1
|
Tagami K, Okuzawa T, Yoshida K, Mishima R, Obara N, Kunimatsu A, Koide M, Teranishi T, Itakura K, Ikeda K, Murohara T, Nagata K. L-arginine ameliorates hypertension and cardiac mitochondrial abnormalities but not cardiac injury in male metabolic syndrome rats. Physiol Rep 2025; 13:e70183. [PMID: 39980190 PMCID: PMC11842508 DOI: 10.14814/phy2.70183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 02/22/2025] Open
Abstract
L-Arginine supplementation has beneficial effects on metabolic disorders in rodents. We here investigated the effects of exogenous L-arginine on cardiac pathology and mitochondrial reactive oxygen species (ROS) production and dynamics in DahlS.Z-Leprfa/Leprfa (DS/obese) rats, a model of metabolic syndrome (MetS). DS/obese rats and their lean homozygous littermate (DahlS.Z-Lepr+/Lepr+, or DS/lean) controls were provided with drinking water containing 0.50% L-arginine-HCl or 0.85% L-alanine (isonitrogenous control) from 13 to 17 weeks of age. L-Arginine supplementation markedly alleviated hypertension without affecting cardiac injury in MetS rats. It also attenuated the increase in ROS production apparent in cardiac mitochondria isolated from MetS rats as well as suppressed the associated upregulation of Nox4 mRNA and protein in the heart. Furthermore, L-arginine reversed the decrease in the size of cardiac mitochondria as well as changes in the expression of DRP1 and OPA1 proteins apparent in the L-alanine-treated MetS rat heart. Cardiac arginase II gene expression and arginase activity were increased by L-arginine treatment in MetS rats but not CONT rats. L-Arginine supplementation thus ameliorated hypertension and cardiac mitochondrial abnormalities in MetS rats, with the lack of a cardioprotective effect possibly being due to increased arginase activity.
Collapse
Affiliation(s)
- Kaito Tagami
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Touko Okuzawa
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Keisuke Yoshida
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Rin Mishima
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Natsuki Obara
- Department of Medical TechnologyNagoya University School of Health SciencesNagoyaJapan
| | - Asuko Kunimatsu
- Department of Medical TechnologyNagoya University School of Health SciencesNagoyaJapan
| | - Mayako Koide
- Department of Medical TechnologyNagoya University School of Health SciencesNagoyaJapan
| | - Tamami Teranishi
- Department of Medical TechnologyNagoya University School of Health SciencesNagoyaJapan
| | - Koji Itakura
- Division for Medical Research EngineeringNagoya University Graduate School of MedicineNagoyaJapan
| | - Katsuhide Ikeda
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| | - Toyoaki Murohara
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kohzo Nagata
- Pathophysiology Sciences, Department of Integrated Health SciencesNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
2
|
Nagai M, Rommel KP, Po SS, Dasari TW. Autonomic neuromodulation for cardiomyopathy associated with metabolic syndrome - Prevention of precursors for heart failure with preserved ejection fraction. Hypertens Res 2024; 47:3318-3329. [PMID: 39261699 DOI: 10.1038/s41440-024-01886-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024]
Abstract
Metabolic syndrome (MetS) induces a systemic inflammatory state which can lead to cardiomyopathy, manifesting clinically as heart failure (HF) with preserved ejection fraction (HFpEF). MetS components are intricately linked to the pathophysiologic processes of myocardial remodeling. Increased sympathetic nervous system activity, which is noted as an upstream factor of MetS, has been linked to adverse myocardial structural changes. Since renal denervation and vagus nerve stimulation have a sympathoinhibitory effect, attention has been paid to the cardioprotective effects of autonomic neuromodulation. In this review, the pathophysiology underlying the relationship between MetS and HF is elucidated, and the evidence regarding autonomic neuromodulation in HFpEF is summarized.
Collapse
Affiliation(s)
- Michiaki Nagai
- Cardiovascular section, Department of Medicine, University of Oklahoma, Health Science Center, Oklahoma, USA.
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan.
| | - Karl-Philipp Rommel
- Department of Cardiology, University Medical Center Mainz and German Center for Cardiovascular Research, Mainz, Germany
| | - Sunny S Po
- Cardiovascular section, Department of Medicine, University of Oklahoma, Health Science Center, Oklahoma, USA
| | - Tarun W Dasari
- Cardiovascular section, Department of Medicine, University of Oklahoma, Health Science Center, Oklahoma, USA.
| |
Collapse
|
3
|
Evans AJ, Li YL. Remodeling of the Intracardiac Ganglia During the Development of Cardiovascular Autonomic Dysfunction in Type 2 Diabetes: Molecular Mechanisms and Therapeutics. Int J Mol Sci 2024; 25:12464. [PMID: 39596529 PMCID: PMC11594459 DOI: 10.3390/ijms252212464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most significant health issues worldwide, with associated healthcare costs estimated to surpass USD 1054 billion by 2045. The leading cause of death in T2DM patients is the development of cardiovascular disease (CVD). In the early stages of T2DM, patients develop cardiovascular autonomic dysfunction due to the withdrawal of cardiac parasympathetic activity. Diminished cardiac parasympathetic tone can lead to cardiac arrhythmia-related sudden cardiac death, which accounts for 50% of CVD-related deaths in T2DM patients. Regulation of cardiovascular parasympathetic activity is integrated by neural circuitry at multiple levels including afferent, central, and efferent components. Efferent control of cardiac parasympathetic autonomic tone is mediated through the activity of preganglionic parasympathetic neurons located in the cardiac extensions of the vagus nerve that signals to postganglionic parasympathetic neurons located in the intracardiac ganglia (ICG) on the heart. Postganglionic parasympathetic neurons exert local control on the heart, independent of higher brain centers, through the release of neurotransmitters, such as acetylcholine. Structural and functional alterations in cardiac parasympathetic postganglionic neurons contribute to the withdrawal of cardiac parasympathetic tone, resulting in arrhythmogenesis and sudden cardiac death. This review provides an overview of the remodeling of parasympathetic postganglionic neurons in the ICG, and potential mechanisms contributing to the withdrawal of cardiac parasympathetic tone, ventricular arrhythmogenesis, and sudden cardiac death in T2DM. Improving cardiac parasympathetic tone could be a therapeutic avenue to reduce malignant ventricular arrhythmia and sudden cardiac death, increasing both the lifespan and improving quality of life of T2DM patients.
Collapse
Affiliation(s)
- Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Park H, Park B, Kim KS, Son YH, Park SJ, Lee K, Park H, Park J. Therapeutic Potential of Intermittent Hypoxia in Atrial Fibrillation. Int J Mol Sci 2024; 25:11085. [PMID: 39456866 PMCID: PMC11508233 DOI: 10.3390/ijms252011085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Intermittent hypoxia (IH) has been extensively studied in recent years, demonstrating adverse and beneficial effects on several physiological systems. However, the precise mechanism underlying its cardiac effects on the heart remains unclear. This study aims to explore the effect of treatment on atrial fibrillation under IH conditions, providing data that can potentially be used in the treatment of heart disease. An atrial fibrillation (AF) model was induced by injecting monocrotaline (MCT, 60 mg/kg) into rats. The study included 32 rats divided into four groups: Control, Control + IH, AF, and AF + IH. We evaluated molecular changes associated with AF using ELISA and Western blot and performed electrophysiological experiments to evaluate AF. Arrhythmia-related calcium and fibrosis markers were investigated. Phosphorylation levels of CaMKII, Phospholamban, and RyR2 all increased in the AF group but decreased in the IH-exposed group. Additionally, fibrosis marker expressions such as SMA, MMP2, MMP9, and TGF-β increased in the AF group but were significantly downregulated with IH treatment. Connexin 43 and AQP4 expression were restored in the IH-treated group. These findings suggest that IH may prevent AF by downregulating the expression of calcium-handling proteins and fibrosis-associated proteins in an AF-induced rat model.
Collapse
Affiliation(s)
- Hyewon Park
- Department of Cardiology, College of Medicine, Ewha Womans University School of Medicine, Seoul 07804, Republic of Korea; (H.P.); (B.P.)
| | - Bokyeong Park
- Department of Cardiology, College of Medicine, Ewha Womans University School of Medicine, Seoul 07804, Republic of Korea; (H.P.); (B.P.)
| | - Kyu-sung Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Inha University School of Medicine, Incheon 22332, Republic of Korea;
- Inha Research Institute for Aerospace Medicine, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Young Hoon Son
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA 30322, USA; (Y.H.S.); (S.J.P.)
| | - Sung Jin Park
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA 30322, USA; (Y.H.S.); (S.J.P.)
| | - Kichang Lee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA;
- Harvard Medical School, Boston, MA 02115, USA
| | - Hyelim Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Inha University School of Medicine, Incheon 22332, Republic of Korea;
- Inha Research Institute for Aerospace Medicine, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Junbeom Park
- Department of Cardiology, College of Medicine, Ewha Womans University School of Medicine, Seoul 07804, Republic of Korea; (H.P.); (B.P.)
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, GA 30322, USA; (Y.H.S.); (S.J.P.)
| |
Collapse
|
5
|
Prathumsap N, Ongnok B, Khuanjing T, Arinno A, Maneechote C, Chunchai T, Arunsak B, Kerdphoo S, Chattipakorn SC, Chattipakorn N. Muscarinic and nicotinic receptors stimulation by vagus nerve stimulation ameliorates trastuzumab-induced cardiotoxicity via reducing programmed cell death in rats. Toxicol Appl Pharmacol 2024; 491:117074. [PMID: 39168189 DOI: 10.1016/j.taap.2024.117074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Despite its efficacy in human epidermal growth factor receptor 2 positive cancer treatment, trastuzumab-induced cardiotoxicity (TIC) has become a growing concern. Due to the lack of cardiomyocyte regeneration and proliferation in adult heart, cell death significantly contributes to cardiovascular diseases. Cardiac autonomic modulation by vagus nerve stimulation (VNS) has shown cardioprotective effects in several heart disease models, while the effects of VNS and its underlying mechanisms against TIC have not been found. Forty adult male Wistar rats were divided into 5 groups: (i) control without VNS (CSham) group, (ii) trastuzumab (4 mg/kg/day, i.p.) without VNS (TSham) group, (iii) trastuzumab + VNS (TVNS) group, (iv) trastuzumab + VNS + mAChR blocker (atropine; 1 mg/kg/day, ip, TVNS + Atro) group, and (v) trastuzumab + VNS + nAChR blocker (mecamylamine; 7.5 mg/kg/day, ip, TVNS + Mec) group. Our results showed that trastuzumab induced cardiac dysfunction by increasing autonomic dysfunction, mitochondrial dysfunction/dynamics imbalance, and cardiomyocyte death including apoptosis, autophagic deficiency, pyroptosis, and ferroptosis, which were notably alleviated by VNS. However, mAChR and nAChR blockers significantly inhibited the beneficial effects of VNS on cardiac autonomic dysfunction, mitochondrial dysfunction, cardiomyocyte apoptosis, pyroptosis, and ferroptosis. Only nAChR could counteract the protective effects of VNS on cardiac mitochondrial dynamics imbalance and autophagy insufficiency. Therefore, VNS prevented TIC by rebalancing autonomic activity, ameliorating mitochondrial dysfunction and cardiomyocyte death through mAChR and nAChR activation. The current study provides a novel perspective elucidating the potential treatment of VNS, thus also offering other pharmacological therapeutic promises in TIC patients.
Collapse
MESH Headings
- Animals
- Vagus Nerve Stimulation/methods
- Male
- Rats, Wistar
- Cardiotoxicity
- Rats
- Trastuzumab/toxicity
- Trastuzumab/pharmacology
- Apoptosis/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Receptors, Muscarinic/metabolism
- Receptors, Muscarinic/drug effects
- Receptors, Nicotinic/metabolism
- Receptors, Nicotinic/drug effects
- Nicotinic Antagonists/pharmacology
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/toxicity
- Vagus Nerve/drug effects
Collapse
Affiliation(s)
- Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
6
|
Xue R, Wu Q, Guo L, Ye D, Cao Q, Zhang M, Xian Y, Chen M, Yan K, Zheng J. Pyridostigmine attenuated high-fat-diet induced liver injury by the reduction of mitochondrial damage and oxidative stress via α7nAChR and M3AChR. J Biochem Mol Toxicol 2024; 38:e23671. [PMID: 38454809 DOI: 10.1002/jbt.23671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 01/18/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Obesity is a major cause of nonalcohol fatty liver disease (NAFLD), which is characterized by hepatic fibrosis, lipotoxicity, inflammation, and apoptosis. Previous studies have shown that an imbalance in the autonomic nervous system is closely related to the pathogenesis of NAFLD. In this study, we investigated the effects of pyridostigmine (PYR), a cholinesterase (AChE) inhibitor, on HFD-induced liver injury and explored the potential mechanisms involving mitochondrial damage and oxidative stress. A murine model of HFD-induced obesity was established using the C57BL/6 mice, and PYR (3 mg/kg/d) or placebo was administered for 20 weeks. PYR reduced the body weight and liver weight of the HFD-fed mice. Additionally, the serum levels of IL-6, TNF-α, cholesterol, and triglyceride were significantly lower in the PYR-treated versus the untreated mice, corresponding to a decrease in hepatic fibrosis, lipid accumulation, and apoptosis in the former. Furthermore, the mitochondrial morphology improved significantly in the PYR-treated group. Consistently, PYR upregulated ATP production and the mRNA level of the mitochondrial dynamic factors OPA1, Drp1 and Fis1, and the mitochondrial unfolded protein response (UPRmt) factors LONP1 and HSP60. Moreover, PYR treatment activated the Keap1/Nrf2 pathway and upregulated HO-1 and NQO-1, which mitigated oxidative injury as indicated by decreased 8-OHDG, MDA and H2 O2 levels, and increased SOD activity. Finally, PYR elevated acetylcholine (ACh) levels by inhibiting AChE, and upregulated the α7nAChR and M3AChR proteins in the HFD-fed mice. PYR alleviated obesity-induced hepatic injury in mice by mitigating mitochondrial damage and oxidative stress via α7nAChR and M3AChR.
Collapse
Affiliation(s)
- Runqing Xue
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Qing Wu
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Lulu Guo
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
- The College of Life Sciences, Northwest University, Xi'an, China
| | - Dan Ye
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Qing Cao
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Meng Zhang
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Yushan Xian
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Minchun Chen
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Kangkang Yan
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Jie Zheng
- Department of Pharmacy, The Affiliated Hospital of Northwest University, Xi'an, China
| |
Collapse
|
7
|
Elkattawy HA, Mahmoud SM, Hassan AES, Behiry A, Ebrahim HA, Ibrahim AM, Zaghamir DEF, El-Sherbiny M, El-Sayed SF. Vagal Stimulation Ameliorates Non-Alcoholic Fatty Liver Disease in Rats. Biomedicines 2023; 11:3255. [PMID: 38137476 PMCID: PMC10741668 DOI: 10.3390/biomedicines11123255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The harmful consequences of non-alcoholic fatty liver disease (NAFLD) are posing an increasing threat to public health as the incidence of diabetes and obesity increases globally. A non-invasive treatment with a range of autonomic and metabolic benefits is transcutaneous vagus nerve stimulation (tVNS). AIM OF THE STUDY To investigate the possible preventive impacts of VNS against adult rats' NAFLD caused by a high-fat diet (HFD) and to clarify the underlying mechanisms. METHODS A total of thirty-two adult male rats were split into two groups: the HFD-induced NAFLD group (n = 24) and the control normal group (n = 8). The obesogenic diet was maintained for 12 weeks to induce hepatic steatosis. The HFD-induced NAFLD group (n = 24) was separated into three groups: the group without treatment (n = 8), the group with sham stimulation (n = 8), and the group with VNS treatment (n = 8). VNS was delivered for 30 min per day for 6 weeks after the establishment of NAFLD using a digital TENS device. The subsequent assessments included hepatic triglyceride, cholesterol content, serum lipid profile, and liver function testing. In this context, inflammatory biomarkers (TNF-α, IL-6) and hepatic oxidative stress (MDA, SOD, and GPx) were also assessed. To clarify the possible mechanisms behind the protective benefits of VNS, additional histological inspection and immunohistochemistry analysis of TNF-α and Caspase-3 were performed. RESULTS In the NAFLD-affected obese rats, VNS markedly decreased the rats' body mass index (BMI) and abdominal circumference (AC). Liver function markers (albumin, ALT, and AST) and the serum lipid profile-which included a notable decrease in the amounts of hepatic triglycerides and cholesterol-were both markedly improved. Additionally, oxidative stress and inflammatory indicators showed a considerable decline with VNS. Notably, the liver tissues examined by histopathologists revealed that there is evidence of the protective impact of VNS on the oxidative and inflammatory states linked to HFD-induced NAFLD while maintaining the architectural and functional condition of the liver. CONCLUSIONS Our findings suggest that VNS may represent a promising therapeutic candidate for managing NAFLD induced by obesity. It can be considered to be an effective adjuvant physiological intervention for the obese population with NAFLD to spare the liver against obesity-induced deleterious injury.
Collapse
Affiliation(s)
- Hany A. Elkattawy
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11579, Saudi Arabia;
- Medical Physiology Department, College of Medicine, Zagazig University, Zagazig P.O. Box 44519, Egypt; (A.E.-S.H.); (S.F.E.-S.)
| | - Samar Mortada Mahmoud
- Department of Human Anatomy and Embryology, College of Medicine, Zagazig University, Zagazig P.O. Box 44519, Egypt;
| | - Ahmed El-Sayed Hassan
- Medical Physiology Department, College of Medicine, Zagazig University, Zagazig P.O. Box 44519, Egypt; (A.E.-S.H.); (S.F.E.-S.)
- Department of Basic Medical Sciences, College of Medicine, Sulaiman Al-Rajhi University, Bukayriah 51941, Saudi Arabia
| | - Ahmed Behiry
- Department of Tropical Medicine and Endemic Diseases, College of Medicine, Zagazig University, Zagazig P.O. Box 44519, Egypt;
| | - Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Ateya Megahed Ibrahim
- Department of Nursing, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.M.I.); (D.E.F.Z.)
- Department of Family and Community Health Nursing, Faculty of Nursing, Port Said University, Port Said P.O. Box 42511, Egypt
| | - Donia Elsaid Fathi Zaghamir
- Department of Nursing, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.M.I.); (D.E.F.Z.)
- Department of Pediatric Nursing, Faculty of Nursing, Port Said University, Port Said P.O. Box 42511, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11579, Saudi Arabia;
| | - Sherein F. El-Sayed
- Medical Physiology Department, College of Medicine, Zagazig University, Zagazig P.O. Box 44519, Egypt; (A.E.-S.H.); (S.F.E.-S.)
| |
Collapse
|
8
|
Gruenewald T, Seeman TE, Choo TH, Scodes J, Snyder C, Pavlicova M, Weinstein M, Schwartz JE, Mukkamala R, Sloan RP. Cardiovascular variability, sociodemographics, and biomarkers of disease: the MIDUS study. Front Physiol 2023; 14:1234427. [PMID: 37693005 PMCID: PMC10484414 DOI: 10.3389/fphys.2023.1234427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction: Like heart rate, blood pressure (BP) is not steady but varies over intervals as long as months to as short as consecutive cardiac cycles. This blood pressure variability (BPV) consists of regularly occurring oscillations as well as less well-organized changes and typically is computed as the standard deviation of multiple clinic visit-to-visit (VVV-BP) measures or from 24-h ambulatory BP recordings (ABPV). BP also varies on a beat-to-beat basis, quantified by methods that parse variation into discrete bins, e.g., low frequency (0.04-0.15 Hz, LF). However, beat-to-beat BPV requires continuous recordings that are not easily acquired. As a result, we know little about the relationship between LF-BPV and basic sociodemographic characteristics such as age, sex, and race and clinical conditions. Methods: We computed LF-BPV during an 11-min resting period in 2,118 participants in the Midlife in the US (MIDUS) study. Results: LF-BPV was negatively associated with age, greater in men than women, and unrelated to race or socioeconomic status. It was greater in participants with hypertension but unrelated to hyperlipidemia, hypertriglyceridemia, diabetes, elevated CRP, or obesity. LF-diastolic BPV (DBPV), but not-systolic BPV (SBPV), was negatively correlated with IL-6 and s-ICAM and positively correlated with urinary epinephrine and cortisol. Finally, LF-DBPV was negatively associated with mortality, an effect was rendered nonsignificant by adjustment by age but not other sociodemographic characteristics. Discussion: These findings, the first from a large, national sample, suggest that LF-BPV differs significantly from VVV-BP and ABPV. Confirming its relationship to sociodemographic risk factors and clinical outcomes requires further study with large and representative samples.
Collapse
Affiliation(s)
- Tara Gruenewald
- Department of Psychology, Chapman University, Orange, CA, United States
| | - Teresa E. Seeman
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Tse-Hwei Choo
- Mental Health Data Science Division, New York State Psychiatric Institute, New York, NY, United States
| | - Jennifer Scodes
- Mental Health Data Science Division, New York State Psychiatric Institute, New York, NY, United States
| | - Clayton Snyder
- Mental Health Data Science Division, New York State Psychiatric Institute, New York, NY, United States
| | - Martina Pavlicova
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, United States
| | | | - Joseph E. Schwartz
- Renaissance School of Medicine, Stony Brook University, New York, NY, United States
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Ramakrishna Mukkamala
- Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard P. Sloan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
- New York State Psychiatric Institute, New York, NY, United States
| |
Collapse
|
9
|
Prathumsap N, Ongnok B, Khuanjing T, Arinno A, Maneechote C, Apaijai N, Chunchai T, Arunsak B, Kerdphoo S, Janjek S, Chattipakorn SC, Chattipakorn N. Vagus nerve stimulation exerts cardioprotection against doxorubicin-induced cardiotoxicity through inhibition of programmed cell death pathways. Cell Mol Life Sci 2022; 80:21. [PMID: 36583785 PMCID: PMC11072695 DOI: 10.1007/s00018-022-04678-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022]
Abstract
The aberration of programmed cell death including cell death associated with autophagy/mitophagy, apoptosis, necroptosis, pyroptosis, and ferroptosis can be observed in the development and progression of doxorubicin-induced cardiotoxicity (DIC). Vagus nerve stimulation (VNS) has been shown to exert cardioprotection against cardiomyocyte death through the release of the neurotransmitter acetylcholine (ACh) under a variety of pathological conditions. However, the roles of VNS and its underlying mechanisms against DIC have never been investigated. Forty adults male Wistar rats were divided into 5 experimental groups: (i) control without VNS (CSham) group, (ii) doxorubicin (3 mg/kg/day, i.p.) without VNS (DSham) group, (iii) doxorubicin + VNS (DVNS) group, (iv) doxorubicin + VNS + mAChR antagonist (atropine; 1 mg/kg/day, ip, DVNS + Atro) group, and (v) doxorubicin + VNS + nAChR antagonist (mecamylamine; 7.5 mg/kg/day, ip, DVNS + Mec) group. Our results showed that doxorubicin insult led to left ventricular (LV) dysfunction through impaired cardiac autonomic balance, decreased mitochondrial function, imbalanced mitochondrial dynamics, and exacerbated cardiomyocyte death including autophagy/mitophagy, apoptosis, necroptosis, pyroptosis, and ferroptosis. However, VNS treatment improved cardiac mitochondrial and autonomic functions, and suppressed excessive autophagy, apoptosis, necroptosis, pyroptosis, and ferroptosis, leading to improved LV function. Consistent with this, ACh effectively improved cell viability and suppressed cell cytotoxicity in doxorubicin-treated H9c2 cells. In contrast, either inhibitors of muscarinic (mAChR) or nicotinic acetylcholine receptor (nAChR) completely abrogated the favorable effects mediated by VNS and acetylcholine. These findings suggest that VNS exerts cardioprotective effects against doxorubicin-induced cardiomyocyte death via activation of both mAChR and nAChR.
Collapse
Affiliation(s)
- Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sornram Janjek
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
10
|
Alhawwash A, Muzquiz MI, Richardson L, Vetter C, Smolik M, Goodwill A, Yoshida K. In vivo peripheral nerve activation using sinusoidal low-frequency alternating currents. Artif Organs 2022; 46:2055-2065. [PMID: 35730955 PMCID: PMC9795871 DOI: 10.1111/aor.14347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/19/2022] [Accepted: 06/09/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The sinusoidal low-frequency alternating current (LFAC) waveform was explored recently as a novel means to evoke nerve conduction block. In the present work, we explored whether increasing the amplitude of the LFAC waveform results in nerve fiber activation in autonomic nerves. In-silico methods and preliminary work in somatic nerves indicated a potential frequency dependency on the threshold of activation. The Hering-Breuer (HB) reflex was used as a biomarker to detect cervical vagus nerve activation. METHODS Experiments were conducted in isoflurane-anesthetized swine (n = 5). Two stimulating bipolar cuff electrodes and a tripolar recording cuff electrode were implanted on the left vagus nerve. To ensure the electrical stimulation affects only the afferent pathways, the nerve was crushed caudal to the electrodes to eliminate cardiac effects. (1) Standard pulse stimulation (Vstim) using a monophasic train of pulses was applied through the caudal electrode to elicit HB reflex and to identify the activated nerve fiber type. (2) Continuous sinusoidal LFAC waveform with a frequency ranging from 5 through 20 Hz was applied to the rostral electrode without Vstim to explore the activation thresholds at each LFAC frequency. In both cases, the activation of nerve fibers was detected by a HB reflex-induced reduction in the breathing rate. RESULTS LFAC was found to be capable of eliciting an HB response. The LFAC activation thresholds were found to be frequency-dependent. The HB threshold was 1.02 ± 0.3 mAp at 5 Hz, 0.66 ± 0.3 mAp at 10 Hz, and 0.44 ± 0.2 mAp at 20 Hz. In comparison, it was 0.7 ± 0.47 mA for a 100 μs pulse. The LFAC amplitude was within the linear limits of the electrode interface. Damage to the cuff electrodes or the nerve tissues was not observed. Analysis of Vstim-based compound nerve action potentials (CNAP) indicated that the decrease in breathing rate was found to be correlated with the activation of slower components of the CNAP suggesting that LFAC reached and elicited responses from these slower fibers associated with afferents projecting to the HB response. CONCLUSIONS These results suggest the feasibility of the LFAC waveform at 5, 10, and 20 Hz to activate autonomic nerve fibers and potentially provide a new modality to the neurorehabilitation field.
Collapse
Affiliation(s)
- Awadh Alhawwash
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA,Biomedical Technology DepartmentKing Saud UniversityRiyadhSaudi Arabia
| | - M. Ivette Muzquiz
- Department of Biomedical EngineeringIndiana University ‐ Purdue University IndianapolisIndianapolisIndianaUSA
| | - Lindsay Richardson
- Department of Biomedical EngineeringIndiana University ‐ Purdue University IndianapolisIndianapolisIndianaUSA
| | - Christian Vetter
- Department of Biomedical EngineeringIndiana University ‐ Purdue University IndianapolisIndianapolisIndianaUSA
| | - Macallister Smolik
- Department of BiologyIndiana University ‐ Purdue University IndianapolisIndianapolisIndianaUSA
| | - Adam Goodwill
- Department of Integrative Medical SciencesNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Ken Yoshida
- Department of Biomedical EngineeringIndiana University ‐ Purdue University IndianapolisIndianapolisIndianaUSA
| |
Collapse
|
11
|
Kim S, Park I, Lee JH, Kim S, Jang DH, Jo YH. Vagus Nerve Stimulation Improves Mitochondrial Dysfunction in Post–cardiac Arrest Syndrome in the Asphyxial Cardiac Arrest Model in Rats. Front Neurosci 2022; 16:762007. [PMID: 35692415 PMCID: PMC9178208 DOI: 10.3389/fnins.2022.762007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral mitochondrial dysfunction during post–cardiac arrest syndrome (PCAS) remains unclear, resulting in a lack of therapeutic options that protect against cerebral ischemia–reperfusion injury. We aimed to assess mitochondrial dysfunction in the hippocampus after cardiac arrest and whether vagus nerve stimulation (VNS) can improve mitochondrial dysfunction and neurological outcomes. In an asphyxial cardiac arrest model, male Sprague–Dawley rats were assigned to the vagus nerve isolation (CA) or VNS (CA + VNS) group. Cardiopulmonary resuscitation was performed 450 s after pulseless electrical activity. After the return of spontaneous circulation (ROSC), left cervical VNS was performed for 3 h in the CA + VNS group. Mitochondrial respiratory function was evaluated using high-resolution respirometry of the hippocampal tissue. The neurologic deficit score (NDS) and overall performance category (OPC) were assessed at 24, 48, and 72 h after resuscitation. The leak respiration and oxidative phosphorylation capacity of complex I (OXPHOS CI) at 6 h after ROSC were significantly higher in the CA + VNS group than in the CA group (p = 0.0308 and 0.0401, respectively). Compared with the trends of NDS and OPC in the CA group, the trends of those in the CA + VNS group were significantly different, thus suggesting a favorable neurological outcome in the CA + VNS group (p = 0.0087 and 0.0064 between times × groups interaction, respectively). VNS ameliorated mitochondrial dysfunction after ROSC and improved neurological outcomes in an asphyxial cardiac arrest rat model.
Collapse
Affiliation(s)
- Seonghye Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
| | - Inwon Park
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, South Korea
- *Correspondence: Jae Hyuk Lee,
| | - Serin Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
| | - Dong-Hyun Jang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam-si, South Korea
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
12
|
Jezdimirovic T, Stajer V, OstojicV SM. Cardiovascular autonomic reflex tests and serum FGF21 levels in overweight and normal-weight men and women. Arch Physiol Biochem 2022; 128:373-377. [PMID: 31686543 DOI: 10.1080/13813455.2019.1683586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We evaluated cardiovascular autonomic reflexes and serum fibroblast growth factor 21 (FGF21), a surrogate marker of mitochondrial function, in a cohort of overweight and normal-weight adults (n = 42). METHODS Indices of autonomic function were monitored during supine rest, autonomic reflex tests and submaximal clinical exercise test, with heart rate variables and blood pressure measured with an automatic system. RESULTS Markers of sympathetic dominance were accentuated in overweight adults, including elevated resting low-frequency to the high-frequency ratio for heart rate variability (203 ± 227 vs. 96 ± 42; p = .01), and handgrip diastolic blood pressure (36 ± 15 mmHg vs. 25 ± 12 mmHg; p = .01). A weak non-significant trend has been found for a negative correlation between blood pressure responses to isometric handgrip test and FGF21 in the overweight group (r = -0.37; p = .09). CONCLUSIONS Excess body weight appears to trigger sympathetic overactivity in overweight adults, yet autonomic dysregulation might not be associated with notable changes in serum FGF21.
Collapse
Affiliation(s)
- Tatjana Jezdimirovic
- Applied Bioenergetics Lab, Faculty of Sport and Physical Education, University of Novi Sad, Novi Sad, Serbia
| | - Valdemar Stajer
- Applied Bioenergetics Lab, Faculty of Sport and Physical Education, University of Novi Sad, Novi Sad, Serbia
| | - Sergej M OstojicV
- Applied Bioenergetics Lab, Faculty of Sport and Physical Education, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
13
|
Sloan RP, Cole SW. Parasympathetic neural activity and the reciprocal regulation of innate antiviral and inflammatory genes in the human immune system. Brain Behav Immun 2021; 98:251-256. [PMID: 34400237 PMCID: PMC8511100 DOI: 10.1016/j.bbi.2021.08.217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/18/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
The vagus nerve mediates parasympathetic nervous system control of peripheral physiological processes including cardiovascular activity and immune response. In mice, tonic vagal activation down-regulates inflammation via nicotinic acetylcholine receptor-mediated inhibition of the pro-inflammatory transcription factor NF-κB in monocyte/macrophages. Because Type I interferon and pro-inflammatory genes are regulated reciprocally at the level of transcription factor activation and cell differentiation, we hypothesized that vagal activity would up-regulate Type I interferon response genes concurrently with inflammatory downregulation in human immune cells. We mapped empirical individual differences in the circulating leukocyte transcriptome and vagal activity indexed by high frequency (0.15-0.40 Hz) heart rate variability (HF-HRV) in 380 participants in the Midlife in the US study. Here we show that promoter-based bioinformatics analyses linked greater HF-HRV to reduced NF-κB activity and increased activity of IRF transcription factors involved in Type I interferon response (independent of β-antagonists, BMI, smoking, heavy alcohol consumption, and demographic factors). Transcript origin analyses implicated myeloid lineage immune cells as targets, representing per-cell alterations in gene transcription as HF-HRV was not associated with differential prevalence of leukocyte subsets. These findings support the concept of parasympathetic inhibition of pro-inflammatory gene expression in humans and up-regulation of Type I interferons that could augment host defense against viral infections.
Collapse
Affiliation(s)
- Richard P Sloan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, 622 West 168(th)St., PH1540, New York, NY 10032, USA; New York State Psychiatric Institute, 1051 Riverside Dr., New York, NY, USA.
| | - Steve W Cole
- Department of Psychiatry & Biobehavioral Sciences, Department of Medicine, Division of Hematology-Oncology, Norman Cousins Center, Jonsson Comprehensive Cancer Center, University of California Los Angeles, 11-934 Factor Building, Los Angeles, CA 90095-1678, USA.
| |
Collapse
|
14
|
Wolf V, Kühnel A, Teckentrup V, Koenig J, Kroemer NB. Does transcutaneous auricular vagus nerve stimulation affect vagally mediated heart rate variability? A living and interactive Bayesian meta-analysis. Psychophysiology 2021; 58:e13933. [PMID: 34473846 DOI: 10.1111/psyp.13933] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022]
Abstract
Non-invasive brain stimulation techniques, such as transcutaneous auricular vagus nerve stimulation (taVNS), have considerable potential for clinical use. Beneficial effects of taVNS have been demonstrated on symptoms in patients with mental or neurological disorders as well as transdiagnostic dimensions, including mood and motivation. However, since taVNS research is still an emerging field, the underlying neurophysiological processes are not yet fully understood, and the replicability of findings on biomarkers of taVNS effects has been questioned. The objective of this analysis was to synthesize the current evidence concerning the effects of taVNS on vagally mediated heart rate variability (vmHRV), a candidate biomarker that has, so far, received most attention in the field. We performed a living Bayesian random effects meta-analysis. To keep the synthesis of evidence transparent and up to date as new studies are being published, we developed a Shiny web app that regularly incorporates new results and enables users to modify study selection criteria to evaluate the robustness of the inference across potential confounds. Our analysis focuses on 16 single-blind studies comparing taVNS versus sham in healthy participants. The meta-analysis provides strong evidence for the null hypothesis (g = 0.014, CIshortest = [-0.103, 0.132], BF01 = 24.678), indicating that acute taVNS does not alter vmHRV compared to sham. To conclude, there is no support for the hypothesis that vmHRV is a robust biomarker for acute taVNS. By increasing transparency and timeliness, the concept of living meta-analyses can lead to transformational benefits in emerging fields such as non-invasive brain stimulation.
Collapse
Affiliation(s)
- Vinzent Wolf
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany.,Department of Psychology, University of Salzburg, Salzburg, Austria
| | - Anne Kühnel
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany.,Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry and International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Vanessa Teckentrup
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Julian Koenig
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils B Kroemer
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
Pavlov VA. The evolving obesity challenge: targeting the vagus nerve and the inflammatory reflex in the response. Pharmacol Ther 2021; 222:107794. [PMID: 33310156 PMCID: PMC8027699 DOI: 10.1016/j.pharmthera.2020.107794] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Obesity and the metabolic syndrome (MetS), which have reached pandemic proportions significantly increase the risk for type 2 diabetes, cardiovascular disease, and other serious conditions. Recent data with COVID-19 patients indicate that obesity also is a significant risk factor for this novel viral disease and poor outcome of associated critical illness. These findings considerably change the view of obesity as a driver of serious, but slowly-progressing chronic diseases, and emphasize the urgency to explore new therapeutic approaches. Inflammation is a recognized driver of metabolic derangements in obesity and MetS, and a core feature of COVID-19 pathobiology. Recent advances in our understanding of inflammatory regulation have highlighted the role of the nervous system and the vagus nerve-based inflammatory reflex. Current bioelectronic and pharmacological therapeutic explorations centered on the inflammatory reflex offer new approaches for conditions characterized by immune and metabolic dysregulation and for ameliorating the escalating burden of obesity, MetS, and COVID-19.
Collapse
Affiliation(s)
- Valentin A Pavlov
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA.
| |
Collapse
|
16
|
Luo B, Wu Y, Liu SL, Li XY, Zhu HR, Zhang L, Zheng F, Liu XY, Guo LY, Wang L, Song HX, Lv YX, Cheng ZS, Chen SY, Wang JN, Tang JM. Vagus nerve stimulation optimized cardiomyocyte phenotype, sarcomere organization and energy metabolism in infarcted heart through FoxO3A-VEGF signaling. Cell Death Dis 2020; 11:971. [PMID: 33184264 PMCID: PMC7665220 DOI: 10.1038/s41419-020-03142-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/30/2022]
Abstract
Vagus nerve stimulation (VNS) restores autonomic balance, suppresses inflammation action and minimizes cardiomyocyte injury. However, little knowledge is known about the VNS’ role in cardiomyocyte phenotype, sarcomere organization, and energy metabolism of infarcted hearts. VNS in vivo and acetylcholine (ACh) in vitro optimized the levels of α/β-MHC and α-Actinin positive sarcomere organization in cardiomyocytes while reducing F-actin assembly of cardiomyocytes. Consistently, ACh improved glucose uptake while decreasing lipid deposition in myocytes, correlating both with the increase of Glut4 and CPT1α and the decrease of PDK4 in infarcted hearts in vivo and myocytes in vitro, attributing to improvement in both glycolysis by VEGF-A and lipid uptake by VEGF-B in response to Ach. This led to increased ATP levels accompanied by the repaired mitochondrial function and the decreased oxygen consumption. Functionally, VNS improved the left ventricular performance. In contrast, ACh-m/nAChR inhibitor or knockdown of VEGF-A/B by shRNA powerfully abrogated these effects mediated by VNS. On mechanism, ACh decreased the levels of nuclear translocation of FoxO3A in myocytes due to phosphorylation of FoxO3A by activating AKT. FoxO3A overexpression or knockdown could reverse the specific effects of ACh on the expression of VEGF-A/B, α/β-MHC, Glut4, and CPT1α, sarcomere organization, glucose uptake and ATP production. Taken together, VNS optimized cardiomyocytes sarcomere organization and energy metabolism to improve heart function of the infarcted heart during the process of delaying and/or blocking the switch from compensated hypertrophy to decompensated heart failure, which were associated with activation of both P13K/AKT-FoxO3A-VEGF-A/B signaling cascade.
Collapse
Affiliation(s)
- Bin Luo
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Yan Wu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Shu-Lin Liu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Xing-Yuan Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Hong-Rui Zhu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China
| | - Lei Zhang
- Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Xiao-Yao Liu
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China
| | - Ling-Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Hong-Xian Song
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Yan-Xia Lv
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China.,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China
| | - Zhong-Shan Cheng
- Applied Bioinformatics Center, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shi-You Chen
- The Department of Surgery, University of Missouri, Columbia, MO, USA
| | - Jia-Ning Wang
- Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Jun-Ming Tang
- Department of Physiology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, 442000, Hubei, China. .,Institute of Biomedicine, Hubei University of Medicine, 442000, Hubei, China. .,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, Hubei, China.
| |
Collapse
|
17
|
Subramanian M, Edwards L, Melton A, Branen L, Herron A, Sivasubramanian MK, Monteiro R, Stansbury S, Balasubramanian P, Morris L, Elkholey K, Niewiadomska M, Stavrakis S. Non-invasive vagus nerve stimulation attenuates proinflammatory cytokines and augments antioxidant levels in the brainstem and forebrain regions of Dahl salt sensitive rats. Sci Rep 2020; 10:17576. [PMID: 33067477 PMCID: PMC7567801 DOI: 10.1038/s41598-020-74257-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022] Open
Abstract
The anti-inflammatory effects of vagus nerve stimulation are well known. It has recently been shown that low-level, transcutaneous stimulation of vagus nerve at the tragus (LLTS) reduces cardiac inflammation in a rat model of heart failure with preserved ejection fraction (HFpEF). The mechanisms by which LLTS affect the central neural circuits within the brain regions that are important for the regulation of cardiac vagal tone are not clear. Female Dahl salt-sensitive rats were initially fed with either low salt (LS) or high salt (HS) diet for a period of 6 weeks, followed by sham or active stimulation (LLTS) for 30 min daily for 4 weeks. To study the central effects of LLTS, four brainstem (SP5, NAb, NTS, and RVLM) and two forebrain sites (PVN and SFO) were examined. HS diet significantly increased the gene expression of proinflammatory cytokines in the SP5 and SFO. LLTS reversed HS diet-induced changes at both these sites. Furthermore, LLTS augmented the levels of antioxidant Nrf2 in the SP5 and SFO. Taken together, these findings suggest that LLTS has central anti-inflammatory and antioxidant properties that could mediate the neuromodulation of cardiac vagal tone in the rat model of HFpEF.
Collapse
Affiliation(s)
- Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA.
| | - Laura Edwards
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Avery Melton
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Lyndee Branen
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Angela Herron
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Mahesh Kumar Sivasubramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Raisa Monteiro
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Samantha Stansbury
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, 277 McElroy Hall, Stillwater, OK, 74078, USA
| | - Priya Balasubramanian
- Reynolds Oklahoma Center On Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lynsie Morris
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 Stanton L Young Blvd, Suite 5400, Oklahoma City, OK, 73104, USA
| | - Khaled Elkholey
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Monika Niewiadomska
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Stavrakis
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 Stanton L Young Blvd, Suite 5400, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
18
|
Pongkan W, Jitnapakarn W, Phetnoi W, Punyapornwithaya V, Boonyapakorn C. Obesity-Induced Heart Rate Variability Impairment and Decreased Systolic Function in Obese Male Dogs. Animals (Basel) 2020; 10:ani10081383. [PMID: 32785083 PMCID: PMC7460129 DOI: 10.3390/ani10081383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Obesity in dogs can induce many adverse health effects including musculoskeletal problems, respiratory distress, metabolic syndrome, and cardiovascular diseases. In humans with obesity, heart rate variability (HRV) is used to identify and predict the risk of cardiovascular diseases. However, this predictive tool has never been used in veterinary medicine, and the relationship between obesity and HRV has rarely been investigated. In this study, we investigated HRV, plasma oxidative stress (MDA), and cardiac function in obese male dogs. We hypothesized that obese male dogs have decreased cardiac function and impaired HRV compared to non-obese dogs. Our study found that obese dogs have decreased cardiac systolic function and impaired HRV, as indicated by reduced percentages of cardiac contraction and impaired cardiac autonomic activity compared to non-obese dogs. We concluded that obesity can decrease systolic function and cause HRV impairment, which might increase the risk of cardiovascular disease in dogs. In addition, HRV might be used as a predictive or prognostic tool in the prevention of cardiovascular disease in obese dogs. Abstract Obesity can induce cardiovascular diseases in both humans and animals. Heart rate variability (HRV) is an indicator of sympathovagal balance and is used to identify cardiovascular diseases in humans. However, HRV and cardiac function have rarely been investigated in obese dogs. This study investigated the effect of obesity on oxidative stress, HRV, and cardiac function in obese and non-obese dogs. The nine-scale body condition score (BCS) system was used to determine obesity. Thirty small breed dogs were divided into a normal weight group (n = 15) and an obese group (n = 15). All dogs underwent physical examination, plasma malondialdehyde (MDA) measurement, electrocardiography, echocardiography, and two hours of Holter monitoring. This study found that obese dogs had increased plasma MDA and sympathovagal imbalance, which was indicated by impaired time and frequency domains compared to normal weight dogs. Although cardiac function was within normal limits, the echocardiographic study found that the obese dogs had reduced cardiac wall thickness and lower systolic function, as indicated by a reduction in %ejection fraction, %fractional shortening, increased left ventricular (LV) internal diameter during systole, and LV end-systolic volume compared to normal weight dogs. This study concluded that obesity in dogs can induce increased plasma oxidative stress, impaired HRV, and reduced cardiac systolic function compared to non-obese dogs.
Collapse
Affiliation(s)
- Wanpitak Pongkan
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (W.P.); (W.J.); (W.P.)
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Wannida Jitnapakarn
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (W.P.); (W.J.); (W.P.)
| | - Warunee Phetnoi
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; (W.P.); (W.J.); (W.P.)
| | - Veerasak Punyapornwithaya
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
| | - Chavalit Boonyapakorn
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Correspondence: ; Tel.: +66-53-948-015
| |
Collapse
|
19
|
Burger AM, D'Agostini M, Verkuil B, Van Diest I. Moving beyond belief: A narrative review of potential biomarkers for transcutaneous vagus nerve stimulation. Psychophysiology 2020; 57:e13571. [PMID: 32202671 DOI: 10.1111/psyp.13571] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/15/2020] [Accepted: 02/01/2020] [Indexed: 12/25/2022]
Abstract
Transcutaneous vagus nerve stimulation (tVNS) is a non-invasive neurostimulation technique that is currently being tested as a potential treatment for a myriad of neurological and psychiatric disorders. However, the working mechanisms underlying tVNS are poorly understood and it remains unclear whether stimulation activates the vagus nerve for every participant. Finding a biological marker of tVNS is imperative, as it can help guide research on clinical applications and can inform researchers on optimal stimulation sites and parameters to further optimize treatment efficacy. In this narrative review, we discuss five potential biomarkers for tVNS and review currently available evidence for these markers for both invasive and tVNS. While some of these biomarkers hold promise from a theoretical perspective, none of the potential biomarkers provide clear and definitive indications that tVNS increases the vagal activity or augments activity in the locus coeruleus-noradrenaline network. We conclude the review by providing several recommendations for how to tackle the challenges and opportunities when researching potential biomarkers for the effects of tVNS.
Collapse
Affiliation(s)
- Andreas Michael Burger
- Health Psychology Research Group, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium.,Biological Psychology Research Group, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| | - Martina D'Agostini
- Health Psychology Research Group, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| | - Bart Verkuil
- Department of Clinical Psychology, Leiden University, Leiden, the Netherlands
| | - Ilse Van Diest
- Health Psychology Research Group, Faculty of Psychology and Educational Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Khuanjing T, Palee S, Chattipakorn SC, Chattipakorn N. The effects of acetylcholinesterase inhibitors on the heart in acute myocardial infarction and heart failure: From cells to patient reports. Acta Physiol (Oxf) 2020; 228:e13396. [PMID: 31595611 DOI: 10.1111/apha.13396] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/30/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases remain a major cause of morbidity and mortality worldwide. Cardiovascular diseases such as acute myocardial infarction, ischaemia/reperfusion injury and heart failure are associated with cardiac autonomic imbalance characterized by sympathetic overactivity and parasympathetic withdrawal from the heart. Increased parasympathetic activity by electrical vagal nerve stimulation has been shown to provide beneficial effects in the case of cardiovascular diseases in both animals and patients by improving autonomic function, cardiac remodelling and mitochondrial function. However, clinical limitations for electrical vagal nerve stimulation exist because of its invasive nature, costly equipment and limited clinical validation. Therefore, novel therapeutic approaches which moderate parasympathetic activities could be beneficial for in the case of cardiovascular disease. Acetylcholinesterase inhibitors inhibit acetylcholinesterase and hence increase cholinergic transmission. Recent studies have reported that acetylcholinesterase inhibitors improve autonomic function and cardiac function in cardiovascular disease models. Despite its potential clinical benefits for cardiovascular disease patients, the role of acetylcholinesterase inhibitors in acute myocardial infarction and heart failure remediation remains unclear. This article comprehensively reviews the effects of acetylcholinesterase inhibitors on the heart in acute myocardial infarction and heart failure scenarios from in vitro and in vivo studies to clinical reports. The mechanisms involved are also discussed in this review.
Collapse
Affiliation(s)
- Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Cardiac Electrophysiology Unit Department of Physiology Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Center of Excellence in Cardiac Electrophysiology Research Chiang Mai University Chiang Mai Thailand
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Center of Excellence in Cardiac Electrophysiology Research Chiang Mai University Chiang Mai Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Center of Excellence in Cardiac Electrophysiology Research Chiang Mai University Chiang Mai Thailand
- Department of Oral Biology and Diagnostic Sciences Faculty of Dentistry Chiang Mai University Chiang Mai Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Cardiac Electrophysiology Unit Department of Physiology Faculty of Medicine Chiang Mai University Chiang Mai Thailand
- Center of Excellence in Cardiac Electrophysiology Research Chiang Mai University Chiang Mai Thailand
| |
Collapse
|
21
|
Xue RQ, Zhao M, Wu Q, Yang S, Cui YL, Yu XJ, Liu J, Zang WJ. Regulation of mitochondrial cristae remodelling by acetylcholine alleviates palmitate-induced cardiomyocyte hypertrophy. Free Radic Biol Med 2019; 145:103-117. [PMID: 31553938 DOI: 10.1016/j.freeradbiomed.2019.09.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/21/2019] [Indexed: 12/31/2022]
Abstract
Mitochondrial dysfunction is associated with obesity-induced cardiac remodelling. Recent research suggests that the cristae are the true bioenergetic components of cells. Acetylcholine (ACh), the major neurotransmitter of the vagus nerve, exerts cardio-protective effects against ischaemia. This study investigated the role of cristae remodelling in palmitate (PA)-induced neonatal rat cardiomyocyte hypertrophy and explored the beneficial effects of ACh. We found loose, fragmented and even lysed cristae in PA-treated neonatal cardiomyocytes along with declines in mitochondrial network and complex expression and overproduction of mitochondrial reactive oxygen species (ROS); these changes ultimately resulted in increased myocardial size. Overexpression of mitofilin by adenoviral infection partly improved cristae shape, mitochondrial network, and ATP content and attenuated cell hypertrophy. Interestingly, siRNA-mediated AMP-activated protein kinase (AMPK) silencing increased the number of cristae with a balloon-like morphology without disturbing mitofilin expression. Furthermore, AMPK knockdown abolished the effects of mitofilin overexpression on cristae remodelling and inhibited the interaction of mitofilin with sorting and assembly machinery 50 (Sam50) and coiled-coil helix coiled-coil helix domain-containing protein 3 (CHCHD3), two core components of the mitochondrial contact site and cristae organizing system (MICOS) complex. Intriguingly, ACh upregulated mitofilin expression and AMPK phosphorylation via the muscarinic ACh receptor (MAChR). Moreover, ACh enhanced protein-protein interactions between mitofilin and other components of the MICOS complex, thereby preventing PA-induced mitochondrial dysfunction and cardiomyocyte hypertrophy; however, these effects were abolished by AMPK silencing. Taken together, our data suggest that ACh improves cristae remodelling to defend against PA-induced myocardial hypertrophy, presumably by increasing mitofilin expression and activating AMPK to form the MICOS complex through MAChR. These results suggest new and promising therapeutic approaches targeting mitochondria to prevent lipotoxic cardiomyopathy.
Collapse
Affiliation(s)
- Run-Qing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Qing Wu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Si Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Yan-Ling Cui
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Jiankang Liu
- Frontier Institute of Science and Technol, and Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China.
| |
Collapse
|
22
|
Vagus nerve stimulation as a promising adjunctive treatment for ischemic stroke. Neurochem Int 2019; 131:104539. [DOI: 10.1016/j.neuint.2019.104539] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/03/2019] [Accepted: 08/21/2019] [Indexed: 12/26/2022]
|
23
|
Boswijk E, Franssen R, Vijgen GHEJ, Wierts R, van der Pol JAJ, Mingels AMA, Cornips EMJ, Majoie MHJM, van Marken Lichtenbelt WD, Mottaghy FM, Wildberger JE, Bucerius J. Short-term discontinuation of vagal nerve stimulation alters 18F-FDG blood pool activity: an exploratory interventional study in epilepsy patients. EJNMMI Res 2019; 9:101. [PMID: 31773320 PMCID: PMC6879675 DOI: 10.1186/s13550-019-0567-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/16/2019] [Indexed: 11/15/2022] Open
Abstract
Background Vagus nerve activation impacts inflammation. Therefore, we hypothesized that vagal nerve stimulation (VNS) influenced arterial wall inflammation as measured by 18F-FDG uptake. Results Ten patients with left-sided VNS for refractory epilepsy were studied during stimulation (VNS-on) and in the hours after stimulation was switched off (VNS-off). In nine patients, 18F-FDG uptake was measured in the right carotid artery, aorta, bone marrow, spleen, and adipose tissue. Target-to-background ratios (TBRs) were calculated to normalize the respective standardized uptake values (SUVs) for venous blood pool activity. Median values are shown with interquartile range and compared using the Wilcoxon signed-rank test. Arterial SUVs tended to be higher during VNS-off than VNS-on [SUVmax all vessels 1.8 (1.5–2.2) vs. 1.7 (1.2–2.0), p = 0.051]. However, a larger difference was found for the venous blood pool at this time point, reaching statistical significance in the vena cava superior [meanSUVmean 1.3 (1.1–1.4) vs. 1.0 (0.8–1.1); p = 0.011], resulting in non-significant lower arterial TBRs during VNS-off than VNS-on. Differences in the remaining tissues were not significant. Insulin levels increased after VNS was switched off [55.0 pmol/L (45.9–96.8) vs. 48.1 pmol/L (36.9–61.8); p = 0.047]. The concurrent increase in glucose levels was not statistically significant [4.8 mmol/L (4.7–5.3) vs. 4.6 mmol/L (4.5–5.2); p = 0.075]. Conclusions Short-term discontinuation of VNS did not show a consistent change in arterial wall 18F-FDG-uptake. However, VNS did alter insulin and 18F-FDG blood levels, possibly as a result of sympathetic activation.
Collapse
Affiliation(s)
- Ellen Boswijk
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Renee Franssen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Guy H E J Vijgen
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.,Department of Surgery, Erasmus Medical Center (EMC), Postbus 2040, 3000 CA, Rotterdam, The Netherlands
| | - Roel Wierts
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Jochem A J van der Pol
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Alma M A Mingels
- Department of Clinical Chemistry, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Erwin M J Cornips
- Department of Neurosurgery, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Marian H J M Majoie
- Department of Research & Development, Epilepsy Center Kempenhaeghe, Sterkselseweg 65, 5591 VE, Heeze, The Netherlands.,Department of Neurology, Academic Center for Epileptology, Epilepsy Center Kempenhaeghe & Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.,MHENS School of Mental Health & Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.,School of Health Professions Education, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 60, 6229 ER, Maastricht, The Netherlands
| | - Wouter D van Marken Lichtenbelt
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Joachim E Wildberger
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Jan Bucerius
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands. .,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands. .,Department of Nuclear Medicine, Georg-August University Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
| |
Collapse
|
24
|
Xue RQ, Yu XJ, Zhao M, Xu M, Wu Q, Cui YL, Yang S, Li DL, Zang WJ. Pyridostigmine alleviates cardiac dysfunction via improving mitochondrial cristae shape in a mouse model of metabolic syndrome. Free Radic Biol Med 2019; 134:119-132. [PMID: 30633969 DOI: 10.1016/j.freeradbiomed.2019.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 01/02/2023]
Abstract
Insulin resistance and autonomic imbalance are important pathological processes in metabolic syndrome-induced cardiac remodeling. Recent studies determined that disruption of mitochondrial cristae shape is associated with myocardial ischemia; however, the change in cristae shape in metabolic syndrome-induced cardiac remodeling remains unclear. This study determined the effect of pyridostigmine (PYR), which reversibly inhibits cholinesterase to improve autonomic imbalance, on high-fat diet (HFD)-induced cardiac insulin resistance and explored the potential effect on the shape of mitochondrial cristae. Feeding of a HFD for 22 weeks led to an irregular and even lysed cristae structure in cardiac mitochondria, which contributed to decreased mitochondrial content and ATP production and increased oxygen species production, ultimately impairing insulin signaling and lipid metabolism. Interestingly, PYR enhanced vagal activity by increasing acetylcholine production and exerted mito-protective effects by activating the LKB1/AMPK/ACC signal pathway. Specifically, PYR upregulated OPA1 and Mfn1/2 expression, promoted the formation of the mitofilin/CHCHD3/Sam50 complex, and decreased p-Drp1 and Fis1 expression, resulting in tight and parallel cristae and increasing cardiac mitochondrial complex subunit expression and ATP generation as well as decreasing release of cytochrome C from mitochondria and oxidative damage. Furthermore, PYR improved glucose and insulin tolerance and insulin-stimulated Akt phosphorylation, decreased lipid toxicity, and ultimately ameliorated HFD-induced cardiac remodeling and dysfunction. In conclusion, PYR prevented cardiac and insulin insensitivity and remodeling by stimulating vagal activity to regulate mitochondrial cristae shape and function in HFD-induced metabolic syndrome in mice. These results provide novel insights for the development of a therapeutic strategy for obesity-induced cardiac dysfunction that targets mitochondrial cristae.
Collapse
Affiliation(s)
- Run-Qing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Man Xu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Qing Wu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Yan-Ling Cui
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Si Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China
| | - Dong-Ling Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China.
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, PO Box 77#, No.76 Yanta West Road, Xi'an City, 710061, Shaanxi Province, PR China.
| |
Collapse
|
25
|
Dai C, Chen G, Chen B, Wang J, Yin C, Wang J, Gong Y, Wei L, Huang Y, Li Y. Repetitive anodal transcranial direct current stimulation improves neurological outcome and survival in a ventricular fibrillation cardiac arrest rat model. Brain Stimul 2018; 12:659-667. [PMID: 30611705 DOI: 10.1016/j.brs.2018.12.974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/15/2018] [Accepted: 12/22/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Transcranial direct current stimulation (tDCS) modulates neuronal activity and is a potential therapeutic tool for many neurological diseases. However, its beneficial effects on post cardiac arrest syndrome remains uncertain. OBJECTIVE/HYPOTHESIS We investigated the effects of repetitive anodal tDCS on neurological outcome and survival in a ventricular fibrillation (VF) cardiac arrest rat model. METHODS Cardiopulmonary resuscitation was initiated after 6 min of VF in 36 Sprague-Dawley rats. The animals were randomized into three groups immediately after resuscitation (n = 12 each): no-treatment control (NTC) group, targeted temperature management (TTM) group, and tDCS group. For tDCS, 1 mA anodal tDCS was applied on the dorsal scalp for 0.5 h. The stimulation was repeated for four sessions with 1-h resting interval under normothermia. Post-resuscitation hemodynamic, cerebral, and myocardial injuries, 96-h neurological outcome, and survival were evaluated. RESULTS Compared with the NTC group, post-resuscitation serum astroglial protein S100 beta and cardiac troponin T levels and 96-h neuronal and myocardial damage scores were markedly reduced in the tDCS and TTM groups. Myocardial ejection fraction, neurological deficit score, and 96-h survival rate were also significantly better for the tDCS and TTM groups. The period of post-resuscitation arrhythmia with hemodynamic instability was considerably shorter in the tDCS group, but no differences were observed in neurological outcome and survival between the tDCS and TTM groups. CONCLUSIONS In this cardiac arrest rat model, repeated anodal tDCS commenced after resuscitation improves 96-h neurological outcome and survival to an extent comparable to TTM by attenuating post-resuscitation cerebral and cardiac injuries.
Collapse
Affiliation(s)
- Chenxi Dai
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Gang Chen
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Bihua Chen
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Juan Wang
- Department of Emergency, Southwest Hospital, Army Medical University, Chongqing, China
| | - Changlin Yin
- Department of Emergency, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianjie Wang
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Yushun Gong
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Liang Wei
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China
| | - Yuanyuan Huang
- Department of Neurology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yongqin Li
- School of Biomedical Engineering and Imaging Medicine, Army Medical University, Chongqing, China.
| |
Collapse
|
26
|
Pelot NA, Grill WM. Effects of vagal neuromodulation on feeding behavior. Brain Res 2018; 1693:180-187. [PMID: 29425906 PMCID: PMC6003853 DOI: 10.1016/j.brainres.2018.02.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/23/2018] [Accepted: 02/01/2018] [Indexed: 02/06/2023]
Abstract
Implanted vagus nerve stimulation (VNS) for obesity was recently approved by the FDA. However, its efficacy and mechanisms of action remain unclear. Herein, we synthesize clinical and preclinical effects of VNS on feeding behavior and energy balance and discuss engineering considerations for understanding and improving the therapy. Clinical cervical VNS (≤30 Hz) to treat epilepsy or depression has produced mixed effects on weight loss as a side effect, albeit in uncontrolled, retrospective studies. Conversely, preclinical studies (cervical and subdiaphragmatic VNS) mostly report decreased food intake and either decreased weight gain or weight loss. More recent clinical studies report weight loss in response to kilohertz frequency VNS applied to the subdiaphragmatic vagi, albeit with a large placebo effect. Rather than eliciting neural activity, this therapy putatively blocks conduction in the vagus nerves. Overall, stimulation parameters lack systematic exploration, optimization, and justification based on target nerve fibers and therapeutic outcomes. The vagus nerve transduces, transmits, and integrates important neural (efferent and afferent), humoral, energetic, and inflammatory information between the gut and brain. Thus, improved understanding of the biophysics, electrophysiology, and (patho)physiology has the potential to advance VNS as an effective therapy for a wide range of diseases.
Collapse
Affiliation(s)
- Nicole A Pelot
- Department of Biomedical Engineering, Duke University, Room 1427, Fitzpatrick CIEMAS, 101 Science Drive, Campus Box 90281, Durham, NC, USA
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Room 1427, Fitzpatrick CIEMAS, 101 Science Drive, Campus Box 90281, Durham, NC, USA; Department of Electrical and Computer Engineering, Duke University, Room 130, Hudson Hall, Campus Box 90291, Durham, NC, USA; Department of Neurobiology, Duke University, Room 101B, Bryan Research Building, 311 Research Drive, Campus Box 3209, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
27
|
Chunchai T, Chattipakorn N, Chattipakorn SC. The possible factors affecting microglial activation in cases of obesity with cognitive dysfunction. Metab Brain Dis 2018; 33:615-635. [PMID: 29164373 DOI: 10.1007/s11011-017-0151-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
Obesity has reached epidemic proportions in many countries around the world. Several studies have reported that obesity can lead to the development of cognitive decline. There is increasing evidence to demonstrate that microglia play a crucial role in cognitive decline in cases of obesity, Alzheimer's disease and also in the aging process. Although there have been several studies into microglia over the past decades, the mechanistic link between microglia and cognitive decline in obese models is still not fully understood. In this review, the current available evidence from both in vitro and in vivo investigations regarding the association between the alteration in microglial activity in different obese models with respect to cognition are included. The metabolite profiles from obesity, adiposity, dietary and hormone affected microglial activation and its function in the brain are comprehensively summarized. In addition, the possible roles of microglial activation in relation to cognitive dysfunction are also presented and discussed. To ensure a balanced perspective controversial reports regarding these issues are included and discussed.
Collapse
Affiliation(s)
- Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
28
|
Nuntaphum W, Pongkan W, Wongjaikam S, Thummasorn S, Tanajak P, Khamseekaew J, Intachai K, Chattipakorn SC, Chattipakorn N, Shinlapawittayatorn K. Vagus nerve stimulation exerts cardioprotection against myocardial ischemia/reperfusion injury predominantly through its efferent vagal fibers. Basic Res Cardiol 2018; 113:22. [PMID: 29744667 DOI: 10.1007/s00395-018-0683-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/17/2018] [Accepted: 05/02/2018] [Indexed: 01/08/2023]
Abstract
Vagus nerve stimulation (VNS) has been shown to exert cardioprotection against myocardial ischemia/reperfusion (I/R) injury. However, whether the cardioprotection of VNS is mainly due to direct activation through its ipsilateral efferent fibers (motor) rather than indirect effects mediated by the afferent fibers (sensory) have not been clearly understood. We hypothesized that VNS exerts cardioprotection predominantly through its efferent vagal fibers. Thirty swine (30-35 kg) were randomized into five groups: I/R no VNS (I/R), and left mid-cervical VNS with both vagal trunks intact (LC-VNS), with left vagus nerve transection (LtVNX), with right vagus nerve transection (RtVNX) and with atropine pretreatment (Atropine), respectively. VNS was applied at the onset of ischemia (60 min) and continued until the end of reperfusion (120 min). Cardiac function, infarct size, arrhythmia score, myocardial connexin43 expression, apoptotic markers, oxidative stress markers, inflammatory markers (TNF-α and IL-10) and cardiac mitochondrial function, dynamics and fatty acid oxidation (MFN2, OPA1, DRP1, PGC1α and CPT1) were determined. LC-VNS exerted cardioprotection against myocardial I/R injury via improvement of mitochondrial function and dynamics and shifted cardiac fatty acid metabolism toward beta oxidation. However, LC-VNS and LtVNX, both efferent vagal fibers are intact, produced more profound cardioprotection, particularly infarct size reduction, decreased arrhythmia score, oxidative stress and apoptosis and attenuated mitochondrial dysfunction compared to RtVNX. These beneficial effects of VNS were abolished by atropine. Our findings suggest that selective efferent VNS may potentially be effective in attenuating myocardial I/R injury. Moreover, VNS required the contralateral efferent vagal activities to fully provide its cardioprotection.
Collapse
Affiliation(s)
- Watthana Nuntaphum
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wanpitak Pongkan
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Suwakon Wongjaikam
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Savitree Thummasorn
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pongpan Tanajak
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Juthamas Khamseekaew
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Kannaporn Intachai
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Krekwit Shinlapawittayatorn
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
29
|
Tanajak P, Sa-Nguanmoo P, Apaijai N, Wang X, Liang G, Li X, Jiang C, Chattipakorn SC, Chattipakorn N. Comparisons of cardioprotective efficacy between fibroblast growth factor 21 and dipeptidyl peptidase-4 inhibitor in prediabetic rats. Cardiovasc Ther 2018; 35. [PMID: 28391633 DOI: 10.1111/1755-5922.12263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/27/2017] [Accepted: 04/05/2017] [Indexed: 01/12/2023] Open
Abstract
AIMS Comparative efficacy between fibroblast growth factor 21 (FGF21) and vildagliptin on metabolic regulation, cardiac mitochondrial function, heart rate variability (HRV), and left ventricular (LV) function is not known. We hypothesized that FGF21 and vildagliptin share a similar efficacy in improving these parameters in high fat diet (HFD)-induced obese-insulin resistant rats. METHODS Twenty-four male Wistar rats were fed with either a normal diet (ND) or a HFD for 12 weeks. Then, ND rats were received vehicle (NDV). Rats in the HFD group were divided into three subgroups to receive either vehicle (HFV), recombinant human FGF21 (rhFGF21, 0.1 mg/kg/d, ip; HFF), or vildagliptin (3 mg/kg/d, PO; HFVil) for 28 days. RESULTS HFV rats developed obese-insulin resistance, increased serum tumor necrosis factors alpha (TNF-α) level, impaired heart rate variability (HRV) together with cardiac mitochondrial dysfunction, and LV dysfunction. Cardiac apoptosis was markedly increased in HFV rats indicated by decreased B-cell lymphoma 2 (Bcl-2) with increased Bcl2-associated X-protein (Bax) and cleaved caspase 3 expression. Cardiac FGF21 signaling pathways were markedly decreased in HFV rats indicated by decreased phosphor-fibroblast growth factor receptors 1 (p-FGFR1), phosphor-extracellular signal-regulated protein kinases 1 (p-ERK1/2), proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and carnitine palmitoyltransferase-1 (CPT-1) expression. Although both FGF21 and vildagliptin similarly attenuated these impairments, only HFF rats had decreased body weight, visceral fat, and serum TNF-α levels. CONCLUSIONS FGF21 exerts better metabolic regulation and inflammation reduction than vildagliptin. However, FGF21 and vildagliptin shared a similar efficacy for cardioprotection by improving HRV and LV function.
Collapse
Affiliation(s)
- Pongpan Tanajak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Piangkwan Sa-Nguanmoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Xiaojie Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chao Jiang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
30
|
Lee SW, Kulkarni K, Annoni EM, Libbus I, KenKnight BH, Tolkacheva EG. Stochastic vagus nerve stimulation affects acute heart rate dynamics in rats. PLoS One 2018; 13:e0194910. [PMID: 29590213 PMCID: PMC5874066 DOI: 10.1371/journal.pone.0194910] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/13/2018] [Indexed: 01/09/2023] Open
Abstract
Vagus nerve stimulation (VNS) is an approved therapy for treatment of epilepsy and depression. While also shown to be promising in several preclinical and clinical studies to treat cardiovascular diseases, optimal therapeutic stimulation paradigms are still under investigation. Traditionally, parameters such as frequency, current, and duty cycle are used to adjust the efficacy of VNS therapy. This study explored the effect of novel stochastic VNS (S-VNS) on acute heart rate (HR) dynamics. The effect of S-VNS was evaluated in Sprague Dawley rats by comparing the acute HR and HR variability (HRV) responses to standard, periodic VNS (P-VNS) across different frequencies (FREQs, 10-30 Hz). Our results demonstrate that both S-VNS and P-VNS produced negative chronotropic effects in a FREQ-dependent manner with S-VNS inducing a significantly smaller drop in HR at 10 Hz and 20 Hz compared to P-VNS (p<0.05). S-VNS demonstrated a FREQ-dependent drop in the SD1/SD2 ratio, a measure of HRV, which was absent in P-VNS, suggesting that S-VNS may acutely modulate the nonlinear relationship between short- and long-term HRV. In conclusion, S-VNS is a novel stimulation procedure that may provide different physiological outcomes from standard P-VNS, as indicated by our analysis of HR dynamics. Our study provides a rationale for further detailed investigations into the therapeutic potential of S-VNS as a novel neuromodulation technique.
Collapse
Affiliation(s)
- Steven W Lee
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Kanchan Kulkarni
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Elizabeth M Annoni
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Imad Libbus
- LivaNova, PLC (Cyberonics, Inc.), Houston, TX, United States of America
| | - Bruce H KenKnight
- LivaNova, PLC (Cyberonics, Inc.), Houston, TX, United States of America
| | - Elena G Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
31
|
Early Weight Loss Independent Effects of Sleeve Gastrectomy on Diet-Induced Cardiac Dysfunction in Obese, Wistar Rats. Obes Surg 2018; 27:2370-2377. [PMID: 28299572 DOI: 10.1007/s11695-017-2632-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Several reports suggest that bariatric surgery significantly improves cardiac function in patients with obesity cardiomyopathy. The mechanism is unknown but may be due to weight-loss independent factors. We predict that the changes in gastrointestinal anatomy after a rodent model of sleeve gastrectomy (SG) will have weight-loss independent effects on cardiac dysfunction. METHODOLOGY Cardiac dysfunction was induced by feeding a 60% kcal from fat diet to male Wistar rats for 10 weeks. Rats underwent either a SG (n = 12) or pair-fed, PF (n = 8) sham surgery. Echocardiograms were performed pre- and post-operatively at 6 and 13 weeks. Blood samples were obtained at 10 weeks post-operatively for assessment of insulin sensitivity and heart failure. RESULTS Forty-four percent of SG rats had a normal ejection fraction (EF) at 13 weeks ("responders") compared to five SG rats who did not recover EF ("non-responders"). Zero percent of the PF rats normalized EF (p = 0.03). SG responders had a smaller left ventricular internal diameter in systole and end systolic volume with improved systolic function compared to SG non-responders (EF 90.7 ± 1.7 vs. 75.4 ± 3.6%, p = <0.001). At 10 weeks post-operatively, plasma glucose and B-type natriuretic peptide levels were significantly lower in SG rats compared to PF rats. CONCLUSIONS A SG significantly improved systolic function in 44% of rats with diet-induced obesity and cardiac dysfunction. This improvement is related to weight-loss independent effects of the surgery on the entero-cardiac axis. These results offer a novel weight-loss independent, metabolic role for bariatric surgery as a potential treatment modality for obesity-associated cardiac dysfunction.
Collapse
|
32
|
Ziegler D, Strom A, Bönhof G, Püttgen S, Bódis K, Burkart V, Müssig K, Szendroedi J, Markgraf DF, Roden M. Differential associations of lower cardiac vagal tone with insulin resistance and insulin secretion in recently diagnosed type 1 and type 2 diabetes. Metabolism 2018; 79:1-9. [PMID: 29113812 DOI: 10.1016/j.metabol.2017.10.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/15/2017] [Accepted: 10/20/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE It is unclear to which extent altered insulin sensitivity/secretion contribute to the development of diabetic cardiovascular autonomic neuropathy (CAN) characterized by diminished heart rate variability (HRV). We hypothesised that lower HRV is differentially associated with measures of insulin resistance and insulin secretion in recent-onset type 1 and type 2 diabetes. MATERIALS/METHODS This cross-sectional study included participants from the German Diabetes Study with type 1 (n=275) or type 2 diabetes (n=450) with known diabetes duration ≤1year and glucose-tolerant controls (n=81). Four time domain and frequency domain HRV measures each, reflecting vagal and/or sympathetic modulation were determined over 3h during a hyperinsulinaemic-euglycaemic clamp. Insulin sensitivity was calculated as the M-value, while insulin secretion was determined by glucagon-stimulated incremental C-peptide (ΔC-peptide). RESULTS After adjustment for sex, age, BMI, smoking, and HbA1c, both M-value and ΔC-peptide were lower in the diabetes groups compared to controls (P<0.05). In multiple linear regression analyses after Bonferroni correction, vagus-mediated HRV indices were positively associated with M-value in both diabetes types (P<0.05) and inversely associated with ΔC-peptide only in participants with type 1 diabetes (P<0.05). In type 2 diabetes, the low-frequency/high-frequency (LF/HF) power as an indicator of sympathovagal balance was weakly inversely associated with M-value. CONCLUSIONS Insulin resistance may contribute to the development of early cardiovagal suppression rather than sympathetic predominance in both diabetes types, while in type 1 diabetes a lower glucagon-stimulated insulin secretion is linked to a possibly compensatory higher parasympathetic tone. Whether interventions aimed at reducing insulin resistance could also reduce the risk of CAN remains to be established.
Collapse
Affiliation(s)
- Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Gidon Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sonja Püttgen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel F Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
33
|
Kiss A, Tratsiakovich Y, Mahdi A, Yang J, Gonon AT, Podesser BK, Pernow J. Vagal nerve stimulation reduces infarct size via a mechanism involving the alpha-7 nicotinic acetylcholine receptor and downregulation of cardiac and vascular arginase. Acta Physiol (Oxf) 2017; 221:174-181. [PMID: 28238218 DOI: 10.1111/apha.12861] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 12/17/2016] [Accepted: 02/21/2017] [Indexed: 02/05/2023]
Abstract
AIMS Vagal nerve stimulation (VNS) protects from myocardial and vascular injury following myocardial ischaemia and reperfusion (IR) via a mechanism involving activation of alpha-7 nicotinic acetylcholine receptor (α7 nAChR) and reduced inflammation. Arginase is involved in development of myocardial IR injury driven by inflammatory mediators. The aim of the study was to clarify whether VNS downregulates myocardial and vascular arginase via a mechanism involving activation of α7 nAChR following myocardial IR. METHODS Anaesthetized rats were randomized to (i) sham-operated, (ii) control IR (30-min ischaemia and 2-h reperfusion, (iii) VNS throughout IR, (iv) the arginase inhibitor nor-NOHA+IR, (v) nor-NOHA+VNS+IR, (vi) selective α7 nAChR blockade by methyllycaconitine (MLA) followed by VNS throughout IR and (vii) MLA+IR. RESULTS Infarct size was reduced by VNS compared to control IR (41 ± 3% vs. 67 ± 2% of the myocardium at risk, P < 0.001). Myocardial IR increased myocardial and aortic arginase activity 1.7- and 3.1-fold respectively (P < 0.05). VNS attenuated the increase in arginase activity compared to control IR both in the myocardium and aorta (P < 0.05). MLA partially abolished the cardioprotective effect of VNS and completely abrogated the effect of VNS on arginase activity. Arginase inhibition combined with VNS did not further reduce infarct size. CONCLUSION Vagal nerve stimulation reduced infarct size and reversed the upregulation of arginase induced by IR both in the myocardium and aorta via a mechanism depending on α7 nAChR activation. The data suggest that the cardioprotective effect of VNS is mediated via reduction in arginase activity.
Collapse
Affiliation(s)
- A. Kiss
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research; Medical University of Vienna; Vienna Austria
| | - Y. Tratsiakovich
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
| | - A. Mahdi
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
| | - J. Yang
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
| | - A. T. Gonon
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
| | - B. K. Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research; Medical University of Vienna; Vienna Austria
| | - J. Pernow
- Department of Medicine; Unit of Cardiology; Karolinska Institutet; Heart and Vascular Theme; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
34
|
Tunapong W, Apaijai N, Yasom S, Tanajak P, Wanchai K, Chunchai T, Kerdphoo S, Eaimworawuthikul S, Thiennimitr P, Pongchaidecha A, Lungkaphin A, Pratchayasakul W, Chattipakorn SC, Chattipakorn N. Chronic treatment with prebiotics, probiotics and synbiotics attenuated cardiac dysfunction by improving cardiac mitochondrial dysfunction in male obese insulin-resistant rats. Eur J Nutr 2017; 57:2091-2104. [PMID: 28608320 DOI: 10.1007/s00394-017-1482-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/04/2017] [Indexed: 01/05/2023]
Abstract
PURPOSE In metabolic syndrome, the composition of gut microbiota has been disrupted, and is associated with left ventricular (LV) dysfunction. Several types of prebiotics, probiotics, and synbiotics have been shown to exert cardioprotection by restoring gut microbiota from dysbiosis and reducing systemic inflammation. However, the effects of prebiotics such as xylooligosaccharides (XOS); probiotics such as Lactobacillus paracasei STII01 HP4, and synbiotics on metabolic and LV function in obese insulin-resistant rats have not been investigated. In this study, we hypothesized that prebiotics and probiotics improve metabolic parameters, heart rate variability (HRV), blood pressure (BP), and LV function by attenuating cardiac mitochondrial dysfunction, systemic inflammation, and oxidative stress, and that synbiotics provide greater efficacy than a single regimen in obese insulin resistance. METHODS Rats were fed with either normal diet or high-fat diet (HFD) for 12 weeks and then rats in each dietary group were randomly subdivided into four subgroups to receive either a vehicle, prebiotics, probiotics, or synbiotics for another 12 weeks. Metabolic parameters, BP, HRV, LV function, cardiac mitochondrial function, systemic inflammation, and oxidative stress were determined. RESULTS HFD-fed rats had obese insulin resistance with markedly increased systemic inflammatory marker [Serum LPS; ND; 0.6 ± 0.1 EU/ml vs. HFD; 5.7 ± 1.2 EU/ml (p < 0.05)], depressed HRV, and increased BP and LV dysfunction [%ejection fraction; ND; 93 ± 2% vs. HFD; 83 ± 2% (p < 0.05)]. Prebiotics, probiotics, and synbiotics attenuated insulin resistance by improving insulin sensitivity and lipid profiles. All interventions also improved HRV, BP, LV function [%ejection fraction; HFV; 81 ± 2% vs. HFPE; 93 ± 3%, HFPO; 92 ± 1%, HFC; 92 ± 2% (p < 0.05)] by attenuating mitochondrial dysfunction, oxidative stress, and systemic inflammation in obese insulin-resistant rats. CONCLUSION Prebiotics, probiotics, and synbiotics shared similar efficacy in reducing insulin resistance and LV dysfunction in obese insulin-resistant rats.
Collapse
Affiliation(s)
- Wannipa Tunapong
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sakawdaurn Yasom
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pongpan Tanajak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Keerati Wanchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sathima Eaimworawuthikul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Parameth Thiennimitr
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Anchalee Pongchaidecha
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Anusorn Lungkaphin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
35
|
Khamseekaew J, Kumfu S, Wongjaikam S, Kerdphoo S, Jaiwongkam T, Srichairatanakool S, Fucharoen S, Chattipakorn SC, Chattipakorn N. Effects of iron overload, an iron chelator and a T-Type calcium channel blocker on cardiac mitochondrial biogenesis and mitochondrial dynamics in thalassemic mice. Eur J Pharmacol 2017; 799:118-127. [PMID: 28192097 DOI: 10.1016/j.ejphar.2017.02.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
Although cardiac mitochondrial dysfunction is involved in the pathophysiology of iron-overload cardiomyopathy, the precise mechanisms of iron-induced mitochondrial dysfunction, and the roles of the iron chelator deferiprone and the T-type calcium channel blocker efonidipine on cardiac mitochondrial biogenesis in thalassemic mice are still unknown. β-thalassemic (HT) mice were fed with a normal diet (ND) or a high iron-diet (FE) for 90 days. Then, the FE-fed mice were treated with deferiprone (75mg/kg/day) or efonidipine (4mg/kg/day) for 30 days. The hearts were used to determine cardiac mitochondrial function, biogenesis, mitochondrial dynamics and protein expressions for oxidative phosphorylation (OXPHOS) and apoptosis. ND-fed HT mice had impaired heart rate variability (HRV), increased mitochondrial dynamic proteins and caspase-3, compared with ND-fed wild-type mice. Iron overload led to increased plasma non-transferrin bound iron, oxidative stress, and the impairments of HRV and left ventricular function, cardiac mitochondrial function and mitochondrial dynamics, and decreased complex IV in thalassemic mice. Our results suggested that deferiprone and efonidipine treatment showed similar benefit in attenuating cardiac iron deposit and oxidative stress, and improved cardiac mitochondrial function, leading to improved left ventricular function, without altering the cardiac mitochondrial biogenesis, and apoptosis proteins in iron-overload thalassemic mice.
Collapse
Affiliation(s)
- Juthamas Khamseekaew
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinart Kumfu
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Suwakon Wongjaikam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | | | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
36
|
Combined Iron Chelator and Antioxidant Exerted Greater Efficacy on Cardioprotection Than Monotherapy in Iron-Overloaded Rats. PLoS One 2016; 11:e0159414. [PMID: 27428732 PMCID: PMC4948821 DOI: 10.1371/journal.pone.0159414] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/02/2016] [Indexed: 01/22/2023] Open
Abstract
Background Iron chelators are used to treat iron overload cardiomyopathy patients. However, a direct comparison of the benefits of three common iron chelators (deferoxamine (DFO), deferiprone (DFP) and deferasirox (DFX)) or an antioxidant (N-acetyl cysteine (NAC)) with a combined DFP and NAC treatments on left ventricular (LV) function with iron overload has not been investigated. Methods and Findings Male Wistar rats were fed with either a normal diet or a high iron diet (HFe group) for 4 months. After 2 months, the HFe-fed rats were divided into 6 groups to receive either: a vehicle, DFO (25 mg/kg/day), DFP (75 mg/kg/day), DFX (20 mg/kg/day), NAC (100 mg/kg/day) or the combined DFP and NAC for 2 months. Our results demonstrated that HFe rats had increased plasma non-transferrin bound iron (NTBI), malondialdehyde (MDA), cardiac iron and MDA levels and cardiac mitochondrial dysfunction, leading to LV dysfunction. Although DFO, DFP, DFX or NAC improved these parameters, leading to improved LV function, the combined DFP and NAC therapy caused greater improvement, leading to more extensively improved LV function. Conclusions The combined DFP and NAC treatment had greater efficacy than monotherapy in cardioprotection through the reduction of cardiac iron deposition and improved cardiac mitochondrial function in iron-overloaded rats.
Collapse
|