of tetrahydrobiopterin for heart failure with preserved ejection fraction: A path forward. Life Sci 2024; 345:122594. [PMID: 38537900 DOI: 10.1016/j.lfs.2024.122594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/10/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
A large number of patients are affected by classical heart failure (HF) symptomatology with preserved ejection fraction (HFpEF) and multiorgan syndrome. Due to high morbidity and mortality rate, hospitalization and mortality remain serious socioeconomic problems, while the lack of effective pharmacological or device treatment means that HFpEF presents a major unmet medical need. Evidence from clinical and basic studies demonstrates that systemic inflammation, increased oxidative stress, and impaired mitochondrial function are the common pathological mechanisms in HFpEF. Tetrahydrobiopterin (BH4), beyond being an endogenous co-factor for catalyzing the conversion of some essential biomolecules, has the capacity to prevent systemic inflammation, enhance antioxidant resistance, and modulate mitochondrial energy production. Therefore, BH4 has emerged in the last decade as a promising agent to prevent or reverse the progression of disorders such as cardiovascular disease. In this review, we cover the clinical progress and limitations of using downstream targets of nitric oxide (NO) through NO donors, soluble guanylate cyclase activators, phosphodiesterase inhibitors, and sodium-glucose co-transporter 2 inhibitors in treating cardiovascular diseases, including HFpEF. We discuss the use of BH4 in association with HFpEF, providing new evidence for its potential use as a pharmacological option for treating HFpEF.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Miao Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Guangdong, China
| | - Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
4
|
Robinson P, Sparrow AJ, Psaras Y, Steeples V, Simon JN, Broyles CN, Chang YF, Brook FA, Wang YJ, Blease A, Zhang X, Abassi YA, Geeves MA, Toepfer CN, Watkins H, Redwood C, Daniels MJ. Comparing the effects of chemical Ca 2+ dyes and R-GECO on contractility and Ca 2+ transients in adult and human iPSC cardiomyocytes. J Mol Cell Cardiol 2023; 180:44-57. [PMID: 37127261 PMCID: PMC10659987 DOI: 10.1016/j.yjmcc.2023.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/13/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
We compared commonly used BAPTA-derived chemical Ca2+ dyes (fura2, Fluo-4, and Rhod-2) with a newer genetically encoded indicator (R-GECO) in single cell models of the heart. We assessed their performance and effects on cardiomyocyte contractility, determining fluorescent signal-to-noise ratios and sarcomere shortening in primary ventricular myocytes from adult mouse and guinea pig, and in human iPSC-derived cardiomyocytes. Chemical Ca2+ dyes displayed dose-dependent contractile impairment in all cell types, and we observed a negative correlation between contraction and fluorescence signal-to-noise ratio, particularly for fura2 and Fluo-4. R-GECO had no effect on sarcomere shortening. BAPTA-based dyes, but not R-GECO, inhibited in vitro acto-myosin ATPase activity. The presence of fura2 accentuated or diminished changes in contractility and Ca2+ handling caused by small molecule modulators of contractility and intracellular ionic homeostasis (mavacamten, levosimendan, and flecainide), but this was not observed when using R-GECO in adult guinea pig left ventricular cardiomyocytes. Ca2+ handling studies are necessary for cardiotoxicity assessments of small molecules intended for clinical use. Caution should be exercised when interpreting small molecule studies assessing contractile effects and Ca2+ transients derived from BAPTA-like chemical Ca2+ dyes in cellular assays, a common platform for cardiac toxicology testing and mechanistic investigation of cardiac disease physiology and treatment.
Collapse
Affiliation(s)
- Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK.
| | - Alexander J Sparrow
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Yiangos Psaras
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Violetta Steeples
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Connor N Broyles
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Yu-Fen Chang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Frances A Brook
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Ying-Jie Wang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Andrew Blease
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Xiaoyu Zhang
- Agilent Biosciences, Inc., San Diego, CA 92121, USA
| | | | | | - Christopher N Toepfer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK; Department of Cardiology, Oxford University NHS Hospitals Trust, Oxford, UK
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Matthew J Daniels
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; BHF Centre of Research Excellence, University of Oxford, Oxford, UK; Department of Cardiology, Oxford University NHS Hospitals Trust, Oxford, UK; Department of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
5
|
Chuaiphichai S, Chu SM, Carnicer R, Kelly M, Bendall JK, Simon JN, Douglas G, Crabtree MJ, Casadei B, Channon KM. Endothelial cell-specific roles for tetrahydrobiopterin in myocardial function, cardiac hypertrophy, and response to myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2023; 324:H430-H442. [PMID: 36735402 PMCID: PMC9988535 DOI: 10.1152/ajpheart.00562.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/15/2022] [Accepted: 12/31/2022] [Indexed: 02/04/2023]
Abstract
The cofactor tetrahydrobiopterin (BH4) is a critical regulator of nitric oxide synthase (NOS) function and redox signaling, with reduced BH4 implicated in multiple cardiovascular disease states. In the myocardium, augmentation of BH4 levels can impact on cardiomyocyte function, preventing hypertrophy and heart failure. However, the specific role of endothelial cell BH4 biosynthesis in the coronary circulation and its role in cardiac function and the response to ischemia has yet to be elucidated. Endothelial cell-specific Gch1 knockout mice were generated by crossing Gch1fl/fl with Tie2cre mice, generating Gch1fl/flTie2cre mice and littermate controls. GTP cyclohydrolase protein and BH4 levels were reduced in heart tissues from Gch1fl/flTie2cre mice, localized to endothelial cells, with normal cardiomyocyte BH4. Deficiency in coronary endothelial cell BH4 led to NOS uncoupling, decreased NO bioactivity, and increased superoxide and hydrogen peroxide productions in the hearts of Gch1fl/flTie2cre mice. Under physiological conditions, loss of endothelial cell-specific BH4 led to mild cardiac hypertrophy in Gch1fl/flTie2cre hearts. Endothelial cell BH4 loss was also associated with increased neuronal NOS protein, loss of endothelial NOS protein, and increased phospholamban phosphorylation at serine-17 in cardiomyocytes. Loss of cardiac endothelial cell BH4 led to coronary vascular dysfunction, reduced functional recovery, and increased myocardial infarct size following ischemia-reperfusion injury. Taken together, these studies reveal a specific role for endothelial cell Gch1/BH4 biosynthesis in cardiac function and the response to cardiac ischemia-reperfusion injury. Targeting endothelial cell Gch1 and BH4 biosynthesis may provide a novel therapeutic target for the prevention and treatment of cardiac dysfunction and ischemia-reperfusion injury.NEW & NOTEWORTHY We demonstrate a critical role for endothelial cell Gch1/BH4 biosynthesis in coronary vascular function and cardiac function. Loss of cardiac endothelial cell BH4 leads to coronary vascular dysfunction, reduced functional recovery, and increased myocardial infarct size following ischemia/reperfusion injury. Targeting endothelial cell Gch1 and BH4 biosynthesis may provide a novel therapeutic target for the prevention and treatment of cardiac dysfunction, ischemia injury, and heart failure.
Collapse
Affiliation(s)
- Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sandy M Chu
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthew Kelly
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jenifer K Bendall
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jillian N Simon
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mark J Crabtree
- Department of Biochemical Sciences, School of Bioscience and Medicine, University of Surrey, Guildford, United Kingdom
| | - Barbara Casadei
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Novoa U, Soto K, Valdés C, Villaseñor J, Treuer AV, González DR. Tetrahydrobiopterin (BH 4) Supplementation Prevents the Cardiorenal Effects of Diabetes in Mice by Reducing Oxidative Stress, Inflammation and Fibrosis. Biomedicines 2022; 10:biomedicines10102479. [PMID: 36289741 PMCID: PMC9599239 DOI: 10.3390/biomedicines10102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background: The effects of diabetes on the cardiovascular system as well as in the kidney are profound, which include hypertrophy and fibrosis. Diabetes also induces oxidative stress, at least in part due to the uncoupling of nitric oxide synthase (NOS); this is a shift in NO production toward superoxide production due to reduced levels of the NOS cofactor tetrahydrobiopterin (BH4). With this in mind, we tested the hypothesis that BH4 supplementation may prevent the development of diabetic cardiomyopathy and nephropathy. Methods: Diabetes was induced in Balb/c mice with streptozotocin. Then, diabetic mice were divided into two groups: one group provided with BH4 (sapropterin) in drinking water (daily doses of 15 mg/kg/day, during eight weeks) and the other that received only water. A third group of normoglycemic mice that received only water were used as the control. Results: Cardiac levels of BH4 were increased in mice treated with BH4 (p = 0.0019). Diabetes induced cardiac hypertrophy, which was prevented in the group that received BH4 (p < 0.05). In addition, hypertrophy was evaluated as cardiomyocyte cross-sectional area. This was reduced in diabetic mice that received BH4 (p = 0.0012). Diabetes induced cardiac interstitial fibrosis that was reduced in mice that received BH4 treatment (p < 0.05). We also evaluated in the kidney the impact of BH4 treatment on glomerular morphology. Diabetes induced glomerular hypertrophy compared with normoglycemic mice and was prevented by BH4 treatment. In addition, diabetic mice presented glomerular fibrosis, which was prevented in mice that received BH4. Conclusions: These results suggest that chronic treatment with BH4 in mice ameliorates the cardiorenal effects of diabetes,, probably by restoring the nitroso−redox balance. This offers a possible new alternative to explore a BH4-based treatment for the organ damage caused by diabetes.
Collapse
Affiliation(s)
- Ulises Novoa
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Avenida Lircay s/n, Talca 3460000, Chile
| | - Karen Soto
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Avenida Lircay s/n, Talca 3460000, Chile
| | - Cristian Valdés
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca 3466706, Chile
| | - Jorge Villaseñor
- Instituto de Química de Recursos Naturales, Universidad de Talca, Talca 3460000, Chile
| | - Adriana V. Treuer
- Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Catolica del Maule, Talca 3466706, Chile
| | - Daniel R. González
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Avenida Lircay s/n, Talca 3460000, Chile
- Correspondence: ; Tel.: +56-71-2-418856
| |
Collapse
|
7
|
Xu X, Zhao X, Chen L, Liu M, Hu Z, Ke J, Fu B, Zhou Y, Wei H. CD158a + /CD158b + NK cell imbalance correlates with hypertension in patients with pre-eclampsia. Am J Reprod Immunol 2022; 87:e13532. [PMID: 35253311 DOI: 10.1111/aji.13532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/13/2022] [Accepted: 03/02/2022] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Preeclampsia, a pregnancy complication with hypertension and proteinuria, seriously threats the health and lives of the mother and the baby. The pathogenesis of pre-eclampsia remains incompletely understood. The role of peripheral natural killer cells (NK cells) in the pre-eclampsia is unclear. METHOD OF STUDY Flow cytometry was performed to detect the expression of CD158a (KIR2DL1) and CD158b (KIR2DL2/3) in peripheral NK cells of healthy pregnant women (HP) and patients with pre-eclampsia (PE). Differentially expressed genes (DEGs) in CD158a+ and CD158b+ NK cells were identified by RNA-sequencing and real-time PCR. Protein array analysis was used to identify altered protein levels in the serum of study participants. RESULTS CD158a+ NK cell numbers were increased in the peripheral blood of patients while the number of CD158b+ NK cells was reduced. In addition, the percentage of CD158a+ NK cells within the peripheral NK subset was positively correlated with systolic blood pressure while the percentage of CD158b+ NK cells was negatively correlated with systolic blood pressure. RNA-seq and real-time PCR showed that the expression of ERAP2 and GCH1, the genes that regulate blood pressure and angiogenesis, was decreased in CD158a+ compared to CD158b+ NK cells. Consistently, the level of proteins involved in angiogenesis was altered in the serum of pre-eclampsia patients compared to healthy individuals. CONCLUSIONS CD158a+ NK cells increased while CD158b+ NK cells decreased in the peripheral blood of patients with pre-eclampsia compared to healthy individuals. The change in the frequency of CD158a+ /CD158b+ NK cells is related to the increase in blood pressure.
Collapse
Affiliation(s)
- Xiuxiu Xu
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Xirui Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Ling Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Muziying Liu
- Anhui Institute of Pediatric Research, Anhui Provincial Children's Hospital, Hefei, P.R. China
| | - Ziming Hu
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Jieqi Ke
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Binqing Fu
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Yonggang Zhou
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Haiming Wei
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P.R. China
| |
Collapse
|
8
|
Jayaram R, Jones M, Reilly S, Crabtree MJ, Pal N, Goodfellow N, Nahar K, Simon J, Carnicer R, DeSilva R, Ratnatunga C, Petrou M, Sayeed R, Roalfe A, Channon KM, Bashir Y, Betts T, Hill M, Casadei B. Atrial nitroso-redox balance and refractoriness following on-pump cardiac surgery: a randomized trial of atorvastatin. Cardiovasc Res 2022; 118:184-195. [PMID: 33098411 PMCID: PMC8752359 DOI: 10.1093/cvr/cvaa302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/07/2020] [Accepted: 10/12/2020] [Indexed: 01/19/2023] Open
Abstract
AIMS Systemic inflammation and increased activity of atrial NOX2-containing NADPH oxidases have been associated with the new onset of atrial fibrillation (AF) after cardiac surgery. In addition to lowering LDL-cholesterol, statins exert rapid anti-inflammatory and antioxidant effects, the clinical significance of which remains controversial. METHODS AND RESULTS We first assessed the impact of cardiac surgery and cardiopulmonary bypass (CPB) on atrial nitroso-redox balance by measuring NO synthase (NOS) and GTP cyclohydrolase-1 (GCH-1) activity, biopterin content, and superoxide production in paired samples of the right atrial appendage obtained before (PRE) and after CPB and reperfusion (POST) in 116 patients. The effect of perioperative treatment with atorvastatin (80 mg once daily) on these parameters, blood biomarkers, and the post-operative atrial effective refractory period (AERP) was then evaluated in a randomized, double-blind, placebo-controlled study in 80 patients undergoing cardiac surgery on CPB. CPB and reperfusion led to a significant increase in atrial superoxide production (74% CI 71-76%, n = 46 paired samples, P < 0.0001) and a reduction in atrial tetrahydrobiopterin (BH4) (34% CI 33-35%, n = 36 paired samples, P < 0.01), and in GCH-1 (56% CI 55-58%, n = 26 paired samples, P < 0.001) and NOS activity (58% CI 52-67%, n = 20 paired samples, P < 0.001). Perioperative atorvastatin treatment prevented the effect of CPB and reperfusion on all parameters but had no significant effect on the postoperative right AERP, troponin release, or NT-proBNP after cardiac surgery. CONCLUSION Perioperative statin therapy prevents post-reperfusion atrial nitroso-redox imbalance in patients undergoing on-pump cardiac surgery but has no significant impact on postoperative atrial refractoriness, perioperative myocardial injury, or markers of postoperative LV function. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT01780740.
Collapse
Affiliation(s)
- Raja Jayaram
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Michael Jones
- Cardiology, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Mark J Crabtree
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Nikhil Pal
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Nicola Goodfellow
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Keshav Nahar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Jillian Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Ravi DeSilva
- Cardiothoracic Surgery, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Chandana Ratnatunga
- Cardiothoracic Surgery, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Mario Petrou
- Cardiothoracic Surgery, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rana Sayeed
- Cardiothoracic Surgery, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrea Roalfe
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| | - Yaver Bashir
- Cardiology, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Timothy Betts
- Cardiology, Oxford Heart Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, L6, West Wing, Oxford OX3 9DU, UK
| |
Collapse
|
9
|
Zhong GC, Zhao ZB, Cheng Y, Wang YB, Qiu C, Mao LH, Hu JJ, Cai D, Liu Y, Gong JP, Li SW. Epigenetic silencing of GCH1promotes hepatocellular carcinoma growth by activating superoxide anion-mediated ASK1/p38 signaling via inhibiting tetrahydrobiopterin de novo biosynthesis. Free Radic Biol Med 2021; 168:81-94. [PMID: 33781891 DOI: 10.1016/j.freeradbiomed.2021.03.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/21/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer, including hepatocellular carcinoma (HCC). However, its role in HCC remains to be elucidated. Herein, we identified GTP cyclohydrolase 1 (GCH1), the first rate-limiting enzyme in tetrahydrobiopterin (BH4) de novo biosynthesis, as a novel metabolic regulator of HCC. GCH1 was frequently down-regulated in HCC tissues and cell lines by promoter methylation. Low GCH1 expression was associated with larger tumor size, increased tumor number, and worse prognosis in two independent cohorts of HCC patients. Functionally, GCH1 silencing promoted HCC growth in vitro and in vivo, while GCH1 overexpression exerted an opposite effect. The metabolite BH4 inhibited HCC growth in vitro and in vivo. GCH1 silencing exerted its growth-promoting effect through directly inhibiting BH4 de novo biosynthesis. Mechanistically, GCH1 silencing activated ASK1/p38 signaling; pharmacological or genetic inhibition of ASK1 or p38 abolished GCH1 silencing-induced growth-promoting effect. Further mechanistic studies found that GCH1 silencing-induced BH4 reduction resulted in an increase of intracellular superoxide anion levels in a dose-dependent manner, which mediated the activation of ASK1/p38 signaling. Collectively, our study reveals that epigenetic silencing of GCH1 promotes HCC growth by activating superoxide anion-mediated ASK1/p38 signaling via inhibiting BH4 de novo biosynthesis, suggesting that targeting GCH1/BH4 pathway may be a promising therapeutic strategy to combat HCC.
Collapse
Affiliation(s)
- Guo-Chao Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Bo Zhao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Cheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Bing Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chan Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Hong Mao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie-Jun Hu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Liu
- Department of Gastroenterology, The Fifth People's Hospital of Chengdu, Chengdu, China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Sheng-Wei Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
10
|
Carnicer R, Duglan D, Ziberna K, Recalde A, Reilly S, Simon JN, Mafrici S, Arya R, Rosello-Lleti E, Chuaiphichai S, Tyler D, Lygate CA, Channon KM, Casadei B. BH4 Increases nNOS Activity and Preserves Left Ventricular Function in Diabetes. Circ Res 2021; 128:585-601. [PMID: 33494625 PMCID: PMC7612785 DOI: 10.1161/circresaha.120.316656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
RATIONALE In diabetic patients, heart failure with predominant left ventricular (LV) diastolic dysfunction is a common complication for which there is no effective treatment. Oxidation of the NOS (nitric oxide synthase) cofactor tetrahydrobiopterin (BH4) and dysfunctional NOS activity have been implicated in the pathogenesis of the diabetic vascular and cardiomyopathic phenotype. OBJECTIVE Using mice models and human myocardial samples, we evaluated whether and by which mechanism increasing myocardial BH4 availability prevented or reversed LV dysfunction induced by diabetes. METHODS AND RESULTS In contrast to the vascular endothelium, BH4 levels, superoxide production, and NOS activity (by liquid chromatography) did not differ in the LV myocardium of diabetic mice or in atrial tissue from diabetic patients. Nevertheless, the impairment in both cardiomyocyte relaxation and [Ca2+]i (intracellular calcium) decay and in vivo LV function (echocardiography and tissue Doppler) that developed in wild-type mice 12 weeks post-diabetes induction (streptozotocin, 42-45 mg/kg) was prevented in mGCH1-Tg (mice with elevated myocardial BH4 content secondary to trangenic overexpression of GTP-cyclohydrolase 1) and reversed in wild-type mice receiving oral BH4 supplementation from the 12th to the 18th week after diabetes induction. The protective effect of BH4 was abolished by CRISPR/Cas9-mediated knockout of nNOS (the neuronal NOS isoform) in mGCH1-Tg. In HEK (human embryonic kidney) cells, S-nitrosoglutathione led to a PKG (protein kinase G)-dependent increase in plasmalemmal density of the insulin-independent glucose transporter GLUT-1 (glucose transporter-1). In cardiomyocytes, mGCH1 overexpression induced a NO/sGC (soluble guanylate cyclase)/PKG-dependent increase in glucose uptake via GLUT-1, which was instrumental in preserving mitochondrial creatine kinase activity, oxygen consumption rate, LV energetics (by 31phosphorous magnetic resonance spectroscopy), and myocardial function. CONCLUSIONS We uncovered a novel mechanism whereby myocardial BH4 prevents and reverses LV diastolic and systolic dysfunction associated with diabetes via an nNOS-mediated increase in insulin-independent myocardial glucose uptake and utilization. These findings highlight the potential of GCH1/BH4-based therapeutics in human diabetic cardiomyopathy. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
| | - Drew Duglan
- Cardiovascular Medicine, University of Oxford
| | | | | | | | | | | | - Ritu Arya
- Cardiovascular Medicine, University of Oxford
| | | | | | - Damian Tyler
- Physiology, Anatomy and Genetics, University of Oxford
| | | | | | | |
Collapse
|
11
|
Simon JN, Vrellaku B, Monterisi S, Chu SM, Rawlings N, Lomas O, Marchal GA, Waithe D, Syeda F, Gajendragadkar PR, Jayaram R, Sayeed R, Channon KM, Fabritz L, Swietach P, Zaccolo M, Eaton P, Casadei B. Oxidation of Protein Kinase A Regulatory Subunit PKARIα Protects Against Myocardial Ischemia-Reperfusion Injury by Inhibiting Lysosomal-Triggered Calcium Release. Circulation 2021; 143:449-465. [PMID: 33185461 PMCID: PMC7846288 DOI: 10.1161/circulationaha.120.046761] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Kinase oxidation is a critical signaling mechanism through which changes in the intracellular redox state alter cardiac function. In the myocardium, PKARIα (type-1 protein kinase A) can be reversibly oxidized, forming interprotein disulfide bonds in the holoenzyme complex. However, the effect of PKARIα disulfide formation on downstream signaling in the heart, particularly under states of oxidative stress such as ischemia and reperfusion (I/R), remains unexplored. METHODS Atrial tissue obtained from patients before and after cardiopulmonary bypass and reperfusion and left ventricular (LV) tissue from mice subjected to I/R or sham surgery were used to assess PKARIα disulfide formation by immunoblot. To determine the effect of disulfide formation on PKARIα catalytic activity and subcellular localization, live-cell fluorescence imaging and stimulated emission depletion super-resolution microscopy were performed in prkar1 knock-out mouse embryonic fibroblasts, neonatal myocytes, or adult LV myocytes isolated from "redox dead" (Cys17Ser) PKARIα knock-in mice and their wild-type littermates. Comparison of intracellular calcium dynamics between genotypes was assessed in fura2-loaded LV myocytes, whereas I/R-injury was assessed ex vivo. RESULTS In both humans and mice, myocardial PKARIα disulfide formation was found to be significantly increased (2-fold in humans, P=0.023; 2.4-fold in mice, P<0.001) in response to I/R in vivo. In mouse LV cardiomyocytes, disulfide-containing PKARIα was not found to impact catalytic activity, but instead led to enhanced AKAP (A-kinase anchoring protein) binding with preferential localization of the holoenzyme to the lysosome. Redox-dependent regulation of lysosomal two-pore channels by PKARIα was sufficient to prevent global calcium release from the sarcoplasmic reticulum in LV myocytes, without affecting intrinsic ryanodine receptor leak or phosphorylation. Absence of I/R-induced PKARIα disulfide formation in "redox dead" knock-in mouse hearts resulted in larger infarcts (2-fold, P<0.001) and a concomitant reduction in LV contractile recovery (1.6-fold, P<0.001), which was prevented by administering the lysosomal two-pore channel inhibitor Ned-19 at the time of reperfusion. CONCLUSIONS Disulfide modification targets PKARIα to the lysosome, where it acts as a gatekeeper for two-pore channel-mediated triggering of global calcium release. In the postischemic heart, this regulatory mechanism is critical for protection from extensive injury and offers a novel target for the design of cardioprotective therapeutics.
Collapse
Affiliation(s)
- Jillian N. Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics (S.M., P.S., M.Z.), University of Oxford, United Kingdom
| | - Sandy M. Chu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Nadiia Rawlings
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Oliver Lomas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Gerard A. Marchal
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Dominic Waithe
- Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine (D.W.), University of Oxford, United Kingdom
| | - Fahima Syeda
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom (F.S., L.F.)
| | - Parag R. Gajendragadkar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Raja Jayaram
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Rana Sayeed
- Cardiothoracic Surgery, Oxford Heart Centre, Oxford University Hospitals National Health Service Foundation Trust, United Kingdom (R.S.)
| | - Keith M. Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom (F.S., L.F.)
- Department of Cardiology, University Hospitals Birmingham, United Kingdom (L.F.)
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics (S.M., P.S., M.Z.), University of Oxford, United Kingdom
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics (S.M., P.S., M.Z.), University of Oxford, United Kingdom
| | - Philip Eaton
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, United Kingdom (P.E.)
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.N.S., B.V., S.M.C., N.R., O.L., G.A.M., P.R.G., R.J., K.M.C., B.C.), University of Oxford, United Kingdom
| |
Collapse
|
12
|
Yan T, Zhang T, Mu W, Qi Y, Guo S, Hu N, Zhao W, Zhang S, Wang Q, Shi L, Liu L. Ionizing radiation induces BH 4 deficiency by downregulating GTP-cyclohydrolase 1, a novel target for preventing and treating radiation enteritis. Biochem Pharmacol 2020; 180:114102. [PMID: 32562786 DOI: 10.1016/j.bcp.2020.114102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/22/2020] [Accepted: 06/15/2020] [Indexed: 01/22/2023]
Abstract
Radiation enteritis (RE) is a common side effect after radiotherapy for abdominal cancer. RE pathogenesis is complicated, with no drugs available for prevention or treatments. Intestinal ischemia is a key factor in the occurrence and development of enteritis. The effect of ionizing radiation (IR) on intestinal ischemia is unknown. Deficiency of tetrahydrobiopterin (BH4) produced by GTP-cyclohydrolase 1 (Gch1) is important in ischemic diseases. This study focused on the relationship of Gch1/BH4 between intestinal ischemia in radiation enteritis. BH4 levels were analyzed by high-performance liquid chromatography in humans and rats after radiotherapy. Intestinal blood perfusion was measured by laser doppler flow imaging. Vascular ring tests determined the diastolic functions of rat mesenteric arteries. Gene, protein, and immunohistochemical staining experiments and inhibitor interventions were used to investigate Gch1 and endothelial NOS (eNOS) in rat mesenteric arteries and endothelial cells. The results showed that IR decreased BH4 levels in patients and rats after radiotherapy and decreased intestinal blood perfusion in rats. The degree of change in intestinal ischemia was consistent with intestinal villus injury. Gch1 mRNA and protein levels and nitric oxide (NO) production significantly decreased, while eNOS uncoupling in arterial and vascular endothelial cells strongly increased. BH4 supplementation improved eNOS uncoupling and NO levels in vascular endothelia after IR. The results of this study showed that downregulation of Gch1 in intestinal blood vessels after IR is an important target in RE. BH4 supplementation may prevent intestinal ischemia and improve vascular endothelial function after IR. These findings have clinical significance for the prevention and treatment of RE.
Collapse
Affiliation(s)
- Tao Yan
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Tian Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Wei Mu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Yuhong Qi
- Department of Radiotherapy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Shun Guo
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Na Hu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Weihe Zhao
- Department of Radiotherapy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Song Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Qinhui Wang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Lei Shi
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China.
| | - Linna Liu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China.
| |
Collapse
|
13
|
Oikawa S, Kai Y, Mano A, Nakamura S, Kakinuma Y. S-Nitroso-N-Pivaloyl-D-Penicillamine, a novel non-neuronal ACh system activator, modulates cardiac diastolic function to increase cardiac performance under pathophysiological conditions. Int Immunopharmacol 2020; 84:106459. [PMID: 32325404 DOI: 10.1016/j.intimp.2020.106459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 01/06/2023]
Abstract
We have previously reported the development of a novel chemical compound, S-Nitroso-N-Pivaloyl-D-Penicillamine (SNPiP), for the upregulation of the non-neuronal cardiac cholinergic system (NNCCS), a cardiac acetylcholine (ACh) synthesis system, which is different from the vagus nerve releasing of ACh as a neurotransmitter. However, it remains unclear how SNPiP could influence cardiac function positively, and whether SNPiP could improve cardiac function under various pathological conditions. SNPiP-injected control mice demonstrated a gradual upregulation in diastolic function without changes in heart rate. In contrast to some parameters in cardiac function that were influenced by SNPiP 24 h or 48 h after a single intraperitoneal (IP) injection, 72 h later, end-systolic pressure, cardiac output, end-diastolic volume, stroke volume, and ejection fraction increased. IP SNPiP injection also improved impaired cardiac function, which is a characteristic feature of the db/db heart, in a delayed fashion, including diastolic and systolic function, following either several consecutive injections or a single injection. SNPiP, a novel NNCCS activator, could be applied as a therapeutic agent for the upregulation of NNCCS and as a unique tool for modulating cardiac function via improvement in diastolic function.
Collapse
Affiliation(s)
- Shino Oikawa
- Department of Bioregulatory Science (Physiology), Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Yuko Kai
- Department of Bioregulatory Science (Physiology), Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Asuka Mano
- Department of Bioregulatory Science (Physiology), Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Shigeo Nakamura
- Department of Chemistry, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo 180-0023, Japan
| | - Yoshihiko Kakinuma
- Department of Bioregulatory Science (Physiology), Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| |
Collapse
|
14
|
Lee I, Kim S, Nagar H, Choi SJ, Jeon BH, Piao S, Kim CS. CR6-interacting factor 1 deficiency reduces endothelial nitric oxide synthase activity by inhibiting biosynthesis of tetrahydrobiopterin. Sci Rep 2020; 10:842. [PMID: 31964986 PMCID: PMC6972730 DOI: 10.1038/s41598-020-57673-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/31/2019] [Indexed: 01/07/2023] Open
Abstract
Downregulation of CR6 interacting factor 1 (CRIF1) has been reported to induce mitochondrial dysfunction, resulting in reduced activity of endothelial nitric oxide synthase (eNOS) and NO production in endothelial cells. Tetrahydrobiopterin (BH4) is an important cofactor in regulating the balance between NO (eNOS coupling) and superoxide production (eNOS uncoupling). However, whether the decreased eNOS and NO production in CRIF1-deficient cells is associated with relative BH4 deficiency-induced eNOS uncoupling remains completely unknown. Our results showed that CRIF1 deficiency increased eNOS uncoupling and depleted levels of total biopterin and BH4 by reducing the enzymes of BH4 biosynthesis (GCH-1, PTS, SPR, and DHFR) in vivo and vitro, respectively. Supplementation of CRIF1-deficient cells with BH4 significantly increased the recovery of Akt and eNOS phosphorylation and NO synthesis. In addition, scavenging ROS with MitoTEMPO treatment replenished BH4 levels by elevating levels of GCH-1, PTS, and SPR, but with no effect on the level of DHFR. Downregulation of DHFR synthesis regulators p16 or p21 in CRIF1-deficient cells partially recovered the DHFR expression. In summary, CRIF1 deficiency inhibited BH4 biosynthesis and exacerbated eNOS uncoupling. This resulted in reduced NO production and increased oxidative stress, which contributes to endothelial dysfunction and is involved in the pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Ikjun Lee
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Seonhee Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Harsha Nagar
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Su-Jeong Choi
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Shuyu Piao
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| | - Cuk-Seong Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| |
Collapse
|
15
|
Antonopoulos AS, Goliopoulou A, Oikonomou E, Tsalamandris S, Papamikroulis GA, Lazaros G, Tsiamis E, Latsios G, Brili S, Papaioannou S, Gennimata V, Tousoulis D. Redox State in Atrial Fibrillation Pathogenesis and Relevant Therapeutic Approaches. Curr Med Chem 2019; 26:765-779. [PMID: 28721830 DOI: 10.2174/0929867324666170718130408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/04/2016] [Accepted: 12/04/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Myocardial redox state is a critical determinant of atrial biology, regulating cardiomyocyte apoptosis, ion channel function, and cardiac hypertrophy/fibrosis and function. Nevertheless, it remains unclear whether the targeting of atrial redox state is a rational therapeutic strategy for atrial fibrillation prevention. OBJECTIVE To review the role of atrial redox state and anti-oxidant therapies in atrial fibrillation. METHOD Published literature in Medline was searched for experimental and clinical evidence linking myocardial redox state with atrial fibrillation pathogenesis as well as studies looking into the role of redoxtargeting therapies in the prevention of atrial fibrillation. RESULTS Data from animal models have shown that altered myocardial nitroso-redox balance and NADPH oxidases activity are causally involved in the pathogenesis of atrial fibrillation. Similarly experimental animal data supports that increased reactive oxygen / nitrogen species formation in the atrial tissue is associated with altered electrophysiological properties of atrial myocytes and electrical remodeling, favoring atrial fibrillation development. In humans, randomized clinical studies using redox-related therapeutic approaches (e.g. statins or antioxidant agents) have not documented any benefits in the prevention of atrial fibrillation development (mainly post-operative atrial fibrillation risk). CONCLUSION Despite strong experimental and translational data supporting the role of atrial redox state in atrial fibrillation pathogenesis, such mechanistic evidence has not been translated to clinical benefits in atrial fibrillation risk in randomized clinical studies using redox-related therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - George Lazaros
- 1st Cardiology Department, Athens Medical School, Athens, Greece
| | | | - George Latsios
- 1st Cardiology Department, Athens Medical School, Athens, Greece
| | - Stella Brili
- 1st Cardiology Department, Athens Medical School, Athens, Greece
| | | | | | | |
Collapse
|
16
|
Heikal L, Starr A, Hussein D, Prieto-Lloret J, Aaronson P, Dailey LA, Nandi M. l-Phenylalanine Restores Vascular Function in Spontaneously Hypertensive Rats Through Activation of the GCH1-GFRP Complex. JACC Basic Transl Sci 2018; 3:366-377. [PMID: 29963647 PMCID: PMC6018612 DOI: 10.1016/j.jacbts.2018.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/27/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Tetrahydrobiopterin is an essential cofactor for NO production. Limitation of endogenous tetrahydrobiopterin reduces NO bioavailability, enhances oxidative stress, and impairs vascular function. Orally supplemented tetrahydrobiopterin has therapeutic challenges because it is rapidly oxidized in vivo. Here, the authors demonstrate that l-phenylalanine, when administered orally, raises vascular tetrahydrobiopterin, restores NO, reduces superoxide, and enhances vascular function in spontaneously hypertensive rats. This effect is achieved by activation of a protein complex (GCH1-GFRP) involved in the biosynthesis of tetrahydrobiopterin. Activation of this protein complex by l-phenylalanine or its analogues represents a novel therapeutic target for vascular disorders underpinned by reduced NO bioavailability.
Reduced nitric oxide (NO) bioavailability correlates with impaired cardiovascular function. NO is extremely labile and has been challenging to develop as a therapeutic agent. However, NO bioavailability could be enhanced by pharmacologically targeting endogenous NO regulatory pathways. Tetrahydrobiopterin, an essential cofactor for NO production, is synthesized by GTP cyclohydrolase-1 (GCH1), which complexes with GCH1 feedback regulatory protein (GFRP). The dietary amino acid l-phenylalanine activates this complex, elevating vascular BH4. Here, the authors demonstrate that l-phenylalanine administration restores vascular function in a rodent model of hypertension, suggesting the GCH1-GFRP complex represents a rational therapeutic target for diseases underpinned by endothelial dysfunction.
Collapse
Key Words
- ACh, acetylcholine
- ANOVA, analysis of variance
- BH2, dihydrobiopterin
- BH4, tetrahydrobiopterin
- EC50, effective concentration for 50% maximal response
- EDHF, endothelium derived hyperpolarizing factor
- GCH1, GTP cyclohydrolase-1
- GFRP, GCH1 feedback regulatory protein
- L-phe, l-phenylalanine
- L-tyr, l-tyrosine
- NO, nitric oxide
- ROS, reactive oxygen species
- SHR, spontaneously hypertensive rat(s)
- WKY, Wistar Kyoto rat(s)
- cardiovascular disease
- eNOS, endothelial nitric oxide synthase
- endothelium
- l-phenylalanine
- nitric oxide
- tetrahydrobiopterin
- vascular activity
Collapse
Affiliation(s)
- Lamia Heikal
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Anna Starr
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Dania Hussein
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Jesus Prieto-Lloret
- Division of Asthma, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Phil Aaronson
- Division of Asthma, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Lea Ann Dailey
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Manasi Nandi
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.,Cardiovascular Division, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
17
|
Francis BN, Salameh M, Khamisy-Farah R, Farah R. Tetrahydrobiopterin (BH 4 ): Targeting endothelial nitric oxide synthase as a potential therapy for pulmonary hypertension. Cardiovasc Ther 2018; 36. [PMID: 29151278 DOI: 10.1111/1755-5922.12312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/18/2017] [Accepted: 11/11/2017] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Pulmonary Hypertension (PH) is complex disease which is associated with endothelial and cardiac dysfunction. Tetrahydrobiopterin (BH4 ) regulates endothelial nitric oxide synthase (eNOS) to produce nitric oxide rather than superoxide which maintains normal endothelial and cardiac function. This study explores the therapeutic potential of BH4 in experimental PH. METHODS Monocrotaline-induced PH in rats and Hph-1 deficiency in mice were used for animal experiments. Hemodynamic measurements using pressure transducers were conducted for pulmonary and cardiac pressures, and Langendorff apparatus was used for isolated heart experiments; preventive as well as rescue treatment protocols were conducted; tissues were collected for histological and biochemical studies. RESULTS In vivo acute BH4 administration reduced pulmonary artery pressure (PAP) only in the MCT rat. In a Langendorff preparation, BH4 increased right ventricular systolic pressure (RVSP) in right ventricular hypertrophy (RVH) but not in control. In "prevention" therapy, BH4 (10 and 100 mg/kg) attenuated the development of PH in rat MCT model. eNOS protein levels in lung homogenates were maintained and cGMP levels were increased. In "rescue" therapy, BH4 (10 and 100 mg/kg) ameliorated pulmonary vascular muscularization in a dose-dependent manner. RVSP was reduced in RVH and pulmonary vascular muscularization was attenuated. BH4 at 10 mg/kg reduced RV myocyte diameter while BH4 at 100 mg/kg reversed it to control level. BH4 restored normal levels of eNOS protein and in a dose of 100 mg/kg enhanced lung tissue levels of BH4 , cGMP, and NO compared to placebo. CONCLUSION The current study provides scientific evidence for a therapeutic potential of BH4 in PH and invites further investigation.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/pharmacology
- Arterial Pressure/drug effects
- Biopterins/analogs & derivatives
- Biopterins/pharmacology
- Cyclic GMP/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- GTP Cyclohydrolase/deficiency
- GTP Cyclohydrolase/genetics
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/prevention & control
- Isolated Heart Preparation
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocrotaline
- Myocardial Contraction/drug effects
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/antagonists & inhibitors
- Nitric Oxide Synthase Type III/metabolism
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Time Factors
- Ventricular Function, Right/drug effects
- Ventricular Pressure/drug effects
Collapse
Affiliation(s)
- Bahaa N Francis
- Experimental Medicine and Toxicology, Imperial College London, Hammersmith Hospital, London, UK
- Department of Internal Medicine B, Ziv Medical Center, Safad, Israel
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Maram Salameh
- Pharmacy Department, Carmel Medical Center, Haifa, Israel
| | | | - Raymond Farah
- Department of Internal Medicine B, Ziv Medical Center, Safad, Israel
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| |
Collapse
|
18
|
High-Intensity Exercise Reduces Cardiac Fibrosis and Hypertrophy but Does Not Restore the Nitroso-Redox Imbalance in Diabetic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7921363. [PMID: 28698769 PMCID: PMC5494101 DOI: 10.1155/2017/7921363] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/18/2017] [Accepted: 04/27/2017] [Indexed: 01/25/2023]
Abstract
Diabetic cardiomyopathy refers to the manifestations in the heart as a result of altered glucose homeostasis, reflected as fibrosis, cellular hypertrophy, increased oxidative stress, and apoptosis, leading to ventricular dysfunction. Since physical exercise has been indicated as cardioprotective, we tested the hypothesis that high-intensity exercise training could reverse the cardiac maladaptations produced by diabetes. For this, diabetes was induced in rats by a single dose of alloxan. Diabetic rats were randomly assigned to a sedentary group or submitted to a program of exercise on a treadmill for 4 weeks at 80% of maximal performance. Another group of normoglycemic rats was used as control. Diabetic rat hearts presented cardiomyocyte hypertrophy and interstitial fibrosis. Chronic exercise reduced both parameters but increased apoptosis. Diabetes increased the myocardial levels of the mRNA and proteins of NADPH oxidases NOX2 and NOX4. These altered levels were not reduced by exercise. Diabetes also increased the level of uncoupled endothelial nitric oxide synthase (eNOS) that was not reversed by exercise. Finally, diabetic rats showed a lower degree of phosphorylated phospholamban and reduced levels of SERCA2 that were not restored by high-intensity exercise. These results suggest that high-intensity chronic exercise was able to reverse remodeling in the diabetic heart but was unable to restore the nitroso-redox imbalance imposed by diabetes.
Collapse
|
19
|
Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR, Ge ZD. Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep 2017; 7:3093. [PMID: 28596578 PMCID: PMC5465102 DOI: 10.1038/s41598-017-03234-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
GTP cyclohydrolase 1 (GCH1) and its product tetrahydrobiopterin play crucial roles in cardiovascular health and disease, yet the exact regulation and role of GCH1 in adverse cardiac remodeling after myocardial infarction are still enigmatic. Here we report that cardiac GCH1 is degraded in remodeled hearts after myocardial infarction, concomitant with increases in the thickness of interventricular septum, interstitial fibrosis, and phosphorylated p38 mitogen-activated protein kinase and decreases in left ventricular anterior wall thickness, cardiac contractility, tetrahydrobiopterin, the dimers of nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and the expression of sarcoplasmic reticulum Ca2+ handling proteins. Intriguingly, transgenic overexpression of GCH1 in cardiomyocytes reduces the thickness of interventricular septum and interstitial fibrosis and increases anterior wall thickness and cardiac contractility after infarction. Moreover, we show that GCH1 overexpression decreases phosphorylated p38 mitogen-activated protein kinase and elevates tetrahydrobiopterin levels, the dimerization and phosphorylation of neuronal nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and sarcoplasmic reticulum Ca2+ handling proteins in post-infarction remodeled hearts. Our results indicate that the pivotal role of GCH1 overexpression in post-infarction cardiac remodeling is attributable to preservation of neuronal nitric oxide synthase and sarcoplasmic reticulum Ca2+ handling proteins, and identify a new therapeutic target for cardiac remodeling after infarction.
Collapse
Affiliation(s)
- Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Medicine, Columbia University, 630 W. 168th Street, New York, New York, 10032, USA
| | - Shelley L Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, Wisconsin, 53234, USA
| | - Shigang Qiao
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zeljko J Bosnjak
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Physiology, Medical College of Wiscosin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, People's Republic of China
| | - David C Warltier
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Judy R Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.
| |
Collapse
|
20
|
Shen JS, Arning E, West ML, Day TS, Chen S, Meng XL, Forni S, McNeill N, Goker-Alpan O, Wang X, Ashcraft P, Moore DF, Cheng SH, Schiffmann R, Bottiglieri T. Tetrahydrobiopterin deficiency in the pathogenesis of Fabry disease. Hum Mol Genet 2017; 26:1182-1192. [PMID: 28158561 DOI: 10.1093/hmg/ddx032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/19/2017] [Indexed: 02/07/2023] Open
Abstract
Fabry disease is caused by deficient activity of α-galactosidase A and subsequent accumulation of glycosphingolipids (mainly globotriaosylceramide, Gb3), leading to multisystem organ dysfunction. Oxidative stress and nitric oxide synthase (NOS) uncoupling are thought to contribute to Fabry cardiovascular diseases. We hypothesized that decreased tetrahydrobiopterin (BH4) plays a role in the pathogenesis of Fabry disease. We found that BH4 was decreased in the heart and kidney but not in the liver and aorta of Fabry mice. BH4 was also decreased in the plasma of female Fabry patients, which was not corrected by enzyme replacement therapy (ERT). Gb3 levels were inversely correlated with BH4 levels in animal tissues and cultured patient cells. To investigate the role of BH4 deficiency in disease phenotypes, 12-month-old Fabry mice were treated with gene transfer-mediated ERT or substrate reduction therapy (SRT) for 6 months. In the Fabry mice receiving SRT but not ERT, BH4 deficiency was restored, concomitant with ameliorated cardiac and renal hypertrophy. Additionally, glutathione levels were decreased in Fabry mouse tissues in a sex-dependent manner. Renal BH4 levels were closely correlated with glutathione levels and inversely correlated with cardiac and kidney weight. In conclusion, this study showed that BH4 deficiency occurs in Fabry disease and may contribute to the pathogenesis of the disease through oxidative stress associated with a reduced antioxidant capacity of cells and NOS uncoupling. This study also suggested dissimilar efficacy of ERT and SRT in correcting pre-existing pathologies in Fabry disease.
Collapse
Affiliation(s)
- Jin-Song Shen
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Erland Arning
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Michael L West
- Division of Nephrology, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Taniqua S Day
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | | | - Xing-Li Meng
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Sabrina Forni
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Nathan McNeill
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA, USA
| | - Xuan Wang
- Baylor Research Institute, Dallas, TX, USA
| | - Paula Ashcraft
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - David F Moore
- Sanford Health and University of North Dakota, Fargo, ND, USA
| | | | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| | - Teodoro Bottiglieri
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX 75226, USA
| |
Collapse
|
21
|
Shabeeh H, Khan S, Jiang B, Brett S, Melikian N, Casadei B, Chowienczyk PJ, Shah AM. Blood Pressure in Healthy Humans Is Regulated by Neuronal NO Synthase. Hypertension 2017; 69:970-976. [PMID: 28264923 PMCID: PMC5389591 DOI: 10.1161/hypertensionaha.116.08792] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 12/27/2016] [Accepted: 01/10/2017] [Indexed: 01/22/2023]
Abstract
NO is physiologically generated by endothelial and neuronal NO synthase (nNOS) isoforms. Although nNOS was first identified in brain, it is expressed in other tissues, including perivascular nerves, cardiac and skeletal muscle. Increasing experimental evidence suggests that nNOS has important effects on cardiovascular function, but its composite effects on systemic hemodynamics in humans are unknown. We undertook the first human study to assess the physiological effects of systemic nNOS inhibition on basal hemodynamics. Seventeen healthy normotensive men aged 24±4 years received acute intravenous infusions of an nNOS-selective inhibitor, S-methyl-l-thiocitrulline, and placebo on separate occasions. An initial dose-escalation study showed that S-methyl-l-thiocitrulline (0.1–3.0 µmol/kg) induced dose-dependent changes in systemic hemodynamics. The highest dose of S-methyl-l-thiocitrulline (3.0 µmol/kg over 10 minutes) significantly increased systemic vascular resistance (+42±6%) and diastolic blood pressure (67±1 to 77±3 mm Hg) when compared with placebo (both P<0.01). There were significant decreases in heart rate (60±4 to 51±3 bpm; P<0.01) and left ventricular stroke volume (59±6 to 51±6 mL; P<0.01) but ejection fraction was unaltered. S-methyl-l-thiocitrulline had no effect on radial artery flow-mediated dilatation, an index of endothelial NOS activity. These results suggest that nNOS-derived NO has an important role in the physiological regulation of basal systemic vascular resistance and blood pressure in healthy humans.
Collapse
Affiliation(s)
- Husain Shabeeh
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Sitara Khan
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Benyu Jiang
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Sally Brett
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Narbeh Melikian
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Barbara Casadei
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Philip J Chowienczyk
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.)
| | - Ajay M Shah
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, United Kingdom (H.S., S.K., B.J., S.B., N.M., P.J.C., A.M.S.); and Department of Cardiovascular Medicine, University of Oxford, United Kingdom (B.C.).
| |
Collapse
|
22
|
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: 20] [Impact Index Per Article: 2.5] [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
|
23
|
Vielma AZ, León L, Fernández IC, González DR, Boric MP. Nitric Oxide Synthase 1 Modulates Basal and β-Adrenergic-Stimulated Contractility by Rapid and Reversible Redox-Dependent S-Nitrosylation of the Heart. PLoS One 2016; 11:e0160813. [PMID: 27529477 PMCID: PMC4986959 DOI: 10.1371/journal.pone.0160813] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022] Open
Abstract
S-nitrosylation of several Ca2+ regulating proteins in response to β-adrenergic stimulation was recently described in the heart; however the specific nitric oxide synthase (NOS) isoform and signaling pathways responsible for this modification have not been elucidated. NOS-1 activity increases inotropism, therefore, we tested whether β-adrenergic stimulation induces NOS-1-dependent S-nitrosylation of total proteins, the ryanodine receptor (RyR2), SERCA2 and the L-Type Ca2+ channel (LTCC). In the isolated rat heart, isoproterenol (10 nM, 3-min) increased S-nitrosylation of total cardiac proteins (+46±14%) and RyR2 (+146±77%), without affecting S-nitrosylation of SERCA2 and LTCC. Selective NOS-1 blockade with S-methyl-L-thiocitrulline (SMTC) and Nω-propyl-l-arginine decreased basal contractility and relaxation (−25–30%) and basal S-nitrosylation of total proteins (−25–60%), RyR2, SERCA2 and LTCC (−60–75%). NOS-1 inhibition reduced (−25–40%) the inotropic response and protein S-nitrosylation induced by isoproterenol, particularly that of RyR2 (−85±7%). Tempol, a superoxide scavenger, mimicked the effects of NOS-1 inhibition on inotropism and protein S-nitrosylation; whereas selective NOS-3 inhibitor L-N5-(1-Iminoethyl)ornithine had no effect. Inhibition of NOS-1 did not affect phospholamban phosphorylation, but reduced its oligomerization. Attenuation of contractility was abolished by PKA blockade and unaffected by guanylate cyclase inhibition. Additionally, in isolated mouse cardiomyocytes, NOS-1 inhibition or removal reduced the Ca2+-transient amplitude and sarcomere shortening induced by isoproterenol or by direct PKA activation. We conclude that 1) normal cardiac performance requires basal NOS-1 activity and S-nitrosylation of the calcium-cycling machinery; 2) β-adrenergic stimulation induces rapid and reversible NOS-1 dependent, PKA and ROS-dependent, S-nitrosylation of RyR2 and other proteins, accounting for about one third of its inotropic effect.
Collapse
Affiliation(s)
- Alejandra Z. Vielma
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Luisa León
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Ignacio C. Fernández
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Daniel R. González
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Av. Lircay S.N., Talca, Chile
| | - Mauricio P. Boric
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
- * E-mail:
| |
Collapse
|
24
|
Roselló-Lletí E, Tarazón E, Ortega A, Gil-Cayuela C, Carnicer R, Lago F, González-Juanatey JR, Portolés M, Rivera M. Protein Inhibitor of NOS1 Plays a Central Role in the Regulation of NOS1 Activity in Human Dilated Hearts. Sci Rep 2016; 6:30902. [PMID: 27481317 PMCID: PMC4969592 DOI: 10.1038/srep30902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/08/2016] [Indexed: 12/22/2022] Open
Abstract
An essential factor for the production of nitric oxide by nitric oxide synthase 1 (NOS1), major modulator of cardiac function, is the cofactor tetrahydrobiopterin (BH4). BH4 is regulated by GTP cyclohydrolase 1, the rate-limiting enzyme in BH4 biosynthesis which catalyses the formation of dihydroneopterin 3'triphosfate from GTP, producing BH4 after two further steps catalyzed by 6-pyruvoyltetrahydropterin synthase and sepiapterin reductase. However, there are other essential factors involved in the regulation of NOS1 activity, such as protein inhibitor of NOS1 (PIN), calmodulin, heat shock protein 90, and NOS interacting protein. All these molecules have never been analysed in human non-ischemic dilated hearts (DCM). In this study we demonstrated that the upregulation of cardiac NOS1 is not accompanied by increased NOS1 activity in DCM, partly due to the elevated PIN levels and not because of alterations in biopterin biosynthesis. Notably, the PIN concentration was significantly associated with impaired ventricular function, highlighting the importance of this NOS1 activity inhibitor in Ca(2+) homeostasis. These results take a central role in the current list of targets for future studies focused on the complex cardiac dysfunction processes through more efficient harnessing of NOS1 signalling.
Collapse
Affiliation(s)
- Esther Roselló-Lletí
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Estefanía Tarazón
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Ana Ortega
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Carolina Gil-Cayuela
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Ricardo Carnicer
- Department of Cardiovascular Medicine, University of Oxford, United Kingdom
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Jose Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| |
Collapse
|
25
|
Wu HE, Baumgardt SL, Fang J, Paterson M, Liu Y, Du J, Shi Y, Qiao S, Bosnjak ZJ, Warltier DC, Kersten JR, Ge ZD. Cardiomyocyte GTP Cyclohydrolase 1 Protects the Heart Against Diabetic Cardiomyopathy. Sci Rep 2016; 6:27925. [PMID: 27295516 PMCID: PMC4904741 DOI: 10.1038/srep27925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy increases the risk of heart failure and death. At present, there are no effective approaches to preventing its development in the clinic. Here we report that reduction of cardiac GTP cyclohydrolase 1 (GCH1) degradation by genetic and pharmacological approaches protects the heart against diabetic cardiomyopathy. Diabetic cardiomyopathy was induced in C57BL/6 wild-type mice and transgenic mice with cardiomyocyte-specific overexpression of GCH1 with streptozotocin, and control animals were given citrate buffer. We found that diabetes-induced degradation of cardiac GCH1 proteins contributed to adverse cardiac remodeling and dysfunction in C57BL/6 mice, concomitant with decreases in tetrahydrobiopterin, dimeric and phosphorylated neuronal nitric oxide synthase, sarcoplasmic reticulum Ca(2+) handling proteins, intracellular [Ca(2+)]i, and sarcoplasmic reticulum Ca(2+) content and increases in phosphorylated p-38 mitogen-activated protein kinase and superoxide production. Interestingly, GCH-1 overexpression abrogated these detrimental effects of diabetes. Furthermore, we found that MG 132, an inhibitor for 26S proteasome, preserved cardiac GCH1 proteins and ameliorated cardiac remodeling and dysfunction during diabetes. This study deepens our understanding of impaired cardiac function in diabetes, identifies GCH1 as a modulator of cardiac remodeling and function, and reveals a new therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MA 21224, USA
| | - Shelley L. Baumgardt
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Mark Paterson
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yanan Liu
- Department of Medicine, Columbia University, 630 W. 168th Street, New York, NY 10032, USA
| | - Jianhai Du
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, WI 53234, USA
| | - Shigang Qiao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zeljko J. Bosnjak
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - David C. Warltier
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Judy R. Kersten
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
26
|
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: 1.9] [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
|
27
|
Roof SR, Boslett J, Russell D, del Rio C, Alecusan J, Zweier JL, Ziolo MT, Hamlin R, Mohler PJ, Curran J. Insulin-like growth factor 1 prevents diastolic and systolic dysfunction associated with cardiomyopathy and preserves adrenergic sensitivity. Acta Physiol (Oxf) 2016; 216:421-34. [PMID: 26399932 DOI: 10.1111/apha.12607] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/03/2015] [Accepted: 09/15/2015] [Indexed: 12/11/2022]
Abstract
AIMS Insulin-like growth factor 1 (IGF-1)-dependent signalling promotes exercise-induced physiological cardiac hypertrophy. However, the in vivo therapeutic potential of IGF-1 for heart disease is not well established. Here, we test the potential therapeutic benefits of IGF-1 on cardiac function using an in vivo model of chronic catecholamine-induced cardiomyopathy. METHODS Rats were perfused with isoproterenol via osmotic pump (1 mg kg(-1) per day) and treated with 2 mg kg(-1) IGF-1 (2 mg kg(-1) per day, 6 days a week) for 2 or 4 weeks. Echocardiography, ECG, and blood pressure were assessed. In vivo pressure-volume loop studies were conducted at 4 weeks. Heart sections were analysed for fibrosis and apoptosis, and relevant biochemical signalling cascades were assessed. RESULTS After 4 weeks, diastolic function (EDPVR, EDP, tau, E/A ratio), systolic function (PRSW, ESPVR, dP/dtmax) and structural remodelling (LV chamber diameter, wall thickness) were all adversely affected in isoproterenol-treated rats. All these detrimental effects were attenuated in rats treated with Iso+IGF-1. Isoproterenol-dependent effects on BP were attenuated by IGF-1 treatment. Adrenergic sensitivity was blunted in isoproterenol-treated rats but was preserved by IGF-1 treatment. Immunoblots indicate that cardioprotective p110α signalling and activated Akt are selectively upregulated in Iso+IGF-1-treated hearts. Expression of iNOS was significantly increased in both the Iso and Iso+IGF-1 groups; however, tetrahydrobiopterin (BH4) levels were decreased in the Iso group and maintained by IGF-1 treatment. CONCLUSION IGF-1 treatment attenuates diastolic and systolic dysfunction associated with chronic catecholamine-induced cardiomyopathy while preserving adrenergic sensitivity and promoting BH4 production. These data support the potential use of IGF-1 therapy for clinical applications for cardiomyopathies.
Collapse
Affiliation(s)
| | - J. Boslett
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - D. Russell
- Department of Veterinary Clinical Sciences; College of Veterinarian Medicine; The Ohio State University; Columbus OH USA
| | | | - J. Alecusan
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - J. L. Zweier
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - M. T. Ziolo
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
- Department of Physiology and Cell Biology; The Ohio State University Wexner Medical Center; Columbus OH USA
| | | | - P. J. Mohler
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
- Department of Internal Medicine; The Ohio State University Wexner Medical Center; Columbus OH USA
- Department of Physiology and Cell Biology; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - J. Curran
- The Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
- Department of Internal Medicine; The Ohio State University Wexner Medical Center; Columbus OH USA
| |
Collapse
|
28
|
Sethumadhavan S, Whitsett J, Bennett B, Ionova IA, Pieper GM, Vasquez-Vivar J. Increasing tetrahydrobiopterin in cardiomyocytes adversely affects cardiac redox state and mitochondrial function independently of changes in NO production. Free Radic Biol Med 2016; 93:1-11. [PMID: 26826575 PMCID: PMC5498285 DOI: 10.1016/j.freeradbiomed.2016.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
Tetrahydrobiopterin (BH4) represents a potential strategy for the treatment of cardiac remodeling, fibrosis and/or diastolic dysfunction. The effects of oral treatment with BH4 (Sapropterin™ or Kuvan™) are however dose-limiting with high dose negating functional improvements. Cardiomyocyte-specific overexpression of GTP cyclohydrolase I (mGCH) increases BH4 several-fold in the heart. Using this model, we aimed to establish the cardiomyocyte-specific responses to high levels of BH4. Quantification of BH4 and BH2 in mGCH transgenic hearts showed age-based variations in BH4:BH2 ratios. Hearts of mice (<6 months) have lower BH4:BH2 ratios than hearts of older mice while both GTPCH activity and tissue ascorbate levels were higher in hearts of young than older mice. No evident changes in nitric oxide (NO) production assessed by nitrite and endogenous iron-nitrosyl complexes were detected in any of the age groups. Increased BH4 production in cardiomyocytes resulted in a significant loss of mitochondrial function. Diminished oxygen consumption and reserve capacity was verified in mitochondria isolated from hearts of 12-month old compared to 3-month old mice, even though at 12 months an improved BH4:BH2 ratio is established. Accumulation of 4-hydroxynonenal (4-HNE) and decreased glutathione levels were found in the mGCH hearts and isolated mitochondria. Taken together, our results indicate that the ratio of BH4:BH2 does not predict changes in neither NO levels nor cellular redox state in the heart. The BH4 oxidation essentially limits the capacity of cardiomyocytes to reduce oxidant stress. Cardiomyocyte with chronically high levels of BH4 show a significant decline in redox state and mitochondrial function.
Collapse
Affiliation(s)
- Savitha Sethumadhavan
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jennifer Whitsett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Brian Bennett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Physics, Marquette University, Milwaukee, 1250 W Wisconsin Ave, Milwaukee, WI 53233, USA
| | - Irina A Ionova
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Galen M Pieper
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
29
|
Hashimoto T, Sivakumaran V, Carnicer R, Zhu G, Hahn VS, Bedja D, Recalde A, Duglan D, Channon KM, Casadei B, Kass DA. Tetrahydrobiopterin Protects Against Hypertrophic Heart Disease Independent of Myocardial Nitric Oxide Synthase Coupling. J Am Heart Assoc 2016; 5:e003208. [PMID: 27001967 PMCID: PMC4943286 DOI: 10.1161/jaha.116.003208] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/11/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nitric oxide synthase uncoupling occurs under conditions of oxidative stress modifying the enzyme's function so it generates superoxide rather than nitric oxide. Nitric oxide synthase uncoupling occurs with chronic pressure overload, and both are ameliorated by exogenous tetrahydrobiopterin (BH4)-a cofactor required for normal nitric oxide synthase function-supporting a pathophysiological link. Genetically augmenting BH4 synthesis in endothelial cells fails to replicate this benefit, indicating that other cell types dominate the effects of exogenous BH4 administration. We tested whether the primary cellular target of BH4 is the cardiomyocyte or whether other novel mechanisms are invoked. METHODS AND RESULTS Mice with cardiomyocyte-specific overexpression of GTP cyclohydrolase 1 (mGCH1) and wild-type littermates underwent transverse aortic constriction. The mGCH1 mice had markedly increased myocardial BH4 and, unlike wild type, maintained nitric oxide synthase coupling after transverse aortic constriction; however, the transverse aortic constriction-induced abnormalities in cardiac morphology and function were similar in both groups. In contrast, exogenous BH4 supplementation improved transverse aortic constricted hearts in both groups, suppressed multiple inflammatory cytokines, and attenuated infiltration of inflammatory macrophages into the heart early after transverse aortic constriction. CONCLUSIONS BH4 protection against adverse remodeling in hypertrophic cardiac disease is not driven by its prevention of myocardial nitric oxide synthase uncoupling, as presumed previously. Instead, benefits from exogenous BH4 are mediated by a protective effect coupled to suppression of inflammatory pathways and myocardial macrophage infiltration.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Biopterins/analogs & derivatives
- Biopterins/pharmacology
- Cardiovascular Agents/pharmacology
- Cytokines/metabolism
- Cytoprotection
- Disease Models, Animal
- GTP Cyclohydrolase/genetics
- GTP Cyclohydrolase/metabolism
- Humans
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Inflammation Mediators/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Mice, Transgenic
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/metabolism
- Oxidation-Reduction
- Signal Transduction
- Superoxides/metabolism
- Time Factors
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Toru Hashimoto
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Vidhya Sivakumaran
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | | | - Guangshuo Zhu
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Virginia S Hahn
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Djahida Bedja
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Alice Recalde
- Department of Cardiovascular Medicine, University of Oxford, UK
| | - Drew Duglan
- Department of Cardiovascular Medicine, University of Oxford, UK
| | - Keith M Channon
- Department of Cardiovascular Medicine, University of Oxford, UK
| | - Barbara Casadei
- Department of Cardiovascular Medicine, University of Oxford, UK
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
30
|
Abstract
Despite the growing number of patients affected, the understanding of diastolic dysfunction and heart failure with preserved ejection fraction (HFpEF) is still poor. Clinical trials, largely based on successful treatments for systolic heart failure, have been disappointing, suggesting that HFpEF has a different pathology to that of systolic dysfunction. In this review, general concepts, epidemiology, diagnosis, and treatment of diastolic dysfunction are summarized, with an emphasis on new experiments suggesting that oxidative stress plays a crucial role in the pathogenesis of at least some forms of the disease. This observation has lead to potential new diagnostics and therapeutics for diastolic dysfunction and heart failure caused by diastolic dysfunction.
Collapse
Affiliation(s)
- Euy-Myoung Jeong
- Cardiovascular Research Center and Cardiovascular Institute of Lifespan, The Warren Alpert Medical School, Brown University
| | | |
Collapse
|
31
|
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: 1.5] [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
|
32
|
Bendall JK, Douglas G, McNeill E, Channon KM, Crabtree MJ. Tetrahydrobiopterin in cardiovascular health and disease. Antioxid Redox Signal 2014; 20:3040-77. [PMID: 24294830 PMCID: PMC4038990 DOI: 10.1089/ars.2013.5566] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/01/2013] [Accepted: 12/02/2013] [Indexed: 01/03/2023]
Abstract
Tetrahydrobiopterin (BH4) functions as a cofactor for several important enzyme systems, and considerable evidence implicates BH4 as a key regulator of endothelial nitric oxide synthase (eNOS) in the setting of cardiovascular health and disease. BH4 bioavailability is determined by a balance of enzymatic de novo synthesis and recycling, versus degradation in the setting of oxidative stress. Augmenting vascular BH4 levels by pharmacological supplementation has been shown in experimental studies to enhance NO bioavailability. However, it has become more apparent that the role of BH4 in other enzymatic pathways, including other NOS isoforms and the aromatic amino acid hydroxylases, may have a bearing on important aspects of vascular homeostasis, inflammation, and cardiac function. This article reviews the role of BH4 in cardiovascular development and homeostasis, as well as in pathophysiological processes such as endothelial and vascular dysfunction, atherosclerosis, inflammation, and cardiac hypertrophy. We discuss the therapeutic potential of BH4 in cardiovascular disease states and attempt to address how this modulator of intracellular NO-redox balance may ultimately provide a powerful new treatment for many cardiovascular diseases.
Collapse
Affiliation(s)
- Jennifer K Bendall
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford , John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | |
Collapse
|
33
|
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: 5.3] [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
|
34
|
Hong KU, Moore JB. Recent advances in cardiac myocyte biology and function. Circ Res 2013; 113:e121-4. [PMID: 24311621 DOI: 10.1161/circresaha.113.302990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kyung U Hong
- From the Department of Medicine, Institute of Molecular Cardiology (K.U.H., J.B.M.) and Department of Medicine, Diabetes and Obesity Center (K.U.H.), University of Louisville, Louisville, KY
| | | |
Collapse
|
35
|
Idigo WO, Reilly S, Zhang MH, Zhang YH, Jayaram R, Carnicer R, Crabtree MJ, Balligand JL, Casadei B. Regulation of endothelial nitric-oxide synthase (NOS) S-glutathionylation by neuronal NOS: evidence of a functional interaction between myocardial constitutive NOS isoforms. J Biol Chem 2012; 287:43665-73. [PMID: 23091050 DOI: 10.1074/jbc.m112.412031] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Myocardial constitutive No production depends on the activity of both endothelial and neuronal NOS (eNOS and nNOS, respectively). Stimulation of myocardial β(3)-adrenergic receptor (β(3)-AR) produces a negative inotropic effect that is dependent on eNOS. We evaluated whether nNOS also plays a role in β(3)-AR signaling and found that the β(3)-AR-mediated reduction in cell shortening and [Ca(2+)](i) transient amplitude was abolished both in eNOS(-/-) and nNOS(-/-) left ventricular (LV) myocytes and in wild type LV myocytes after nNOS inhibition with S-methyl-L-thiocitrulline. LV superoxide (O(2)(·-)) production was increased in nNOS(-/-) mice and reduced by L-N(ω)-nitroarginine methyl ester (L-NAME), indicating uncoupling of eNOS activity. eNOS S-glutathionylation and Ser-1177 phosphorylation were significantly increased in nNOS(-/-) myocytes, whereas myocardial tetrahydrobiopterin, eNOS Thr-495 phosphorylation, and arginase activity did not differ between genotypes. Although inhibitors of xanthine oxidoreductase (XOR) or NOX2 NADPH oxidase caused a similar reduction in myocardial O(2)(·-), only XOR inhibition reduced eNOS S-glutathionylation and Ser-1177 phosphorylation and restored both eNOS coupled activity and the negative inotropic and [Ca(2+)](i) transient response to β(3)-AR stimulation in nNOS(-/-) mice. In summary, our data show that increased O(2)(·-) production by XOR selectively uncouples eNOS activity and abolishes the negative inotropic effect of β(3)-AR stimulation in nNOS(-/-) myocytes. These findings provide unequivocal evidence of a functional interaction between the myocardial constitutive NOS isoforms and indicate that aspects of the myocardial phenotype of nNOS(-/-) mice result from disruption of eNOS signaling.
Collapse
Affiliation(s)
- Winifred O Idigo
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Niggli E, Ullrich ND, Gutierrez D, Kyrychenko S, Poláková E, Shirokova N. Posttranslational modifications of cardiac ryanodine receptors: Ca(2+) signaling and EC-coupling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:866-75. [PMID: 22960642 DOI: 10.1016/j.bbamcr.2012.08.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 10/27/2022]
Abstract
In cardiac muscle, a number of posttranslational protein modifications can alter the function of the Ca(2+) release channel of the sarcoplasmic reticulum (SR), also known as the ryanodine receptor (RyR). During every heartbeat RyRs are activated by the Ca(2+)-induced Ca(2+) release mechanism and contribute a large fraction of the Ca(2+) required for contraction. Some of the posttranslational modifications of the RyR are known to affect its gating and Ca(2+) sensitivity. Presently, research in a number of laboratories is focused on RyR phosphorylation, both by PKA and CaMKII, or on RyR modifications caused by reactive oxygen and nitrogen species (ROS/RNS). Both classes of posttranslational modifications are thought to play important roles in the physiological regulation of channel activity, but are also known to provoke abnormal alterations during various diseases. Only recently it was realized that several types of posttranslational modifications are tightly connected and form synergistic (or antagonistic) feed-back loops resulting in additive and potentially detrimental downstream effects. This review summarizes recent findings on such posttranslational modifications, attempts to bridge molecular with cellular findings, and opens a perspective for future work trying to understand the ramifications of crosstalk in these multiple signaling pathways. Clarifying these complex interactions will be important in the development of novel therapeutic approaches, since this may form the foundation for the implementation of multi-pronged treatment regimes in the future. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
Affiliation(s)
- Ernst Niggli
- Department of Physiology, University of Bern, Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|