1
|
Wang Y, Li W, Li C, Zhou T, Herrmann J, Wang W. New insights into therapeutic strategies, drugs, and targets for advancing cancer therapy-related cardiovascular toxicity. Sci Bull (Beijing) 2025; 70:991-993. [PMID: 40016034 DOI: 10.1016/j.scib.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Affiliation(s)
- Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Tingting Zhou
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902, USA.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
2
|
Wang Z, Yang Y, Wang N, Lu L, Xu C, Ren J, Yang L. RIP3 orchestrates oxidative stress and pyroptosis in doxorubicin-induced cardiotoxicity through regulation of AKT/Nrf2 signaling cascade. Mol Cell Biochem 2025; 480:2331-2343. [PMID: 38955910 DOI: 10.1007/s11010-024-05029-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/04/2024] [Indexed: 07/04/2024]
Abstract
This study was designed to explore the role of RIP3 in DOX-induced cardiotoxicity and its underlying molecular mechanisms. Our results demonstrate that RIP3 exacerbates DOX-induced cardiotoxicity through promoting oxidative stress and pyroptosis by regulating the AKT/Nuclear factor erythroid 2-related factor 2 (Nrf2) signal pathway. Inhibition of RIP3 using GSK-872 attenuated DOX-induced cardiac remodeling and contractile dysfunction. Moreover, using GSK-872 in vivo, the results revealed that inhibition of RIP3 alleviated DOX-induced cardiotoxicity by the resulting inhibition of oxidative stress and pyroptosis. In addition, inhibition of RIP3 increased the protein levels of AKT and Nrf2 in DOX-treated mouse hearts. Furthermore, the AKT inhibitor LY294002 lessened RIP3 reduction-offered protection against DOX-induced H9c2 cell injury by moderating oxidative stress and pyroptosis. Taken together, these data demonstrate that RIP3 activation orchestrates DOX-induced cardiotoxicity through elevated oxidative stress and pyroptosis in an AKT/Nrf2-dependent manner. Those findings highlight the clinical relevance and therapeutic potential of targeting RIP3 for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhenyi Wang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China
| | - Yitong Yang
- Department of Children's Respiratory Asthma, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xian Yang, 712046, Shaanxi, China
| | - Nisha Wang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China
| | - Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Chennian Xu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhong Shan Hospital Fudan University, Shanghai, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Lifang Yang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
3
|
Barbetti M, Ottaviani C, Thayer JF, Sgoifo A, Carnevali L. Sex differences in heart rate and heart rate variability responses to transcutaneous auricular vagal nerve stimulation in rats. Auton Neurosci 2025; 257:103237. [PMID: 39693842 DOI: 10.1016/j.autneu.2024.103237] [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: 11/15/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
The identification of reliable biomarkers of transcutaneous auricular vagus nerve stimulation (taVNS) responsiveness is a key challenge both at the clinical and preclinical level. Vagally-mediated heart rate variability (vmHRV), a surrogate measure of cardiac vagal efferent activity, is an ideal candidate. Yet, the effects of taVNS on vmHRV remain inconclusive, likely due to the high degree of heterogeneity in stimulation protocols (e.g., taVNS parameters and side of the ear target), and little consideration of contributing factors such as sex differences. This study investigates sex differences in heart rate and vmHRV responses to different protocols of taVNS in adult rats. Male and female wild-type Groningen rats received sham or active stimulation (6 Hz or 20 Hz, 1 ms, 6 V) on the left or right auricular concha region. ECG signals were recorded before (10 min), during (20 min) and after (10 min) each session in a between-subject design. We found differential side-, frequency- and sex-specific chronotropic responses to taVNS, whereby heart rate decreased and vmHRV indexes increased at 6 Hz in males and at 20 Hz in females. Also, increases in vmHRV were only observed for right-side taVNS. The current findings suggest that biological sex should be considered for fine-tuning regulation of taVNS-induced cardiac responses and provide information regarding the side-specific effects of taVNS on vmHRV. These results will likely guide future rodent research to the choice of the most appropriate stimulation protocol in both sexes for generating information that can be translated into taVNS-related strategies in humans.
Collapse
Affiliation(s)
- Margherita Barbetti
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Cristina Ottaviani
- Department of Psychology, Sapienza University of Rome, Rome, Italy; Neuroimaging Laboratory, IRCCS, Santa Lucia Foundation, Rome, Italy
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, CA, USA
| | - Andrea Sgoifo
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Luca Carnevali
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
| |
Collapse
|
4
|
Tan W, Wang Y, Cheng S, Liu Z, Xie M, Song L, Qiu Q, Wang X, Li Z, Liu T, Guo F, Wang J, Zhou X. AdipoRon ameliorates the progression of heart failure with preserved ejection fraction via mitigating lipid accumulation and fibrosis. J Adv Res 2025; 68:299-315. [PMID: 38382593 PMCID: PMC11785573 DOI: 10.1016/j.jare.2024.02.015] [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: 12/12/2023] [Revised: 02/17/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Obesity and imbalance in lipid homeostasis contribute greatly to heart failure with preserved ejection fraction (HFpEF), the dominant form of heart failure. Few effective therapies exist to control metabolic alterations and lipid homeostasis. OBJECTIVES We aimed to investigate the cardioprotective roles of AdipoRon, the adiponectin receptor agonist, in regulating lipid accumulation in the two-hit HFpEF model. METHODS HFpEF mouse model was induced using 60 % high-fat diet plus L-NAME drinking water. Then, AdipoRon (50 mg/kg) or vehicle were administered by gavage to the two-hit HFpEF mouse model once daily for 4 weeks. Cardiac function was evaluated using echocardiography, and Postmortem analysis included RNA-sequencing, untargeted metabolomics, transmission electron microscopy and molecular biology methods. RESULTS Our study presents the pioneering evidence that AdipoR was downregulated and impaired fatty acid oxidation in the myocardia of HFpEF mice, which was associated with lipid metabolism as indicated by untargeted metabolomics. AdipoRon, orally active synthetic adiponectin receptor agonist, could upregulate AdipoR1/2 (independently of adiponectin) and reduce lipid droplet accumulation, and alleviate fibrosis to restore HFpEF phenotypes. Finally, AdipoRon primarily exerted its effects through restoring the balance of myocardial fatty acid intake, transport, and oxidation via the downstream AMPKα or PPARα signaling pathways. The protective effects of AdipoRon in HFpEF mice were reversed by compound C and GW6471, inhibitors of AMPKα and PPARα, respectively. CONCLUSIONS AdipoRon ameliorated the HFpEF phenotype by promoting myocardial fatty acid oxidation, decreasing fatty acid transport, and inhibiting fibrosis via the upregulation of AdipoR and the activation of AdipoR1/AMPKα and AdipoR2/PPARα-related downstream pathways. These findings underscore the therapeutic potential of AdipoRon in HFpEF. Importantly, all these parameters get restored in the context of continued mechanical and metabolic stressors associated with HFpEF.
Collapse
Affiliation(s)
- Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Yijun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Siyi Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Lingpeng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Qinfang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zeyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Tianyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| | - Jun Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, China; Institute of Molecular Medicine, Renmin Hospital of Wuhan University, China; Hubei Key Laboratory of Autonomic Nervous System Modulation, China; Taikang Center for Life and Medical Sciences, Wuhan University, China; Cardiac Autonomic Nervous System Research Center of Wuhan University, China; Hubei Key Laboratory of Cardiology, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
5
|
Zhang S, Huang Y, Han C, Wang F, Chen M, Yang Z, Yang S, Wang C. Central SGLT2 mediate sympathoexcitation in hypertensive heart failure via attenuating subfornical organ endothelial cGAS ubiquitination to amplify neuroinflammation: Molecular mechanism behind sympatholytic effect of Empagliflozin. Int Immunopharmacol 2025; 145:113711. [PMID: 39647283 DOI: 10.1016/j.intimp.2024.113711] [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: 10/13/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Sodium/glucose co-transporter 2 (SGLT2) inhibitors have transformed heart failure (HF) treatment, offering sympatholytic effects whose mechanisms are not fully understood. Our previous studies identified Cyclic GMP-AMP synthase (cGAS)-derived neuroinflammation in the Subfornical organ (SFO) as a promoter of sympathoexcitation, worsening myocardial remodeling in HF. This research explored the role of central SGLT2 in inducing endothelial cGAS-driven neuroinflammation in the SFO during HF and assessed the impact of SGLT2 inhibitors on this process. METHODS Hypertensive HF was induced in mice via Angiotensin II infusion for four weeks. SGLT2 expression and localization in the SFO were determined through immunoblotting and double-immunofluorescence staining. AAV9-TIE-shRNA (SGLT2) facilitated targeted SGLT2 knockdown in SFO endothelial cells (ECs), with subsequent analyses via immunoblotting, staining, and co-immunoprecipitation to investigate interactions with cGAS, mitochondrial alterations, and pro-inflammatory pathway activation. Renal sympathetic nerve activity and heart rate variability were measured to assess sympathetic output, alongside evaluations of cardiac function in HF mice. RESULTS In HF model mice, SGLT2 levels are markedly raised in SFO ECs, disrupting mitochondrial function and elevating oxidative stress. SGLT2 knockdown preserved mitochondrial integrity and function, reduced inflammation, and highlighted the influence of SGLT2 on mitochondrial health. SGLT2's interaction with cGAS prevented its ubiquitination and degradation, amplifying neuroinflammation and HF progression. Conversely, Empagliflozin counteracted these effects, suggesting that targeting the SGLT2-cGAS interaction as a novel HF treatment avenue. CONCLUSION This study revealed that SGLT2 directly reduced cGAS degradation in brain ECs, enhancing neuroinflammation in the SFO, and promoting sympathoexcitation and myocardial remodeling. The significance of the central SGLT2-cGAS interaction in cardiovascular disease mechanisms is emphasized.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Yijun Huang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chengzhi Han
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Fanshun Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Maoxiang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Shouguo Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
6
|
Zhang J, Ding W, Yin Z, Liu S, Zhao M, Xu Y, Liu J, Pan W, Peng S, Wei C, Zheng Z, Qin JJ, Wan J, Wang M. Interleukin-12p40 deficiency attenuates myocardial ferroptosis in doxorubicin-induced chronic cardiomyopathy by inhibiting Th17 differentiation and interleukin-17A production. Cardiovasc Res 2024; 120:2117-2133. [PMID: 39298642 DOI: 10.1093/cvr/cvae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/07/2024] [Accepted: 08/06/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Interleukin (IL)-12p40 is a common subunit of the bioactive cytokines IL-12 and IL-23, and it also has its own intrinsic functional activity. However, its role in doxorubicin-induced chronic cardiomyopathy (DICCM) as well as the underlying mechanisms are still unknown. METHODS AND RESULTS In this study, we used IL-12p40-knockout mice, IL-23p19-knockout mice, Rag1-knockout mice, a ferroptosis inhibitor, recombinant IL-12 (rIL-12), rIL-23, rIL-12p40, rIL-12p80, and anti-IL17A to investigate the effects of IL-12p40 on DICCM and elucidate the underlying mechanisms. We found that myocardial ferroptosis were increased in DICCM and that the inhibition of ferroptosis protected against DICCM. The expression of IL-12p40 was upregulated, and IL-12p40 was predominantly expressed by CD4+ T cells in the hearts of mice with DICCM. IL-12p40 knockout attenuated cardiac dysfunction, fibrosis and ferroptosis in DICCM, and similar results were observed in the context of CD4+ T cell IL-12p40 deficiency in Rag1-/- mice. Treatment with rIL-23, but not rIL-12, rIL-12p40 monomer or rIL-12p80, abolished the protective effects of IL-12p40 knockout. Moreover, rIL-23 treatment and IL-23p19 knockout exacerbated and ameliorated DICCM, respectively. IL-12p40 knockout might protect against DICCM by inhibiting Th17 differentiation and IL-17A production but not Th1, Th2 and Treg differentiation. Neutralizing IL-17A with an antibody also attenuated cardiac dysfunction, fibrosis, and ferroptosis. The IL-12p40/Th17/IL-17A axis might promote cardiomyocyte ferroptosis by activating TNF receptor-associated factor 6 (TRAF6)/mitogen-activated protein kinase (MAPK)/P53 signalling in DICCM. CONCLUSION Interleukin-12p40 deficiency protects against DICCM by inhibiting Th17 differentiation and the production of IL-17A, which plays critical roles in cardiomyocyte ferroptosis in DICCM via activating TRAF6/MAPK/P53 signalling. Our study may provide novel insights for the identification of therapeutic targets for treating DICCM in the clinic.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
7
|
Wang C, Fan P, Wang Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat Rev 2024; 130:102819. [PMID: 39216183 DOI: 10.1016/j.ctrv.2024.102819] [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: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Defined as scarce expression of hormone receptors and human epidermal growth factor receptor 2, triple-negative breast cancer (TNBC) is labeled as the most heterogeneous subtype of breast cancer with poorest prognosis. Despite rapid advancements in precise subtyping and tailored therapeutics, the ensuing cancer therapy-related cardiovascular toxicity (CTR-CVT) could exert detrimental impacts to TNBC survivors. Nowadays, this interdisciplinary issue is incrementally concerned by cardiologists, oncologists and other pertinent experts, propelling cardio-oncology as a booming field focusing on the whole-course management of cancer patients with potential cardiovascular threats. Here in this review, we initially profile the evolving molecular subtyping and therapeutic landscape of TNBC. Further, we introduce various monitoring approaches of CTR-CVT. In the main body, we elaborate on typical cardiotoxicities ensuing anti-TNBC treatments in detail, ranging from chemotherapy (especially anthracyclines), surgery, anesthetics, radiotherapy to immunotherapy, with future perspectives on promising directions in the era of artificial intelligence and traditional Chinese medicine.
Collapse
Affiliation(s)
- Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong 226007, Jiangsu, China
| | - Pinchao Fan
- The First Clinical Medical College, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211112, Jiangsu, China
| | - Qingqing Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
8
|
Yang Y, Wang Z, Wang N, Yang J, Yang L. CaMKII Exacerbates Doxorubicin-Induced Cardiotoxicity by Promoting Ubiquitination Through USP10 Inhibition. Cancer Med 2024; 13:e70286. [PMID: 39517125 PMCID: PMC11549063 DOI: 10.1002/cam4.70286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an effective anticancer drug, but it has a problem of cardiotoxicity that cannot be ignored. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is tightly associated with the pathological progression of DOX-induced cardiotoxicity. Ubiquitin-specific protease 10 (USP10) plays an important role in many biological processes and cancers. However, its association with DOX-induced cardiotoxicity and CaMKII remains unclear. METHODS H9C2 cells, HL-1 cells and C57BL/6 mice were used to establish the DOX-induced cardiotoxicity model, and the CaMKII-specific inhibitor KN-93 and USP10 specific inhibitor Spautin-1 were used to observe the CaMKII and USP10 effect. In cell experiments, CCK-8 method was used to assess cell viability, LDH kit was used to assess lactate dehydrogenase expression, DCFH-DA staining was used to observe changes in active oxygen content, TUNEL staining was used to observe cell apoptosis, and Western blotting method was used to detect relevant protein markers. The expression of p-CaMKII and USP10 was assessed by immunofluorescence staining. In animal experiments, mouse echocardiograph was used were used to evaluate cardiac function, and HE staining and Masson staining were used to evaluate myocardial injury. Cardiomyocyte apoptosis was detected by TUNEL staining. Western blotting method was used to detect relevant protein markers. RESULTS Our results demonstrated that activation of CaMKII and inhibition of USP10 pathway related to DOX-induced cardiotoxicity. Inhibition of CaMKII with KN-93 ameliorated DOX-induced cardiac dysfunction and cytotoxicity. In addition, CaMKII inhibition prevented DOX-induced apoptosis and ubiquitination. Furthermore, CaMKII inhibition increased USP10 expression in DOX-treated mouse hearts, H9C2 cells and HL-1 cells. At last, the USP10 inhibitor, Spautin-1, blocked the regulatory effect of CaMKII inhibition on apoptosis and ubiquitination in DOX-induced cardiotoxicity. CONCLUSION Our findings revealed that DOX-induced myocardial apoptosis and activated CaMKII through cellular and animal levels, while providing a novel probe into the mechanism of CaMKII action: promoting ubiquitination by inhibiting USP10 aggravated apoptosis.
Collapse
Affiliation(s)
- Yitong Yang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
- Department of Children's Respiratory AsthmaSecond Affiliated Hospital of Shaanxi University of Chinese MedicineXianyangShaanxiChina
| | - Zhenyi Wang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Nisha Wang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Lifang Yang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
9
|
Li Y, Yan J, Yang P. The mechanism and therapeutic strategies in doxorubicin-induced cardiotoxicity: Role of programmed cell death. Cell Stress Chaperones 2024; 29:666-680. [PMID: 39343295 PMCID: PMC11490929 DOI: 10.1016/j.cstres.2024.09.001] [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: 07/15/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Doxorubicin (DOX) is the most commonly used anthracycline anticancer agent, while its clinical utility is limited by harmful side effects like cardiotoxicity. Numerous studies have elucidated that programmed cell death plays a significant role in DOX-induced cardiotoxicity (DIC). This review summarizes several kinds of programmed cell death, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, oxidative stress, inflammation, and mitochondrial dysfunction are also important factors in the molecular mechanisms of DIC. Besides, a comprehensive understanding of specific signal pathways of DIC can be helpful to its treatment. Therefore, the related signal pathways are elucidated in this review, including sirtuin deacetylase (silent information regulator 2 [Sir2]) 1 (SIRT1)/nuclear factor erythroid 2-related factor 2, SIRT1/Klotho, SIRT1/Recombinant Sestrin 2, adenosine monophosphate-activated protein kinase, AKT, and peroxisome proliferator-activated receptor. Heat shock proteins function as chaperones, which play an important role in various stressful situations, especially in the heart. Thus, some of heat shock proteins involved in DIC are also included. Hence, the last part of this review focuses on the therapeutic research based on the mechanisms above.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of Second Clinical Medical College, Southern Medical University, Guangzhou, China.
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Lazar DR, Cainap S, Lazar FL, Maniu D, Blag C, Bota M, Colceriu MC, Zdrenghea M. The Effects of Pediatric Acute Lymphoblastic Leukemia Treatment on Cardiac Repolarization. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1158. [PMID: 39457123 PMCID: PMC11505846 DOI: 10.3390/children11101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Background: In recent years, cardiac dysfunction in childhood cancer survivors has become an important issue. Studies are focusing on identifying means for the early identification of patients at risk. Considering this, our study aims to investigate 24-hour Holter electrocardiogram (ECG) repolarization changes throughout doxorubicin (DOX) and cyclophosphamide (CPM) administration in pediatric patients treated for acute lymphoblastic leukemia (ALL). Methods: This was an investigator-driven, single-center, prospective, observational study. Enrolled children had a baseline bedside ECG examination performed before starting chemotherapy (T0). Serial Holter ECG examinations were conducted at three moments during their treatment protocol: day 8 (T1), day 29 (T2), and day 36 (T3). This study evaluated several ECG repolarization parameters, such as the QT interval, corrected QT interval (QTc), and QTc dispersion, as well as ST segment variations. Results: We evaluated 37 children diagnosed with ALL. The T0 examination revealed that over a third of patients had a resting heart rate (HR) outside the normal range for their age and sex. During chemotherapy, statistically significant increases in both HR as well as QT and QTc dispersion values were noticed, especially during the first DOX administration. What is more, a significant increase in the percentage of patients with ST segment depression from T1 to T2 and T3 was noticed. Rhythm disturbances were rare in the study population, with only a few patients presenting ventricular or supraventricular extrasystoles. Conclusions: This study reveals silent repolarization changes occurring early during anticancer treatment in children treated for ALL. These findings could aid in a better understanding of the cardiac toxicity mechanism, and they could potentially improve cardiac risk stratification for oncologic patients. Because of the small number of patients, our results need to be validated by larger studies.
Collapse
Affiliation(s)
- Diana R. Lazar
- Department No. 11, Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Department of Pediatric Cardiology, Emergency Clinical Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Simona Cainap
- Department of Pediatric Cardiology, Emergency Clinical Hospital for Children, 400394 Cluj-Napoca, Romania
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Florin Leontin Lazar
- Department No. 5, Internal Medicine, Medical Clinic Number 1, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dana Maniu
- Biomolecular Physics Department, Faculty of Physics, “Babes-Bolyai” University, 400084 Cluj-Napoca, Romania
| | - Cristina Blag
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pediatric Oncology and Hematology, Emergency Clinical Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Madalina Bota
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pediatric Oncology and Hematology, Emergency Clinical Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Marius C. Colceriu
- Department of Functional Biosciences, Discipline of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department No. 11, Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Department of Hematology, “Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
11
|
Nagai M, Ewbank H, Po SS, Dasari TW. Neuromodulation of Cardiovascular Risks Associated With Cardiotoxic Chemotherapy: A First-in-Human Randomized Pilot Study. Neuromodulation in Cancer Study (NCAN). Am J Clin Oncol 2024; 47:425-430. [PMID: 38800981 DOI: 10.1097/coc.0000000000001111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
OBJECTIVES Cardiotoxic chemotherapy is used to treat malignancies such as breast cancer and lymphoma. These treatments predispose patients to cardiotoxicity that can lead to cancer treatment-related cardiac dysfunction (CTRCD). The use of high doses of anthracyclines or in combination with human epidermal growth factor receptor 2 antagonists is associated with a progressively higher risk of CTRCD. CTRCD is preceded by increased activation of the sympathetic nervous system and abnormal left ventricular mechanical deformation as measured by abnormal global longitudinal strain (GLS). Low-level tragus stimulation (LLTS) is a new, safe, noninvasive technique that offers great potential to reduce increased sympathetic activation and improve GLS. Here, we describe a study method to examine the effects of LLTS on autonomic balance and cardiac function in breast cancer or lymphoma patients treated with anthracyclines. METHODS A first-in-human pilot, randomized, double-blind feasibility study will evaluate 104 patients (age >50 y) with breast cancer or lymphoma who receive anthracyclines with one additional CTRCD risk factor. Patients undergo 2 weeks of LLTS daily (1 h/d). Autonomic balance will be measured using heart rate variability metrics. Strain imaging using GLS will be performed pre and post-LLTS. Endothelial inflammation and oxidative stress measures will be performed using in vitro assays at baseline and after 2 weeks. CONCLUSION We hypothesize that LLTS stabilizes sympathovagal imbalance and improves cardiac performance in anthracycline-treated patients with breast cancer or lymphoma.
Collapse
Affiliation(s)
- Michiaki Nagai
- Department of Medicine, Cardiovascular Section, University of Oklahoma, Health Science Center, OK
| | | | | | | |
Collapse
|
12
|
Wang C, Zhou L, Liu C, Qiao J, Han X, Wang L, Liu Y, Xu B, Qiu Q, Zhang Z, Wang J, Zhou X, Zeng M, Yu L, Fu L. Pt nanoshells with a high NIR-II photothermal conversion efficiency mediates multimodal neuromodulation against ventricular arrhythmias. Nat Commun 2024; 15:6362. [PMID: 39069566 DOI: 10.1038/s41467-024-50557-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
Autonomic nervous system disorders play a pivotal role in the pathophysiology of cardiovascular diseases. Regulating it is essential for preventing and treating acute ventricular arrhythmias (VAs). Photothermal neuromodulation is a nonimplanted technique, but the response temperature ranges of transient receptor potential vanilloid 1 (TRPV1) and TWIK-related K+ Channel 1 (TREK1) exhibit differences while being closely aligned, and the acute nature of VAs require that it must be rapid and precise. However, the low photothermal conversion efficiency (PCE) still poses limitations in achieving rapid and precise treatment. Here, we achieve a nearly perfect blackbody absorption and a high PCE in the second near infrared (NIR-II) window (73.7% at 1064 nm) via a Pt nanoparticle shell (PtNP-shell). By precisely manipulating the photothermal effect, we successfully achieve rapid and precise multimodal neuromodulation encompassing neural activation (41.0-42.9 °C) and inhibition (45.0-46.9 °C) in a male canine model. The NIR-II photothermal modulation additionally achieves multimodal reversible autonomic modulation and confers protection against acute VAs associated with myocardial ischemia and reperfusion injury in interventional therapy.
Collapse
Affiliation(s)
- Chenlu Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chengzhe Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiaming Qiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinrui Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Luyang Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Yaxi Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Bi Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Qinfang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zizhuo Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiale Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China.
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Cardiology, Wuhan, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Mengqi Zeng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China.
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Cardiology, Wuhan, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Lei Fu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
- The Institute for Advanced Studies, Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Bo W, Cai M, Ma Y, Di L, Geng Y, Li H, Tang C, Tai F, He Z, Tian Z. Manipulation of Glutamatergic Neuronal Activity in the Primary Motor Cortex Regulates Cardiac Function in Normal and Myocardial Infarction Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305581. [PMID: 38488323 PMCID: PMC11132081 DOI: 10.1002/advs.202305581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/28/2024] [Indexed: 05/29/2024]
Abstract
Cardiac function is under neural regulation; however, brain regions in the cerebral cortex responsible for regulating cardiac function remain elusive. In this study, retrograde trans-synaptic viral tracing is used from the heart to identify a specific population of the excitatory neurons in the primary motor cortex (M1) that influences cardiac function in mice. Optogenetic activation of M1 glutamatergic neurons increases heart rate, ejection fraction, and blood pressure. By contrast, inhibition of M1 glutamatergic neurons decreased cardiac function and blood pressure as well as tyrosine hydroxylase (TH) expression in the heart. Using viral tracing and optogenetics, the median raphe nucleus (MnR) is identified as one of the key relay brain regions in the circuit from M1 that affect cardiac function. Then, a mouse model of cardiac injury is established caused by myocardial infarction (MI), in which optogenetic activation of M1 glutamatergic neurons impaired cardiac function in MI mice. Moreover, ablation of M1 neurons decreased the levels of norepinephrine and cardiac TH expression, and enhanced cardiac function in MI mice. These findings establish that the M1 neurons involved in the regulation of cardiac function and blood pressure. They also help the understanding of the neural mechanisms underlying cardiovascular regulation.
Collapse
Affiliation(s)
- Wenyan Bo
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Mengxin Cai
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Yixuan Ma
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Lingyun Di
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Yanbin Geng
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Hangzhuo Li
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Caicai Tang
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Fadao Tai
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Zhixiong He
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Institute of Brain and Behavioral SciencesShaanxi Normal UniversityXi'an710119China
| |
Collapse
|
14
|
Xie M, Guo F, Song L, Tan W, Han X, Xu S, Li X, Wang Y, Wang Y, Zhou L, Zhou X, Jiang H, Yu L. Noninvasive neuromodulation protects against doxorubicin-induced cardiotoxicity and inhibits tumor growth. iScience 2024; 27:109163. [PMID: 38425841 PMCID: PMC10904274 DOI: 10.1016/j.isci.2024.109163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Doxorubicin (Dox) poses a considerable threat to patients owing to its cardiotoxicity, thus limiting its clinical utility. Optimal cardioprotective intervention strategies are needed to suppress tumor growth but also minimize cardiac side effects. Here, we showed that tragus vagus nerve stimulation (tVNS) improved the imbalanced autonomic tone, ameliorated impaired cardiac function and fibrosis, attenuated myocyte apoptosis, and mitochondrial dysfunction compared to those in the Dox group. The beneficial effects were attenuated by methyllycaconitine citrate (MLA). The transcript profile revealed that there were 312 differentially expressed genes and the protection of tVNS and retardation of MLA were related to inflammatory response and NADPH oxidase activity. In addition, tVNS synergizing with Dox inhibited tumor growth and lung metastasis and promoted apoptosis of tumor cells in an anti-tumor immunity manner. These results indicated that non-invasive neuromodulation can play a dual role in preventing Dox-induced cardiotoxicity and suppressing tumor growth through inflammation and oxidative stress.
Collapse
Affiliation(s)
- Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Lingpeng Song
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Xinrui Han
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Saiting Xu
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Xujun Li
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Yijun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University; Institute of Molecular Medicine, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Hubei Key Laboratory of Cardiology; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| |
Collapse
|
15
|
Chen L, Tang C, Wang Z, Zhang L, Gu B, Liu X, Ming D. Enhancing Motor Sequence Learning via Transcutaneous Auricular Vagus Nerve Stimulation (taVNS): An EEG Study. IEEE J Biomed Health Inform 2024; 28:1285-1296. [PMID: 38109248 DOI: 10.1109/jbhi.2023.3344176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Motor learning plays a crucial role in human life, and various neuromodulation methods have been utilized to strengthen or improve it. Transcutaneous auricular vagus nerve stimulation (taVNS) has gained increasing attention due to its non-invasive nature, affordability and ease of implementation. Although the potential of taVNS on regulating motor learning has been suggested, its actual regulatory effect has yet been fully explored. Electroencephalogram (EEG) analysis provides an in-depth understanding of cognitive processes involved in motor learning so as to offer methodological support for regulation of motor learning. To investigate the effect of taVNS on motor learning, this study recruited 22 healthy subjects to participate a single-blind, sham-controlled, and within-subject serial reaction time task (SRTT) experiment. Every subject involved in two sessions at least one week apart and received a 20-minute active/sham taVNS in each session. Behavioral indicators as well as EEG characteristics during the task state, were extracted and analyzed. The results revealed that compared to the sham group, the active group showed higher learning performance. Additionally, the EEG results indicated that after taVNS, the motor-related cortical potential amplitudes and alpha-gamma modulation index decreased significantly and functional connectivity based on partial directed coherence towards frontal lobe was enhanced. These findings suggest that taVNS can improve motor learning, mainly through enhancing cognitive and memory functions rather than simple movement learning. This study confirms the positive regulatory effect of taVNS on motor learning, which is particularly promising as it offers a potential avenue for enhancing motor skills and facilitating rehabilitation.
Collapse
|
16
|
Shi H, Lu H, Zheng Y, Pu P, Wei L, Hu D, Tang H, Wang L. Bioinformatics and experimental studies jointly reveal that Sacubitril Valsartan improves myocardial oxidative stress and inflammation by regulating the MAPK signaling pathway to treat chemotherapy related cardiotoxicity. Biochem Biophys Res Commun 2024; 690:149244. [PMID: 38029488 DOI: 10.1016/j.bbrc.2023.149244] [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: 08/30/2023] [Revised: 10/28/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND CRC is a common but serious complication or sequela of tumor treatment, and new coping strategies are urgently needed. SV is a classic clinical cardiovascular protective drug, which has been widely used in the treatment of heart failure, hypertension and other diseases. It has good therapeutic effect in other cardiovascular diseases such as diabetes cardiomyopathy, ischemic cardiomyopathy and vascular disease, but it has not been proved by research that SV can prevent and treat CRC. METHOD In this study, DOX was used to induce a rat CRC model and evaluate the therapeutic effect of SV on it. Subsequently, R software was applied to analyze the control group, SV group, and DOX group in databases GSE207283 and GSE22369, and to screen for common differentially expressed genes. Use the DAVID website for enrichment analysis and visualization. Use STRING website to analyze and visualize protein interaction networks of key genes. Finally, experimental verification was conducted on key genes. RESULT Our research results show that SV has a protective effect on DOX induced myocardial injury by alleviating Weight loss, increasing Ejection fraction, and reducing the level of biomarkers of myocardial injury. Meanwhile, SV can effectively alleviate the above abnormalities. Bioinformatics and KEGG pathway analysis showed significant enrichment of metabolic and MAPK signaling pathways, suggesting that they may be the main regulatory pathway for SV treatment of CRC. Subsequent studies have also confirmed that SV can inhibit DOX induced myocardial injury through the MAPK signaling pathway, and alleviate DOX induced oxidative stress and inflammatory states. CONCLUSION Our research indicates that SV is a potential drug for treating CRC and preliminarily elucidates its molecular mechanism of regulating the MAPK pathway to improve oxidative stress and inflammation.
Collapse
Affiliation(s)
- Hongwei Shi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanlei Zheng
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lai Wei
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
17
|
Chen S, Zou Y, Song C, Cao K, Cai K, Wu Y, Zhang Z, Geng D, Sun W, Ouyang N, Zhang N, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches. Basic Res Cardiol 2023; 118:48. [PMID: 37938421 PMCID: PMC10632287 DOI: 10.1007/s00395-023-01018-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Nanxiang Ouyang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
18
|
Zhu H, He M, Wang Y, Zhang Y, Dong J, Chen B, Li Y, Zhou L, Du L, Liu Y, Zhang W, Ta D, Duan S. Low-intensity pulsed ultrasound alleviates doxorubicin-induced cardiotoxicity via inhibition of S100a8/a9-mediated cardiac recruitment of neutrophils. Bioeng Transl Med 2023; 8:e10570. [PMID: 38023700 PMCID: PMC10658545 DOI: 10.1002/btm2.10570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/25/2023] [Indexed: 12/01/2023] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity limits its broad use as a chemotherapy agent. The development of effective and non-invasive strategies to prevent DOX-associated adverse cardiac events is urgently needed. We aimed to examine whether and how low-intensity pulsed ultrasound (LIPUS) plays a protective role in DOX-induced cardiotoxicity. Male C57BL/6J mice were used to establish models of both acute and chronic DOX-induced cardiomyopathy. Non-invasive LIPUS therapy was conducted for four consecutive days after DOX administration. Cardiac contractile function was evaluated by echocardiography. Myocardial apoptosis, oxidative stress, and fibrosis were analyzed using terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) staining, dihydroethidium (DHE) staining, and picrosirius red staining assays. RNA-seq analysis was performed to unbiasedly explore the possible downstream regulatory mechanisms. Neutrophil recruitment and infiltration in the heart were analyzed by flow cytometry. The S100a8/a9 inhibitor ABR-238901 was utilized to identify the effect of S100a8/a9 signaling. We found that LIPUS therapy elicited a great benefit on DOX-induced heart contractile dysfunction in both acute and chronic DOX models. Chronic DOX administration increased serum creatine kinase and lactate dehydrogenase levels, as well as myocardial apoptosis, all of which were significantly mitigated by LIPUS. In addition, LIPUS treatment prevented chronic DOX-induced cardiac oxidative stress and fibrosis. RNA-seq analysis revealed that LIPUS treatment partially reversed alterations of gene expression induced by DOX. Gene ontology (GO) analysis of the downregulated genes between DOX-LIPUS and DOX-Sham groups indicated that inhibition of neutrophil chemotaxis might be involved in the protective effects of LIPUS therapy. Flow cytometry analysis illustrated the inhibitory effects of LIPUS on DOX-induced neutrophil recruitment and infiltration in the heart. Moreover, S100 calcium binding protein A8/A9 (S100a8/a9) was identified as a potential key target of LIPUS therapy. S100a8/a9 inhibition by ABR-238901 showed a similar heart protective effect against DOX-induced cardiomyopathy to LIPUS treatment. LIPUS therapy prevents DOX-induced cardiotoxicity through inhibition of S100a8/a9-mediated neutrophil recruitment to the heart, suggesting its potential application in cancer patients undergoing chemotherapy with DOX.
Collapse
Affiliation(s)
- Hong Zhu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Min He
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Yong‐Li Wang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuanxin Zhang
- Department of CardiologyNinth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingsong Dong
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Bo‐Yan Chen
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yu‐Lin Li
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lu‐Jun Zhou
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lin‐Juan Du
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Wu‐Chang Zhang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
- Department of Rehabilitation MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Sheng‐Zhong Duan
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| |
Collapse
|
19
|
Increased sympathetic outflow induced by emotional stress aggravates myocardial ischemia-reperfusion injury via activation of TLR7/MyD88/IRF5 signaling pathway. Inflamm Res 2023; 72:901-913. [PMID: 36933018 DOI: 10.1007/s00011-023-01708-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 02/10/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Emotional stress substantially increases the risk of ischemic cardiovascular diseases. Previous study indicates that sympathetic outflow is increased under emotional stress. We aim to investigate the role of increased sympathetic outflow induced by emotional stress in myocardial ischemia-reperfusion (I/R) injury, and explore the underlying mechanisms. METHODS AND RESULTS We used Designer Receptors Exclusively Activated by Designer Drugs technique to activate the ventromedial hypothalamus (VMH), a critical emotion-related nucleus. The results revealed that emotional stress stimulated by VMH activation increased sympathetic outflow, enhanced blood pressure, aggravated myocardial I/R injury, and exacerbated infarct size. The RNA-seq and molecular detection demonstrated that toll-like receptor 7 (TLR7), myeloid differentiation factor 88 (MyD88), interferon regulatory factor 5 (IRF5), and downstream inflammatory markers in cardiomyocytes were significantly upregulated. Emotional stress-induced sympathetic outflow further exacerbated the disorder of the TLR7/MyD88/IRF5 inflammatory signaling pathway. While inhibition of the signaling pathway partially alleviated myocardial I/R injury aggravated by emotional stress-induced sympathetic outflow. CONCLUSION Increased sympathetic outflow induced by emotional stress activates TLR7/MyD88/IRF5 signaling pathway, ultimately aggravating I/R injury.
Collapse
|
20
|
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
|
21
|
Hu Z, Gao S, Yang J, Xu B, Tang W, Bradley JL, Peberdy MA, Ornato JP. IVABRADINE-INDUCED HEART RATE REDUCTION INCREASES THE SEVERITY OF POSTRESUSCITATION MYOCARDIAL DYSFUNCTION IN A RAT MODEL OF CARDIOPULMONARY RESUSCITATION. Shock 2022; 58:573-581. [PMID: 36548647 PMCID: PMC9803391 DOI: 10.1097/shk.0000000000002020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 10/19/2022] [Indexed: 12/24/2022]
Abstract
ABSTRACT Aims: A rapid heart rate (HR) that occurs after cardiopulmonary resuscitation (CPR) is a short-term compensatory mechanism preserving cardiac output. However, if of long duration, it is unfavorable for myocardial function postresuscitation because of disrupted balance between myocardial oxygen supply and demand. This raises the assumption that such a sustained fast HR should be regulated. The present study aimed to investigate the follow-on effect of ivabradine (a specific inhibitor of the I f current of the sinoatrial node)-induced HR reduction (HRR) on postresuscitation myocardial function in a rat model of CPR. Methods and results: Six minutes of ventricular fibrillation and 8 min of CPR were performed on Sprague-Dawley rats. All 32 resuscitated animals were then randomized into saline and ivabradine groups, each group having nonsurvival and survival subgroups (n = 8 each). Saline or ivabradine (0.5 mL/kg) was administered at 1 h postresuscitation. Heart rate, myocardial function as expressed by cardiac output, ejection fraction, and myocardial performance index were assessed at baseline and hourly from 1 to 5 h postresuscitation. Heart rate variability was analyzed at baseline and at 1, 3, and 5 h postresuscitation. Serum epinephrine and cardiac troponin I at baseline and at 1, 3, and 5 h postresuscitation in nonsurvival subgroup were measured. Survival duration in the survival subgroup was observed. The baseline HR was approximately 390 beats/min (bpm). After resuscitation, an average increase of Δ ≈ +15 bpm (relative ratio ≈ +3.8%) with a resultant HR of 405 bpm lasting more than 5 h occurred. Ivabradine group achieved a steady HRR of Δ ≈ -30 bpm (relative ratio ≈ -7.4%) as compared with saline group ( P < 0.01). Postresuscitation myocardial function was significantly worse in the ivabradine group (all P < 0.01). Heart rate variability was significantly impaired in the ivabradine group (all P < 0.05). Serum cardiac troponin I and epinephrine concentration were significantly higher in the ivabradine group (all P < ?0.01). Survival duration was significantly shortened in the ivabradine group as compared with the saline group (388 vs. 526 min, P < ?0.01). Conclusions: Ivabradine-induced HRR increases the severity of postresuscitation myocardial dysfunction and shortens survival duration in a rat model of CPR.
Collapse
Affiliation(s)
- Zhangle Hu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
- Department of Cardiology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jin Yang
- Department of Respiratory Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Banglong Xu
- Department of Cardiology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wanchun Tang
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Jennifer L. Bradley
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Mary Ann Peberdy
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| | - Joseph P. Ornato
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
22
|
Bacova BS, Andelova K, Sykora M, Egan Benova T, Barancik M, Kurahara LH, Tribulova N. Does Myocardial Atrophy Represent Anti-Arrhythmic Phenotype? Biomedicines 2022; 10:2819. [PMID: 36359339 PMCID: PMC9687767 DOI: 10.3390/biomedicines10112819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2023] Open
Abstract
This review focuses on cardiac atrophy resulting from mechanical or metabolic unloading due to various conditions, describing some mechanisms and discussing possible strategies or interventions to prevent, attenuate or reverse myocardial atrophy. An improved awareness of these conditions and an increased focus on the identification of mechanisms and therapeutic targets may facilitate the development of the effective treatment or reversion for cardiac atrophy. It appears that a decrement in the left ventricular mass itself may be the central component in cardiac deconditioning, which avoids the occurrence of life-threatening arrhythmias. The depressed myocardial contractility of atrophied myocardium along with the upregulation of electrical coupling protein, connexin43, the maintenance of its topology, and enhanced PKCƐ signalling may be involved in the anti-arrhythmic phenotype. Meanwhile, persistent myocardial atrophy accompanied by oxidative stress and inflammation, as well as extracellular matrix fibrosis, may lead to severe cardiac dysfunction, and heart failure. Data in the literature suggest that the prevention of heart failure via the attenuation or reversion of myocardial atrophy is possible, although this requires further research.
Collapse
Affiliation(s)
| | - Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Tamara Egan Benova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Lin Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Japan
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| |
Collapse
|
23
|
Evans PC, Davidson SM, Wojta J, Bäck M, Bollini S, Brittan M, Catapano AL, Chaudhry B, Cluitmans M, Gnecchi M, Guzik TJ, Hoefer I, Madonna R, Monteiro JP, Morawietz H, Osto E, Padró T, Sluimer JC, Tocchetti CG, Van der Heiden K, Vilahur G, Waltenberger J, Weber C. From novel discovery tools and biomarkers to precision medicine-basic cardiovascular science highlights of 2021/22. Cardiovasc Res 2022; 118:2754-2767. [PMID: 35899362 PMCID: PMC9384606 DOI: 10.1093/cvr/cvac114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Here, we review the highlights of cardiovascular basic science published in 2021 and early 2022 on behalf of the European Society of Cardiology Council for Basic Cardiovascular Science. We begin with non-coding RNAs which have emerged as central regulators cardiovascular biology, and then discuss how technological developments in single-cell 'omics are providing new insights into cardiovascular development, inflammation, and disease. We also review recent discoveries on the biology of extracellular vesicles in driving either protective or pathogenic responses. The Nobel Prize in Physiology or Medicine 2021 recognized the importance of the molecular basis of mechanosensing and here we review breakthroughs in cardiovascular sensing of mechanical force. We also summarize discoveries in the field of atherosclerosis including the role of clonal haematopoiesis of indeterminate potential, and new mechanisms of crosstalk between hyperglycaemia, lipid mediators, and inflammation. The past 12 months also witnessed major advances in the field of cardiac arrhythmia including new mechanisms of fibrillation. We also focus on inducible pluripotent stem cell technology which has demonstrated disease causality for several genetic polymorphisms in long-QT syndrome and aortic valve disease, paving the way for personalized medicine approaches. Finally, the cardiovascular community has continued to better understand COVID-19 with significant advancement in our knowledge of cardiovascular tropism, molecular markers, the mechanism of vaccine-induced thrombotic complications and new anti-viral therapies that protect the cardiovascular system.
Collapse
Affiliation(s)
| | | | | | | | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, L.go R. Benzi 10, 16132 Genova, Italy
| | - Mairi Brittan
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences, University of Edinburgh, Scotland
| | | | - Bill Chaudhry
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthijs Cluitmans
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Philips Research, Eindhoven, Netherlands
| | - Massimiliano Gnecchi
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia Division of Cardiology, Unit of Translational Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Medicine, University of Cape Town, South Africa
| | - Tomasz J Guzik
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Imo Hoefer
- Central Diagnostic Laboratory, UMC Utrecht, the Netherlands
| | - Rosalinda Madonna
- Institute of Cardiology, Department of Surgical, Medical, Molecular and Critical Care Area, University of Pisa, Pisa, 56124 Italy
- Department of Internal Medicine, Cardiology Division, University of Texas Medical School, Houston, TX, USA
| | - João P Monteiro
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences, University of Edinburgh, Scotland
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Elena Osto
- Institute of Clinical Chemistry and Department of Cardiology, Heart Center, University Hospital & University of Zurich, Switzerland
| | - Teresa Padró
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, and CIBERCV-Instituto de Salud Carlos III, Barcelona, Spain
| | - Judith C Sluimer
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, Netherland
- University/BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, 80131 Napoli, Italy
| | - Kim Van der Heiden
- Biomedical Engineering, Thoraxcenter, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, and CIBERCV-Instituto de Salud Carlos III, Barcelona, Spain
| | - Johannes Waltenberger
- Cardiovascular Medicine, Medical Faculty, University of Muenster, Muenster, Germany
- Diagnostic and Therapeutic Heart Center, Zurich, Switzerland
| | | |
Collapse
|
24
|
Zhou Z, Liu C, Xu S, Wang J, Guo F, Duan S, Deng Q, Sun J, Yu F, Zhou Y, Wang M, Wang Y, Zhou L, Jiang H, Yu L. Metabolism regulator adiponectin prevents cardiac remodeling and ventricular arrhythmias via sympathetic modulation in a myocardial infarction model. Basic Res Cardiol 2022; 117:34. [PMID: 35819552 DOI: 10.1007/s00395-022-00939-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/31/2023]
Abstract
The stellate ganglia play an important role in cardiac remodeling after myocardial infarction (MI). This study aimed to investigate whether adiponectin (APN), an adipokine mainly secreted by adipose tissue, could modulate the left stellate ganglion (LSG) and exert cardioprotective effects through the sympathetic nervous system (SNS) in a canine model of MI. APN microinjection and APN overexpression with recombinant adeno-associated virus vector in the LSG were performed in acute and chronic MI models, respectively. The results showed that acute APN microinjection decreased LSG function and neural activity, and suppressed ischemia-induced ventricular arrhythmia. Chronic MI led to a decrease in the effective refractory period and action potential duration at 90% and deterioration in echocardiography performance, all of which was blunted by APN overexpression. Moreover, APN gene transfer resulted in favorable heart rate variability alteration, and decreased cardiac SNS activity, serum noradrenaline and neuropeptide Y, which were augmented after MI. APN overexpression also decreased the expression of nerve growth factor and growth associated protein 43 in the LSG and peri-infarct myocardium, respectively. Furthermore, RNA sequencing of LSG indicated that 4-week MI up-regulated the mRNA levels of macrophage/microglia activation marker Iba1, chemokine ligands (CXCL10, CCL20), chemokine receptor CCR5 and pro-inflammatory cytokine IL6, and downregulated IL1RN and IL10 mRNA, which were reversed by APN overexpression. Our results reveal that APN inhibits cardiac sympathetic remodeling and mitigates cardiac remodeling after MI. APN-mediated gene therapy may provide a potential therapeutic strategy for the treatment of MI.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Chengzhe Liu
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Saiting Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Jun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Shoupeng Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Qiang Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Fu Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Yuyang Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Meng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China.,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China. .,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China. .,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China.
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei Province, People's Republic of China. .,Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, 430060, People's Republic of China. .,Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
25
|
Choline Protects the Heart from Doxorubicin-Induced Cardiotoxicity through Vagal Activation and Nrf2/HO-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4740931. [PMID: 35422894 PMCID: PMC9005275 DOI: 10.1155/2022/4740931] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/14/2022] [Indexed: 12/15/2022]
Abstract
Choline is a precursor of the major neurotransmitter acetylcholine and has been demonstrated beneficial in diverse models of cardiovascular disease. Here, we sought to verify that choline protects the heart from DOX-induced cardiotoxicity and the underlying mechanisms. The results showed that DOX treatment decreased left ventricular ejection fraction and fractional shortening and increased serum cardiac markers and myocardial fibrosis, which were alleviated by cotreatment with choline. DOX-induced cardiotoxicity was accompanied by increases in oxidative stress, inflammation, and apoptosis, which were rectified by choline cotreatment. Levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1), which are antioxidant markers, were lowered by DOX and upregulated by choline. Moreover, DOX significantly decreased serum acetylcholine levels and the high-frequency component of heart rate variability and increased serum norepinephrine levels and the low-frequency component; these effects were rescued by choline administration. Interestingly, the protective effects of choline could be partially reversed by administration of the muscarinic receptor antagonist atropine. This suggests that choline might be a promising adjunct therapeutic agent to alleviate DOX-induced cardiotoxicity.
Collapse
|
26
|
Arinno A, Maneechote C, Khuanjing T, Ongnok B, Prathumsap N, Chunchai T, Arunsak B, Kerdphoo S, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Cardioprotective effects of melatonin and metformin against doxorubicin-induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem Pharmacol 2021; 192:114743. [PMID: 34453902 DOI: 10.1016/j.bcp.2021.114743] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023]
Abstract
Doxorubicin (Dox) is widely used in chemotherapy regimens for several malignant conditions. Unfortunately, cumulative and irreversible cardiotoxicity of Dox is the most prominent adverse effect which limits its use. Several pharmacological interventions which exert antioxidant properties, including melatonin and metformin, have demonstrated beneficial effects against various cardiac pathological conditions. However, the exact molecular mechanisms underlying their cardioprotective effects are not completely understood. We hypothesized that treatment with either melatonin or metformin provides cardioprotection against Dox-induced cardiotoxicity through mitochondrial protection. Thirty-two male Wistar rats received 6 doses of either 0.9% normal saline solution (0.9% NSS, n = 8) or Dox (3 mg/kg, i.p., n = 24). The Dox-treated rats (n = 8/group) were co-treated with: 1) Vehicle (0.9% NSS), 2) Melatonin (10 mg/kg/day), and 3) Metformin (250 mg/kg/day) for 30 consecutive days via oral gavage. Following the treatment, left ventricular (LV) function, oxidative stress, inflammation, mitochondrial function, dynamics, biogenesis and bioenergetics, mitophagy, autophagy, and apoptosis were determined. Dox induced excessive oxidative stress, inflammation, autophagy, apoptosis, reduced mitochondrial function, dynamics balance, biogenesis, and bioenergetics leading to LV dysfunction. Treatment with either melatonin or metformin exerted equal measures of cardioprotection via reducing oxidative stress, inflammation, autophagy, apoptosis, and improved mitochondrial function, dynamics balance, biogenesis, and bioenergetics in the Dox-treated rats. Melatonin and metformin exerted both anti-cancer and cardioprotective properties, suggesting they have potential roles in concomitant therapy in cancer patients receiving Dox treatment.
Collapse
Affiliation(s)
- Apiwan Arinno
- 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
| | - Chayodom Maneechote
- 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
| | - Thawatchai Khuanjing
- 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
| | - Benjamin Ongnok
- 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
| | - Nanthip Prathumsap
- 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
| | - Busarin Arunsak
- 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
| | - 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
| | - Krekwit Shinlapawittayatorn
- 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
| | - 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
|