1
|
Nakajima T, Haruyama A, Fukuda T, Minami K, Nakajima T, Hasegawa T, Tokoi S, Obi S, Oguri G, Sakuma M, Shibasaki I, Fukuda H, Toyoda S. Increased neprilysin expression is linked to atrial fibrotic remodeling in cardiovascular surgery patients. IJC HEART & VASCULATURE 2025; 57:101647. [PMID: 40201407 PMCID: PMC11976568 DOI: 10.1016/j.ijcha.2025.101647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/01/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025]
Abstract
Background Neprilysin (NEP) is a membrane-bound neutral endopeptidase with various physiological functions. We investigated the roles of NEP in atrial fibrotic remodeling and atrial fibrillation (AF) in patients undergoing cardiovascular surgery. Methods Atrial tissue samples were obtained from left atrium (LA) appendages, and mRNA expression level was analyzed by real-time reverse transcription polymerase chain reaction in 61 cases (25 paroxysmal fibrillation (PAF), 36 AF). Adipose tissue (AT) mRNA expression levels were also analyzed. Western blotting and immunohistochemical staining were used for determining tissue protein expression. Serum NEP levels were measured by enzyme-linked immunosorbent assay (ELISA). Two-dimensional speckle tracking echocardiography was performed to measure mean left atrial reservoir strain (mLASr) to evaluate atrial remodeling in pre-operative patients and control participants. Results Immunohistochemical staining and western blotting revealed NEP expression in both AT and LA. Serum NEP levels did not correlate with NEP mRNA or protein expression in AT and LA. NEP mRNA expression levels correlated with fibrosis-related gene expression. NEP mRNA, protein, and fibrosis-related gene expression levels increased in PAF patients with low mLASr compared with high mLASr. PAF patients with high NEP mRNA expression showed increased fibrosis-related gene expression compared with those with low NEP expression. Multiple regression analysis revealed that NEP mRNA expression level was an independent variable for predicting fibrosis-related gene expression, whereas NOX4 and NLRP3 were independent variables for predicting NEP expression levels. Conclusions Increased atrial expression of NEP is linked to atrial fibrotic remodeling, and the development of AF in patients undergoing cardiovascular surgery.
Collapse
Affiliation(s)
- Toshiaki Nakajima
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Akiko Haruyama
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Taira Fukuda
- Department of Liberal Arts and Sciences, Kanagawa University of Human Services, Yokosuka, Kanagawa, Japan
| | - Kentaro Minami
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Takafumi Nakajima
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Takaaki Hasegawa
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Seiko Tokoi
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Syotaro Obi
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Gaku Oguri
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Ikuko Shibasaki
- Department of Cardiovascular Surgery, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Hirotsugu Fukuda
- Department of Cardiovascular Surgery, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, Shimotsuga-gun, Tochigi, Japan
| |
Collapse
|
2
|
Castillo RL, Farías J, Sandoval C, González-Candia A, Figueroa E, Quezada M, Cruz G, Llanos P, Jorquera G, Kostin S, Carrasco R. Role of NLRP3 Inflammasome in Heart Failure Patients Undergoing Cardiac Surgery as a Potential Determinant of Postoperative Atrial Fibrillation and Remodeling: Is SGLT2 Cotransporter Inhibition an Alternative for Cardioprotection? Antioxidants (Basel) 2024; 13:1388. [PMID: 39594530 PMCID: PMC11591087 DOI: 10.3390/antiox13111388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
In heart failure (HF) patients undergoing cardiac surgery, an increased activity of mechanisms related to cardiac remodeling may determine a higher risk of postoperative atrial fibrillation (POAF). Given that atrial fibrillation (AF) has a negative impact on the course and management of HF, including the need for anticoagulation therapy, identifying the factors associated with AF occurrence after cardiac surgery is crucial for the prognosis of these patients. POAF is thought to occur when various clinical and biochemical triggers act on susceptible cardiac tissue (first hit), with oxidative stress and inflammation during cardiopulmonary bypass (CPB) surgery being potential contributing factors (second hit). However, the molecular mechanisms involved in these processes remain poorly characterized. Recent research has shown that patients who later develop POAF often have pre-existing abnormalities in calcium handling and activation of NLRP3-inflammasome signaling in their atrial cardiomyocytes. These molecular changes may make cardiomyocytes more susceptible to spontaneous Ca2+-releases and subsequent arrhythmias, particularly when exposed to inflammatory mediators. Additionally, some clinical studies have linked POAF with elevated preoperative inflammatory markers, but there is a need for further research in order to better understand the impact of CPB surgery on local and systemic inflammation. This knowledge would make it possible to determine whether patients susceptible to POAF have pre-existing inflammatory conditions or cellular electrophysiological factors that make them more prone to developing AF and cardiac remodeling. In this context, the NLRP3 inflammasome, expressed in cardiomyocytes and cardiac fibroblasts, has been identified as playing a key role in the development of HF and AF, making patients with pre-existing HF with reduced ejection fraction (HFrEF) the focus of several clinical studies with interventions that act at this level. On the other hand, HFpEF has been linked to metabolic and non-ischemic risk factors, but more research is needed to better characterize the myocardial remodeling events associated with HFpEF. Therefore, since ventricular remodeling may differ between HFrEF and HFpEF, it is necessary to perform studies in both groups of patients due to their pathophysiological variations. Clinical evidence has shown that pharmacological therapies that are effective for HFrEF may not provide the same anti-remodeling benefits in HFpEF patients, particularly compared to traditional adrenergic and renin-angiotensin-aldosterone system inhibitors. On the other hand, there is growing interest in medications with pleiotropic or antioxidant/anti-inflammatory effects, such as sodium-glucose cotransporter 2 inhibitors (SGLT-2is). These drugs may offer anti-remodeling effects in both HFrEF and HFpEF by inhibiting pro-inflammatory, pro-oxidant, and NLRP3 signaling pathways and their mediators. The anti-inflammatory, antioxidant, and anti-remodeling effects of SGLT-2 i have progressively expanded from HFrEF and HFpEF to other forms of cardiac remodeling. However, these advances in research have not yet encompassed POAF despite its associations with inflammation, oxidative stress, and remodeling. Currently, the direct or indirect effects of NLRP3-dependent pathway inhibition on the occurrence of POAF have not been clinically assessed. However, given that NLRP3 pathway inhibition may also indirectly affect other pathways, such as inhibition of NF-kappaB or inhibition of matrix synthesis, which are strongly linked to POAF and cardiac remodeling, it is reasonable to hypothesize that this type of intervention could play a role in preventing these events.
Collapse
Affiliation(s)
- Rodrigo L. Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago 7500922, Chile
| | - Jorge Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Alejandro González-Candia
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Esteban Figueroa
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Mauricio Quezada
- Facultad de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile;
| | - Gonzalo Cruz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago 8380544, Chile
| | - Gonzalo Jorquera
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8331051, Chile;
| | - Sawa Kostin
- Faculty of Health Sciences, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany;
| | - Rodrigo Carrasco
- Departamento de Cardiología, Clínica Alemana de Santiago, Santiago 7500922, Chile;
| |
Collapse
|
3
|
Șoșdean R, Dănilă MD, Ionică LN, Pescariu AS, Mircea M, Ionac A, Mornoș C, Luca CT, Feier HB, Muntean DM, Sturza A. Monoamine Oxidase Contributes to Valvular Oxidative Stress: A Prospective Observational Pilot Study in Patients with Severe Mitral Regurgitation. Int J Mol Sci 2024; 25:10307. [PMID: 39408637 PMCID: PMC11477003 DOI: 10.3390/ijms251910307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Monoamine oxidases (MAOs), mitochondrial enzymes that constantly produce hydrogen peroxide (H2O2) as a byproduct of their activity, have been recently acknowledged as contributors to oxidative stress in cardiometabolic pathologies. The present study aimed to assess whether MAOs are mediators of valvular oxidative stress and interact in vitro with angiotensin 2 (ANG2) to mimic the activation of the renin-angiotensin system. To this aim, valvular tissue samples were harvested from 30 patients diagnosed with severe primary mitral regurgitation and indication for surgical repair. Their reactive oxygen species (ROS) levels were assessed by means of a ferrous oxidation xylenol orange (FOX) assay, while MAO expression was assessed by immune fluorescence (protein) and qRT-PCR (mRNA). The experiments were performed using native valvular tissue acutely incubated or not with angiotensin 2 (ANG2), MAO inhibitors (MAOI) and the angiotensin receptor blocker, irbesartan (Irb). Correlations between oxidative stress and echocardiographic parameters were also analyzed. Ex vivo incubation with ANG2 increased MAO-A and -B expression and ROS generation. The level of valvular oxidative stress was negatively correlated with the left ventricular ejection fraction. MAOI and Irb reduced valvular H2O2. production. In conclusion, both MAO isoforms are expressed in pathological human mitral valves and contribute to local oxidative stress and ventricular functional impairment and can be modulated by the local renin-angiotensin system.
Collapse
Affiliation(s)
- Raluca Șoșdean
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Maria D. Dănilă
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
| | - Loredana N. Ionică
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
- Department X—Medical Semiotics I, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania
| | - Alexandru S. Pescariu
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Monica Mircea
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Adina Ionac
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Cristian Mornoș
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Constantin T. Luca
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Horea B. Feier
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
- Department VI—Cardiovascular Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Square no 2, 300041 Timișoara, Romania
| | - Danina M. Muntean
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
| | - Adrian Sturza
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania
| |
Collapse
|
4
|
Nakajima T, Haruyama A, Fukuda T, Minami K, Hirose S, Yazawa H, Nakajima T, Hasegawa T, Kitagawa Y, Obi S, Inami S, Oguri G, Shibasaki I, Amano H, Arikawa T, Sakuma M, Abe S, Fukuda H, Toyoda S. Left atrial reservoir strain is a marker of atrial fibrotic remodeling in patients undergoing cardiovascular surgery: Analysis of gene expression. PLoS One 2024; 19:e0306323. [PMID: 38976680 PMCID: PMC11230549 DOI: 10.1371/journal.pone.0306323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024] Open
Abstract
Left atrial strain (LAS) measured by two-dimensional speckle tracking echocardiography (2DSTE) is considered to be a marker of LA structural remodeling, but it remains unsettled. We investigated the potential usefulness and clinical relevance of LAS to detect atrial remodeling including fibrosis by analyzing gene expression in cardiovascular surgery patients. Preoperative 2DSTE was performed in 131 patients (92 patients with sinus rhythm [SR] patients including paroxysmal AF [PAF], 39 atrial fibrillation [AF]) undergoing cardiovascular surgery. Atrial samples were obtained from the left atrial appendages, and mRNA expression level was analyzed by real-time reverse transcription polymerase chain reaction (RT-PCR) in 59 cases (24 PAF, 35 AF). Mean value of left atrial reservoir strain (mLASr) correlated with left atrial volume index (LAVI), and left atrial conduit strain (mLAScd). mLASr also correlated with left atrial contractile strain (mLASct) in SR patients including PAF. mLASr was significantly lower, and LAVI was higher, in the AF group, compared with SR patients including PAF. The expression of COL1A1 mRNA encoding collagen type I α1 significantly increased in AF patients (p = 0.031). mLASr negatively correlated with COL1A1 expression level, and multivariate regression analysis showed that mLASr was an independent predictor of atrial COL1A1 expression level, even after adjusting for age, sex, and BMI. But, neither mLAScd / mLASct nor LAVI (bp) correlated with COL1A1 gene expression. The expression level of COL1A1 mRNA strongly correlated with ECM-related genes (COL3A1, FN1). It also correlated ECM degradation-related genes (MMP2, TIMP1, and TIMP2), pro-fibrogenic cytokines (TGFB1 encoding TGFβ1, END1, PDGFD, CTGF), oxidant stress-related genes (NOX2, NOX4), ACE, inflammation-related genes (NLRP, IL1B, MCP-1), and apoptosis (BAX). Among the fibrosis-related genes examined, univariable regression analysis showed that log (COL1A1) was associated with log (TGFB1) (adjusted R2 = 0.685, p<0.001), log (NOX4) (adjusted R2 = 0.622, p<0.001), log (NOX2) (adjusted R2 = 0.611, p<0.001), suggesting that TGFB1 and NOX4 was the potent independent determinants of COL1A1 expression level. mLASr negatively correlated with the ECM-related genes, and fibrosis-related gene expression level including TGFB1, NOX2, and NLRP3 in PAF patients. PAF patients with low mLASr had higher expression of the fibrosis-related gene expression, compared with those with high mLASr. These results suggest that LASr correlates with atrial COL1A1 gene expression associated with fibrosis-related gene expression. Patients with low LASr exhibit increased atrial fibrosis-related gene expression, even those with PAF, highlighting the utility of LAS as a marker for LA fibrosis in cardiovascular surgery patients.
Collapse
Affiliation(s)
- Toshiaki Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Akiko Haruyama
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Taira Fukuda
- Department of Liberal Arts and Human Development, Kanagawa University of Human Services, Kanagawa, Japan
| | - Kentaro Minami
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Suguru Hirose
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Hiroko Yazawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takafumi Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takaaki Hasegawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Yoshiyuki Kitagawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Syotaro Obi
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Shu Inami
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Gaku Oguri
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Ikuko Shibasaki
- Department of Cardiovascular Surgery, Dokkyo Medical University Hospital, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Hirohisa Amano
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Takuo Arikawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Shichiro Abe
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Hirotsugu Fukuda
- Department of Cardiovascular Surgery, Dokkyo Medical University Hospital, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| |
Collapse
|
5
|
Ninni S, Algalarrondo V, Brette F, Lemesle G, Fauconnier J. Left atrial cardiomyopathy: Pathophysiological insights, assessment methods and clinical implications. Arch Cardiovasc Dis 2024; 117:283-296. [PMID: 38490844 DOI: 10.1016/j.acvd.2024.02.001] [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: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Atrial cardiomyopathy is defined as any complex of structural, architectural, contractile or electrophysiological changes affecting atria, with the potential to produce clinically relevant manifestations. Most of our knowledge about the mechanistic aspects of atrial cardiomyopathy is derived from studies investigating animal models of atrial fibrillation and atrial tissue samples obtained from individuals who have a history of atrial fibrillation. Several noninvasive tools have been reported to characterize atrial cardiomyopathy in patients, which may be relevant for predicting the risk of incident atrial fibrillation and its related outcomes, such as stroke. Here, we provide an overview of the pathophysiological mechanisms involved in atrial cardiomyopathy, and discuss the complex interplay of these mechanisms, including aging, left atrial pressure overload, metabolic disorders and genetic factors. We discuss clinical tools currently available to characterize atrial cardiomyopathy, including electrocardiograms, cardiac imaging and serum biomarkers. Finally, we discuss the clinical impact of atrial cardiomyopathy, and its potential role for predicting atrial fibrillation, stroke, heart failure and dementia. Overall, this review aims to highlight the critical need for a clinically relevant definition of atrial cardiomyopathy to improve treatment strategies.
Collapse
Affiliation(s)
- Sandro Ninni
- CHU de Lille, Université de Lille, 59000 Lille, France.
| | - Vincent Algalarrondo
- Department of Cardiology, Bichat University Hospital, AP-HP, 75018 Paris, France
| | - Fabien Brette
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | | | - Jérémy Fauconnier
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| |
Collapse
|
6
|
Balan AI, Halațiu VB, Scridon A. Oxidative Stress, Inflammation, and Mitochondrial Dysfunction: A Link between Obesity and Atrial Fibrillation. Antioxidants (Basel) 2024; 13:117. [PMID: 38247541 PMCID: PMC10812976 DOI: 10.3390/antiox13010117] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
The adipose tissue has long been thought to represent a passive source of triglycerides and fatty acids. However, extensive data have demonstrated that the adipose tissue is also a major endocrine organ that directly or indirectly affects the physiological functions of almost all cell types. Obesity is recognized as a risk factor for multiple systemic conditions, including metabolic syndrome, type 2 diabetes mellitus, sleep apnea, cardiovascular disorders, and many others. Obesity-related changes in the adipose tissue induce functional and structural changes in cardiac myocytes, promoting a wide range of cardiovascular disorders, including atrial fibrillation (AF). Due to the wealth of epidemiologic data linking AF to obesity, the mechanisms underlying AF occurrence in obese patients are an area of rich ongoing investigation. However, progress has been somewhat slowed by the complex phenotypes of both obesity and AF. The triad inflammation, oxidative stress, and mitochondrial dysfunction are critical for AF pathogenesis in the setting of obesity via multiple structural and functional proarrhythmic changes at the level of the atria. The aim of this paper is to provide a comprehensive view of the close relationship between obesity-induced oxidative stress, inflammation, and mitochondrial dysfunction and the pathogenesis of AF. The clinical implications of these mechanistic insights are also discussed.
Collapse
Affiliation(s)
- Alkora Ioana Balan
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Vasile Bogdan Halațiu
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Alina Scridon
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|
7
|
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
|
8
|
Han X, Liu Y, Li G, Tse G, Liu T. A narrative review on prediabetes or diabetes and atrial fibrillation: From molecular mechanisms to clinical practice. HEART AND MIND 2023; 7:207-216. [DOI: 10.4103/hm.hm-d-23-00028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/19/2023] [Indexed: 01/11/2025] Open
Abstract
Based on glucose levels, people fall into three groups, normal individuals, prediabetic patients, and diabetic mellitus (DM) patients. Prediabetes (pre-DM) is an intermediate condition that exists between normal glucose levels and DM. Atrial fibrillation (AF), one of the most prevalent cardiac arrhythmias in medical practice, contributes to a considerable morbidity and mortality rate. In this review, we looked at the clinical symptoms, pathological alterations, molecular mechanisms, and associated risk factors of pre-DM, type 2 DM (T2DM), and AF. In clinical practice, pre-DM can increase the prevalence of AF. In the hyperglycemic state, oxidative stress, inflammation, and endoplasmic reticulum stress can cause alterations in atrial cell or cardiac fibroblast function through tumor necrosis factor-α/nuclear factor-κB (NF-κB)/transforming growth factor-β, mitogen-activated protein kinase-matrix metalloproteinase-9 and PARP-1 is poly (ADP-ribose) polymerase 1. IκB kinase-α/NF-κB pathways, and further cause atria undergo structural, electrical, and neural remodeling which lead to the occurrence and persistence of AF. In addition, pre-DM and T2DM may worsen as a result of obesity, obstructive sleep apnea, and arterial hypertension. Furthermore, clinical researches have demonstrated that lifestyle interventions and/or pharmacotherapy in pre-DM patients can effectively delay the progresssion of pre-DM to T2DM. Individualized glycemic management and AF management should be provided to AF patients with pre-DM or DM.
Collapse
Affiliation(s)
- Xuyao Han
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ying Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- Kent and Medway Medical School, University of Kent, Canterbury, United Kingdom
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Tong Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
9
|
Molnár AÁ, Sánta A, Pásztor DT, Merkely B. Atrial Cardiomyopathy in Valvular Heart Disease: From Molecular Biology to Clinical Perspectives. Cells 2023; 12:1796. [PMID: 37443830 PMCID: PMC10340254 DOI: 10.3390/cells12131796] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
This review discusses the evolving topic of atrial cardiomyopathy concerning valvular heart disease. The pathogenesis of atrial cardiomyopathy involves multiple factors, such as valvular disease leading to atrial structural and functional remodeling due to pressure and volume overload. Atrial enlargement and dysfunction can trigger atrial tachyarrhythmia. The complex interaction between valvular disease and atrial cardiomyopathy creates a vicious cycle of aggravating atrial enlargement, dysfunction, and valvular disease severity. Furthermore, atrial remodeling and arrhythmia can predispose to atrial thrombus formation and stroke. The underlying pathomechanism of atrial myopathy involves molecular, cellular, and subcellular alterations resulting in chronic inflammation, atrial fibrosis, and electrophysiological changes. Atrial dysfunction has emerged as an essential determinant of outcomes in valvular disease and heart failure. Despite its predictive value, the detection of atrial fibrosis and dysfunction is challenging and is not included in the clinical routine. Transthoracic echocardiography and cardiac magnetic resonance imaging are the main diagnostic tools for atrial cardiomyopathy. Recently published data have revealed that both left atrial volumes and functional parameters are independent predictors of cardiovascular events in valvular disease. The integration of atrial function assessment in clinical practice might help in early cardiovascular risk estimation, promoting early therapeutic intervention in valvular disease.
Collapse
|
10
|
Liu X, Zhang W, Luo J, Shi W, Zhang X, Li Z, Qin X, Liu B, Wei Y. TRIM21 deficiency protects against atrial inflammation and remodeling post myocardial infarction by attenuating oxidative stress. Redox Biol 2023; 62:102679. [PMID: 36996623 PMCID: PMC10074251 DOI: 10.1016/j.redox.2023.102679] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/05/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Atrial remodeling is a major contributor to the onset of atrial fibrillation (AF) after myocardial infarction (MI). Tripartite motif-containing protein 21 (TRIM21), an E3 ubiquitin protein ligase, is associated with pathological cardiac remodeling and dysfunction. However, the role of TRIM21 in postmyocardial infarction atrial remodeling and subsequent AF remains unclear. This study investigated the role of TRIM21 in post myocardial infarction atrial remodeling using TRIM21 knockout mice and explored the underlying mechanisms by overexpressing TRIM21 in HL-1 atrial myocytes using a lentiviral vector. The expression of TRIM21 in the left atrium of the mouse MI model was significantly elevated. TRIM21 deficiency alleviated MI-induced atrial oxidative damage, Cx43 downregulation, atrial fibrosis and enlargement, and abnormalities in electrocardiogram parameters (prolongation of the P-wave and PR interval). TRIM21 overexpression in atrial myocyte HL-1 cells further enhanced oxidative damage and Cx43 downregulation, whereas these effects were reversed by the reactive oxygen species scavenger N-acetylcysteine. The findings suggest that TRIM21 likely induces Nox2 expression mechanistically by activating the NF-κB pathway, which in turn leads to myocardial oxidative damage, inflammation, and atrial remodeling.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Wenming Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Wentao Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xingxu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
11
|
Chin CG, Chen YC, Lin YK, Lu YY, Cheng WL, Chung CC, Chen SA, Chen YJ. Effect of macrophage migration inhibitory factor on pulmonary vein arrhythmogenesis through late sodium current. Europace 2023; 25:698-706. [PMID: 36056883 PMCID: PMC10103572 DOI: 10.1093/europace/euac152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Macrophage migration inhibitory factor (MIF), a pleiotropic inflammatory cytokine, is highly expressed in patients with atrial fibrillation (AF). Inflammation increases the risk of AF and is primarily triggered by pulmonary vein (PV) arrhythmogenesis. This study investigated whether MIF can modulate the electrical activity of the PV and examined the underlying mechanisms of MIF. METHODS AND RESULTS A conventional microelectrode, a whole-cell patch clamp, western blotting, and immunofluorescent confocal microscopy were used to investigate electrical activity, calcium (Ca2+) regulation, protein expression, ionic currents, and cytosolic reactive oxygen species (ROS) in rabbit PV tissue and isolated single cardiomyocytes with and without MIF incubation (100 ng/mL, treated for 6 h). The MIF (100 ng/mL)-treated PV tissue (n = 8) demonstrated a faster beating rate (1.8 ± 0.2 vs. 2.6 ± 0.1 Hz, P < 0.05), higher incidence of triggered activity (12.5 vs. 100%, P < 0.05), and premature atrial beat (0 vs. 100%, P < 0.05) than the control PV tissue (n = 8). Compared with the control PV cardiomyocytes, MIF-treated single PV cardiomyocytes had larger Ca2+ transients (0.6 ± 0.1 vs. 1.0 ± 0.1, ΔF/F0, P < 0.05), sarcoplasmic reticulum Ca2+ content (0.9 ± 0.20 vs. 1.7 ± 0.3 mM of cytosol, P < 0.05), and cytosolic ROS (146.8 ± 5.3 vs. 163.7 ± 3.8, ΔF/F0, P < 0.05). Moreover, MIF-treated PV cardiomyocytes exhibited larger late sodium currents (INa-Late), L-type Ca2+ currents, and Na+/Ca2+ exchanger currents than the control PV cardiomyocytes. KN93 [a selective calcium/calmodulin-dependent protein kinase II (CaMKII) blocker, 1 μM], ranolazine (an INa-Late inhibitor, 10 μM), and N-(mercaptopropionyl) glycine (ROS inhibitor, 10 mM) reduced the beating rates and the incidence of triggered activity and premature captures in the MIF-treated PV tissue. CONCLUSION Macrophage migration inhibitory factor increased PV arrhythmogenesis through Na+ and Ca2+ dysregulation through the ROS activation of CaMKII signalling, which may contribute to the genesis of AF during inflammation. Anti-CaMKII treatment may reverse PV arrhythmogenesis. Our results clearly reveal a key link between MIF and AF and offer a viable therapeutic target for AF treatment.
Collapse
Affiliation(s)
- Chye-Gen Chin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Wan-Li Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chih Chung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Cardiology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Zhong X, Jiao H, Zhao D, Yang M, Teng J. Association between serum uric acid levels and atrial fibrillation in different fasting glucose patterns: A case-control study. Front Endocrinol (Lausanne) 2023; 14:1021267. [PMID: 36755929 PMCID: PMC9899926 DOI: 10.3389/fendo.2023.1021267] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Previous studies have shown both dysglycaemia and hyperuricemia are associated with an increased risk of atrial fibrillation (AF), while the relationship between serum uric acid (SUA) levels and AF in different fasting glucose patterns (FBG) is unclear. Therefore, this study aimed to determine the association between SUA and AF in different FBG patterns. METHODS A total of 1840 patients in this case-control study were enrolled, including 920 AF patients and 920 controls. Patients were divided into three groups according to the different FBG patterns: normoglycemic, impaired fasting glucose (IFG), and diabetes mellitus (DM). Multivariate logistic regression models were performed to evaluate the relationship between SUA and AF in different FBG patterns. Pearson correlation analysis was used to explore the correlation between SUA and metabolic factors. Receiver operating characteristic (ROC) curve models indicated the diagnostic efficiency of SUA for diagnosing AF. RESULTS SUA was independently associated with AF after adjusting for all confounding factors in different FBG patterns(normoglycemic: OR=1.313, 95% CI:1.120-1.539; IFG: OR=1.386, 95% CI:1.011-1.898; DM: OR=1.505, 95% CI:1.150-1.970). Pearson's correlation analysis suggested that SUA in AF patients was correlated with several different metabolic factors in different FBG patterns (p<0.05). ROC curve analysis showed that SUA in the normoglycemic group combined with CHD and APOB [AUC: 0.906 (95% CI: 0.888-0.923)], in the IFG group combined with CHD and Scr [AUC: 0.863 (95% CI: 0.820-0.907)], in the DM group combined with CHD and SBP [AUC: 0.858 (95% CI: 0.818-0.898)] had the highest AUC for predicting AF. CONCLUSION Findings implied a significant association between SUA and AF in different FBG patterns and provide specific models combined with other factors (CHD, APOB, SCr, SBP), which might contribute to the diagnosis of AF.
Collapse
Affiliation(s)
- Xia Zhong
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huachen Jiao
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Huachen Jiao,
| | - Dongsheng Zhao
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengqi Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jing Teng
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
13
|
Chang M, Gada KD, Chidipi B, Tsalatsanis A, Gibbons J, Remily-Wood E, Logothetis DE, Oberstaller J, Noujaim SF. I KACh is constitutively active via PKC epsilon in aging mediated atrial fibrillation. iScience 2022; 25:105442. [PMID: 36388956 PMCID: PMC9650037 DOI: 10.1016/j.isci.2022.105442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 09/22/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
Atrial fibrillation (AF), the most common abnormal heart rhythm, is a major cause for stroke. Aging is a significant risk factor for AF; however, specific ionic pathways that can elucidate how aging leads to AF remain elusive. We used young and old wild-type and PKC epsilon- (PKCϵ) knockout mice, whole animal, and cellular electrophysiology, as well as whole heart, and cellular imaging to investigate how aging leads to the aberrant functioning of a potassium current, and consequently to AF facilitation. Our experiments showed that knocking out PKCϵ abrogates the effects of aging on AF by preventing the development of a constitutively active acetylcholine sensitive inward rectifier potassium current (IKACh). Moreover, blocking this abnormal current in the old heart reduces AF inducibility. Our studies demonstrate that in the aging heart, IKACh is constitutively active in a PKCϵ-dependent manner, contributing to the perpetuation of AF.
Collapse
Affiliation(s)
- Mengmeng Chang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kirin D. Gada
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Science, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA 02115, USA
| | - Bojjibabu Chidipi
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Athanasios Tsalatsanis
- College of Medicine Office of Research, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Justin Gibbons
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Elizabeth Remily-Wood
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Diomedes E. Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Science, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, MA 02115, USA
| | - Jenna Oberstaller
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Sami F. Noujaim
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
14
|
The Role of Mitochondrial Dysfunction in Atrial Fibrillation: Translation to Druggable Target and Biomarker Discovery. Int J Mol Sci 2021; 22:ijms22168463. [PMID: 34445167 PMCID: PMC8395135 DOI: 10.3390/ijms22168463] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent and progressive cardiac arrhythmia worldwide and is associated with serious complications such as heart failure and ischemic stroke. Current treatment modalities attenuate AF symptoms and are only moderately effective in halting the arrhythmia. Therefore, there is an urgent need to dissect molecular mechanisms that drive AF. As AF is characterized by a rapid atrial activation rate, which requires a high energy metabolism, a role of mitochondrial dysfunction in AF pathophysiology is plausible. It is well known that mitochondria play a central role in cardiomyocyte function, as they produce energy to support the mechanical and electrical function of the heart. Details on the molecular mechanisms underlying mitochondrial dysfunction are increasingly being uncovered as a contributing factor in the loss of cardiomyocyte function and AF. Considering the high prevalence of AF, investigating the role of mitochondrial impairment in AF may guide the path towards new therapeutic and diagnostic targets. In this review, the latest evidence on the role of mitochondria dysfunction in AF is presented. We highlight the key modulators of mitochondrial dysfunction that drive AF and discuss whether they represent potential targets for therapeutic interventions and diagnostics in clinical AF.
Collapse
|
15
|
Li Y, Zhang YX, Ning DS, Chen J, Li SX, Mo ZW, Peng YM, He SH, Chen YT, Zheng CJ, Gao JJ, Yuan HX, Ou JS, Ou ZJ. Simvastatin inhibits POVPC-mediated induction of endothelial-to-mesenchymal cell transition. J Lipid Res 2021; 62:100066. [PMID: 33711324 PMCID: PMC8063863 DOI: 10.1016/j.jlr.2021.100066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT), the process by which an endothelial cell (EC) undergoes a series of molecular events that result in a mesenchymal cell phenotype, plays an important role in atherosclerosis. 1-Palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC), derived from the oxidation of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine, is a proinflammatory lipid found in atherosclerotic lesions. Whether POVPC promotes EndMT and how simvastatin influences POVPC-mediated EndMT remains unclear. Here, we treated human umbilical vein ECs with POVPC, simvastatin, or both, and determined their effect on EC viability, morphology, tube formation, proliferation, and generation of NO and superoxide anion (O2•-). Expression of specific endothelial and mesenchymal markers was detected by immunofluorescence and immunoblotting. POVPC did not affect EC viability but altered cellular morphology from cobblestone-like ECs to a spindle-like mesenchymal cell morphology. POVPC increased O2- generation and expression of alpha-smooth muscle actin, vimentin, Snail-1, Twist-1, transforming growth factor-beta (TGF-β), TGF-β receptor II, p-Smad2/3, and Smad2/3. POVPC also decreased NO production and expression of CD31 and endothelial NO synthase. Simvastatin inhibited POVPC-mediated effects on cellular morphology, production of O2•- and NO, and expression of specific endothelial and mesenchymal markers. These data demonstrate that POVPC induces EndMT by increasing oxidative stress, which stimulates TGF-β/Smad signaling, leading to Snail-1 and Twist-1 activation. Simvastatin inhibited POVPC-induced EndMT by decreasing oxidative stress, suppressing TGF-β/Smad signaling, and inactivating Snail-1 and Twist-1. Our findings reveal a novel mechanism of atherosclerosis that can be inhibited by simvastatin.
Collapse
Affiliation(s)
- Yan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yi-Xin Zhang
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing Chen
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shang-Xuan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Shi-Hui He
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Chun-Juan Zheng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Hao-Xiang Yuan
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
16
|
Pastori D, Baratta F, Di Rocco A, Farcomeni A, Del Ben M, Angelico F, Violi F, Pignatelli P, Lip GYH. Statin use and mortality in atrial fibrillation: A systematic review and meta-analysis of 100,287 patients. Pharmacol Res 2021; 165:105418. [PMID: 33450384 DOI: 10.1016/j.phrs.2021.105418] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/06/2020] [Accepted: 01/03/2021] [Indexed: 01/24/2023]
Abstract
Statins are effective for reducing cardiovascular disease in patients at risk or with cardiovascular disease. The benefit of statin therapy on adverse cardiovascular outcomes in patients with non-valvular atrial fibrillation (AF) is not clear. We performed a systematic review and meta-analysis of studies retrieved from MEDLINE via PubMed and Cochrane (CENTRAL) database of studies investigating the efficacy of statins in AF patients. The principal endpoint was all-cause mortality. Other endpoints were cardiovascular mortality, ischemic stroke, composite endpoints and any bleeding. We included 14 studies (2 post-hoc analysis of randomized clinical trials, 8 prospective and 4 retrospective) with 100,287 AF patients, of whom 23,228 were on statins. The pooled hazard ratio (HR) for all-cause mortality was 0.59 (95 % Confidence Interval [CI] 0.54-0.65). This association was consistent by aging, sex and prevalent cardiovascular or cerebrovascular disease. and the beneficial effect was evident already after 12 months of therapy. The absolute risk reduction for all-cause mortality in patients treated with statins was 10 % (95 % CI 9-10). The pooled HR for statins against cardiovascular mortality was 0.75 (95 % CI 0.58-0.96). No association was found with other secondary endpoints. Regarding bleeding events, the pooled HR for statin use was 0.60 (95 % CI 0.48-0.76). Our meta-analysis shows that in AF patients, statin therapy was associated with a reduction in all-cause and cardiovascular mortality are reduced by 41 % and 25 %, respectively. Randomized clinical trials in AF patients are necessary, as well as clarity on AF-specific LDL cholesterol targets.
Collapse
Affiliation(s)
- Daniele Pastori
- I Clinica Medica, Atherothrombosis Centre, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; Liverpool Centre for Cardiovascular Science University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.
| | - Francesco Baratta
- I Clinica Medica, Atherothrombosis Centre, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Arianna Di Rocco
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessio Farcomeni
- Department of Economics and Finance, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Del Ben
- I Clinica Medica, Atherothrombosis Centre, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Francesco Violi
- I Clinica Medica, Atherothrombosis Centre, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Pasquale Pignatelli
- I Clinica Medica, Atherothrombosis Centre, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| |
Collapse
|
17
|
Wang P, Tian X, Tang J, Duan X, Wang J, Cao H, Qiu X, Wang W, Mai M, Yang Q, Liao R, Yan F. Artemisinin protects endothelial function and vasodilation from oxidative damage via activation of PI3K/Akt/eNOS pathway. Exp Gerontol 2021; 147:111270. [PMID: 33556535 DOI: 10.1016/j.exger.2021.111270] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Previous studies showed that artemisinin (ART) may be useful in the protection against the early development of atherosclerosis, but the effects of ART on vasodilation and eNOS remained unclear. OBJECTIVES AND METHODS In the current study, we investigated the protective effect of ART on endothelial cell injury induced by oxidative stress and its underlying mechanism via MTT assay, Flow Cytometry Assay, Vasodilation study, Western blotting and vivo assay. RESULTS We found that pretreatment of human umbilical vein endothelial cells (HUVECs) with ART significantly suppressed H2O2-induced cell death by decreasing the extent of oxidation and MDA activity, activating SOD, increasing NO production and inhibiting caspase 3/7 activity. Meanwhile, we also found that ART was able to activate PI3K/Akt/eNOS pathway. PI3K inhibitor LY294002 or Akt kinase specific inhibitor Akt inhibitor VIII blocked the protective effect of ART. To explore the effect of ART in the damage of vasodilation induced by H2O2 in mice, we treated the aortic ring from C57BL/6 mice with H2O2 with or without ART, the results demonstrated that ART ameliorated endothelium-dependent vasodilation damage induced by H2O2. CONCLUSION Taken together, these data suggest that ART is able to protect endothelial function and vasodilation from oxidative damage, at least in part through activation of PI3K/Akt/eNOS pathway. Our findings indicate that artemisinin maybe as a potential therapeutic agent for patients with atherosclerosis.
Collapse
Affiliation(s)
- Peng Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoying Tian
- School of Medical Science, Jinan University, Guangzhou, China
| | - Juxian Tang
- Department of Hematology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Xiao Duan
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jinying Wang
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Huan Cao
- School of Medical Science, Jinan University, Guangzhou, China
| | - Xiaoyuan Qiu
- School of Medical Science, Jinan University, Guangzhou, China
| | - Wenxuan Wang
- School of Medical Science, Jinan University, Guangzhou, China
| | - Mengfei Mai
- School of Medical Science, Jinan University, Guangzhou, China
| | - Qiaohong Yang
- School of Medical Science, Jinan University, Guangzhou, China.
| | - Rifang Liao
- Department of pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Fengxia Yan
- School of Medical Science, Jinan University, Guangzhou, China.
| |
Collapse
|
18
|
Butts B, Ahmed MI, Bajaj NS, Cox Powell P, Pat B, Litovsky S, Gupta H, Lloyd SG, Denney TS, Zhang X, Aban I, Sadayappan S, McNamara JW, Watson MJ, Ferrario CM, Collawn JF, Lewis C, Davies JE, Dell'Italia LJ. Reduced Left Atrial Emptying Fraction and Chymase Activation in Pathophysiology of Primary Mitral Regurgitation. JACC Basic Transl Sci 2020; 5:109-122. [PMID: 32140620 PMCID: PMC7046515 DOI: 10.1016/j.jacbts.2019.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 11/17/2022]
Abstract
Increasing left atrial (LA) size predicts outcomes in patients with isolated mitral regurgitation (MR). Chymase is plentiful in the human heart and affects extracellular matrix remodeling. Chymase activation correlates to LA fibrosis, LA enlargement, and a decreased total LA emptying fraction in addition to having a potential intracellular role in mediating myofibrillar breakdown in LA myocytes. Because of the unreliability of the left ventricular ejection fraction in predicting outcomes in MR, LA size and the total LA emptying fraction may be more suitable indicators for timing of surgical intervention.
Collapse
Affiliation(s)
- Brittany Butts
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mustafa I Ahmed
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Navkaranbir S Bajaj
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pamela Cox Powell
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Betty Pat
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Silvio Litovsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Himanshu Gupta
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
- Department of Cardiology, Valley Health System, Paramus, New Jersey
| | - Steven G Lloyd
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University School of Engineering, Auburn, Alabama
| | - Xiaoxia Zhang
- Department of Electrical and Computer Engineering, Auburn University School of Engineering, Auburn, Alabama
| | - Inmaculada Aban
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sakthivel Sadayappan
- Division of Cardiovascular Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James W McNamara
- Division of Cardiovascular Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael J Watson
- Division of Cardiothoracic Surgery, Department of Surgery, Duke University, Durham, North Carolina
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest University Health Science Center, Winston-Salem, North Carolina
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Clifton Lewis
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - James E Davies
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Louis J Dell'Italia
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
19
|
Effects of N-acetyl cysteine, vitamin E and vitamin C on liver glutathione levels following amiodarone treatment in rats. POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2019; 16:88-92. [PMID: 31410096 PMCID: PMC6690152 DOI: 10.5114/kitp.2019.86361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 04/28/2019] [Indexed: 11/18/2022]
Abstract
Introduction Amiodarone, a pharmaceutical extensively used to suppress atrial and ventricular tachyarrhythmias, is also known to cause many side effects on many tissues. N-acetyl-cysteine (NAC), vitamin E and vitamin C are known as antioxidants for their ability to minimize oxidative stress. In the peer-reviewed literature, there is no study reporting on the protective effects of these antioxidant agents against its hepatotoxicity. Aim We investigated the oxidative effects of NAC, vitamins E and C on liver tissue after amiodarone treatment. Material and methods Rats were randomly assigned to: control; amiodarone group; amiodarone + NAC treated group; amiodarone + Vit. E group and amiodarone + Vit. C group. Liver tissues were isolated from animals and total glutathione levels were measured. Results In all time intervals, the level of glutathione increased. When all time intervals were compared, the amiodarone group revealed the lowest levels. The antioxidant co-administered group was studied; the glutathione levels were statistically significantly higher than the sole amiodarone group. When vitamins E, C or N-acetyl cysteine were examined, there was no statistically significant difference among them. Conclusions In this study we found that hepatotoxicity capacity of amiodarone may be reduced by taking up antioxidants. In addition, the effect documented here may be reproducible and may be applied to clinical settings.
Collapse
|
20
|
Fang CY, Chen MC, Chang TH, Wu CC, Chang JP, Huang HD, Ho WC, Wang YZ, Pan KL, Lin YS, Huang YK, Chen CJ, Lee WC. Idi1 and Hmgcs2 Are Affected by Stretch in HL-1 Atrial Myocytes. Int J Mol Sci 2018; 19:ijms19124094. [PMID: 30567295 PMCID: PMC6321625 DOI: 10.3390/ijms19124094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 01/27/2023] Open
Abstract
Background: Lipid expression is increased in the atrial myocytes of mitral regurgitation (MR) patients. This study aimed to investigate key regulatory genes and mechanisms of atrial lipotoxic myopathy in MR. Methods: The HL-1 atrial myocytes were subjected to uniaxial cyclic stretching for eight hours. Fatty acid metabolism, lipoprotein signaling, and cholesterol metabolism were analyzed by PCR assay (168 genes). Results: The stretched myocytes had significantly larger cell size and higher lipid expression than non-stretched myocytes (all p < 0.001). Fatty acid metabolism, lipoprotein signaling, and cholesterol metabolism in the myocytes were analyzed by PCR assay (168 genes). In comparison with their counterparts in non-stretched myocytes, seven genes in stretched monocytes (Idi1, Olr1, Nr1h4, Fabp2, Prkag3, Slc27a5, Fabp6) revealed differential upregulation with an altered fold change >1.5. Nine genes in stretched monocytes (Apoa4, Hmgcs2, Apol8, Srebf1, Acsm4, Fabp1, Acox2, Acsl6, Gk) revealed differential downregulation with an altered fold change <0.67. Canonical pathway analysis, using Ingenuity Pathway Analysis software, revealed that the only genes in the “superpathway of cholesterol biosynthesis” were Idi1 (upregulated) and Hmgcs2 (downregulated). The fraction of stretched myocytes expressing Nile red was significantly decreased by RNA interference of Idi1 (p < 0.05) and was significantly decreased by plasmid transfection of Hmgcs2 (p = 0.004). Conclusions: The Idi1 and Hmgcs2 genes have regulatory roles in atrial lipotoxic myopathy associated with atrial enlargement.
Collapse
Affiliation(s)
- Chih-Yuan Fang
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Mien-Cheng Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 110, Taiwan.
| | - Chia-Chen Wu
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Hsien-Da Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 300, Taiwan.
| | - Wan-Chun Ho
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Yi-Zhen Wang
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Kuo-Li Pan
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Yao-Kuang Huang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Chien-Jen Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Wei-Chieh Lee
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
21
|
Abstract
BACKGROUND Atrial fibrillation (AF) is initiated through arrhythmic atrial excitation from outside the sinus node or remodeling of atrial tissue that allows reentry of excitation. Angiotensin II (AngII) has been implicated in the initiation and maintenance of AF through changes in Ca2+ handling and production of reactive oxygen species (ROS). OBJECTIVE We aimed to determine the role of p21-activated kinase 1 (Pak1), a downstream target in the AngII signaling cascade, in atrial electrophysiology and arrhythmia. METHODS Wild-type and Pak1-/- mice were used to determine atrial function in vivo on the organ and cellular level by quantification of electrophysiological and Ca2+ handling properties. RESULTS We demonstrate that reduced Pak1 activity increases the inducibility of atrial arrhythmia in vivo and in vitro. On the cellular level, Pak1-/- atrial myocytes (AMs) exhibit increased basal and AngII (1 μM)-induced ROS production, sensitivity to the NADPH oxidase-2 (NOX2) inhibitors gp91ds-tat and apocynin (1 μM), and enhanced membrane translocation of Ras-related C3 substrate 1 (Rac1) that is part of the multimolecular NOX2 complex. Upon stimulation with AngII, Pak1-/- AMs exhibit an exaggerated increase in the intracellular Calcium concentration ([Ca2+]i) and arrhythmic events that were sensitive to sodium-calcium exchanger (NCX) inhibitors (KB-R7943 and SEA0400; 1 μM) and suppressed in AMs from NOX2-deficient (gp91phox-/-) mice. Pak1 stimulation (FTY720; 200 nM) in wild-type AMs and AMs from a canine model of ventricular tachypacing-induced AF prevented AngII-induced arrhythmic Ca2+ overload by attenuating NCX activity in a NOX2-dependent manner. CONCLUSION The experimental results support that Pak1 stimulation can attenuate NCX-dependent Ca2+ overload and prevent triggered arrhythmic activity by suppressing NOX2-dependent ROS production.
Collapse
|
22
|
Liang X, Zhang Q, Wang X, Yuan M, Zhang Y, Xu Z, Li G, Liu T. Reactive oxygen species mediated oxidative stress links diabetes and atrial fibrillation. Mol Med Rep 2018; 17:4933-4940. [PMID: 29393403 PMCID: PMC5865952 DOI: 10.3892/mmr.2018.8472] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 11/13/2017] [Indexed: 01/01/2023] Open
Abstract
Diabetes is an independent risk factor for atrial fibrillation (AF); however, the underlying mechanism linking diabetes and AF remains to be clarified. The present study aimed to explore the molecular mechanism of increased reactive oxygen species (ROS) production in AF and the ROS‑mediated downstream events in diabetes. Firstly, the atrial fibroblasts were isolated from the left atrium of rabbits using enzyme digestion and differential adhesion. Then, the isolated cells were identified by morphology analysis under a microscope, collagen distribution using Masson trichrome staining and vimentin by immunofluorescence. Following this, the collected atrial fibroblasts were randomly divided into 7 groups and administered with high glucose (25 mM glucose), H2O2 stimulation (100 nmol/l), glucose + apocynin (100 µg/ml), H2O2 + apocynin, glucose + H2O2, and a combination of glucose, apocynin and H2O2, as well as the negative control (NC). An MTS assay was performed to investigate cell proliferation following the different treatments, and western blotting was conducted to explore the expression of several proteins including NAD(P)H oxidative (NOX) subunits, key factors involved in mitogen‑activated protein kinase (MAPK) signaling pathways and matrix metalloproteinases (MMPs). The atrial fibroblasts were spindle‑shaped with one or more protuberances. Vimentin was positively expressed in collected cells under confocal laser scanning microscopy. This result indicated that the atrial fibroblasts were successfully prepared. High glucose and H2O2 stimulation significantly increased the proliferation of atrial fibroblasts and apocynin markedly attenuated the promoting effects on cell proliferation induced by high glucose and H2O2 treatment (P<0.05). Additionally, high glucose and H2O2 stimulation increased the expression of Rac1, phospho(p)‑c‑Jun N‑terminal kinase 1, p38, p‑p38 and MMP9, which was markedly decreased by the addition of apocynin (P<0.05). The mechanism associated with diabetes and AF may be attributed to oxidative stress (ROS production) derived from NOX activity, and then induced activation of the MAPK signaling pathways and MMP9 expression.
Collapse
Affiliation(s)
- Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Qitong Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xinghua Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Meng Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yue Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Zhao Xu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
23
|
Barangi S, Hayes AW, Karimi G. The more effective treatment of atrial fibrillation applying the natural compounds; as NADPH oxidase and ion channel inhibitors. Crit Rev Food Sci Nutr 2017; 58:1230-1241. [PMID: 28925721 DOI: 10.1080/10408398.2017.1379000] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia that occurs because of several different risk factors, e.g., valvular heart disease, coronary artery disease, age ≥75 years, hypertension and diabetes mellitus. One key risk factor that results in AF, is oxidative stress. Evidence suggests that there is a correlation between oxidative processes and the genesis of AF. Oxidative stress occurs when the generation of reactive oxygen species (ROS) increase due to excessive activity of enzymes including NADPH oxidase (NOX) and xanthine oxidase; or its degradation decrease by dysfunctional antioxidant enzyme systems, such as superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx). Afterwards, elevated ROS may shift ion channel activity to increase AF susceptibility. The outbreak of AF continues to grow. Unfortunately, current treatment strategies may have limited efficacy or adverse effects. On the other hand, the inhibition of ROS formation and alteration of ion channel activity could be important therapeutic targets for prevention or treatments of AF. Additionally, many studies have been shown that several natural compounds have the ability to inhibit NADPH oxidases directly. This review focuses on natural compounds which specially inhibit NOX isoforms and have direct effects on ion channels, suggesting these compounds can be helpful in AF treatment.
Collapse
Affiliation(s)
- Samira Barangi
- a Department of Pharmacodynamics and Toxicology , School of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - A Wallace Hayes
- b Harvard University, Cambridge, MA, USA; Michigan State University , East Lansing , MI , USA
| | - Gholamreza Karimi
- a Department of Pharmacodynamics and Toxicology , School of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran.,c Pharmaceutical Research Center , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
24
|
Atrial Fibrillation and Sleep Apnoea: Guilt by Association? Heart Lung Circ 2017; 26:902-910. [DOI: 10.1016/j.hlc.2017.05.127] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 12/18/2022]
|
25
|
Moris D, Spartalis M, Spartalis E, Karachaliou GS, Karaolanis GI, Tsourouflis G, Tsilimigras DI, Tzatzaki E, Theocharis S. The role of reactive oxygen species in the pathophysiology of cardiovascular diseases and the clinical significance of myocardial redox. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:326. [PMID: 28861423 PMCID: PMC5566734 DOI: 10.21037/atm.2017.06.27] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 05/25/2017] [Indexed: 01/24/2023]
Abstract
Acute and chronic excessive intracellular increase of reactive oxygen species (ROS) is involved in the development and progression of cardiovascular diseases. ROS are by-products of various oxidative physiological and biochemical processes. Sources of ROS are mitochondrial respiration, NADH/NADPH oxidase, xanthine oxidoreductase or the uncoupling of nitric oxide synthase (NOS) in vascular cells. ROS mediate various signaling pathways that underlie cardiovascular pathophysiology. The delicate equilibrium between free-radical generation and antioxidant defense is altered in favor of the former, thus leading to redox imbalance, oxidative stress, and increased cellular injury. An understanding of the pathophysiological mechanisms mediated by oxidative stress is crucial to the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Demetrios Moris
- Department of Surgery, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael Spartalis
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Eleftherios Spartalis
- Laboratory of Experimental Surgery and Surgical Research, Medical School, University of Athens, Athens, Greece
| | - Georgia-Sofia Karachaliou
- Laboratory of Experimental Surgery and Surgical Research, Medical School, University of Athens, Athens, Greece
| | | | - Gerasimos Tsourouflis
- Laboratory of Experimental Surgery and Surgical Research, Medical School, University of Athens, Athens, Greece
| | | | - Eleni Tzatzaki
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | | |
Collapse
|
26
|
Dong Z, Lin C, Liu Y, Jin H, Wu H, Li Z, Sun L, Zhang L, Hu X, Wei Y, Wang C, Han W. Upregulation of sestrins protect atriums against oxidative damage and fibrosis in human and experimental atrial fibrillation. Sci Rep 2017; 7:46307. [PMID: 28397812 PMCID: PMC5387733 DOI: 10.1038/srep46307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/15/2017] [Indexed: 01/18/2023] Open
Abstract
Atrial Fibrillation (AF) is common in the elderly and Sestrins (Sesns) have been suggested to prevent age-related pathologies. The aim of this study was to investigate the effects of Sesns in AF. Clinical data were collected and a small sample of atrial appendage and atrium was obtained from patients undergoing valve repairment. The expression of Sesn1, Sesn2, and Sesn3 was significantly higher in patients with permanent atrial fibrillation (PmAF) than that in sinus rhythm (SR), and further greater in the left atrium than the right in PmAF patients. Superoxide anion and malondialdehyde were enhanced and positively correlated to the protein expression of Sesn1/2/3. Reactive oxygen species (ROS) production and Ca2+ overload were significantly decreased and cell survival was enhanced by overexpression of Sesns 1/2/3 in cultured HL-1 cells. Conversely, knockdown of Sesn1/2/3 resulted in significantly increased ROS and Ca2+ overload. In addition, the overexpression of Sesn1/2 significantly reduced the proliferation of fibroblasts, as well as decreased the protein expression of collagen and fibronectin1 in angiotensin II-stimulated cardiac fibroblasts. Our study demonstrated for the first time that Sesns expression is significantly up-regulated in AF, which therefore may protect hearts against oxidative damage and atrial fibrosis.
Collapse
Affiliation(s)
- Zengxiang Dong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaolan Lin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yujiao Liu
- Intensive Care Unit, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Hongbo Jin
- Laboratory of Physiology, Harbin Medical University, Harbin, China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenjun Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liping Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xi Hu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingying Wei
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengcheng Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Han
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Li JY, He Y, Ke HH, Jin Y, Jiang ZY, Zhong GQ. Plasma oxidative stress and inflammatory biomarkers are associated with the sizes of the left atrium and pulmonary vein in atrial fibrillation patients. Clin Cardiol 2017; 40:89-94. [PMID: 28207162 DOI: 10.1002/clc.22633] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/13/2016] [Accepted: 09/19/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Oxidative stress and inflammatory processes are responsible for the pathogenesis of AF, but their relationship with the sizes of the LA and PVs in AF patients remains unclear. HYPOTHESIS Oxidative stress and inflammatory processes are associated with the sizes of the LA and PVs in AF patients. METHODS 82 AF patients were compared to 30 control patients by using a case-control study design. Oxidative stress, inflammatory biomarkers and the sizes of the LA and PVs were detected. RESULTS (1) Hs-CRP, IL-6, IL-8, TNF-α, MDA and ox-LDL were higher, and SOD was lower in AF patients than in control patients. Hs-CRP, MDA and ox-LDL were higher in permanent AF patients than in paroxysmal and persistent AF patients. (2) CsA of LSPV, RSPV, RIPV, LAA and LAV were statistically higher in AF patients than in control patients. CsA of RSPV, LSPV, LIPV and LAV were higher in permanent AF patients than in paroxysmal and persistent AF patients. (3) In the AF group, hs-CRP and TNF-α were positively correlated with LAV; MDA was positively correlated with CsA of LAA, LSPV and LAV; SOD was passively correlated with CsA of LAA and LAV; ox-LDL was positively correlated with CsA of LAA and LAV. Multivariate logistic regression analysis showed hs-CRP, ox-LDL, RSPV CsA, LIPV CsA and LAV were associated with AF. CONCLUSIONS Oxidative stress, inflammatory biomarkers and the sizes of the LA and PVs were significantly increased in AF patients. Hs-CRP, ox-LDL, RSPV CsA, LIPV CsA and LAV were associated with AF persistence.
Collapse
Affiliation(s)
- Jin-Yi Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hong-Hong Ke
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Jin
- Department of Cardiology, People's Hospital of Zhengzhou, Zhengzhou, China
| | - Zhi-Yuan Jiang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guo-Qiang Zhong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
28
|
Ou ZJ, Chen J, Dai WP, Liu X, Yang YK, Li Y, Lin ZB, Wang TT, Wu YY, Su DH, Cheng TP, Wang ZP, Tao J, Ou JS. 25-Hydroxycholesterol impairs endothelial function and vasodilation by uncoupling and inhibiting endothelial nitric oxide synthase. Am J Physiol Endocrinol Metab 2016; 311:E781-E790. [PMID: 27600825 DOI: 10.1152/ajpendo.00218.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022]
Abstract
Endothelial dysfunction is a key early step in atherosclerosis. 25-Hydroxycholesterol (25-OHC) is found in atherosclerotic lesions. However, whether 25-OHC promotes atherosclerosis is unclear. Here, we hypothesized that 25-OHC, a proinflammatory lipid, can impair endothelial function, which may play an important role in atherosclerosis. Bovine aortic endothelial cells were incubated with 25-OHC. Endothelial cell proliferation, migration, and tube formation were measured. Nitric oxide (NO) production and superoxide anion generation were determined. The expression and phosphorylation of endothelial NO synthase (eNOS) and Akt as well as the association of eNOS and heat shock protein (HSP)90 were detected by immunoblot analysis and immunoprecipitation. Endothelial cell apoptosis was monitored by TUNEL staining and caspase-3 activity, and expression of Bcl-2, Bax, cleaved caspase-9, and cleaved caspase-3 were detected by immunoblot analysis. Finally, aortic rings from Sprague-Dawley rats were isolated and treated with 25-OHC, and endothelium-dependent vasodilation was evaluated. 25-OHC significantly inhibited endothelial cell proliferation, migration, and tube formation. 25-OHC markedly decreased NO production and increased superoxide anion generation. 25-OHC reduced the phosphorylation of Akt and eNOS and the association of eNOS and HSP90. 25-OHC also enhanced endothelial cell apoptosis by decreasing Bcl-2 expression and increasing cleaved caspase-9 and cleaved caspase-3 expressions as well as caspase-3 activity. 25-OHC impaired endothelium-dependent vasodilation. These data demonstrated that 25-OHC could impair endothelial function by uncoupling and inhibiting eNOS activity as well as by inducing endothelial cell apoptosis. Our findings indicate that 25-OHC may play an important role in regulating atherosclerosis.
Collapse
Affiliation(s)
- Zhi-Jun Ou
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jing Chen
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Wei-Ping Dai
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Xiang Liu
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Yin-Ke Yang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Yan Li
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Ze-Bang Lin
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Tian-Tian Wang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Ying-Ying Wu
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Dan-Hong Su
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Tian-Pu Cheng
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Zhi-Ping Wang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jun Tao
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
29
|
Wang Y, Wang YL, Huang X, Yang Y, Zhao YJ, Wei CX, Zhao M. Ibutilide protects against cardiomyocytes injury via inhibiting endoplasmic reticulum and mitochondrial stress pathways. Heart Vessels 2016; 32:208-215. [PMID: 27639990 PMCID: PMC5288448 DOI: 10.1007/s00380-016-0891-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is a complex disease with multiple inter-relating causes culminating in rapid atrial activation and atrial structural remodeling. The contribution of endoplasmic reticulum and mitochondria stress to AF has been highlighted. As the class III antiarrhythmic agent, ibutilide are widely used to AF. This study was designed to explore whether ibutilide could treat AF by inhibiting endoplasmic reticulum stress pathways and mitochondria stress. The neonatal rat cardiomyocytes were isolated and exposed to H2O2, ibutilide was add to the culture medium 12 h. Then the cell viability, oxidative stress levels and apoptotic rate were analyzed. In addition, endoplasmic reticulum stress related protein (GRP78, GRP94, CHOP), mitochondria-dependent protein (Bax, Bcl-2) and caspase-3/9/12 were identified by real-time PCR and western blot analysis. In our results, remarkable decreased cell viability and oxidative stress levels were detected in cardiomyocytes after treating with H2O2. The apoptotic rate and the expression of proteins involved in mitochondrial stress and endoplasmic reticulum stress pathways increased. While ibutilide significantly inhibited these changes. These data suggested that ibutilide serves a protective role against H2O2-induced apoptosis of neonatal rat cardiomyocytes, and the mechanism is related to suppression of mitochondrial stress and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Yu Wang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, No. 22 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China
| | - Yi-Li Wang
- Affiliated Hospital of Inner Mongolia University for Nationalities, No. 1472 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China
| | - Xia Huang
- Affiliated Hospital of Inner Mongolia University for Nationalities, No. 1472 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China
| | - Yang Yang
- Affiliated Hospital of Inner Mongolia University for Nationalities, No. 1472 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China
| | - Ya-Jun Zhao
- Affiliated Hospital of Inner Mongolia University for Nationalities, No. 1472 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China
| | - Cheng-Xi Wei
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, No. 22 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China.
| | - Ming Zhao
- Affiliated Hospital of Inner Mongolia University for Nationalities, No. 1472 Holin He Street, Tongliao, Inner Mongolia, 028002, People's Republic of China.
| |
Collapse
|
30
|
Goette A, Kalman JM, Aguinaga L, Akar J, Cabrera JA, Chen SA, Chugh SS, Corradi D, D'Avila A, Dobrev D, Fenelon G, Gonzalez M, Hatem SN, Helm R, Hindricks G, Ho SY, Hoit B, Jalife J, Kim YH, Lip GYH, Ma CS, Marcus GM, Murray K, Nogami A, Sanders P, Uribe W, Van Wagoner DR, Nattel S. EHRA/HRS/APHRS/SOLAECE expert consensus on Atrial cardiomyopathies: Definition, characterisation, and clinical implication. J Arrhythm 2016; 32:247-78. [PMID: 27588148 PMCID: PMC4996910 DOI: 10.1016/j.joa.2016.05.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Andreas Goette
- Departement of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Germany
| | - Jonathan M Kalman
- University of Melbourne, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | | | | | | | | | - Sumeet S Chugh
- The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | | | - Mario Gonzalez
- Penn State Heart and Vascular Institute, Penn State University, Hershey, PA, USA
| | - Stephane N Hatem
- Department of Cardiology, Assistance Publique - Hô pitaux de Paris, Pitié-Salpêtrière Hospital, Sorbonne University, INSERM UMR_S1166, Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Robert Helm
- Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | | | - Siew Yen Ho
- Royal Brompton Hospital and Imperial College London, London, UK
| | - Brian Hoit
- UH Case Medical Center, Cleveland, OH, USA
| | | | | | | | | | | | | | | | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - William Uribe
- Electrophysiology Deparment at Centros Especializados de San Vicente Fundació n and Clínica CES. Universidad CES, Universidad Pontificia Bolivariana (UPB), Medellin, Colombia
| | | | - Stanley Nattel
- Université de Montréal, Montreal Heart Institute Research Center and McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
31
|
Goette A, Kalman JM, Aguinaga L, Akar J, Cabrera JA, Chen SA, Chugh SS, Corradi D, D'Avila A, Dobrev D, Fenelon G, Gonzalez M, Hatem SN, Helm R, Hindricks G, Ho SY, Hoit B, Jalife J, Kim YH, Lip GYH, Ma CS, Marcus GM, Murray K, Nogami A, Sanders P, Uribe W, Van Wagoner DR, Nattel S. EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication. Europace 2016; 18:1455-1490. [PMID: 27402624 DOI: 10.1093/europace/euw161] [Citation(s) in RCA: 507] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Andreas Goette
- Departement of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Germany
| | - Jonathan M Kalman
- University of Melbourne, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | | | | | | | | | - Sumeet S Chugh
- The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | | | - Mario Gonzalez
- Penn State Heart and Vascular Institute, Penn State University, Hershey, PA, USA
| | - Stephane N Hatem
- Department of Cardiology, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital; Sorbonne University; INSERM UMR_S1166; Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Robert Helm
- Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | | | - Siew Yen Ho
- Royal Brompton Hospital and Imperial College London, London, UK
| | - Brian Hoit
- UH Case Medical Center, Cleveland, OH, USA
| | | | | | | | | | | | | | | | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - William Uribe
- Electrophysiology Deparment at Centros Especializados de San Vicente Fundación and Clínica CES. Universidad CES, Universidad Pontificia Bolivariana (UPB), Medellin, Colombia
| | | | - Stanley Nattel
- Université de Montréal, Montreal Heart Institute Research Center and McGill University, Montreal, Quebec, Canada .,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
32
|
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: Definition, characterization, and clinical implication. Heart Rhythm 2016; 14:e3-e40. [PMID: 27320515 DOI: 10.1016/j.hrthm.2016.05.028] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/21/2022]
|
33
|
Chen MC, Chang JP, Lin YS, Pan KL, Ho WC, Liu WH, Chang TH, Huang YK, Fang CY, Chen CJ. Deciphering the gene expression profile of peroxisome proliferator-activated receptor signaling pathway in the left atria of patients with mitral regurgitation. J Transl Med 2016; 14:157. [PMID: 27250500 PMCID: PMC4890244 DOI: 10.1186/s12967-016-0871-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/19/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Differentially expressed genes in the left atria of mitral regurgitation (MR) pigs have been linked to peroxisome proliferator-activated receptor (PPAR) signaling pathway in the KEGG pathway. However, specific genes of the PPAR signaling pathway in the left atria of MR patients have never been explored. METHODS This study enrolled 15 MR patients with heart failure, 7 patients with aortic valve disease and heart failure, and 6 normal controls. We used PCR assay (84 genes) for PPAR pathway and quantitative RT-PCR to study specific genes of the PPAR pathway in the left atria. RESULTS Gene expression profiling analysis through PCR assay identified 23 genes to be differentially expressed in the left atria of MR patients compared to normal controls. The expressions of APOA1, ACADM, FABP3, ETFDH, ECH1, CPT1B, CPT2, SLC27A6, ACAA2, SMARCD3, SORBS1, EHHADH, SLC27A1, PPARGC1B, PPARA and CPT1A were significantly up-regulated, whereas the expression of PLTP was significantly down-regulated in the MR patients compared to normal controls. The expressions of HMGCS2, ACADM, FABP3, MLYCD, ECH1, ACAA2, EHHADH, CPT1A and PLTP were significantly up-regulated in the MR patients compared to patients with aortic valve disease. Notably, only ACADM, FABP3, ECH1, ACAA2, EHHADH, CPT1A and PLTP of the PPAR pathway were significantly differentially expressed in the MR patients compared to patients with aortic valve disease and normal controls. CONCLUSIONS Differentially expressed genes of the PPAR pathway have been identified in the left atria of MR patients compared with patients with aortic valve disease and normal controls.
Collapse
Affiliation(s)
- Mien-Cheng Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung, 83301, Taiwan.
| | - Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Kuo-Li Pan
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Wan-Chun Ho
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung, 83301, Taiwan
| | - Wen-Hao Liu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung, 83301, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Yao-Kuang Huang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chih-Yuan Fang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung, 83301, Taiwan
| | - Chien-Jen Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 123 Ta Pei Road, Niao Sung District, Kaohsiung, 83301, Taiwan
| |
Collapse
|
34
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
35
|
|
36
|
The electrical heart: 25 years of discovery in cardiac electrophysiology, arrhythmias and sudden death. Cardiovasc Pathol 2016; 25:149-57. [DOI: 10.1016/j.carpath.2015.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 12/16/2022] Open
|
37
|
Geng X, Yan L, Dong J, Liang Y, Deng Y, Li T, Luo T, Lin H, Zhang S. Role of Nox2 and p22phox in Persistent Postoperative Hypertension in Aldosterone-Producing Adenoma Patients after Adrenalectomy. Int J Endocrinol 2016; 2016:2395634. [PMID: 27057164 PMCID: PMC4771902 DOI: 10.1155/2016/2395634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022] Open
Abstract
Adrenal aldosterone-producing adenoma (APA), producing the salt-retaining hormone aldosterone, commonly causes secondary hypertension, which often persists after unilateral adrenalectomy. Although persistent hypertension was correlated with residual hormone aldosterone, the in vivo mechanism remains unclear. NADPH oxidase is the critical cause of aldosterone synthesis in vitro. Nox2 and p22phox comprise the NADPH oxidase catalytic core, serving to initiate a reactive oxygen species (ROS) cascade that may participate in the pathology. mRNAs of seven NADPH oxidase isoforms in APA were evaluated by RT-PCR and Q-PCR and their proteins by immunohistochemistry and Western blotting. NADPH oxidase activity was also detected. Nox2 and p22phox were especially abundant in APA. Particularly higher Nox2 and p22phox gene and protein levels were seen in APA than controls. Significant correlations between Nox2 mRNA and aldosterone synthase (CYP11B2) mRNA (R = 0.66, P < 0.01) and Nox2 protein and baseline plasma aldosterone concentration (PAC) (R = 0.503, P < 0.01) were detected in APA; however, none were found between p22phox mRNA, CYP11B2 mRNA, p22phox protein, and baseline PAC. Importantly, we found that Nox2 localized specifically in hyperplastic zona glomerulosa cells. In conclusion, our results highlight that Nox2 and p22phox may be directly involved in pathological aldosterone production and zona glomerulosa cell proliferation after APA resection.
Collapse
Affiliation(s)
- Xiaojing Geng
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Endocrinology Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Li Yan
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Dong
- Department of General Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Liang
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yajuan Deng
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ting Li
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tongfeng Luo
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Endocrinology Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Hailun Lin
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shaoling Zhang
- Department of Endocrinology Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- *Shaoling Zhang:
| |
Collapse
|
38
|
Chang JP, Chen MC, Liu WH, Lin YS, Huang YK, Pan KL, Ho WC, Fang CY, Chen CJ, Chen HC. Mitochondrial apoptotic pathway activation in the atria of heart failure patients due to mitral and tricuspid regurgitation. Exp Mol Pathol 2015; 99:65-73. [PMID: 26004742 DOI: 10.1016/j.yexmp.2015.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/05/2015] [Accepted: 05/20/2015] [Indexed: 11/19/2022]
Abstract
Apoptosis occurs in atrial cardiomyocytes in mitral and tricuspid valve disease. The purpose of this study was to examine the respective roles of the mitochondrial and tumor necrosis factor-α receptor associated death domain (TRADD)-mediated death receptor pathways for apoptosis in the atrial cardiomyocytes of heart failure patients due to severe mitral and moderate-to-severe tricuspid regurgitation. This study comprised eighteen patients (7 patients with persistent atrial fibrillation and 11 in sinus rhythm). Atrial appendage tissues were obtained during surgery. Three purchased normal human left atrial tissues served as normal controls. Moderately-to-severely myolytic cardiomyocytes comprised 59.7±22.1% of the cardiomyocytes in the right atria and 52.4±12.9% of the cardiomyocytes in the left atria of mitral and tricuspid regurgitation patients with atrial fibrillation group and comprised 58.4±24.8% of the cardiomyocytes in the right atria of mitral and tricuspid regurgitation patients with sinus rhythm. In contrast, no myolysis was observed in the normal human adult left atrial tissue samples. Immunohistochemical analysis showed expression of cleaved caspase-9, an effector of the mitochondrial pathways, in the majority of right atrial cardiomyocytes (87.3±10.0%) of mitral and tricuspid regurgitation patients with sinus rhythm, and right atrial cardiomyocytes (90.6±31.4%) and left atrial cardiomyocytes (70.7±22.0%) of mitral and tricuspid regurgitation patients with atrial fibrillation. In contrast, only 5.7% of cardiomyocytes of the normal left atrial tissues showed strongly positive expression of cleaved caspase-9. Of note, none of the atrial cardiomyocytes in right atrial tissue in sinus rhythm and in the fibrillating right and left atria of mitral and tricuspid regurgitation patients, and in the normal human adult left atrial tissue samples showed cleaved caspase-8 expression, which is a downstream effector of TRADD of the death receptor pathway. Immunoblotting of atrial extracts showed that there was enhanced expression of cytosolic cytochrome c, an effector of the mitochondrial pathways, but no expression of membrane TRADD and cytosolic caspase-8 in the right atrial tissue of mitral and tricuspid regurgitation patients with sinus rhythm, and right atrial and left atrial tissues of mitral and tricuspid regurgitation patients with atrial fibrillation. Taken together, this study showed that mitochondrial pathway for apoptosis was activated in the right atria in sinus rhythm and in the left and right atria in atrial fibrillation of heart failure patients due to mitral and tricuspid regurgitation, and this mitochondrial pathway activation may contribute to atrial contractile dysfunction and enlargement in this clinical setting.
Collapse
Affiliation(s)
- Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Mien-Cheng Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan.
| | - Wen-Hao Liu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yao-Kuang Huang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Kuo-Li Pan
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Wan-Chun Ho
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chih-Yuan Fang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Chien-Jen Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| | - Huang-Chung Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taiwan
| |
Collapse
|
39
|
Chen MC, Chang JP, Chang TH, Hsu SD, Huang HD, Ho WC, Wang FS, Hsiao CC, Liu WH. Unraveling regulatory mechanisms of atrial remodeling of mitral regurgitation pigs by gene expression profiling analysis: role of type I angiotensin II receptor antagonist. Transl Res 2015; 165:599-620. [PMID: 25500755 DOI: 10.1016/j.trsl.2014.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 12/12/2022]
Abstract
Left atrial enlargement associated with mitral regurgitation (MR) predicts a poor prognosis. However, the underlying regulatory mechanisms of atrial remodeling remain unclear. We used high-density oligonucleotide microarrays and enrichment analysis to identify the alteration of RNA expression pattern and biological processes involved in the atrial remodeling of pigs with and without MR. Gene arrays from left atria tissues were compared in 13 pigs (iatrogenic MR pigs [n = 6], iatrogenic MR pigs treated with valsartan [n = 4], and pigs without MR [n = 3]). A total of 22 genes were differentially upregulated by altered fold change >2.0 (Log2FC > 1), and 49 genes were differentially downregulated by altered fold change <0.5 (Log2FC < -1) in the left atria of the MR pigs compared with the pigs without MR. Enrichment analysis showed that renin-angiotensin system was identified in the Kyoto Encyclopedia of Genes and Genomes pathway. Notably, 12 of the 22 upregulated genes were identified to be downregulated by valsartan and 10 of the 49 downregulated genes were identified to be upregulated by valsartan. The tissue concentrations of angiotensin II and gene expression of hypertrophic gene, myosin regulatory light chain 2, ventricular isoforms, and fibrosis-related genes were significantly increased in the MR pigs compared with pigs without MR. In conclusion, differentially expressed transcriptome and related biological pathways have been identified in the left atria of the MR pigs compared with pigs without MR. Additionally, some of the differentially expressed genes could be regulated by type I angiotensin II receptor blocker.
Collapse
Affiliation(s)
- Mien-Cheng Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Jen-Ping Chang
- Division of Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Da Hsu
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Wan-Chun Ho
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Feng-Sheng Wang
- Department of Medical Research, Graduate institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chang-Chun Hsiao
- Department of Medical Research, Graduate institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Hao Liu
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
40
|
|
41
|
Zhang Y, Shimizu H, Siu KL, Mahajan A, Chen JN, Cai H. NADPH oxidase 4 induces cardiac arrhythmic phenotype in zebrafish. J Biol Chem 2014; 289:23200-23208. [PMID: 24962575 DOI: 10.1074/jbc.m114.587196] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress has been implicated in cardiac arrhythmia, although a causal relationship remains undefined. We have recently demonstrated a marked up-regulation of NADPH oxidase isoform 4 (NOX4) in patients with atrial fibrillation, which is accompanied by overproduction of reactive oxygen species (ROS). In this study, we investigated the impact on the cardiac phenotype of NOX4 overexpression in zebrafish. One-cell stage embryos were injected with NOX4 RNA prior to video recording of a GFP-labeled (myl7:GFP zebrafish line) beating heart in real time at 24-31 h post-fertilization. Intriguingly, NOX4 embryos developed cardiac arrhythmia that is characterized by irregular heartbeats. When quantitatively analyzed by an established LQ-1 program, the NOX4 embryos displayed much more variable beat-to-beat intervals (mean S.D. of beat-to-beat intervals was 0.027 s/beat in control embryos versus 0.038 s/beat in NOX4 embryos). Both the phenotype and the increased ROS in NOX4 embryos were attenuated by NOX4 morpholino co-injection, treatments of the embryos with polyethylene glycol-conjugated superoxide dismutase, or NOX4 inhibitors fulvene-5, 6-dimethylamino-fulvene, and proton sponge blue. Injection of NOX4-P437H mutant RNA had no effect on the cardiac phenotype or ROS production. In addition, phosphorylation of calcium/calmodulin-dependent protein kinase II was increased in NOX4 embryos but diminished by polyethylene glycol-conjugated superoxide dismutase, whereas its inhibitor KN93 or AIP abolished the arrhythmic phenotype. Taken together, our data for the first time uncover a novel pathway that underlies the development of cardiac arrhythmia, namely NOX4 activation, subsequent NOX4-specific NADPH-driven ROS production, and redox-sensitive CaMKII activation. These findings may ultimately lead to novel therapeutics targeting cardiac arrhythmia.
Collapse
Affiliation(s)
- Yixuan Zhang
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA and Los Angeles, California 90095
| | - Hirohito Shimizu
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, California 90095
| | - Kin Lung Siu
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA and Los Angeles, California 90095
| | - Aman Mahajan
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA and Los Angeles, California 90095
| | - Jau-Nian Chen
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, California 90095
| | - Hua Cai
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA and Los Angeles, California 90095.
| |
Collapse
|
42
|
Corradi D. Atrial fibrillation from the pathologist's perspective. Cardiovasc Pathol 2013; 23:71-84. [PMID: 24462196 DOI: 10.1016/j.carpath.2013.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/03/2013] [Accepted: 12/07/2013] [Indexed: 12/18/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia encountered in clinical practice, is associated with increased morbidity and mortality. Electrophysiologically, it is characterized by a high rate of asynchronous atrial cell depolarization causing a loss of atrial contractile function and irregular ventricular rates. For a long time, AF was considered as a pure functional disorder without any structural background. Only in recent years, have new mapping and imaging techniques identified atrial locations, which are very often involved in the initiation and maintenance of this supraventricular arrhythmia (i.e. the distal portion of the pulmonary veins and the surrounding atrial myocardium). Morphological analysis of these myocardial sites has demonstrated significant structural remodeling as well as paved the way for further knowledge of AF natural history, pathogenesis, and treatment. This architectural myocardial disarrangement is induced by the arrhythmia itself and the very frequently associated cardiovascular disorders. At the same time, the structural remodeling is also capable of sustaining AF, thereby creating a sort of pathogenetic vicious circle. This review focuses on current understanding about the structural and genetic bases of AF with reference to their classification, pathogenesis, and clinical implications.
Collapse
Affiliation(s)
- Domenico Corradi
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy.
| |
Collapse
|
43
|
Oxidative stress in atrial fibrillation: an emerging role of NADPH oxidase. J Mol Cell Cardiol 2013; 62:72-9. [PMID: 23643589 DOI: 10.1016/j.yjmcc.2013.04.019] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 03/22/2013] [Accepted: 04/18/2013] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. Patients with AF have up to seven-fold higher risk of suffering from ischemic stroke. Better understanding of etiologies of AF and its thromboembolic complications are required for improved patient care, as current anti-arrhythmic therapies have limited efficacy and off target effects. Accumulating evidence has implicated a potential role of oxidative stress in the pathogenesis of AF. Excessive production of reactive oxygen species (ROS) is likely involved in the structural and electrical remodeling of the heart, contributing to fibrosis and thrombosis. In particular, NADPH oxidase (NOX) has emerged as a potential enzymatic source for ROS production in AF based on growing evidence from clinical and animal studies. Indeed, NOX can be activated by known upstream triggers of AF such as angiotensin II and atrial stretch. In addition, treatments such as statins, antioxidants, ACEI or AT1RB have been shown to prevent post-operative AF; among which ACEI/AT1RB and statins can attenuate NOX activity. On the other hand, detailed molecular mechanisms by which specific NOX isoform(s) are involved in the pathogenesis of AF and the extent to which activation of NOX plays a causal role in AF development remains to be determined. The current review discusses causes and consequences of oxidative stress in AF with a special focus on the emerging role of NOX pathways.
Collapse
|
44
|
Ci HB, Ou ZJ, Chang FJ, Liu DH, He GW, Xu Z, Yuan HY, Wang ZP, Zhang X, Ou JS. Endothelial microparticles increase in mitral valve disease and impair mitral valve endothelial function. Am J Physiol Endocrinol Metab 2013; 304:E695-E702. [PMID: 23384770 DOI: 10.1152/ajpendo.00016.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitral valve endothelial cells are important for maintaining lifelong mitral valve integrity and function. Plasma endothelial microparticles (EMPs) increased in various pathological conditions related to activation of endothelial cells. However, whether EMPs will increase in mitral valve disease and their relationship remains unclear. Here, 81 patients with mitral valve disease and 45 healthy subjects were analyzed for the generation of EMPs by flow cytometry. Human mitral valve endothelial cells (HMVECs) were treated with EMPs. The phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), the association of eNOS and heat shock protein 90 (HSP90), and the generation of nitric oxide (NO) and superoxide anion (O(2)(∙-)) were measured. EMPs were increased significantly in patients with mitral valve disease compared with those in healthy subjects. EMPs were negatively correlated with mitral valve area in patients with isolated mitral stenosis. EMPs were significantly higher in the group with severe mitral regurgitation than those in the group with mild and moderate mitral regurgitation. Furthermore, EMPs were decreased dramatically in both Akt and eNOS phosphorylation and the association of HSP90 with eNOS in HMVECs. EMPs decreased NO production but increased O(2)(∙-) generation in HMVECs. Our data demonstrated that EMPs were significantly increased in patients with mitral valve disease. The increase of EMPs can in turn impair HMVEC function by inhibiting the Akt/eNOS-HSP90 signaling pathway. These findings suggest that EMPs may be a therapeutic target for mitral valve disease.
Collapse
Affiliation(s)
- Hong-Bo Ci
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
PKCβ inhibition with ruboxistaurin reduces oxidative stress and attenuates left ventricular hypertrophy and dysfunction in rats with streptozotocin-induced diabetes. Clin Sci (Lond) 2011; 122:161-73. [PMID: 21892921 DOI: 10.1042/cs20110176] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Oxidative stress plays critical roles in the development of diabetic cardiovascular complications, including myocardial hypertrophy. The β isoform of PKC (protein kinase C) is preferentially overexpressed in the myocardium of diabetic subjects accompanied with increased activation of the pro-oxidant enzyme NADPH oxidase, which may exacerbate oxidative stress. We hypothesized that myocardial PKCβ is a major upstream mediator of oxidative stress in diabetes and that PKCβ inhibition can attenuate myocardial hypertrophy and dysfunction. Control or streptozotocin-induced diabetic rats were treated with the selective PKCβ inhibitor RBX (ruboxistaurin; 1 mg/kg of body weight per day) or the antioxidant NAC (N-acetylcysteine) for 4 weeks. LV (left ventricular) dimensions and functions were detected by echocardiography. 15-F2t-isoprostane (a specific index of oxidative stress) and myocardial activities of superoxide dismutase as well as protein levels of NADPH oxidase were assessed by immunoassay or Western blotting. Echocardiography revealed that the LV mass/body weight ratio was significantly increased in diabetic rats (P<0.01 compared with the control group) in parallel with the impaired LV relaxation. A significant increase in cardiomyocyte cross-sectional area was observed in diabetic rats accompanied by an increased production of O2- (superoxide anion) and 15-F2t-isoprostane (all P<0.05 compared with the control group). RBX normalized these changes with concomitant inhibition of PKCβ2 activation and prevention of NADPH oxidase subunit p67phox membrane translocation and p22phox overexpression. The effects of RBX were comparable with that of NAC, except that NAC was inferior to RBX in attenuating cardiac dysfunction. It is concluded that RBX can ameliorate myocardial hypertrophy and dysfunction in diabetes, which may represent a novel therapy in the prevention of diabetic cardiovascular complications.
Collapse
|
46
|
Xu Q, Guan YQ, Zhang D, Su GH. The effects of statin on atrial fibrillation: a meta-analysis of published data from randomized controlled trials. Curr Med Res Opin 2011; 27:1771-9. [PMID: 21806315 DOI: 10.1185/03007995.2011.605785] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Some clinical and experimental studies have shown the use of statins could protect against AF, but there are not adequate data at present. OBJECTIVES We performed a meta-analysis of randomized trials with statins on the endpoint of incidence of AF to estimate the impact of statin use on AF development. METHODS We searched PUBMED, EMBASE and the Cochrane controlled Trials Register (Cochrane Library Issue 4, 2010) up to November 2010 to identify studies covering the use of statins on atrial fibrillation. RESULTS In published data from nine short term trials (1044 patients, 421 AF), the effect of statins was significantly associated with a decreased risk of recurrence of AF (OR 0.43, 95% CI 0.25 to 0.73, P = 0.002). The result of OR was higher when studies with Jadad score ≤3 were excluded (OR 0.32, 95% CI 0.18 to 0.54, P ≤ 0.0001). Among four long term trials (12,442 patients, 618 AF), the effect of statins was associated with a decreased risk of recurrence of AF (OR 0.81, 95% CI 0.68 to 0.97, P = 0.02). In three long term trials of more intensive versus standard statin (9130 patients, 188 AF), there was no evidence of a reduction in the risk of AF (OR 1.05, 95% CI 0.79 to 1.40, P= 0.74). CONCLUSION Our meta-analysis suggests that the use of statins may be associated with preventing AF in short term trials and long term trials, but in the long term trials of more intensive versus standard statin, there was no evidence of a reduction in the risk of AF. However, we still need large-scale randomized double blind statin trials with AF occurrence as the main endpoint in order to finally confirm the benefits of statin in AF patients.
Collapse
Affiliation(s)
- Qi Xu
- School of Medicine, Shandong University, Shandong Province, China
| | | | | | | |
Collapse
|
47
|
Chen MC, Chang JP, Ho WC, Liu WH, Chen CJ, Yang CH, Chen YL, Tsai TH. Expression of spliceosome assembly factor SC-35 in TUNEL-positive atrial cardiomyocytes in mitral and tricuspid regurgitation: Viability of atrial cardiomyocytes. Int J Cardiol 2011; 151:323-7. [DOI: 10.1016/j.ijcard.2010.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 04/12/2010] [Accepted: 06/04/2010] [Indexed: 12/27/2022]
|