1
|
Pang B, Qi X, Zhang H. Salivary-Gland-Mediated Nitrate Recirculation as a Modulator for Cardiovascular Diseases. Biomolecules 2025; 15:439. [PMID: 40149975 PMCID: PMC11940199 DOI: 10.3390/biom15030439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular diseases (CVDs), which include multiple disorders of the heart and blood vessels, are the leading causes of death. Nitric oxide (NO) is a vasodilator that regulates vascular tension. Endogenous NO is produced via the L-arginine-nitric oxide synthase (NOS) pathway. In conditions of cardiovascular dysfunction, NOS activity is impaired, leading to NO deficiency. In turn, the reduction in NO bioactivity exacerbates the pathogenesis of CVDs. Exogenous intake of inorganic nitrate supplements endogenous production via the nitrate-nitrite-NO pathway to maintain the NO supply. Salivary glands play an essential role in the conversion of nitrate to NO, with approximately 25% of circulating nitrate being absorbed and secreted into saliva. As a result, salivary nitrate concentrations can exceed that in the blood by more than tenfold. This recycled nitrate in saliva serves as a reservoir for NO and performs NO-like functions when endogenous NO production is insufficient. In this review, we summarize the emerging benefits of dietary nitrate in CVDs, with a particular focus on salivary-gland-mediated nitrate recirculation in maintaining NO bioavailability and cardiovascular homeostasis. Salivary-gland-mediated nitrate recirculation provides a novel perspective for potential intervention of CVDs.
Collapse
Affiliation(s)
- Baoxing Pang
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xingyun Qi
- Department of Biology, Rutgers University, Camden, NJ 08103, USA
| | - Huiliang Zhang
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
2
|
Kabanov D, Vrana Klimovic S, Beckerová D, Molcan M, Scurek M, Brat K, Bebarova M, Rotrekl V, Pribyl J, Pesl M. Salbutamol attenuates arrhythmogenic effect of aminophylline in a hPSC-derived cardiac model. Sci Rep 2024; 14:27399. [PMID: 39521810 PMCID: PMC11550379 DOI: 10.1038/s41598-024-76846-4] [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: 02/29/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
The combination of aminophylline and salbutamol is frequently used in clinical practice in the treatment of obstructive lung diseases. While the side effects (including arrhythmias) of the individual bronchodilator drugs were well described previously, the side effects of combined treatment are almost unknown. We aimed to study the arrhythmogenic potential of combined aminophylline and salbutamol treatment in vitro. For this purpose, we used the established atomic force microscopy (AFM) model coupled with cardiac organoids derived from human pluripotent stem cells (hPSC-CMs). We focused on the chronotropic, inotropic, and arrhythmogenic effects of salbutamol alone and aminophylline and salbutamol combined treatment. We used a method based on heart rate/beat rate variability (HRV/BRV) analysis to detect arrhythmic events in the hPSC-CM based AFM recordings. Salbutamol and aminophylline had a synergistic chronotropic and inotropic effect compared to the effects of monotherapy. Our main finding was that salbutamol reduced the arrhythmogenic effect of aminophylline, most likely mediated by endothelial nitric oxide synthase activated by beta-2 adrenergic receptors. These findings were replicated and confirmed using hPSC-CM derived from two cell lines (CCTL4 and CCTL12). Data suggest that salbutamol as an add-on therapy may not only deliver a bronchodilator effect but also increase the cardiovascular safety of aminophylline, as salbutamol reduces its arrhythmogenic potential.
Collapse
Grants
- A4L_ACTIONS 964997 Horizon 2020
- A4L_ACTIONS 964997 Horizon 2020
- A4L_ACTIONS 964997 Horizon 2020
- and CIISB, Instruct-CZ Centre of Instruct-ERIC EU consortium LM2023042 Ministerstvo Školství, Mládeže a Tělovýchovy
- and CIISB, Instruct-CZ Centre of Instruct-ERIC EU consortium LM2023042 Ministerstvo Školství, Mládeže a Tělovýchovy
- MUNI/A/1547/2023 Ministerstvo Školství, Mládeže a Tělovýchovy
- MUNI/A/1547/2023 Ministerstvo Školství, Mládeže a Tělovýchovy
- MUNI/A/1547/2023 Ministerstvo Školství, Mládeže a Tělovýchovy
- and CIISB, Instruct-CZ Centre of Instruct-ERIC EU consortium LM2023042 Ministerstvo Školství, Mládeže a Tělovýchovy
- MUNI/A/1547/2023 Ministerstvo Školství, Mládeže a Tělovýchovy
- UP CIISB" (No. CZ.02.1.01/0.0/0.0/18_046/0015974), European Regional Development Fund
- UP CIISB" (No. CZ.02.1.01/0.0/0.0/18_046/0015974), European Regional Development Fund
- UP CIISB" (No. CZ.02.1.01/0.0/0.0/18_046/0015974), European Regional Development Fund
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- NU20-06-001 Ministerstvo Zdravotnictví Ceské Republiky
- A4L_Bridge101136453 HORIZON EUROPE European Research Council
- MUQUABIS GA no. 101070546 HORIZON EUROPE European Research Council
- A4L_Bridge101136453 HORIZON EUROPE European Research Council
- A4L_Bridge101136453 HORIZON EUROPE European Research Council
- EXCELES, No. LX22NPO5104 HORIZON EUROPE Framework Programme
- EXCELES, No. LX22NPO5104 HORIZON EUROPE Framework Programme
Collapse
Affiliation(s)
- Daniil Kabanov
- CEITEC MU, Masaryk University, Brno, Czech Republic
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Simon Vrana Klimovic
- CEITEC MU, Masaryk University, Brno, Czech Republic
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Deborah Beckerová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Martin Molcan
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Martin Scurek
- Department of Respiratory Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kristian Brat
- Department of Respiratory Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marketa Bebarova
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jan Pribyl
- CEITEC MU, Masaryk University, Brno, Czech Republic.
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Martin Pesl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
- First Department of Internal Medicine - Cardioangiology, Faculty of Medicine, Masaryk University, St. Anne's University Hospital, Brno, Czech Republic.
| |
Collapse
|
3
|
Guo YJ, Yao JJ, Guo ZZ, Ding M, Zhang KL, Shen QH, Li Y, Yu SF, Wan T, Xu FP, Wang Y, Qi XX, Wu JJ, Chen JX, Liu ZQ, Lu LL. HBB contributes to individualized aconitine-induced cardiotoxicity in mice via interfering with ABHD5/AMPK/HDAC4 axis. Acta Pharmacol Sin 2024; 45:1224-1236. [PMID: 38467717 PMCID: PMC11130212 DOI: 10.1038/s41401-023-01206-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/19/2023] [Indexed: 03/13/2024]
Abstract
The root of Aconitum carmichaelii Debx. (Fuzi) is an herbal medicine used in China that exerts significant efficacy in rescuing patients from severe diseases. A key toxic compound in Fuzi, aconitine (AC), could trigger unpredictable cardiotoxicities with high-individualization, thus hinders safe application of Fuzi. In this study we investigated the individual differences of AC-induced cardiotoxicities, the biomarkers and underlying mechanisms. Diversity Outbred (DO) mice were used as a genetically heterogeneous model for mimicking individualization clinically. The mice were orally administered AC (0.3, 0.6, 0.9 mg· kg-1 ·d-1) for 7 d. We found that AC-triggered cardiotoxicities in DO mice shared similar characteristics to those observed in clinic patients. Most importantly, significant individual differences were found in DO mice (variation coefficients: 34.08%-53.17%). RNA-sequencing in AC-tolerant and AC-sensitive mice revealed that hemoglobin subunit beta (HBB), a toxic-responsive protein in blood with 89% homology to human, was specifically enriched in AC-sensitive mice. Moreover, we found that HBB overexpression could significantly exacerbate AC-induced cardiotoxicity while HBB knockdown markedly attenuated cell death of cardiomyocytes. We revealed that AC could trigger hemolysis, and specifically bind to HBB in cell-free hemoglobin (cf-Hb), which could excessively promote NO scavenge and decrease cardioprotective S-nitrosylation. Meanwhile, AC bound to HBB enhanced the binding of HBB to ABHD5 and AMPK, which correspondingly decreased HDAC-NT generation and led to cardiomyocytes death. This study not only demonstrates HBB achievement a novel target of AC in blood, but provides the first clue for HBB as a novel biomarker in determining the individual differences of Fuzi-triggered cardiotoxicity.
Collapse
Affiliation(s)
- Ya-Juan Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jing-Jing Yao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhen-Zhen Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ming Ding
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Kun-Lin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qing-Hong Shen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yu Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shao-Fang Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ting Wan
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fu-Ping Xu
- Guandong Provincial hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiao-Xiao Qi
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jin-Jun Wu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jian-Xin Chen
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Lin-Lin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Abolfazli S, Mortazavi P, Kheirandish A, Butler AE, Jamialahmadi T, Sahebkar A. Regulatory effects of curcumin on nitric oxide signaling in the cardiovascular system. Nitric Oxide 2024; 143:16-28. [PMID: 38141926 DOI: 10.1016/j.niox.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
The continuously rising prevalence of cardiovascular disease (CVD) globally substantially impacts the economic growth of developing countries. Indeed, one of the leading causes of death worldwide is unfavorable cardiovascular events. Reduced nitric oxide (NO) generation is the pathogenic foundation of endothelial dysfunction, which is regarded as the first stage in the development of a number of CVDs. Nitric oxide exerts an array of biological effects, including vasodilation, the suppression of vascular smooth muscle cell proliferation and the functional control of cardiac cells. Numerous treatment strategies aim to increase NO synthesis or upregulate downstream NO signaling pathways. The major component of Curcuma longa, curcumin, has long been utilized in traditional medicine to treat various illnesses, especially CVDs. Curcumin improves CV function as well as having important pleiotropic effects, such as anti-inflammatory and antioxidant, through its ability to increase the bioavailability of NO and to positively impact NO-related signaling pathways. In this review, we discuss the scientific literature relating to curcumin's positive effects on NO signaling and vascular endothelial function.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Parham Mortazavi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Kheirandish
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, PO Box, 15503, Adliya, Bahrain
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Pant J, Agarwal R, S S, Mohan L. Acute Exposure to Bisphenol S Decreases In Vitro Right Atrial Contractility in Rats. Cureus 2023; 15:e51387. [PMID: 38292991 PMCID: PMC10826249 DOI: 10.7759/cureus.51387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2023] [Indexed: 02/01/2024] Open
Abstract
AIM/OBJECTIVE Bisphenols are widely used in the manufacturing of polycarbonate material and epoxy resins which constitute the essential component of plastic. Bisphenol A (BPA) has been reported to produce toxicity on organs in both animal and human studies. Therefore, plastic manufacturers are replacing BPA with other analogues that are considered to be safe, and BPA-free products are now available in the market. However, some studies have reported that bisphenol-S (BPS) also possesses toxic properties. It has been reported to depress ventricular contraction as well as produce ventricular arrhythmia on acute exposure. The present study was performed to examine the effect of BPS on in vitro spontaneously-beating right atria in rats. METHODS In the present study, in vitro spontaneous contractions of right atria obtained from adult female rats of the Wistar strain were recorded. The atria were exposed to BPS (10-6-10 mM) and its effects on atrial contractions were recorded in the form of cumulative-concentration response with and without administration of antagonists namely atropine, L-NAME, and methylene blue. RESULTS BPS decreased the rate as well as the force of atrial contractions. The changes produced in the rate and force of atrial contractions were not attributed to ethanol, which was used to prepare BPS solutions. The decrease in right atrial contractility produced by BPS was blocked by L-NAME; however, atropine and methylene blue were not able to antagonize the effects of BPS on atria. CONCLUSIONS The present study indicates the involvement of NO-dependent but cGMP independent pathway responsible for BPS-induced cardio-toxicity.
Collapse
Affiliation(s)
- Jayanti Pant
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Radhika Agarwal
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Srikant S
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Latika Mohan
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| |
Collapse
|
6
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
7
|
Koya T, Watanabe M, Natsui H, Kadosaka T, Koizumi T, Nakao M, Hagiwara H, Kamada R, Temma T, Anzai T. Pharmacological nNOS inhibition modified small-conductance Ca 2+-activated K + channel without altering Ca 2+ dynamics. Am J Physiol Heart Circ Physiol 2022; 323:H869-H878. [PMID: 36149772 DOI: 10.1152/ajpheart.00252.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Atrial fibrillation (AF) is associated with electrical remodeling processes that promote a substrate for the maintenance of AF. Although the small-conductance Ca2+-activated K+ (SK) channel is a key factor in atrial electrical remodeling, the mechanism of its activation remains unclear. Regional nitric oxide (NO) production by neuronal nitric oxide synthase (nNOS) is involved in atrial electrical remodeling. In this study, atrial tachyarrhythmia (ATA) induction and optical mapping were performed on perfused rat hearts. nNOS is pharmacologically inhibited by S-methylthiocitrulline (SMTC). The influence of the SK channel was examined using a specific channel inhibitor, apamin (APA). Parameters such as action potential duration (APD), conduction velocity, and calcium transient (CaT) were evaluated using voltage and calcium optical mapping. The dominant frequency was examined in the analysis of AF dynamics. SMTC (100 nM) increased the inducibility of ATA and apamin (100 nM) mitigated nNOS inhibition-induced arrhythmogenicity. SMTC caused abbreviations and enhanced the spatial dispersion of APD, which was reversed by apamin. By contrast, conduction velocity and other parameters associated with CaT were not affected by SMTC or apamin administration. Apamin reduced the frequency of SMTC-induced ATA. In summary, nNOS inhibition abbreviates APD by modifying the SK channels. A specific SK channel blocker, apamin, mitigated APD abbreviation without alteration of CaT, implying an underlying mechanism of posttranslational modification of SK channels.NEW & NOTEWORTHY We demonstrated that pharmacological nNOS inhibition increased the atrial arrhythmia inducibility and a specific small-conductance Ca2+-activated K+ channel blocker, apamin, reversed the enhanced atrial arrhythmia inducibility. Apamin mitigated APD abbreviation without alteration of Ca2+ transient, implying an underlying mechanism of posttranslational modification of SK channels.
Collapse
Affiliation(s)
- Taro Koya
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaya Watanabe
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Natsui
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Kadosaka
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Koizumi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Motoki Nakao
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hikaru Hagiwara
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rui Kamada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taro Temma
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
8
|
Imani A, Rajani SF, Rakhshan K, Faghihi M, Nemati M, Parsazadegan T. The role of nitric oxide on the antiarrhythmic effects of ketamine/xylazine in a rat model of acute cardiac ischemia-reperfusion. Curr Res Physiol 2022; 5:302-311. [PMID: 35856058 PMCID: PMC9287742 DOI: 10.1016/j.crphys.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/21/2022] [Accepted: 06/30/2022] [Indexed: 11/10/2022] Open
Abstract
The prevalence of ventricular arrhythmias during general anesthesia is about 70%. In experimental studies on the antiarrhythmic effects of different agents, using anesthetic drugs that do not have any protective properties are preferable. The present study was conducted to investigate molecular mechanisms involved in the antiarrhythmic effects of ketamine/xylazine (K/X). Sixty male rats were assigned to eight groups: K/X, L -NAME (25–35 mg/kg) with thiopental (TP), L-NAME (25–35 mg/kg) with ketamine/xylazine, L arginine (100 mg/kg) with thiopental, L-arginine (100 mg/kg) with ketamine/xylazine. After anesthetic induction using TP or K/X, the animals were subjected to 30 min of ischemia. Hemodynamic parameters, ventricular arrhythmias during ischemia, the incidence of ventricular tachycardia (VT), and ventricular fibrillation (VF) were measured. Additionally, in order to assess nitrite/nitrate ratio and LDH after ischemia, serum samples were collected and used. Our results showed that in the K/X group, the number of VT and VF, duration of VT (p = 0.006), the severity of arrhythmias (p = 0.0179). There was no VF incidence in this group. These protective effects were faded by administration of L-NAME with K/X. The combination of L- Arginine in the TP group decreased the number and duration of VT (p < 0.001, p = 0.0013) with no incidence of VF in comparison with TP. L-arginine with K/X groups increased the number and duration of VT (p < 0.0001, p < 0.001) compared to K/X and VF was seen (100%). However, there was no significant difference between TP and K/X groups in terms of this nitrite/nitrate ratio. These findings suggest that the antiarrhythmic effects of ketamine/xylazine might be partially relative to the nitric oxide synthesis pathway. The prevalence of ventricular arrhythmias during general anesthesia is about 70%. ketamine/xylazine as common anesthetic agents have antiarrhythmic properties. The antiarrhythmic effects of ketamine/xylazine might be partially relative to the nitric oxide synthesis pathway.
Collapse
|
9
|
Xiao J, Zhang Y, Tang Y, Dai H, OuYang Y, Li C, Yu M. MiRNA-1202 promotes the TGF-β1-induced proliferation, differentiation and collagen production of cardiac fibroblasts by targeting nNOS. PLoS One 2021; 16:e0256066. [PMID: 34428251 PMCID: PMC8384215 DOI: 10.1371/journal.pone.0256066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
Background Atrial fibrillation (AF) is a clinically common arrhythmia that affects human health. Myocardial fibrosis serves as an important contributor to AF. Recently, miRNA-1202 have been reported to be up-regulated in AF. However, the role of miRNA-1202 and its mechanism in myocardial fibrosis remain unclear. Methods Human cardiac fibroblasts (HCFs) were used to construct a fibrosis model by TGF-β1 induction. The expression of miR-1202 was measured by qRT-PCR. Cell proliferation was assessed by CCK-8 assays. Protein expression levels were measured by western blot. Collagen accumulation was measured by ELISA. The relationship between miR-1202 and nNOS was investigated by luciferase reporter assays. Results MiR-1202 expression was obviously increased in HCFs and was both time- and dose-independent. MiR-1202 could increase the proliferation and collagen I, collagen III, and α-SMA levels with or without TGF-β1. MiR-1202 could also increase TGF-β1 and p-Smad2/3 protein levels in comparison to the control group. However, they were obviously decreased after inhibitor transfection. MiR-1202 targets nNOS for negative regulation of HCFs fibrosis by decreasing cell differentiation, collagen deposition and the activity of the TGF-β1/Smad2/3 pathway. Co-transfection of miR-1202 inhibitor and siRNA of nNOS inhibited nNOS protein expression, thereby enhancing the HCFs proliferation. Furthermore, co-transfection of the miR-1202 inhibitor and siRNA of nNOS significantly promoted collagen I, collagen III, TGF-β1, Smad2/3 and α-SMA protein expression and Smad2/3 protein phosphorylation. These findings suggested that miR-1202 promotes HCFs transformation to a pro-fibrotic phenotype by targeting nNOS through activating the TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Jingwen Xiao
- The Department of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| | - Yan Zhang
- The Department of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
- * E-mail:
| | - Yuan Tang
- The Cardiac Function Laboratory of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| | - Hengfen Dai
- The Department of Clinical Pharmacy, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| | - Yu OuYang
- The Department of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| | - Chuanchuan Li
- The Department of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| | - Meiqin Yu
- The Cardiac Function Laboratory of Cardiovascular Medicine, FuZhou First Hospital, FuZhou, Fujian, P.R. China
| |
Collapse
|
10
|
Hegyi B, Shimkunas R, Jian Z, Izu LT, Bers DM, Chen-Izu Y. Mechanoelectric coupling and arrhythmogenesis in cardiomyocytes contracting under mechanical afterload in a 3D viscoelastic hydrogel. Proc Natl Acad Sci U S A 2021; 118:e2108484118. [PMID: 34326268 PMCID: PMC8346795 DOI: 10.1073/pnas.2108484118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The heart pumps blood against the mechanical afterload from arterial resistance, and increased afterload may alter cardiac electrophysiology and contribute to life-threatening arrhythmias. However, the cellular and molecular mechanisms underlying mechanoelectric coupling in cardiomyocytes remain unclear. We developed an innovative patch-clamp-in-gel technology to embed cardiomyocytes in a three-dimensional (3D) viscoelastic hydrogel that imposes an afterload during regular myocyte contraction. Here, we investigated how afterload affects action potentials, ionic currents, intracellular Ca2+ transients, and cell contraction of adult rabbit ventricular cardiomyocytes. We found that afterload prolonged action potential duration (APD), increased transient outward K+ current, decreased inward rectifier K+ current, and increased L-type Ca2+ current. Increased Ca2+ entry caused enhanced Ca2+ transients and contractility. Moreover, elevated afterload led to discordant alternans in APD and Ca2+ transient. Ca2+ alternans persisted under action potential clamp, indicating that the alternans was Ca2+ dependent. Furthermore, all these afterload effects were significantly attenuated by inhibiting nitric oxide synthase 1 (NOS1). Taken together, our data reveal a mechano-chemo-electrotransduction (MCET) mechanism that acutely transduces afterload through NOS1-nitric oxide signaling to modulate the action potential, Ca2+ transient, and contractility. The MCET pathway provides a feedback loop in excitation-Ca2+ signaling-contraction coupling, enabling autoregulation of contractility in cardiomyocytes in response to afterload. This MCET mechanism is integral to the individual cardiomyocyte (and thus the heart) to intrinsically enhance its contractility in response to the load against which it has to do work. While this MCET is largely compensatory for physiological load changes, it may also increase susceptibility to arrhythmias under excessive pathological loading.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616;
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| |
Collapse
|
11
|
Wang P, Wei M, Zhu X, Liu Y, Yoshimura K, Zheng M, Liu G, Kume S, Morishima M, Kurokawa T, Ono K. Nitric oxide down-regulates voltage-gated Na + channel in cardiomyocytes possibly through S-nitrosylation-mediated signaling. Sci Rep 2021; 11:11273. [PMID: 34050231 PMCID: PMC8163867 DOI: 10.1038/s41598-021-90840-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
Nitric oxide (NO) is produced from endothelial cells and cardiomyocytes composing the myocardium and benefits cardiac function through both vascular-dependent and—independent effects. This study was purposed to investigate the possible adverse effect of NO focusing on the voltage-gated Na+ channel in cardiomyocytes. We carried out patch-clamp experiments on rat neonatal cardiomyocytes demonstrating that NOC-18, an NO donor, significantly reduced Na+ channel current in a dose-dependent manner by a long-term application for 24 h, accompanied by a reduction of Nav1.5-mRNA and the protein, and an increase of a transcription factor forkhead box protein O1 (FOXO1) in the nucleus. The effect of NOC-18 on the Na+ channel was blocked by an inhibitor of thiol oxidation N-ethylmaleimide, a disulfide reducing agent disulfide 1,4-Dithioerythritol, or a FOXO1 activator paclitaxel, suggesting that NO is a negative regulator of the voltage-gated Na+ channel through thiols in regulatory protein(s) for the channel transcription.
Collapse
Affiliation(s)
- Pu Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Mengyan Wei
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Xiufang Zhu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Yangong Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China.,Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Kenshi Yoshimura
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, 050031, Hebei Province, People's Republic of China
| | - Shinichiro Kume
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Masaki Morishima
- Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Tatsuki Kurokawa
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan
| | - Katsushige Ono
- Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, 879-5593, Japan.
| |
Collapse
|
12
|
Infante T, Costa D, Napoli C. Novel Insights Regarding Nitric Oxide and Cardiovascular Diseases. Angiology 2021; 72:411-425. [PMID: 33478246 DOI: 10.1177/0003319720979243] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a powerful mediator with biological activities such as vasodilation and prevention of vascular smooth muscle cell proliferation as well as functional regulation of cardiac cells. Thus, impaired production or reduced bioavailability of NO predisposes to the onset of different cardiovascular (CV) diseases. Alterations in the redox balance associated with excitation-contraction coupling have been identified in heart failure (HF), thus contributing to contractile abnormalities and arrhythmias. For its ability to influence cell proliferation and angiogenesis, NO may be considered a therapeutic option for the management of several CV diseases. Several clinical studies and trials investigated therapeutic NO strategies for systemic hypertension, atherosclerosis, and/or prevention of in stent restenosis, coronary heart disease (CHD), pulmonary arterial hypertension (PAH), and HF, although with mixed results in long-term treatment and effective dose administered in selected groups of patients. Tadalafil, sildenafil, and cinaguat were evaluated for the treatment of PAH, whereas vericiguat was investigated in the treatment of HF patients with reduced ejection fraction. Furthermore, supplementation with hydrogen sulfide, tetrahydrobiopterin, and nitrite/nitrate has shown beneficial effects at the vascular level.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, 18994University of Campania "L. Vanvitelli," Naples, Italy
| | - Claudio Napoli
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
13
|
Boycott HE, Nguyen MN, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Front Physiol 2020; 11:606740. [PMID: 33384614 PMCID: PMC7770138 DOI: 10.3389/fphys.2020.606740] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
The ability§ of the heart to adapt to changes in the mechanical environment is critical for normal cardiac physiology. The role of nitric oxide is increasingly recognized as a mediator of mechanical signaling. Produced in the heart by nitric oxide synthases, nitric oxide affects almost all mechano-transduction pathways within the cardiomyocyte, with roles mediating mechano-sensing, mechano-electric feedback (via modulation of ion channel activity), and calcium handling. As more precise experimental techniques for applying mechanical stresses to cells are developed, the role of these forces in cardiomyocyte function can be further understood. Furthermore, specific inhibitors of different nitric oxide synthase isoforms are now available to elucidate the role of these enzymes in mediating mechano-electrical signaling. Understanding of the links between nitric oxide production and mechano-electrical signaling is incomplete, particularly whether mechanically sensitive ion channels are regulated by nitric oxide, and how this affects the cardiac action potential. This is of particular relevance to conditions such as atrial fibrillation and heart failure, in which nitric oxide production is reduced. Dysfunction of the nitric oxide/mechano-electrical signaling pathways are likely to be a feature of cardiac pathology (e.g., atrial fibrillation, cardiomyopathy, and heart failure) and a better understanding of the importance of nitric oxide signaling and its links to mechanical regulation of heart function may advance our understanding of these conditions.
Collapse
Affiliation(s)
- Hannah E. Boycott
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Piccirillo G, Moscucci F, Bertani G, Lospinuso I, Mastropietri F, Fabietti M, Sabatino T, Zaccagnini G, Crapanzano D, Di Diego I, Corrao A, Rossi P, Magrì D. Short-Period Temporal Dispersion Repolarization Markers Predict 30-Days Mortality in Decompensated Heart Failure. J Clin Med 2020; 9:E1879. [PMID: 32560151 PMCID: PMC7356287 DOI: 10.3390/jcm9061879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Electrocardiographic (ECG) markers of the temporal dispersion of the myocardial repolarization phase have been shown able to identify chronic heart failure (CHF) patients at high mortality risk. The present prospective single-center study sought to investigate in a well-characterized cohort of decompensated heart failure (HF) patients the ability of short-term myocardial temporal dispersion ECG variables in predicting the 30-day mortality, as well as their relationship with N-terminal Pro Brain Natriuretic Peptide (NT-proBNP) plasmatic values. METHOD One hundred and thirteen subjects (male: 59, 67.8%) with decompensated CHF underwent 5 min of ECG recording, via a mobile phone. We obtained QT end (QTe), QT peak (QTp) and T peak to T end (Te) and calculated the mean, standard deviation (SD), and normalized index (VN). RESULTS Death occurred for 27 subjects (24%) within 30 days after admission. Most of the repolarization indexes (QTe mean (p < 0.05), QTeSD (p < 0.01), QTpSD (p < 0.05), mean Te (p < 0.05), TeSD (p < 0.001) QTeVN (p < 0.05) and TeVN (p < 0.01)) were significantly higher in those CHF patients with the highest NT-proBNP (>75th percentile). In all the ECG data, only TeSD was significantly and positively related to the NT-proBNP levels (r: 0.471; p < 0.001). In the receiver operating characteristic (ROC) analysis, the highest accuracy for 30-day mortality was found for QTeSD (area under curve, AUC: 0.705, p < 0.01) and mean Te (AUC: 0.680, p < 0.01), whereas for the NT-proBNP values higher than the 75th percentile, the highest accuracy was found for TeSD (AUC: 0.736, p < 0.001) and QTeSD (AUC: 0.696, p < 0.01). CONCLUSION Both mean Te and TeSD could be considered as reliable markers of worsening HF and of 30-day mortality. Although larger and possibly interventional studies are needed to confirm our preliminary finding, these non-invasive and transmissible ECG parameters could be helpful in the remote monitoring of advanced HF patients and, possibly, in their clinical management. (ClinicalTrials.gov number, NCT04127162).
Collapse
Affiliation(s)
- Gianfranco Piccirillo
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Federica Moscucci
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Gaetano Bertani
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Ilaria Lospinuso
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Fabiola Mastropietri
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Marcella Fabietti
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Teresa Sabatino
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Giulia Zaccagnini
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Davide Crapanzano
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Ilaria Di Diego
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Andrea Corrao
- Anestesiologiche e Cardiovascolari, Dipartimento di Scienze Cliniche Internistiche, Policlinico Umberto I, La Sapienza University of Rome, 00185 Rome, Italy; (G.P.); (G.B.); (I.L.); (F.M.); (M.F.); (T.S.); (G.Z.); (D.C.); (I.D.D.); (A.C.)
| | - Pietro Rossi
- Cardiology Division, Arrhythmology Unit, S. Giovanni Calibita, Isola Tiberina, 00186 Rome, Italy;
| | - Damiano Magrì
- Dipartimento di Medicina Clinica e Molecolare, S. Andrea Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
15
|
Ca 2+ Channels Mediate Bidirectional Signaling between Sarcolemma and Sarcoplasmic Reticulum in Muscle Cells. Cells 2019; 9:cells9010055. [PMID: 31878335 PMCID: PMC7016941 DOI: 10.3390/cells9010055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
The skeletal muscle and myocardial cells present highly specialized structures; for example, the close interaction between the sarcoplasmic reticulum (SR) and mitochondria—responsible for excitation-metabolism coupling—and the junction that connects the SR with T-tubules, critical for excitation-contraction (EC) coupling. The mechanisms that underlie EC coupling in these two cell types, however, are fundamentally distinct. They involve the differential expression of Ca2+ channel subtypes: CaV1.1 and RyR1 (skeletal), vs. CaV1.2 and RyR2 (cardiac). The CaV channels transform action potentials into elevations of cytosolic Ca2+, by activating RyRs and thus promoting SR Ca2+ release. The high levels of Ca2+, in turn, stimulate not only the contractile machinery but also the generation of mitochondrial reactive oxygen species (ROS). This forward signaling is reciprocally regulated by the following feedback mechanisms: Ca2+-dependent inactivation (of Ca2+ channels), the recruitment of Na+/Ca2+ exchanger activity, and oxidative changes in ion channels and transporters. Here, we summarize both well-established concepts and recent advances that have contributed to a better understanding of the molecular mechanisms involved in this bidirectional signaling.
Collapse
|
16
|
Lillo MA, Himelman E, Shirokova N, Xie LH, Fraidenraich D, Contreras JE. S-nitrosylation of connexin43 hemichannels elicits cardiac stress-induced arrhythmias in Duchenne muscular dystrophy mice. JCI Insight 2019; 4:130091. [PMID: 31751316 DOI: 10.1172/jci.insight.130091] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/07/2019] [Indexed: 01/16/2023] Open
Abstract
Patients with Duchenne muscular dystrophy (DMD) commonly present with severe ventricular arrhythmias that contribute to heart failure. Arrhythmias and lethality are also consistently observed in adult Dmdmdx mice, a mouse model of DMD, after acute β-adrenergic stimulation. These pathological features were previously linked to aberrant expression and remodeling of the cardiac gap junction protein connexin43 (Cx43). Here, we report that remodeled Cx43 protein forms Cx43 hemichannels in the lateral membrane of Dmdmdx cardiomyocytes and that the β-adrenergic agonist isoproterenol (Iso) aberrantly activates these hemichannels. Block of Cx43 hemichannels or a reduction in Cx43 levels (using Dmdmdx Cx43+/- mice) prevents the abnormal increase in membrane permeability, plasma membrane depolarization, and Iso-evoked electrical activity in these cells. Additionally, Iso treatment promotes nitric oxide (NO) production and S-nitrosylation of Cx43 hemichannels in Dmdmdx heart. Importantly, inhibition of NO production prevents arrhythmias evoked by Iso. We found that NO directly activates Cx43 hemichannels by S-nitrosylation of cysteine at position 271. Our results demonstrate that opening of remodeled and S-nitrosylated Cx43 hemichannels plays a key role in the development of arrhythmias in DMD mice and that these channels may serve as therapeutic targets to prevent fatal arrhythmias in patients with DMD .
Collapse
Affiliation(s)
| | - Eric Himelman
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | |
Collapse
|
17
|
Demeter-Haludka V, Kovács M, Prorok J, Nagy N, Varró A, Végh Á. Examination of the Changes in Calcium Homeostasis in the Delayed Antiarrhythmic Effect of Sodium Nitrite. Int J Mol Sci 2019; 20:E5687. [PMID: 31766239 PMCID: PMC6888494 DOI: 10.3390/ijms20225687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/09/2019] [Accepted: 11/11/2019] [Indexed: 01/11/2023] Open
Abstract
We have evidence that the intravenous infusion of sodium nitrite (NaNO2) results in an antiarrhythmic effect when given 24 h prior to an ischemia and reperfusion (I/R) insult in anaesthetized dogs. This protection was associated with the reduction of reactive oxygen species resulting from I/R through the attenuation of mitochondrial respiration. Here, we examined whether the changes in calcium, which also contributes to arrhythmia generation, play a role in the NaNO2-induced effect. On the first day, 30 anaesthetized dogs were treated either with saline or NaNO2 (0.2 µmol/kg/min) for 20 min. Some animals were subjected to a 25 min LAD (anterior descending branch of the left coronary artery) occlusion and 2 min reperfusion (I/R = 4; NaNO2-I/R = 6), or the heart was removed 24 h later. We have shown that nitrite prevented the I/R-induced increase in cellular and mitochondrial calcium deposits. During simulated I/R, the amplitude of the calcium transient and the diastolic calcium level were significantly lower in the nitrite-treated hearts and the ERP (effective refractory period) fraction of the action potential was significantly increased. Furthermore, nitrite also enhanced the mitochondrial respiratory response and prevented the MPTPT opening during calcium overload. These results suggest that nitrite can reduce the harmful consequences of calcium overload, perhaps directly by modulating ion channels or indirectly by reducing the mitochondrial ROS (reactive oxygen species) production.
Collapse
Affiliation(s)
- Vivien Demeter-Haludka
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
| | - Mária Kovács
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
- MTA-SZTE Research Group of Cardiovascular Pharmacology, H-6721 Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
| | - Ágnes Végh
- Department of Pharmacology and Pharmacotherapy, University of Szeged, H-6721 Szeged, Hungary; (V.D.-H.); (J.P.); (N.N.); (A.V.); (Á.V.)
| |
Collapse
|
18
|
Bae H, Kim T, Lim I. Effects of nitric oxide on apoptosis and voltage-gated calcium channels in human cardiac myofibroblasts. Clin Exp Pharmacol Physiol 2019; 47:16-26. [PMID: 31519057 DOI: 10.1111/1440-1681.13178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/30/2019] [Accepted: 09/10/2019] [Indexed: 02/05/2023]
Abstract
We characterised the voltage-gated Ca2+ channels (VGCCs) in human cardiac fibroblasts (HCFs) and myofibroblasts (HCMFs) and investigated the effects of nitric oxide (NO) on apoptosis and on these channels. Western blotting and immunofluorescence analyses show that α-smooth muscle actin (a myofibroblast marker) was markedly expressed in passage (P) 12-15 but not in P4 HCF cells, whereas calponin (a fibroblast marker) was expressed only in P4 cells. CaV 1.2 (L-type) and CaV 3.3 (T-type) of VGCCs were highly expressed in P12-15 cells, but only weak CaV 2.3 (R-type) expression was identified in P4 cells using reverse transcription-polymerase chain reaction analysis. S-Nitroso-N-acetylpenicillamine (SNAP, an NO donor) decreased cell viability of HCMFs in a dose-dependent manner and induced apoptotic changes, and nifedipine (an L-type Ca2+ channel blocker) prevented apoptosis as shown with immunofluorescence staining and flow cytometry. Whole-cell mode patch-clamp recordings demonstrate the presence of L-type Ca2+ (IC a,L ) and T-type Ca2+ (IC a,T ) currents in HCMFs. SNAP inhibited IC a,L of HCMFs, but pre-treatment with ODQ (a guanylate cyclase inhibitor) or KT5823 (a PKG inhibitor) prevented it. Pre-treating cells with KT5720 (a PKA inhibitor) or SQ22536 (an adenylate cyclase inhibitor) blocked SNAP-induced inhibition of IC a,L . 8-Bromo-cyclic GMP or 8-bromo-cyclic AMP also inhibited IC a,L . However, pre-treatment with N-ethylmaleimide (a thiol-alkylating reagent) did not block the SNAP effect, nor did DL-dithiothreitol (a reducing agent) reverse it. These data suggest that high concentrations of NO injure HCMFs and inhibit IC a,L through the PKG and PKA signalling pathways but not through the S-nitrosylation pathway.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Mazzei L, Sanz R, Manucha W. Alterations on a key nephrogenic/cardiogenic gene expression linked to hypertension development. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 32:70-78. [PMID: 31472952 DOI: 10.1016/j.arteri.2019.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/11/2019] [Indexed: 10/26/2022]
Abstract
The elevation of blood pressure produces specific organic lesions, including kidney and cardiac damage. On the other hand, cardiovascular disease usually leads to the development of hypertension. Thus, hypertension could be both a cause and a consequence of cardiovascular disease. Previous studies linked the lack of nitric oxide to cardiovascular abnormalities, including hypertension, arteriosclerosis, myocardial infarction, cardiac hypertrophy, diastolic heart failure, and reduced endothelium-derived hyperpolarizing factor responses, with shorter survival. The lack of this gas also leads to renal/cardiac abnormalities. It is widely known that nephrogenic deficiency is a risk factor for kidney disease. Besides, recent evidence suggests that alterations in WT-1, a key nephrogenic factor, could contribute to the development of hypertension. Moreover, some genes involved in the development of hypertension depend on WT-1. This knowledge makes it essential to investigate and understand the mechanisms regulating the expression of these genes during renal/cardiac development, and hypertension. As a consequence, the most in-depth knowledge of the complex aetiopathogenic mechanism responsible for the hypertensive disease will allow us to propose novel therapeutic tools.
Collapse
Affiliation(s)
- Luciana Mazzei
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina; Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina
| | - Raúl Sanz
- Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina; Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina.
| |
Collapse
|
20
|
Abstract
Electromechanical coupling studies have described the intervention of nitric oxide and S-nitrosylation processes in Ca2+ release induced by stretch, with heterogeneous findings. On the other hand, ion channel function activated by stretch is influenced by nitric oxide, and concentration-dependent biphasic effects upon several cellular functions have been described. The present study uses isolated and perfused rabbit hearts to investigate the changes in mechanoelectric feedback produced by two different concentrations of the nitric oxide carrier S-nitrosoglutathione. Epicardial multielectrodes were used to record myocardial activation at baseline and during and after left ventricular free wall stretch using an intraventricular device. Three experimental series were studied: (a) control (n = 10); (b) S-nitrosoglutathione 10 µM (n = 11); and (c) S-nitrosoglutathione 50 µM (n = 11). The changes in ventricular fibrillation (VF) pattern induced by stretch were analyzed and compared. S-nitrosoglutathione 10 µM did not modify VF at baseline, but attenuated acceleration of the arrhythmia (15.6 ± 1.7 vs. 21.3 ± 3.8 Hz; p < 0.0001) and reduction of percentile 5 of the activation intervals (42 ± 3 vs. 38 ± 4 ms; p < 0.05) induced by stretch. In contrast, at baseline using the 50 µM concentration, percentile 5 was shortened (38 ± 6 vs. 52 ± 10 ms; p < 0.005) and the complexity index increased (1.77 ± 0.18 vs. 1.27 ± 0.13; p < 0.0001). The greatest complexity indices (1.84 ± 0.17; p < 0.05) were obtained during stretch in this series. S-nitrosoglutathione 10 µM attenuates the effects of mechanoelectric feedback, while at a concentration of 50 µM the drug alters the baseline VF pattern and accentuates the increase in complexity of the arrhythmia induced by myocardial stretch.
Collapse
|
21
|
Scirè A, Cianfruglia L, Minnelli C, Bartolini D, Torquato P, Principato G, Galli F, Armeni T. Glutathione compartmentalization and its role in glutathionylation and other regulatory processes of cellular pathways. Biofactors 2019; 45:152-168. [PMID: 30561781 DOI: 10.1002/biof.1476] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
Glutathione is considered the major non-protein low molecular weight modulator of redox processes and the most important thiol reducing agent of the cell. The biosynthesis of glutathione occurs in the cytosol from its constituent amino acids, but this tripeptide is also present in the most important cellular districts, such as mitochondria, nucleus, and endoplasmic reticulum, thus playing a central role in several metabolic pathways and cytoprotection mechanisms. Indeed, glutathione is involved in the modulation of various cellular processes and, not by chance, it is a ubiquitous determinant for redox signaling, xenobiotic detoxification, and regulation of cell cycle and death programs. The balance between its concentration and redox state is due to a complex series of interactions between biosynthesis, utilization, degradation, and transport. All these factors are of great importance to understand the significance of cellular redox balance and its relationship with physiological responses and pathological conditions. The purpose of this review is to give an overview on glutathione cellular compartmentalization. Information on its subcellular distribution provides a deeper understanding of glutathione-dependent processes and reflects the importance of compartmentalization in the regulation of specific cellular pathways. © 2018 BioFactors, 45(2):152-168, 2019.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Minnelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Desirée Bartolini
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Galli
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
22
|
Jones DA, Rathod KS, Williamson A, Harrington D, Andiapen M, van Eijl S, Westwood M, Antoniou S, Schilling RJ, Ahluwalia A, Mathur A. The effect of intracoronary sodium nitrite on the burden of ventricular arrhythmias following primary percutaneous coronary intervention for acute myocardial infarction. Int J Cardiol 2019; 266:1-6. [PMID: 29887423 DOI: 10.1016/j.ijcard.2018.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/06/2018] [Accepted: 01/08/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Pre-clinical evidence suggests delivery of nitric oxide (NO) through administration of inorganic nitrite suppresses arrhythmias resulting from acute ischaemia and reperfusion (I/R). To date no assessment of whether inorganic nitrite might limit reperfusion arrhythmia has occurred in man, therefore we explored the effects on I/R-induced ventricular arrhythmias in the NITRITE-AMI cohort. METHODS In the NITRITE-AMI cohort, Holter analysis was performed prior to and for 24 h after primary PCI in 80 patients who received either intra-coronary sodium nitrite (N = 40) or placebo (N = 40) during primary PCI for AMI. RESULTS Ventricular rhythm disturbance was experienced by 100% patients; however, there was no difference in the number between the groups, p = .2196. Non-sustained ventricular tachycardia (NSVT) occurred in 67.5% (27/40) of nitrite-treated patients compared to 89% (35/39) of those treated with placebo (p = .027). There was a significant reduction in both the number of runs (63%, p ≤.0001) and total beats of NSVT (64%, p = .0019) in the nitrite-treated patients compared to placebo. Post-hoc analyses demonstrate a direct correlation of occurrence of NSVT with infarct size, with the correlation stronger in the placebo versus the nitrite group initiating an independent nitrite effect (Nitrite: r = 0.110, p = .499, placebo: r = 0.527, p = .001, p for comparison: 0.004). CONCLUSION Overall no difference in ventricular rhythm disturbance was seen with intra-coronary nitrite treatment during primary PCI in STEMI patients, however nitrite treatment was associated with an important reduction in the incidence and severity of NSVT. In view of the sustained reduction of MACE seen, this effect warrants further study in a large-scale trial.
Collapse
Affiliation(s)
- Daniel A Jones
- Centre of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, United Kingdom; Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom.
| | - Krishnaraj S Rathod
- Centre of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, United Kingdom; Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Anna Williamson
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Deirdre Harrington
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Mervyn Andiapen
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Sven van Eijl
- Centre of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, United Kingdom
| | - Mark Westwood
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Sotiris Antoniou
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Richard J Schilling
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| | - Amrita Ahluwalia
- Centre of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, United Kingdom
| | - Anthony Mathur
- Centre of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, United Kingdom; Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, United Kingdom
| |
Collapse
|
23
|
Wang M, Sun L, Ding W, Cai S, Zhao Q. Ablation alleviates atrial fibrillation by regulating the signaling pathways of endothelial nitric oxide synthase/nitric oxide via miR‐155‐5p and miR‐24‐3p. J Cell Biochem 2018; 120:4451-4462. [PMID: 30302809 DOI: 10.1002/jcb.27733] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Maojing Wang
- Department of Cardiology The Affiliated Hospital of Qingdao University Qingdao China
| | - Libo Sun
- Department of Pharmacy The Affiliated Hospital of Qingdao University Qingdao China
| | - Wei Ding
- Department of Ophthalmology The West Coast New Area People’s Hospital of Qingdao Qingdao China
| | - Shanglang Cai
- Department of Cardiology The Affiliated Hospital of Qingdao University Qingdao China
| | - Qing Zhao
- Department of Cardiology The Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
24
|
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia and is associated with pronounced morbidity and mortality. Its prevalence, expected to further increase for the forthcoming years, and associated frequent hospitalizations turn AF into a major health problem. Structural and electrical atrial remodelling underlie the substrate for AF, but the exact mechanisms driving this remodelling remain incompletely understood. Recent studies have shown that microRNAs (miRNA), short non-coding RNAs that regulate gene expression, may be involved in the pathophysiology of AF. MiRNAs have been implicated in AF-induced ion channel remodelling and fibrosis. MiRNAs could therefore provide insight into AF pathophysiology or become novel targets for therapy with miRNA mimics or anti-miRNAs. Moreover, circulating miRNAs have been suggested as a new class of diagnostic and prognostic biomarkers of AF. However, the origin and function of miRNAs in tissue and plasma frequently remain unknown and studies investigating the role of miRNAs in AF vary in design and focus and even present contradicting results. Here, we provide a systematic review of the available clinical and functional studies investigating the tissue and plasma miRNAs in AF and will thereafter discuss the potential of miRNAs as biomarkers or novel therapeutic targets in AF.
Collapse
|
25
|
Zhang X, Huo Q, Sun W, Zhang C, Wu Z, Xing B, Li Q. Rs2910164 in microRNA‑146a confers an elevated risk of depression in patients with coronary artery disease by modulating the expression of NOS1. Mol Med Rep 2018; 18:603-609. [PMID: 29749487 DOI: 10.3892/mmr.2018.8929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/13/2017] [Indexed: 11/06/2022] Open
Abstract
Depression has been well established as an independent predictor of mortality and cardiac morbidity rates in patients with coronary artery disease (CAD). Evidence has shown that single nucleotide polymorphisms located in pre‑microRNA (miRNA) or mature miRNA may modify various biological processes and affect the process of carcinogenesis, and the downregulation of neuronal nitric oxide synthase 1 (NOS1) can induce depression. It has been shown that NOS1 is the target gene of miR‑146a, and that the rs2910164 G/C polymorphism can downregulate the expression of miR‑146a. In the present study, computational analysis was used to identify the target of miR‑146a, and a luciferase reporter assay system was used to validate NOS1 as a target gene of miR‑146a. In addition, U251 cells were treated with miR‑146a mimics/inhibitors to verify the negative regulatory association between miR‑146a and NOS1. Reverse transcription‑quantitative polymerase chain reaction analysis and western blot analysis were used to estimate the mRNA expression of NOS1 and the expression of miR‑146a. The results showed that the 'seed sequence' was located within the 3'‑untranslated region of NOS1 by searching an online miRNA database (www.mirdb.org), and the luciferase reporter assay confirmed that NOS1 was a direct target gene of miR‑146a. It was also found that the mRNA and protein expression levels of NOS1 in U251 cells treated with miR‑146a mimics and NOS1 small interfering RNA were substantially downregulated, compared with cells treated with the scramble control. The cells treated with miR‑146a inhibitors showed increased expression of NOS1. In addition, the presence of a minor allele of the rs2910164 polymorphism was significantly associated with risk of depression in patients with CAD. Taken together, the findings indicated a decreased risk of depression in the patients with CAD who were carriers of the miR‑146a rs2910164 C allele, and this association may be attributed to its ability to compromise the expression of miR‑146a, and thereby increase the expression of its target gene, NOS1.
Collapse
Affiliation(s)
- Xinling Zhang
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Qianqian Huo
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Wei Sun
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Chunxiang Zhang
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Zongyin Wu
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Bing Xing
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| | - Qiang Li
- Department of Cardiology, The First People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
26
|
Bae H, Choi J, Kim YW, Lee D, Kim JH, Ko JH, Bang H, Kim T, Lim I. Effects of Nitric Oxide on Voltage-Gated K⁺ Currents in Human Cardiac Fibroblasts through the Protein Kinase G and Protein Kinase A Pathways but Not through S-Nitrosylation. Int J Mol Sci 2018. [PMID: 29534509 PMCID: PMC5877675 DOI: 10.3390/ijms19030814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study investigated the expression of voltage-gated K+ (KV) channels in human cardiac fibroblasts (HCFs), and the effect of nitric oxide (NO) on the KV currents, and the underlying phosphorylation mechanisms. In reverse transcription polymerase chain reaction, two types of KV channels were detected in HCFs: delayed rectifier K+ channel and transient outward K+ channel. In whole-cell patch-clamp technique, delayed rectifier K+ current (IK) exhibited fast activation and slow inactivation, while transient outward K+ current (Ito) showed fast activation and inactivation kinetics. Both currents were blocked by 4-aminopyridine. An NO donor, S-nitroso-N-acetylpenicillamine (SNAP), increased the amplitude of IK in a concentration-dependent manner with an EC50 value of 26.4 µM, but did not affect Ito. The stimulating effect of SNAP on IK was blocked by pretreatment with 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or by KT5823. 8-bromo-cyclic GMP stimulated the IK. The stimulating effect of SNAP on IK was also blocked by pretreatment with KT5720 or by SQ22536. Forskolin and 8-bromo-cyclic AMP each stimulated IK. On the other hand, the stimulating effect of SNAP on IK was not blocked by pretreatment of N-ethylmaleimide or by DL-dithiothreitol. Our data suggest that NO enhances IK, but not Ito, among KV currents of HCFs, and the stimulating effect of NO on IK is through the PKG and PKA pathways, not through S-nitrosylation.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jeongyoon Choi
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Donghee Lee
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jung-Ha Kim
- Department of Family Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| |
Collapse
|
27
|
Molsidomine Attenuates Ventricular Electrical Remodeling and Arrhythmogenesis in Rats With Chronic β-Adrenergic Receptor Activation Through the NO/cGMP/PKG Pathway. J Cardiovasc Pharmacol 2017; 68:342-355. [PMID: 27482866 DOI: 10.1097/fjc.0000000000000422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study investigated the effects and associated underlying mechanisms of molsidomine, a nitric oxide (NO) donor, on cardiac electrical remodeling and ventricular tachycardias (VTs) induced by chronic isoprenaline (ISO) stimulation in rats. The rats were randomly divided into groups that were treated with saline (control group), ISO (ISO group), ISO + molsidomine (ISO + M group), and ISO + molsidomine + the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, ISO + M + O group) for 14 days. An electrophysiological study was performed to assess cardiac repolarization, action potential duration restitution, and the induction of action potential duration alternans and VTs in vitro. The properties of the Ca transients, Ca handling-related proteins, and NO/guanosine 3'5'-cyclic monophosphate (cGMP)/protein kinase G (PKG) pathway were examined. Compared with the control group, chronic ISO stimulation prolonged the cardiac repolarization, decreased the Ca transient alternans and action potential duration alternans thresholds, and increased the maximum slope (Smax) of the action potential duration restitution curve and incidence of VTs in vitro. All these effects were attenuated by molsidomine treatment (P < 0.05). Moreover, molsidomine activated cGMP/PKG signaling and stabilized the expression of calcium handling-related proteins compared with the ISO group. However, the protective effects of molsidomine were partially inhibited by ODQ. Our results suggest that molsidomine stabilizes calcium handling and attenuates cardiac electrical remodeling and arrhythmogenesis in rats with chronic β-adrenergic receptor activation. These effects are at least partially mediated by the activation of NO/cGMP/PKG pathway.
Collapse
|
28
|
Lin YK, Chen YA, Lee TI, Chen YC, Chen SA, Chen YJ. Aging Modulates the Substrate and Triggers Remodeling in Atrial Fibrillation. Circ J 2017; 82:1237-1244. [PMID: 28904308 DOI: 10.1253/circj.cj-17-0242] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aging plays a critical role in the genesis of atrial fibrillation (AF) and also increases the risks of cardiac dysfunction and stroke in AF patients. AF is caused by increased AF triggering from abnormalities of the thoracic vein and/or modulated substrate (atrial) with enhancement of AF maintenance. Clinical and laboratory evidence indicates that aging is significant in the creation of atrial electrical and structural remodeling that leads to increased susceptibility to AF occurrence. Aging is commonly associated with cardiovascular comorbidities, oxidative stress, calcium dysregulation, atrial myopathy with apoptosis, and fibrosis, which all contribute to the genesis of AF. This review updates the current understanding of the effects of aging on the pathophysiology of AF.
Collapse
Affiliation(s)
- Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University
| | - Yi-Ann Chen
- Division of Nephrology, Sijhih Cathay General Hospital
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Wan Fang Hospital, Taipei Medical University
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center
| | - Shih-Ann Chen
- Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University
| |
Collapse
|
29
|
Morris G, Walder K, Carvalho AF, Tye SJ, Lucas K, Berk M, Maes M. The role of hypernitrosylation in the pathogenesis and pathophysiology of neuroprogressive diseases. Neurosci Biobehav Rev 2017; 84:453-469. [PMID: 28789902 DOI: 10.1016/j.neubiorev.2017.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/02/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022]
Abstract
There is a wealth of data indicating that de novo protein S-nitrosylation in general and protein transnitrosylation in particular mediates the bulk of nitric oxide signalling. These processes enable redox sensing and facilitate homeostatic regulation of redox dependent protein signalling, function, stability and trafficking. Increased S-nitrosylation in an environment of increasing oxidative and nitrosative stress (O&NS) is initially a protective mechanism aimed at maintaining protein structure and function. When O&NS becomes severe, mechanisms governing denitrosylation and transnitrosylation break down leading to the pathological state referred to as hypernitrosylation (HN). Such a state has been implicated in the pathogenesis and pathophysiology of several neuropsychiatric and neurodegenerative diseases and we investigate its potential role in the development and maintenance of neuroprogressive disorders. In this paper, we propose a model whereby the hypernitrosylation of a range of functional proteins and enzymes lead to changes in activity which conspire to produce at least some of the core abnormalities contributing to the development and maintenance of pathology in these illnesses.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW, Wales, United Kingdom
| | - Ken Walder
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil
| | - Susannah J Tye
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia; Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil; Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, 55128 Mainz, Germany
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia.
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
30
|
Barnett SD, Buxton ILO. The role of S-nitrosoglutathione reductase (GSNOR) in human disease and therapy. Crit Rev Biochem Mol Biol 2017; 52:340-354. [PMID: 28393572 PMCID: PMC5597050 DOI: 10.1080/10409238.2017.1304353] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR), or ADH5, is an enzyme in the alcohol dehydrogenase (ADH) family. It is unique when compared to other ADH enzymes in that primary short-chain alcohols are not its principle substrate. GSNOR metabolizes S-nitrosoglutathione (GSNO), S-hydroxymethylglutathione (the spontaneous adduct of formaldehyde and glutathione), and some alcohols. GSNOR modulates reactive nitric oxide (•NO) availability in the cell by catalyzing the breakdown of GSNO, and indirectly regulates S-nitrosothiols (RSNOs) through GSNO-mediated protein S-nitrosation. The dysregulation of GSNOR can significantly alter cellular homeostasis, leading to disease. GSNOR plays an important regulatory role in smooth muscle relaxation, immune function, inflammation, neuronal development and cancer progression, among many other processes. In recent years, the therapeutic inhibition of GSNOR has been investigated to treat asthma, cystic fibrosis and interstitial lung disease (ILD). The direct action of •NO on cellular pathways, as well as the important regulatory role of protein S-nitrosation, is closely tied to GSNOR regulation and defines this enzyme as an important therapeutic target.
Collapse
Affiliation(s)
- Scott D Barnett
- a Department of Pharmacology , University of Nevada, Reno School of Medicine , Reno , NV , USA
| | - Iain L O Buxton
- a Department of Pharmacology , University of Nevada, Reno School of Medicine , Reno , NV , USA
| |
Collapse
|
31
|
Sigurdsson MI, Saddic L, Heydarpour M, Chang TW, Shekar P, Aranki S, Couper GS, Shernan SK, Muehlschlegel JD, Body SC. Post-operative atrial fibrillation examined using whole-genome RNA sequencing in human left atrial tissue. BMC Med Genomics 2017; 10:25. [PMID: 28464817 PMCID: PMC5414158 DOI: 10.1186/s12920-017-0270-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 04/25/2017] [Indexed: 01/02/2023] Open
Abstract
Background Both ambulatory atrial fibrillation (AF) and post-operative AF (poAF) are associated with substantial morbidity and mortality. Analyzing the tissue-specific gene expression in the left atrium (LA) can identify novel genes associated with AF and further the understanding of the mechanism by which previously identified genetic variants associated with AF mediate their effects. Methods LA free wall samples were obtained intraoperatively immediately prior to mitral valve surgery in 62 Caucasian individuals. Gene expression was quantified on mRNA harvested from these samples using RNA sequencing. An expression quantitative trait loci (eQTL) analysis was performed, comparing gene expression between different genotypes of 1.0 million genetic markers, emphasizing genomic regions and genes associated with AF. Results Comparison of whole-genome expression between patients who later developed poAF and those who did not identified 23 differentially expressed genes. These included genes associated with the resting membrane potential modified by potassium currents, as well as genes within Wnt signaling and cyclic GMP metabolism. The eQTL analysis identified 16,139 cis eQTL relationships in the LA, including several involving genes and single nucleotide polymorphisms (SNPs) linked to AF. A previous relationship between rs3744029 and MYOZ1 expression was confirmed, and a novel relationship between rs6795970 and the expression of the SCN10A gene was identified. Conclusions The current study is the first analysis of the human LA expression landscape using high-throughput RNA sequencing. Several novel genes and variants likely involved in AF pathogenesis were identified, thus furthering the understanding of how variants associated with AF mediate their effects via altered gene expression. Trial registration ClinicalTrials.gov ID: NCT00833313, registered 5. January 2009 Electronic supplementary material The online version of this article (doi:10.1186/s12920-017-0270-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin I Sigurdsson
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Louis Saddic
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Tzuu-Wang Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Prem Shekar
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Sary Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Gregory S Couper
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Stanton K Shernan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| |
Collapse
|
32
|
Zhao CY, Greenstein JL, Winslow RL. Mechanisms of the cyclic nucleotide cross-talk signaling network in cardiac L-type calcium channel regulation. J Mol Cell Cardiol 2017; 106:29-44. [PMID: 28365422 PMCID: PMC5508987 DOI: 10.1016/j.yjmcc.2017.01.013] [Citation(s) in RCA: 6] [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: 10/26/2016] [Revised: 12/16/2016] [Accepted: 01/20/2017] [Indexed: 10/19/2022]
Abstract
Regulation of L-type Calcium (Ca2+) Channel (LCC) gating is critical to shaping the cardiac action potential (AP) and triggering the initiation of excitation-contraction (EC) coupling in cardiac myocytes. The cyclic nucleotide (cN) cross-talk signaling network, which encompasses the β-adrenergic and the Nitric Oxide (NO)/cGMP/Protein Kinase G (PKG) pathways and their interaction (cross-talk) through distinctively-regulated phosphodiesterase isoenzymes (PDEs), regulates LCC current via Protein Kinase A- (PKA) and PKG-mediated phosphorylation. Due to the tightly-coupled and intertwined biochemical reactions involved, it remains to be clarified how LCC gating is regulated by the signaling network from receptor to end target. In addition, the large number of EC coupling-related phosphorylation targets of PKA and PKG makes it difficult to quantify and isolate changes in L-type Ca2+ current (ICaL) responses regulated by the signaling network. We have developed a multi-scale, biophysically-detailed computational model of LCC regulation by the cN signaling network that is supported by experimental data. LCCs are modeled with functionally distinct PKA- and PKG-phosphorylation dependent gating modes. The model exhibits experimentally observed single channel characteristics, as well as whole-cell LCC currents upon activation of the cross-talk signaling network. Simulations show 1) redistribution of LCC gating modes explains changes in whole-cell current under various stimulation scenarios of the cN cross-talk network; 2) NO regulation occurs via potentiation of a gating mode characterized by prolonged closed times; and 3) due to compensatory actions of cross-talk and antagonizing functions of PKA- and PKG-mediated phosphorylation of LCCs, the effects of individual inhibitions of PDEs 2, 3, and 4 on ICaL are most pronounced at low levels of β-adrenergic stimulation. Simulations also delineate the contribution of the following two mechanisms to overall LCC regulation, which have otherwise been challenging to distinguish: 1) regulation of PKA and PKG activation via cN cross-talk (Mechanism 1); and 2) LCC interaction with activated PKA and PKG (Mechanism 2). These results provide insights into how cN signals transduced via the cN cross-talk signaling network are integrated via LCC regulation in the heart.
Collapse
Affiliation(s)
- Claire Y Zhao
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD, 21218, USA.
| | - Joseph L Greenstein
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD, 21218, USA.
| | - Raimond L Winslow
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
33
|
Dimitroulas T, Hodson J, Panoulas VF, Sandoo A, Smith J, Kitas G. Genetic variations in the alanine-glyoxylate aminotransferase 2 (AGXT2) gene and dimethylarginines levels in rheumatoid arthritis. Amino Acids 2017; 49:1133-1141. [PMID: 28357606 DOI: 10.1007/s00726-017-2413-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/22/2017] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is associated with high rates of cardiovascular events mainly due to coronary and cerebrovascular atherosclerotic disease. Asymmetric (ADMA) and symmetric (SDMA) dimethylarginines are endogenous inhibitors of nitric oxide synthase and have been repeatedly linked with adverse cardiovascular outcomes in the general population and various disease settings. Alanine-glyoxylate aminotransferase 2 (AGTX2) is considered an alternative metabolic pathway contributing to the clearance of dimethylarginines in humans. The aim of the current study was to investigate the effect of specific AGXT-2 gene polymorphisms on circulating levels of ADMA or SDMA in patients with RA. Serum ADMA and SDMA levels were measured in 201 individuals with RA [median age: 67 years (IQR: 59-73), 155 females]. Two single nucleotide polymorphisms (SNPs) in the AGXT-2 gene-rs37369 and rs28305-were genotyped. Distributions of SDMA and ADMA were skewed, hence comparisons across the gene polymorphisms were performed using Kruskal-Wallis tests, and summarized using medians and interquartile ranges. Univariable analysis did not demonstrate a significant difference in the levels of SDMA or ADMA amongst the different genotypic groups of either rs37369AGXT2 (p = 0.800, 0.977) or rs28305AGXT2 (p = 0.463, 0.634). In multivariable analyses, ADMA levels were found to be significantly associated with erythrocyte sedimentation rate and estimated glomerular filtration rate, whilst SDMA levels were significantly associated with estimated glomerular filtration rate and quantitative insulin sensitivity check index. After adjustments for these factors, the relationship between the AGXT2 gene variants and both ADMA and SDMA remained non-significant. Our study in a well-characterized RA population did not show an association between serum concentrations of dimethylarginines and genetic variants of the AGXT2 gene.
Collapse
Affiliation(s)
- Theodoros Dimitroulas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK. .,4th Department of Internal Medicine, School of Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, 54642, Thessaloniki, Greece.
| | - James Hodson
- Institute of Translational Medicine, Queen Elizabeth Hospital Birmingham, University Hospital Birmingham NHS Foundation Trust, Mindelsohn Way, Birmingham, B15 2WB, UK
| | - Vasileios F Panoulas
- Cardiovascular Sciences, Imperial College London, National Heart and Lung Institute, London, UK
| | - Aamer Sandoo
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK.,School of Sport, Health and Exercise Sciences, Bangor University, George Building, Bangor, Gwynedd, LL57 2PZ, Wales, UK
| | - Jacqueline Smith
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK
| | - George Kitas
- Arthritis Research UK Epidemiology Unit, University of Manchester, Oxford Road, Manchester, UK
| |
Collapse
|
34
|
Ahmed SM, Abdelrahman SA, Salama AE. Efficacy of gold nanoparticles against isoproterenol induced acute myocardial infarction in adult male albino rats. Ultrastruct Pathol 2017; 41:168-185. [PMID: 28277146 DOI: 10.1080/01913123.2017.1281367] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This study was undertaken to investigate the role of gold nanoparticles (GNPs) of 50 nm diameter on isoproterenol (ISO) induced acute myocardial infarction in adult male albino rats. Forty five adult Wistar male albino rats were equally divided into three groups. Control (group I) was further subdivided into three subgroups. In group II, the rats received ISO subcutaneously at a dose of 100 mg/kg for three days. In group III, rats received ISO as group II and then GNPs (400 μg/kg/day) intravenously for 14 consecutive days. Echocardiography was performed. Left ventricular specimens were prepared for H&E, van Gieson staining, immunohistochemical analysis for (eNOs and Bcl-2), and Electron microscope examination. Energy dispersive X-ray microanalysis was also performed. Cardiac markers such as creatine Kinase-MB (CK-MB), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and cardiac troponin T (cTnT) were measured. Group II revealed cardiomyocytes with deeply stained acidophilic cytoplasm, small dark nuclei, intracellular vacuolations, wide intercellular spaces, and extravasated red blood cells. Increased collagen fibers were observed. Electron microscope examination showed cardiomyocyte with small and irregular outlined nuclei, mitochondria with irregular cristae and others with ruptured mitochondrial membrane, abnormal alignment of myofibrils, dilated cisternae of smooth endoplasmic reticulum, and disorganized intercalated discs. Group III showed that most cardiomyocytes preserved the normal architecture. Increased expression of eNOs immunoreaction and decreased Bcl-2 immunoreaction were detected in group II as compared to the control and GNP-treated groups. These findings suggested that GNPs of 50 nm diameter improved myocardial injury after ISO-induced myocardial infarction in rats. ABBREVIATIONS Myocardial infarction (MI), Isoproterenol (ISO), Nitric oxide (NO), Neuronal NOS (nNOs), Endothelial NOs (eNOs), Gold nanoparticle (GNPs), Diamiobenzidine (DAB), Serum Creatine Kinase-MB (CK-MB), Alanine aminotransferase (ALT), Cardiac troponin T (cTnT), Electrochemiluminiscence (ECLIA), Cardiomyocytes (CMC), Peroxisomal proliferator activated receptor (PPARs), Reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Samah M Ahmed
- a Histology and Cell Biology Department, Faculty of Human Medicine , Zagazig University , Zagazig , Egypt
| | - Shaimaa Ali Abdelrahman
- a Histology and Cell Biology Department, Faculty of Human Medicine , Zagazig University , Zagazig , Egypt
| | | |
Collapse
|
35
|
Blinatumomab provoked fatal heart failure. Int Immunopharmacol 2016; 41:42-46. [DOI: 10.1016/j.intimp.2016.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 11/18/2022]
|
36
|
Gasparova I, Kubatka P, Opatrilova R, Caprnda M, Filipova S, Rodrigo L, Malan L, Mozos I, Rabajdova M, Nosal V, Kobyliak N, Valentova V, Petrovic D, Adamek M, Kruzliak P. Perspectives and challenges of antioxidant therapy for atrial fibrillation. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:1-14. [PMID: 27900409 DOI: 10.1007/s00210-016-1320-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/18/2016] [Indexed: 12/26/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia associated with significant morbidity and mortality. The mechanisms underlying the pathogenesis of AF are poorly understood, although electrophysiological remodeling has been described as an important initiating step. There is growing evidence that oxidative stress is involved in the pathogenesis of AF. Many known triggers of oxidative stress, such as age, diabetes, smoking, and inflammation, are linked with an increased risk of arrhythmia. Numerous preclinical studies and clinical trials reported the importance of antioxidant therapy in the prevention of AF, using vitamins C and E, polyunsaturated fatty acids, statins, or nitric oxide donors. The aim of our work is to give a current overview and analysis of opportunities, challenges, and benefits of antioxidant therapy in AF.
Collapse
Affiliation(s)
- Iveta Gasparova
- Institute of Biology, Genetics and Medical Genetics, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovak Republic, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovak Republic, Slovakia
| | - Radka Opatrilova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Martin Caprnda
- 2nd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Slavomira Filipova
- Department of Cardiology, National Institute of Cardiovascular Diseases, Bratislava, Slovakia
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University of Asturias (HUCA), Oviedo, Spain
| | - Leone Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Ioana Mozos
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Miroslava Rabajdova
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Vladimir Nosal
- Clinic of Neurology, Jessenius Faculty of Medicine, Comenius University and University Hospital in Martin, Martin, Slovak Republic
| | - Nazarii Kobyliak
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Vanda Valentova
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovak Republic, Slovakia
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljublana, Ljublana, Slovenia
| | - Mariusz Adamek
- Department of Thoracic Surgery, Medical University of Silesia, Zabrze, Poland
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic. .,2nd Department of Surgery, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
37
|
Wu W, Perrin-Sarrado C, Ming H, Lartaud I, Maincent P, Hu XM, Sapin-Minet A, Gaucher C. Polymer nanocomposites enhance S-nitrosoglutathione intestinal absorption and promote the formation of releasable nitric oxide stores in rat aorta. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1795-1803. [DOI: 10.1016/j.nano.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 04/08/2016] [Accepted: 05/05/2016] [Indexed: 02/02/2023]
|
38
|
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
|
39
|
Reilly SN, Liu X, Carnicer R, Recalde A, Muszkiewicz A, Jayaram R, Carena MC, Wijesurendra R, Stefanini M, Surdo NC, Lomas O, Ratnatunga C, Sayeed R, Krasopoulos G, Rajakumar T, Bueno-Orovio A, Verheule S, Fulga TA, Rodriguez B, Schotten U, Casadei B. Up-regulation of miR-31 in human atrial fibrillation begets the arrhythmia by depleting dystrophin and neuronal nitric oxide synthase. Sci Transl Med 2016; 8:340ra74. [PMID: 27225184 PMCID: PMC4993239 DOI: 10.1126/scitranslmed.aac4296] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 04/22/2016] [Indexed: 01/06/2023]
Abstract
Atrial fibrillation (AF) is a growing public health burden, and its treatment remains a challenge. AF leads to electrical remodeling of the atria, which in turn promotes AF maintenance and resistance to treatment. Although remodeling has long been a therapeutic target in AF, its causes remain poorly understood. We show that atrial-specific up-regulation of microRNA-31 (miR-31) in goat and human AF depletes neuronal nitric oxide synthase (nNOS) by accelerating mRNA decay and alters nNOS subcellular localization by repressing dystrophin translation. By shortening action potential duration and abolishing rate-dependent adaptation of the action potential duration, miR-31 overexpression and/or disruption of nNOS signaling recapitulates features of AF-induced remodeling and significantly increases AF inducibility in mice in vivo. By contrast, silencing miR-31 in atrial myocytes from patients with AF restores dystrophin and nNOS and normalizes action potential duration and its rate dependency. These findings identify atrial-specific up-regulation of miR-31 in human AF as a key mechanism causing atrial dystrophin and nNOS depletion, which in turn contributes to the atrial phenotype begetting this arrhythmia. miR-31 may therefore represent a potential therapeutic target in AF.
Collapse
Affiliation(s)
- Svetlana N. Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Xing Liu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Alice Recalde
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Anna Muszkiewicz
- Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Raja Jayaram
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Maria Cristina Carena
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Rohan Wijesurendra
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Matilde Stefanini
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Nicoletta C. Surdo
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Oliver Lomas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Chandana Ratnatunga
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Rana Sayeed
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - George Krasopoulos
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Timothy Rajakumar
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | - Sander Verheule
- Department of Physiology, University of Maastricht, 6211 LK Maastricht, Netherlands
| | - Tudor A. Fulga
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Ulrich Schotten
- Department of Physiology, University of Maastricht, 6211 LK Maastricht, Netherlands
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
40
|
Seppälä I, Kleber ME, Bevan S, Lyytikäinen LP, Oksala N, Hernesniemi JA, Mäkelä KM, Rothwell PM, Sudlow C, Dichgans M, Mononen N, Vlachopoulou E, Sinisalo J, Delgado GE, Laaksonen R, Koskinen T, Scharnagl H, Kähönen M, Markus HS, März W, Lehtimäki T. Associations of functional alanine-glyoxylate aminotransferase 2 gene variants with atrial fibrillation and ischemic stroke. Sci Rep 2016; 6:23207. [PMID: 26984639 PMCID: PMC4794714 DOI: 10.1038/srep23207] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/02/2016] [Indexed: 12/04/2022] Open
Abstract
Asymmetric and symmetric dimethylarginines (ADMA and SDMA) impair nitric oxide bioavailability and have been implicated in the pathogenesis of atrial fibrillation (AF). Alanine–glyoxylate aminotransferase 2 (AGXT2) is the only enzyme capable of metabolizing both of the dimethylarginines. We hypothesized that two functional AGXT2 missense variants (rs37369, V140I; rs16899974, V498L) are associated with AF and its cardioembolic complications. Association analyses were conducted using 1,834 individulas with AF and 7,159 unaffected individuals from two coronary angiography cohorts and a cohort comprising patients undergoing clinical exercise testing. In coronary angiography patients without structural heart disease, the minor A allele of rs16899974 was associated with any AF (OR = 2.07, 95% CI 1.59-2.68), and with paroxysmal AF (OR = 1.98, 95% CI 1.44–2.74) and chronic AF (OR = 2.03, 95% CI 1.35–3.06) separately. We could not replicate the association with AF in the other two cohorts. However, the A allele of rs16899974 was nominally associated with ischemic stroke risk in the meta-analysis of WTCCC2 ischemic stroke cohorts (3,548 cases, 5,972 controls) and with earlier onset of first-ever ischemic stroke (360 cases) in the cohort of clinical exercise test patients. In conclusion, AGXT2 variations may be involved in the pathogenesis of AF and its age-related thromboembolic complications.
Collapse
Affiliation(s)
- Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Steve Bevan
- School of Life Science, University of Lincoln, Lincoln, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Niku Oksala
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland.,Division of Vascular Surgery, Department of Surgery, Tampere University Hospital, Tampere, Finland
| | - Jussi A Hernesniemi
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland.,Heart Hospital, Tampere University Hospital, Tampere, Finland
| | - Kari-Matti Mäkelä
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Peter M Rothwell
- Stroke Prevention Research Unit, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Cathie Sudlow
- Division of Clinical Neurosciences and Insititute of Genetics and Molecular Medicine, University of Edinburgh, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany &Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Efthymia Vlachopoulou
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Reijo Laaksonen
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Tuomas Koskinen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Satakunta Central Hospital, Department of Surgery, Pori, Finland
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Tampere, Finland
| | - Hugh S Markus
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Services GmbH, Mannheim, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| |
Collapse
|
41
|
Zhao CY, Greenstein JL, Winslow RL. Roles of phosphodiesterases in the regulation of the cardiac cyclic nucleotide cross-talk signaling network. J Mol Cell Cardiol 2016; 91:215-27. [PMID: 26773602 PMCID: PMC4764497 DOI: 10.1016/j.yjmcc.2016.01.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 12/12/2015] [Accepted: 01/04/2016] [Indexed: 12/27/2022]
Abstract
The balanced signaling between the two cyclic nucleotides (cNs) cAMP and cGMP plays a critical role in regulating cardiac contractility. Their degradation is controlled by distinctly regulated phosphodiesterase isoenzymes (PDEs), which in turn are also regulated by these cNs. As a result, PDEs facilitate communication between the β-adrenergic and Nitric Oxide (NO)/cGMP/Protein Kinase G (PKG) signaling pathways, which regulate the synthesis of cAMP and cGMP respectively. The phenomena in which the cAMP and cGMP pathways influence the dynamics of each other are collectively referred to as cN cross-talk. However, the cross-talk response and the individual roles of each PDE isoenzyme in shaping this response remain to be fully characterized. We have developed a computational model of the cN cross-talk network that mechanistically integrates the β-adrenergic and NO/cGMP/PKG pathways via regulation of PDEs by both cNs. The individual model components and the integrated network model replicate experimentally observed activation-response relationships and temporal dynamics. The model predicts that, due to compensatory interactions between PDEs, NO stimulation in the presence of sub-maximal β-adrenergic stimulation results in an increase in cytosolic cAMP accumulation and corresponding increases in PKA-I and PKA-II activation; however, the potentiation is small in magnitude compared to that of NO activation of the NO/cGMP/PKG pathway. In a reciprocal manner, β-adrenergic stimulation in the presence of sub-maximal NO stimulation results in modest cGMP elevation and corresponding increase in PKG activation. In addition, we demonstrate that PDE2 hydrolyzes increasing amounts of cAMP with increasing levels of β-adrenergic stimulation, and hydrolyzes increasing amounts of cGMP with decreasing levels of NO stimulation. Finally, we show that PDE2 compensates for inhibition of PDE5 both in terms of cGMP and cAMP dynamics, leading to cGMP elevation and increased PKG activation, while maintaining whole-cell β-adrenergic responses similar to that prior to PDE5 inhibition. By defining and quantifying reactions comprising cN cross-talk, the model characterizes the cross-talk response and reveals the underlying mechanisms of PDEs in this non-linear, tightly-coupled reaction system.
Collapse
Affiliation(s)
- Claire Y Zhao
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| | - Joseph L Greenstein
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| | - Raimond L Winslow
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
42
|
Shim J, Park JH, Lee JY, Uhm JS, Joung B, Lee MH, Ellinor PT, Pak HN. eNOS3 Genetic Polymorphism Is Related to Post-Ablation Early Recurrence of Atrial Fibrillation. Yonsei Med J 2015; 56:1244-50. [PMID: 26256966 PMCID: PMC4541653 DOI: 10.3349/ymj.2015.56.5.1244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Previous studies have demonstrated an association between eNOS polymorphisms and atrial fibrillation (AF). We sought to determine whether eNOS polymorphisms are associated with AF recurrence after a radiofrequency catheter ablation (RFCA). MATERIALS AND METHODS A total of 500 consecutive patients (56±11 years, 77% male) with paroxysmal (68%) or persistent (32%) AF who underwent RFCA and 500 age, gender-matched controls were genotyped for the eNOS3 single nucleotide polymorphism (rs1799983). AF recurrence was monitored according to 2012 ACC/AHA/ESC guidelines. RESULTS The frequencies of the rs1799983 variant alleles (T) in the case and control group were not significantly different (OR 1.05, 95% CI 0.75-1.46, p=0.798). AF patients with rs1799983 variants were more likely to have coronary artery disease or stroke than those without genetic variant at this gene (31.0% vs. 17.3%, p=0.004). During mean 17 months follow-up, early recurrence of AF (ERAF; within 3 months) and clinical recurrence (CR) of AF were 31.8% and 24.8%, respectively. The rs1799983 variant was associated with higher risk of ERAF (OR 1.71, 95% CI 1.06-2.79, p=0.028), but not with CR. ERAF occurred earlier (11±16 days) in variant group than those without variant allele (20±25 days, p=0.016). A multiple logistic regression analysis showed that presence of the rs1799983 variant (OR 1.75, 95% CI 1.07-2.86, p=0.026) and persistent AF were independent predictors for ERAF after AF ablation. CONCLUSION The rs1799983 variant of the eNOS3 gene was associated with ERAF, but not with CR, after RFCA. eNOS3 gene variants may have a potential role for stratification of post-ablation management.
Collapse
Affiliation(s)
- Jaemin Shim
- Department of Cardiology, Korea University Anam Hospital, Seoul, Korea
| | - Jae Hyung Park
- Department of Cardiology, Yonsei University Health System, Seoul, Korea
- Cardiovascular Genome Center, Yonsei University Health System, Seoul, Korea
| | - Ji-Young Lee
- Department of Cardiology, Yonsei University Health System, Seoul, Korea
- Cardiovascular Genome Center, Yonsei University Health System, Seoul, Korea
| | - Jae Sun Uhm
- Department of Cardiology, Yonsei University Health System, Seoul, Korea
| | - Boyoung Joung
- Department of Cardiology, Yonsei University Health System, Seoul, Korea
| | - Moon-Hyoung Lee
- Department of Cardiology, Yonsei University Health System, Seoul, Korea
| | - Patrick T Ellinor
- Cardiac Arrhythmia Service & Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Hui-Nam Pak
- Cardiovascular Genome Center, Yonsei University Health System, Seoul, Korea
- Department of Cardiology, Yonsei University Health System, Seoul, Korea.
| |
Collapse
|
43
|
Kovács M, Kiss A, Gönczi M, Miskolczi G, Seprényi G, Kaszaki J, Kohr MJ, Murphy E, Végh Á. Effect of sodium nitrite on ischaemia and reperfusion-induced arrhythmias in anaesthetized dogs: is protein S-nitrosylation involved? PLoS One 2015; 10:e0122243. [PMID: 25909651 PMCID: PMC4409072 DOI: 10.1371/journal.pone.0122243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/18/2015] [Indexed: 11/21/2022] Open
Abstract
Background and Purpose To provide evidence for the protective role of inorganic nitrite against acute ischaemia and reperfusion-induced ventricular arrhythmias in a large animal model. Experimental Approach Dogs, anaesthetized with chloralose and urethane, were administered intravenously with sodium nitrite (0.2 µmolkg-1min-1) in two protocols. In protocol 1 nitrite was infused 10 min prior to and during a 25 min occlusion of the left anterior descending (LAD) coronary artery (NaNO2-PO; n = 14), whereas in protocol 2 the infusion was started 10 min prior to reperfusion of the occluded vessel (NaNO2-PR; n = 12). Control dogs (n = 15) were infused with saline and subjected to the same period of ischaemia and reperfusion. Severities of ischaemia and ventricular arrhythmias, as well as changes in plasma nitrate/nitrite (NOx) levels in the coronary sinus blood, were assessed throughout the experiment. Myocardial superoxide and nitrotyrosine (NT) levels were determined during reperfusion. Changes in protein S-nitrosylation (SNO) and S-glutathionylation were also examined. Key Results Compared with controls, sodium nitrite administered either pre-occlusion or pre-reperfusion markedly suppressed the number and severity of ventricular arrhythmias during occlusion and increased survival (0% vs. 50 and 92%) upon reperfusion. There were also significant decreases in superoxide and NT levels in the nitrite treated dogs. Compared with controls, increased SNO was found only in NaNO2-PR dogs, whereas S-glutathionylation occurred primarily in NaNO2-PO dogs. Conclusions Intravenous infusion of nitrite profoundly reduced the severity of ventricular arrhythmias resulting from acute ischaemia and reperfusion in anaesthetized dogs. This effect, among several others, may result from an NO-mediated reduction in oxidative stress, perhaps through protein SNO and/or S-glutathionylation.
Collapse
Affiliation(s)
- Mária Kovács
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Attila Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Márton Gönczi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gottfried Miskolczi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - György Seprényi
- Department of Medical Biology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - József Kaszaki
- Institute of Surgical Research, Albert Szent-Györgyi Medical Center, University of Szeged, Szeged, Hungary
| | - Mark J Kohr
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ágnes Végh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
44
|
Sedova K, Bernikova O, Azarov J, Shmakov D, Vityazev V, Kharin S. Effects of echinochrome on ventricular repolarization in acute ischemia. J Electrocardiol 2015; 48:181-6. [DOI: 10.1016/j.jelectrocard.2015.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Indexed: 11/27/2022]
|
45
|
Balance of nitric oxide and reactive oxygen species in myocardial reperfusion injury and protection. J Cardiovasc Pharmacol 2014; 62:567-75. [PMID: 23921313 DOI: 10.1097/fjc.0b013e3182a50c45] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Depending on their concentrations, both nitric oxide (NO) and reactive oxygen species (ROS) take part either in myocardial ischemia reperfusion injury or in protection by ischemic and pharmacological preconditioning (Ipre) and postconditioning (Ipost). At the beginning of reperfusion, a transient release of NO is promptly scavenged by ROS to form the highly toxic peroxynitrite, which is responsible for a further increase of ROS through endothelial nitric oxide synthase uncoupling. The protective role of NO has suggested the use of NO donors to mimic Ipre and Ipost. However, NO donors have not always given the expected protection, possibly because they are responsible for the production of different amounts of ROS that depend on the amount of released NO. This review is focused on the role of the balance of NO and ROS in myocardial injury and its prevention by Ipre and Ipost and after the use of NO donors given with or without antioxidant compounds to mimic Ipre and Ipost.
Collapse
|
46
|
Roggelin L, Pelletier D, Hill JN, Feldt T, Hoffmann S, Ansong D, Sylverken J, Burhenne J, Fischer-Herr J, Mehrfar P, Thiel C, Burchard GD, Nguah SB, Cramer JP. Disease-associated QT-shortage versus quinine associated QT-prolongation: age dependent ECG-effects in Ghanaian children with severe malaria. Malar J 2014; 13:219. [PMID: 24902591 PMCID: PMC4067506 DOI: 10.1186/1475-2875-13-219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/20/2014] [Indexed: 01/08/2023] Open
Abstract
Background While several anti-malarials are known to affect the electric conduction system of the heart, less is known on the direct effects of Plasmodium falciparum infection. Some earlier studies point to a direct impact of Plasmodium falciparum infection on the electric conduction system of the heart. The aim of this study was to analyse infection- and drug-induced effects on the electric conduction system. Methods Children aged 12 months to 108 months with severe malaria were included in Kumasi, Ghana. In addition to basic demographic, clinical, biochemical and parasitological, biochemical data were measured data upon hospitalization (day 0) and 12-lead electrocardiograms were recorded before (day 0) and after (day 1) initiation of quinine therapy as well as after 42 (±3) days. Results A total of 180 children were included. Most children were tachycardic on day 0 but heart rate declined on day 1 and during follow up. The corrected QT intervals were longest on day 1 and shortest on day 0. Comparison of QT intervals with day 42 (healthy status) after stratification for age demonstrated that in the youngest (<24 months) this was mainly due to a QT shortage on day 0 while a QT prolongation on day 1 was most pronounced in the oldest (≥48 months). Nearly one third of the participating children had measurable 4-aminoquinoline levels upon admission, but no direct effect on the corrected QT intervals could be shown. Conclusion Severe P. falciparum infection itself can provoke changes in the electrophysiology of the heart, independent of anti-malarial therapy. Especially in young - thus non immune - children the effect of acute disease associated pre-treatment QT-shortage is more pronounced than quinine associated QT-prolongation after therapy. Nevertheless, neither malaria nor anti-malarial induced effects on the electrophysiology of the heart were associated with clinically relevant arrhythmias in the present study population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jakob P Cramer
- Section Tropical Medicine, Department of Internal Medicine, University Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
47
|
Single-nucleotide variations in cardiac arrhythmias: prospects for genomics and proteomics based biomarker discovery and diagnostics. Genes (Basel) 2014; 5:254-69. [PMID: 24705329 PMCID: PMC4094932 DOI: 10.3390/genes5020254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/19/2014] [Accepted: 02/19/2014] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular diseases are a large contributor to causes of early death in developed countries. Some of these conditions, such as sudden cardiac death and atrial fibrillation, stem from arrhythmias—a spectrum of conditions with abnormal electrical activity in the heart. Genome-wide association studies can identify single nucleotide variations (SNVs) that may predispose individuals to developing acquired forms of arrhythmias. Through manual curation of published genome-wide association studies, we have collected a comprehensive list of 75 SNVs associated with cardiac arrhythmias. Ten of the SNVs result in amino acid changes and can be used in proteomic-based detection methods. In an effort to identify additional non-synonymous mutations that affect the proteome, we analyzed the post-translational modification S-nitrosylation, which is known to affect cardiac arrhythmias. We identified loss of seven known S-nitrosylation sites due to non-synonymous single nucleotide variations (nsSNVs). For predicted nitrosylation sites we found 1429 proteins where the sites are modified due to nsSNV. Analysis of the predicted S-nitrosylation dataset for over- or under-representation (compared to the complete human proteome) of pathways and functional elements shows significant statistical over-representation of the blood coagulation pathway. Gene Ontology (GO) analysis displays statistically over-represented terms related to muscle contraction, receptor activity, motor activity, cystoskeleton components, and microtubule activity. Through the genomic and proteomic context of SNVs and S-nitrosylation sites presented in this study, researchers can look for variation that can predispose individuals to cardiac arrhythmias. Such attempts to elucidate mechanisms of arrhythmia thereby add yet another useful parameter in predicting susceptibility for cardiac diseases.
Collapse
|
48
|
Trenor B, Cardona K, Saiz J, Rajamani S, Belardinelli L, Giles WR. Carbon monoxide effects on human ventricle action potential assessed by mathematical simulations. Front Physiol 2013; 4:282. [PMID: 24146650 PMCID: PMC3797961 DOI: 10.3389/fphys.2013.00282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/18/2013] [Indexed: 12/15/2022] Open
Abstract
Carbon monoxide (CO) that is produced in a number of different mammalian tissues is now known to have significant effects on the cardiovascular system. These include: (i) vasodilation, (ii) changes in heart rate and strength of contractions, and (iii) modulation of autonomic nervous system input to both the primary pacemaker and the working myocardium. Excessive CO in the environment is toxic and can initiate or mediate life threatening cardiac rhythm disturbances. Recent reports link these ventricular arrhythmias to an increase in the slowly inactivating, or “late” component of the Na+ current in the mammalian heart. The main goal of this paper is to explore the basis of this pro-arrhythmic capability of CO by incorporating changes in CO-induced ion channel activity with intracellular signaling pathways in the mammalian heart. To do this, a quite well-documented mathematical model of the action potential and intracellular calcium transient in the human ventricular myocyte has been employed. In silico iterations based on this model provide a useful first step in illustrating the cellular electrophysiological consequences of CO that have been reported from mammalian heart experiments. Specifically, when the Grandi et al. model of the human ventricular action potential is utilized, and after the Na+ and Ca2+ currents in a single myocyte are modified based on the experimental literature, early after-depolarization (EAD) rhythm disturbances appear, and important elements of the underlying causes of these EADs are revealed/illustrated. Our modified mathematical model of the human ventricular action potential also provides a convenient digital platform for designing future experimental work and relating these changes in cellular cardiac electrophysiology to emerging clinical and epidemiological data on CO toxicity.
Collapse
Affiliation(s)
- Beatriz Trenor
- Instituto Interuniversitario de Investigación en Bioingeniería y Tecnología Orientada al Ser Humano, Universitat Politècnica de València Valencia, Spain
| | | | | | | | | | | |
Collapse
|
49
|
Martínez-Ruiz A, Araújo IM, Izquierdo-Álvarez A, Hernansanz-Agustín P, Lamas S, Serrador JM. Specificity in S-nitrosylation: a short-range mechanism for NO signaling? Antioxid Redox Signal 2013; 19:1220-35. [PMID: 23157283 PMCID: PMC3785806 DOI: 10.1089/ars.2012.5066] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) classical and less classical signaling mechanisms (through interaction with soluble guanylate cyclase and cytochrome c oxidase, respectively) operate through direct binding of NO to protein metal centers, and rely on diffusibility of the NO molecule. S-Nitrosylation, a covalent post-translational modification of protein cysteines, has emerged as a paradigm of nonclassical NO signaling. RECENT ADVANCES Several nonenzymatic mechanisms for S-nitrosylation formation and destruction have been described. Enzymatic mechanisms for transnitrosylation and denitrosylation have been also studied as regulators of the modification of specific subsets of proteins. The advancement of modification-specific proteomic methodologies has allowed progress in the study of diverse S-nitrosoproteomes, raising clues and questions about the parameters for determining the protein specificity of the modification. CRITICAL ISSUES We propose that S-nitrosylation is mainly a short-range mechanism of NO signaling, exerted in a relatively limited range of action around the NO sources, and tightly related to the very controlled regulation of subcellular localization of nitric oxide synthases. We review the nonenzymatic and enzymatic mechanisms that support this concept, as well as physiological examples of mammalian systems that illustrate well the precise compartmentalization of S-nitrosylation. FUTURE DIRECTIONS Individual and proteomic studies of protein S-nitrosylation-based signaling should take into account the subcellular localization in order to gain further insight into the functional role of this modification in (patho)physiological settings.
Collapse
Affiliation(s)
- Antonio Martínez-Ruiz
- 1 Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP) , Madrid, Spain
| | | | | | | | | | | |
Collapse
|
50
|
TNF-α provokes electrical abnormalities in rat atrial myocardium via a NO-dependent mechanism. Pflugers Arch 2013; 465:1741-52. [DOI: 10.1007/s00424-013-1320-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/09/2013] [Accepted: 06/22/2013] [Indexed: 01/06/2023]
|