1
|
Cao Y, Sun J, Wang X, Zhang X, Tian H, Huang L, Huang Z, Zhang Y, Zhang J, Li L, Zhou S. The double-edged nature of nicotine: toxicities and therapeutic potentials. Front Pharmacol 2024; 15:1427314. [PMID: 39206262 PMCID: PMC11350241 DOI: 10.3389/fphar.2024.1427314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Nicotine is the primary addictive component of cigarette smoke and is associated with various smoking-related diseases. However, recent research has revealed its broader cognitive-enhancing and anti-inflammatory properties, suggesting its potential therapeutic applications in several conditions. This review aims to examine the double-edged nature of nicotine, encompassing its positive and negative effects. We provide a concise overview of the physiochemical properties and pharmacology of nicotine, including insights into nicotine receptors. Therefore, the article is divided into two main sections: toxicity and therapeutic potential. We comprehensively explored nicotine-related diseases, focusing on specific signaling pathways and the underlying mechanisms that contribute to its effects. Furthermore, we addressed the current research challenges and future development perspectives. This review aims to inspire future researchers to explore the full medical potential of nicotine, which holds significant promise for the clinical management of specific diseases.
Collapse
Affiliation(s)
- Yun Cao
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Jiali Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Xiaofeng Wang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Xiaoyu Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Huijuan Tian
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Lingling Huang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Yaping Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
- Key Laboratory of Aerosol Analysis Regulation and Biological Effects of Anhui Province, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Jin Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Shun Zhou
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
- Key Laboratory of Aerosol Analysis Regulation and Biological Effects of Anhui Province, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| |
Collapse
|
2
|
Liu T, Fu Y, Shi J, He S, Chen D, Li W, Chen Y, Zhang L, Lv Q, Yang Y, Jin Q, Wang J, Xie M. Noninvasive ultrasound stimulation to treat myocarditis through splenic neuro-immune regulation. J Neuroinflammation 2023; 20:94. [PMID: 37069636 PMCID: PMC10108488 DOI: 10.1186/s12974-023-02773-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway (CAP) has been widely studied to modulate the immune response. Current stimulating strategies are invasive or imprecise. Noninvasive low-intensity pulsed ultrasound (LIPUS) has become increasingly appreciated for targeted neuronal modulation. However, its mechanisms and physiological role on myocarditis remain poorly defined. METHODS The mouse model of experimental autoimmune myocarditis was established. Low-intensity pulsed ultrasound was targeted at the spleen to stimulate the spleen nerve. Under different ultrasound parameters, histological tests and molecular biology were performed to observe inflammatory lesions and changes in immune cell subsets in the spleen and heart. In addition, we evaluated the dependence of the spleen nerve and cholinergic anti-inflammatory pathway of low-intensity pulsed ultrasound in treating autoimmune myocarditis in mice through different control groups. RESULTS The echocardiography and flow cytometry of splenic or heart infiltrating immune cells revealed that splenic ultrasound could alleviate the immune response, regulate the proportion and function of CD4+ Treg and macrophages by activating cholinergic anti-inflammatory pathway, and finally reduce heart inflammatory injury and improve cardiac remodeling, which is as effective as an acetylcholine receptor agonists GTS-21. Transcriptome sequencing showed significant differential expressed genes due to ultrasound modulation. CONCLUSIONS It is worth noting that the ultrasound therapeutic efficacy depends greatly on acoustic pressure and exposure duration, and the effective targeting organ was the spleen but not the heart. This study provides novel insight into the therapeutic potentials of LIPUS, which are essential for its future application.
Collapse
Affiliation(s)
- Tianshu Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanan Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shukun He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dandan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
3
|
Niu XH, Liu RH, Lv X, He RL, Lv FZ, Wu SJ, Li XQ, Li L, Lin JF. Activating α7nAChR helps post-myocardial infarction healing by regulating macrophage polarization via the STAT3 signaling pathway. Inflamm Res 2023; 72:879-892. [PMID: 36912917 DOI: 10.1007/s00011-023-01714-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/11/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Monocytes/macrophages play critical roles in inflammation and cardiac remodeling following myocardial infarction (MI). The cholinergic anti-inflammatory pathway (CAP) modulates local and systemic inflammatory responses by activating α7 nicotinic acetylcholine receptors (α7nAChR) in monocytes/macrophages. We investigated the effect of α7nAChR on MI-induced monocyte/macrophage recruitment and polarization and its contribution to cardiac remodeling and dysfunction. METHODS Adult male Sprague Dawley rats underwent coronary ligation and were intraperitoneally injected with the α7nAChR-selective agonist PNU282987 or the antagonist methyllycaconitine (MLA). RAW264.7 cells were stimulated with lipopolysaccharide (LPS) + interferon-gamma (IFN-γ) and treated with PNU282987, MLA, and S3I-201 (a STAT3 inhibitor). Cardiac function was evaluated by echocardiography. Masson's trichrome and immunofluorescence were used to detect cardiac fibrosis, myocardial capillary density, and M1/M2 macrophages. Western blotting was used to detect protein expression, and the proportion of monocytes was measured using flow cytometry. RESULTS Activating the CAP with PNU282987 significantly improved cardiac function and reduced cardiac fibrosis and 28-day mortality after MI. On days 3 and 7 post-MI, PNU282987 reduced the percentage of peripheral CD172a + CD43low monocytes and the infiltration of M1 macrophages in the infarcted hearts, whereas it increased the recruitment of peripheral CD172a + CD43high monocytes and M2 macrophages. Conversely, MLA exerted the opposite effects. In vitro, PNU282987 inhibited M1 macrophage polarization and promoted M2 macrophage polarization in LPS + IFN-γ-stimulated RAW264.7 cells. These PNU282987-induced changes in LPS + IFN-γ-stimulated RAW264.7 cells were reversed by administering S3I-201. CONCLUSION Activating α7nAChR inhibits the early recruitment of pro-inflammatory monocytes/macrophages during MI and improves cardiac function and remodeling. Our findings suggest a promising therapeutic target for regulating monocyte/macrophage phenotypes and promoting healing after MI.
Collapse
Affiliation(s)
- Xiao-Hui Niu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China
| | - Rong-Hua Liu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China
| | - Xiao Lv
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China
| | - Rui-Lin He
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China
| | - Fang-Zhou Lv
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shu-Jie Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China
| | - Xu-Qing Li
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China.
| | - Jia-Feng Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, China.
| |
Collapse
|
4
|
The Association between Inflammatory Biomarkers and Cardiovascular Autonomic Dysfunction after Bacterial Infection. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12073484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart rate variability (HRV) is a known measure of cardiac autonomic function. A cardiovascular autonomic dysfunction (CAD), measured as changes in HRV, is usually presented after an infectious process. The aim of the present study is to assess the association between serum inflammatory markers and CAD. For this purpose, 50 volunteers (13 of them recovering from an infection) were recruited and followed-up for 6 weeks. Their serum inflammatory biomarkers (CRP, IL1, IL4, IL6, IL10, and TNFalpha) were quantified throughout those weeks, along with their HRV resting, in response to the Valsalva maneuver, metronome breathing, standing and sustained handgrip. The correlation of within-subject changes in both HRV and inflammatory biomarkers was assessed to evaluate the concurrent changes. An inverse within-subject correlation was found between CRP and HRV in response to the Valsalva maneuver (rho (95% CI): −0.517 (−0.877 to −0.001); p = 0.032) and HRV standing (rho (95% CI): −0.490 (−0.943 to −0.036); p = 0.034). At the beginning, increased values of CRP are found along with reduced levels of HRV. Then, the CRP was reduced, accompanied by an improvement (increase) in HRV. These results suggest that CRP is a potential marker of CAD. Whether it is the cause, the consequence or a risk indicator non-causally associated is still to be determined.
Collapse
|
5
|
Zhang W, Lin H, Zou M, Yuan Q, Huang Z, Pan X, Zhang W. Nicotine in Inflammatory Diseases: Anti-Inflammatory and Pro-Inflammatory Effects. Front Immunol 2022; 13:826889. [PMID: 35251010 PMCID: PMC8895249 DOI: 10.3389/fimmu.2022.826889] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
As an anti-inflammatory alkaloid, nicotine plays dual roles in treating diseases. Here we reviewed the anti-inflammatory and pro-inflammatory effects of nicotine on inflammatory diseases, including inflammatory bowel disease, arthritis, multiple sclerosis, sepsis, endotoxemia, myocarditis, oral/skin/muscle inflammation, etc., mainly concerning the administration methods, different models, therapeutic concentration and duration, and relevant organs and tissues. According to the data analysis from recent studies in the past 20 years, nicotine exerts much more anti-inflammatory effects than pro-inflammatory ones, especially in ulcerative colitis, arthritis, sepsis, and endotoxemia. On the other hand, in oral inflammation, nicotine promotes and aggravates some diseases such as periodontitis and gingivitis, especially when there are harmful microorganisms in the oral cavity. We also carefully analyzed the nicotine dosage to determine its safe and effective range. Furthermore, we summarized the molecular mechanism of nicotine in these inflammatory diseases through regulating immune cells, immune factors, and the vagus and acetylcholinergic anti-inflammatory pathways. By balancing the “beneficial” and “harmful” effects of nicotine, it is meaningful to explore the effective medical value of nicotine and open up new horizons for remedying acute and chronic inflammation in humans.
Collapse
Affiliation(s)
- Wenji Zhang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hui Lin
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Mingmin Zou
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Qinghua Yuan
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenrui Huang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xiaoying Pan
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Xiaoying Pan, ; Wenjuan Zhang,
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Xiaoying Pan, ; Wenjuan Zhang,
| |
Collapse
|
6
|
Lu J, Wu W. Cholinergic modulation of the immune system - A novel therapeutic target for myocardial inflammation. Int Immunopharmacol 2021; 93:107391. [PMID: 33548577 DOI: 10.1016/j.intimp.2021.107391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/26/2020] [Accepted: 01/09/2021] [Indexed: 12/11/2022]
Abstract
The immune system and the nervous system depend on each other for their fine tuning and working, thus cooperating to maintain physiological homeostasis and prevent infections. The cholinergic system regulates the mobilization, differentiation, secretion, and antigen presentation of adaptive and innate immune cells mainly through α7 nicotinic acetylcholine receptors (α7nAChRs). The neuro-immune interactions are established and maintained by the following mechanisms: colocalization of immune and neuronal cells at defined anatomical sites, expression of the non-neuronal cholinergic system by immune cells, and the acetylcholine receptor-mediated activation of intracellular signaling pathways. Based on these immunological mechanisms, the protective effects of cholinergic system in animal models of diseases were summarized in this paper, such as myocardial infarction/ischemia-reperfusion, viral myocarditis, and endotoxin-induced myocardial damage. In addition to maintaining hemodynamic stability and improving the energy metabolism of the heart, both non-neuronal acetylcholine and neuronal acetylcholine in the heart can alleviate myocardial inflammation and remodeling to exert a significant cardioprotective effect. The new findings on the role of cholinergic agonists and vagus nerve stimulation in immune regulation are updated, so as to develop improved approaches to treat inflammatory heart disease.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China.
| | - Weifeng Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine, Guangxi Medical University, Shuangyong Road 22, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China.
| |
Collapse
|
7
|
Yue-Chun L, Gu XH, Li-Sha G, Zhou DP, Xing C, Guo XL, Pan LL, Song SY, Yu LL, Chen GY, Lin JF, Chu MP. Vagus nerve plays a pivotal role in CD4+ T cell differentiation during CVB3-induced murine acute myocarditis. Virulence 2021; 12:360-376. [PMID: 33380272 PMCID: PMC7834089 DOI: 10.1080/21505594.2020.1869384] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Abnormalities in CD4+ T cell (Th cell) differentiation play an important role in the pathogenesis of viral myocarditis (VMC). Our previous studies demonstrated that activation of the cholinergic anti-inflammatory pathway (CAP) alleviated the inflammatory response. In addition, we observed that right cervical vagotomy aggravates VMC by inhibiting CAP. However, the vagus nerve’s effect on differentiation of CD4+ T cells has not been studied in VMC mice to date. In this study, we investigated the effects of cervical vagotomy and the α7nAChR agonist pnu282987 on CD4+ T cell differentiation in a murine myocarditis model (BALB/c) infected with coxsackievirus B3 (CVB3). Splenic CD4+ T cells from CVB3-induced mice obtained and cultured to investigate the potential mechanism of CD4+ T cell differentiation. Each Th cell subset was analyzed by flow cytometry. Our results showed that right cervical vagotomy increased proportions of Th1 and Th17 cells and decreased proportions of Th2 and Treg cells in the spleen. Vagotomy-induced upregulation of T-bet, Ror-γ, IFN-γ, and IL-17 expression while downregulating the expression of Gata3, Foxp3, and IL-4 in the heart. In addition, we observed upregulated levels of proinflammatory cytokines, aggravated myocardial lesions and cellular infiltration, and worsened cardiac function in VMC mice. Pnu282987 administration reversed these outcomes. Furthermore, vagotomy inhibited JAK2-STAT3 activation and enhanced NF-κB activation in splenic CD4+ T cells. The CD4+ T cell differentiation was related to JAK2-STAT3 and NF-κB signal pathways. In conclusion, vagus nerve modulates the inflammatory response by regulating CD4+ T cell differentiation in response to VMC.
Collapse
Affiliation(s)
- Li Yue-Chun
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Xiao-Hong Gu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Ge Li-Sha
- Department of Pediatric Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - De-Pu Zhou
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Chao Xing
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Xiao-Ling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Lu-Lu Pan
- Child Health Manage Department, Maternal and Child Health Care Institution , Wenzhou, China
| | - Shi-Yang Song
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Li-Li Yu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Guang-Yi Chen
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Jia-Feng Lin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Mao-Ping Chu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| |
Collapse
|
8
|
Baptista AF, Baltar A, Okano AH, Moreira A, Campos ACP, Fernandes AM, Brunoni AR, Badran BW, Tanaka C, de Andrade DC, da Silva Machado DG, Morya E, Trujillo E, Swami JK, Camprodon JA, Monte-Silva K, Sá KN, Nunes I, Goulardins JB, Bikson M, Sudbrack-Oliveira P, de Carvalho P, Duarte-Moreira RJ, Pagano RL, Shinjo SK, Zana Y. Applications of Non-invasive Neuromodulation for the Management of Disorders Related to COVID-19. Front Neurol 2020; 11:573718. [PMID: 33324324 PMCID: PMC7724108 DOI: 10.3389/fneur.2020.573718] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Novel coronavirus disease (COVID-19) morbidity is not restricted to the respiratory system, but also affects the nervous system. Non-invasive neuromodulation may be useful in the treatment of the disorders associated with COVID-19. Objective: To describe the rationale and empirical basis of the use of non-invasive neuromodulation in the management of patients with COVID-10 and related disorders. Methods: We summarize COVID-19 pathophysiology with emphasis of direct neuroinvasiveness, neuroimmune response and inflammation, autonomic balance and neurological, musculoskeletal and neuropsychiatric sequela. This supports the development of a framework for advancing applications of non-invasive neuromodulation in the management COVID-19 and related disorders. Results: Non-invasive neuromodulation may manage disorders associated with COVID-19 through four pathways: (1) Direct infection mitigation through the stimulation of regions involved in the regulation of systemic anti-inflammatory responses and/or autonomic responses and prevention of neuroinflammation and recovery of respiration; (2) Amelioration of COVID-19 symptoms of musculoskeletal pain and systemic fatigue; (3) Augmenting cognitive and physical rehabilitation following critical illness; and (4) Treating outbreak-related mental distress including neurological and psychiatric disorders exacerbated by surrounding psychosocial stressors related to COVID-19. The selection of the appropriate techniques will depend on the identified target treatment pathway. Conclusion: COVID-19 infection results in a myriad of acute and chronic symptoms, both directly associated with respiratory distress (e.g., rehabilitation) or of yet-to-be-determined etiology (e.g., fatigue). Non-invasive neuromodulation is a toolbox of techniques that based on targeted pathways and empirical evidence (largely in non-COVID-19 patients) can be investigated in the management of patients with COVID-19.
Collapse
Affiliation(s)
- Abrahão Fontes Baptista
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Brazilian Institute of Neuroscience and Neurotechnology Centros de Pesquisa, Investigação e Difusão - Fundação de Amparo à Pesquisa do Estado de São Paulo (BRAINN/CEPID-FAPESP), University of Campinas, Campinas, Brazil
- Laboratory of Medical Investigations 54 (LIM-54), São Paulo University, São Paulo, Brazil
| | - Adriana Baltar
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Specialized Neuromodulation Center—Neuromod, Recife, Brazil
| | - Alexandre Hideki Okano
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Brazilian Institute of Neuroscience and Neurotechnology Centros de Pesquisa, Investigação e Difusão - Fundação de Amparo à Pesquisa do Estado de São Paulo (BRAINN/CEPID-FAPESP), University of Campinas, Campinas, Brazil
- Graduate Program in Physical Education, State University of Londrina, Londrina, Brazil
| | - Alexandre Moreira
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | | | - Ana Mércia Fernandes
- Centro de Dor, LIM-62, Departamento de Neurologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - André Russowsky Brunoni
- Serviço Interdisciplinar de Neuromodulação, Laboratório de Neurociências (LIM-27), Instituto Nacional de Biomarcadores em Neuropsiquiatria, São Paulo, Brazil
- Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bashar W. Badran
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC, United States
| | - Clarice Tanaka
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Laboratory of Medical Investigations 54 (LIM-54), São Paulo University, São Paulo, Brazil
- Instituto Central, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Daniel Ciampi de Andrade
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Centro de Dor, LIM-62, Departamento de Neurologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | - Edgard Morya
- Edmond and Lily Safra International Neuroscience Institute, Santos Dumont Institute, Macaiba, Brazil
| | - Eduardo Trujillo
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
| | - Jaiti K. Swami
- Department of Biomedical Engineering, The City College of New York of CUNY, New York, NY, United States
| | - Joan A. Camprodon
- Laboratory for Neuropsychiatry and Neuromodulation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Katia Monte-Silva
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Applied Neuroscience Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Katia Nunes Sá
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Isadora Nunes
- Department of Physiotherapy, Pontifícia Universidade Católica de Minas Gerais, Betim, Brazil
| | - Juliana Barbosa Goulardins
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
- Laboratory of Medical Investigations 54 (LIM-54), São Paulo University, São Paulo, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
- Universidade Cruzeiro do Sul (UNICSUL), São Paulo, Brazil
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York of CUNY, New York, NY, United States
| | | | - Priscila de Carvalho
- Instituto Central, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Jardim Duarte-Moreira
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
- NAPeN Network (Rede de Núcleos de Assistência e Pesquisa em Neuromodulação), Brazil
| | | | - Samuel Katsuyuki Shinjo
- Division of Rheumatology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Yossi Zana
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
| |
Collapse
|
9
|
Pham GS, Shimoura CG, Chaudhari S, Kulp DV, Mathis KW. Chronic unilateral cervical vagotomy reduces renal inflammation, blood pressure, and renal injury in a mouse model of lupus. Am J Physiol Renal Physiol 2020; 319:F155-F161. [PMID: 32538149 DOI: 10.1152/ajprenal.00201.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by hypertension that results from chronic renal inflammation and dysautonomia in the form of dampened vagal tone. In health, the vagus nerve regulates inflammatory processes through mechanisms like the cholinergic anti-inflammatory pathway; so in the case of SLE, reduced efferent vagus nerve activity may indirectly affect renal inflammation and therefore hypertension. In this study, we sought to investigate the impact of disrupting vagal neurotransmission on renal inflammation and hypertension in the setting of chronic inflammatory disease. Female SLE (NZBWF1) and control (NZW) mice were subjected to a right unilateral cervical vagotomy or sham surgery and 3 wk later were implanted with indwelling catheters to measure blood pressure. Indices of splenic and renal inflammation, as well as renal injury, were assessed. Unilateral vagotomy blunted SLE-induced increases in mean arterial pressure, albumin excretion rate, and glomerulosclerosis. This protection was associated with reduced splenic T cells and attenuated SLE-induced increases in renal proinflammatory mediators. In summary, these data indicate that unilateral vagotomy reduces renal inflammation and reduces blood pressure in SLE mice. The vagus nerves have myriad functions, and perhaps other neuroimmune interactions compensate for the ligation of one nerve.
Collapse
Affiliation(s)
- G S Pham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas.,Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, Texas
| | - C G Shimoura
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - S Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - D V Kulp
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, Texas
| | - K W Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
10
|
Dexmedetomidine attenuates inflammation and pancreatic injury in a rat model of experimental severe acute pancreatitis via cholinergic anti-inflammatory pathway. Chin Med J (Engl) 2020; 133:1073-1079. [PMID: 32265428 PMCID: PMC7213633 DOI: 10.1097/cm9.0000000000000766] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Excessive inflammatory responses play a critical role in the development of severe acute pancreatitis (SAP), and controlling such inflammation is vital for managing this often fatal disease. Dexmedetomidine has been reported to possess protective properties in inflammatory diseases. Therefore, this study aimed to investigate whether dexmedetomidine pre-treatment exerts an anti-inflammatory effect in rats with SAP induced by sodium taurocholate, and if so, to determine the potential mechanism. METHODS SAP was induced with sodium taurocholate. Rats received an intraperitoneal injection of dexmedetomidine 30 min before sodium taurocholate administration. α-bungarotoxin, a selective alpha-7 nicotinic acetylcholine receptor (α7nAchR) antagonist, was injected intra-peritoneally 30 min before dexmedetomidine administration. The role of the vagus nerve was evaluated by performing unilateral cervical vagotomy before the administration of dexmedetomidine. Efferent discharge of the vagal nerve was recorded by the BL-420F Data Acquisition & Analysis System. Six hours after onset, serum pro-inflammatory cytokine (tumor necrosis factor α [TNF-α] and interleukin 6 [IL-6]) levels and amylase levels were determined using an enzyme-linked immunosorbent assay and an automated biochemical analyzer, respectively. Histopathological changes in the pancreas were observed after hematoxylin and eosin staining and scored according to Schmidt criteria. RESULTS Pre-treatment with dexmedetomidine significantly decreased serum levels of TNF-α, IL-6, and amylase, strongly alleviating pathological pancreatic injury in the rat model of SAP (TNF-α: 174.2 ± 30.2 vs. 256.1±42.4 pg/ml; IL-6: 293.3 ± 46.8 vs. 421.7 ± 48.3 pg/ml; amylase: 2102.3 ± 165.3 vs. 3186.4 ± 245.2 U/L). However, the anti-inflammatory and pancreatic protective effects were abolished after vagotomy or pre-administration of α-bungarotoxin. Dexmedetomidine also significantly increased the discharge frequency and amplitude of the cervical vagus nerve in the SAP rat model (discharge frequency: 456.8 ± 50.3 vs. 332.4 ± 25.1 Hz; discharge amplitude: 33.4 ± 5.3 vs. 20.5 ± 2.9 μV). CONCLUSIONS Dexmedetomidine administration attenuated the systemic inflammatory response and local pancreatic injury caused by SAP in rats through the cholinergic anti-inflammatory pathway involving vagus- and α7nAChR-dependent mechanisms.
Collapse
|
11
|
Xia W, Li G, Pan Z, Zhou Q. Hypercapnia attenuates ventilator-induced lung injury through vagus nerve activation. Acta Cir Bras 2019; 34:e201900902. [PMID: 31778524 PMCID: PMC6887097 DOI: 10.1590/s0102-865020190090000002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/10/2019] [Indexed: 11/21/2022] Open
Abstract
Purpose: To investigate the role of vagus nerve activation in the protective effects
of hypercapnia in ventilator-induced lung injury (VILI) rats. Methods: Male Sprague-Dawley rats were randomized to either high-tidal volume or
low-tidal volume ventilation (control) and monitored for 4h. The high-tidal
volume group was further divided into either a vagotomy or sham-operated
group and each surgery group was further divided into two subgroups:
normocapnia and hypercapnia. Injuries were assessed hourly through
hemodynamics, respiratory mechanics and gas exchange. Protein concentration,
cell count and cytokines (TNF-α and IL-8) in bronchoalveolar lavage fluid
(BALF), lung wet-to-dry weight and pathological changes were examined. Vagus
nerve activity was recorded for 1h. Results: Compared to the control group, injurious ventilation resulted in a decrease
in PaO2/FiO2 and greater lung static compliance, MPO
activity, enhanced BALF cytokines, protein concentration, cell count, and
histology injury score. Conversely, hypercapnia significantly improved VILI
by decreasing the above injury parameters. However, vagotomy abolished the
protective effect of hypercapnia on VILI. In addition, hypercapnia enhanced
efferent vagus nerve activity compared to normocapnia. Conclusion: These results indicate that the vagus nerve plays an important role in
mediating the anti-inflammatory effect of hypercapnia on VILI.
Collapse
Affiliation(s)
- Wenfang Xia
- MD, Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China. Conception of the study, analysis of data, manuscript writing, critical revision
| | - Guang Li
- MD, Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China. Conception of the study, analysis of data, manuscript writing, critical revision
| | - Zhou Pan
- MD, Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China. Technical procedures, critical revision
| | - Qingshan Zhou
- MD, Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China. Conception of the study, analysis of data, critical revision
| |
Collapse
|
12
|
Guo G, Sun L, Yang L, Xu H. IDO1 depletion induces an anti-inflammatory response in macrophages in mice with chronic viral myocarditis. Cell Cycle 2019; 18:2598-2613. [PMID: 31416389 DOI: 10.1080/15384101.2019.1652471] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inflammation and myocardial weakness, two major hallmarks of chronic viral myocarditis (VMC), often lead to dilated cardiomyopathy or chronic heart failure. It has been reported that indoleamine 2,3-dioxygenase-1 (IDO1) may play a pathogenic role in the progression of inflammatory diseases. Hence, the study is set out to investigate the potential role of IDO1 in chronic VMC by establishing a mouse model of VMC by intraperitoneally injected with coxsackievirus B3 (CVB3). After model establishment, the expression of IDO1 was determined by RT-qPCR and Western blot analysis. IDO1 was identified as an up-regulated gene in CVB3-induced VMC. Then, in order to elucidate the potential role of IDO1 in VMC, macrophages were isolated and treated with the overexpression plasmid of IDO1 or IDO1 inhibitor (1-MT). After that, these transfected macrophages were co-cultured with normal cardiomyocytes, followed by measurement of inflammatory factors and evaluation of cardiomyocyte injury. The overexpression of IDO1 was observed to significantly enhance the levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α (TNF-α), as well as lactate dehydrogenase (LDH) activity and malondialdehyde (MDA) content. By contrast, the treatment of 1-MT in macrophages reversed the promoting effects of IDO1 on cardiomyocyte injury. Co-culture experiment showed that overexpressed IDO1 impaired cardiomyocyte, which was alleviated upon treatment of 1-MT. Taken together, the key findings of the present study provide evidence that 1-MT-mediated IDO1 suppression could potentially reduce inflammatory response in macrophages and consequently ameliorate cardiomyocyte injury in mice with VMC.
Collapse
Affiliation(s)
- Gongliang Guo
- Department of Cardiology, China-Japan Union Hospital of Jilin University , Changchun , P.R. China
| | - Liqun Sun
- Department of Pediatric, The First Hospital of Jilin University , Changchun , P.R. China
| | - Lili Yang
- Department of Obstetrics, The First Hospital of Jilin University , Changchun , P.R. China
| | - Haiming Xu
- Department of Cardiology, China-Japan Union Hospital of Jilin University , Changchun , P.R. China
| |
Collapse
|
13
|
Chen X, Ge W, Hu J, Dong T, Yao H, Chen L, Geng B, Zhou H. Inhibition of prostaglandin E2 receptor 4 by lnc000908 to promote the endothelial-mesenchymal transition participation in cardiac remodelling. J Cell Mol Med 2019; 23:6355-6367. [PMID: 31297954 PMCID: PMC6714495 DOI: 10.1111/jcmm.14524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/25/2019] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) have emerged as potent regulators of cardiac disease; however, the role of lncRNA in cardiac fibrosis remains partially understood. In this study, we identified a cardiac endothelial‐enriched lncRNA‐lnc000908, which was markedly up‐regulated in rats with cardiac fibrosis. In addition, the expression of prostaglandin E2 receptor 4 (EP4) was decreased in cardiac fibrosis. In vivo lnc000908 silencing by lentivirus increased the EP4 level, decreased endothelial‐mesenchymal transition (EndMT) and improved cardiac fibrosis and cardiac function. Consistently, the lnc000908 knockdown also up‐regulated EP4 and suppressed transforming growth factor‐beta (TGF‐β)‐induced EndMT in cardiac microvascular endothelial cells. In contrast, the lnc000908 overexpression by lentivirus decreased the EP4 level and induced EndMT. Of note, these pro‐ or anti‐EndMT effects were reversed by the EP4 overexpression or the EP4 antagonist AH‐23848, respectively. This study demonstrates that lnc000908 is a novel regulator of cardiac fibrosis by modulating the EP4 expression and EndMT.
Collapse
Affiliation(s)
- Xingxing Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenhua Ge
- Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jie Hu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tiancheng Dong
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Yao
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingzhi Chen
- Department of Clinical Laboratory, Wenzhou Central Hospital, Wenzhou, China
| | - Bin Geng
- Hypertension Center of Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Zhou
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
De-Pu Z, Li-Sha G, Guang-Yi C, Xiaohong G, Chao X, Cheng Z, Wen-Wu Z, Jia L, Jia-Feng L, Maoping C, Yue-Chun L. The cholinergic anti-inflammatory pathway ameliorates acute viral myocarditis in mice by regulating CD4 + T cell differentiation. Virulence 2019; 9:1364-1376. [PMID: 30176160 PMCID: PMC6141146 DOI: 10.1080/21505594.2018.1482179] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many studies have found that abnormalities in the proportion and differentiation of CD4+ T cells (Th cells) are closely related to the pathogenesis of viral myocarditis (VMC). Our previous research indicates that the cholinergic anti-inflammatory pathway (CAP) attenuates the inflammatory response of VMC and downregulates the expression of cytokines in Th1 and Th17 cells. This suggests that the cholinergic anti-inflammatory pathway likely attenuates the inflammatory response in VMC by altering Th cell differentiation. The aim of this study is to investigate the effect of CAP on CD4+ T cell differentiation in VMC mice. CD4+ T cells in the spleen of VMC mice were obtained and cultured in the presence of nicotine or methyllycaconitine (MLA). Cells were harvested and analyzed for the percentage of each Th cell subset by flow cytometry and transcription factor release by Western blot. Then, we detected the effect of CAP on the differentiation of Th cells in vivo. Nicotine or MLA was used to activate and block CAP, respectively, in acute virus-induced myocarditis. Nicotine treatment increased the proportion of Th2 and Treg cells, decreased the proportion of Th1 and Th17 cells in the spleen, reduced the level of proinflammatory cytokines, and attenuated the severity of myocardium lesions and cellular infiltration in viral myocarditis. MLA administration had the opposite effect. Our result demonstrated that CAP effectively protects the myocardium from virus infection, which may be attributable to the regulation of Th cell differentiation.
Collapse
Affiliation(s)
- Zhou De-Pu
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Ge Li-Sha
- b Department of Pediatric Emergency , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Chen Guang-Yi
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Gu Xiaohong
- c Children's Heart Center and Department of Pediatrics , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Xing Chao
- d Department of Clinical Laboratory , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Zheng Cheng
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Zhang Wen-Wu
- e Department of Intensive Care Unit , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Li Jia
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Lin Jia-Feng
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Chu Maoping
- c Children's Heart Center and Department of Pediatrics , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Li Yue-Chun
- a Department of Cardiology , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
15
|
Nicotinic Agonist Inhibits Cardiomyocyte Apoptosis in CVB3-Induced Myocarditis via α3 β4-nAChR/PI3K/Akt-Dependent Survivin Upregulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9496419. [PMID: 30984342 PMCID: PMC6431489 DOI: 10.1155/2019/9496419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/05/2018] [Indexed: 12/27/2022]
Abstract
Background Cardiomyocyte apoptosis is critical for the development of coxsackievirus B3- (CVB3-) induced myocarditis, which is a common cardiac disease that may result in heart failure or even sudden death. Previous studies have associated CVB3-induced apoptosis with the downregulation of antiapoptotic proteins. Here, attempts were made to examine whether nicotinic acetylcholine receptors (nAChRs), especially α3β4-nAChRs, were a novel therapeutic antiapoptotic target via the activation of survivin, a strong antiapoptotic protein, in viral myocarditis (VMC). Methods and Results In the present study, we demonstrated that nAChRs, α3β4-nAChR subunits in particular, were present and upregulated in CVB3-infected neonatal rat cardiomyocytes (NRC) and H9c2 cells by RT-qPCR. The function of α3β4-nAChRs was next examined using its specific blocker α-CTX AuIB in vitro. The results of the TUNEL assay and western blot experiments showed that the block of α3β4-nAChRs abrogated nicotine-mediated protection of NRC from CVB3-induced apoptosis, and this effect displayed a substantial correlation with the protein expressions of pAkt, survivin, and Cleaved Caspase-3. Hence, the involvement of the PI3K/Akt pathway was further verified by LY294002, a selective inhibitor of PI3K. As a result, nicotine-mediated induction of pAkt and survivin was abolished by LY294002; meanwhile, apoptotic NRC were increased accompanied by an increase of Cleaved Caspase-3 expression. Regarding CVB3-infected BALB/c mice, the α-CTX AuIB- and LY294002-treated groups had a lower survival rate, deteriorative ventricular systolic function, and more severe inflammation than the nicotine-treated group and the modulation of pAkt, survivin, and Cleaved Caspase-3 protein expressions was similar to that in CVB3-infected NRC. In addition, we found that a nicotinic agonist reduced CVB3 replication in a dose-dependent manner in vitro, which indicates that nAChR activation may serve as a possible protection mechanism of CVB3-induced myocarditis. Conclusions Our study demonstrated that α3β4-nAChR subunits are essential in the nicotine-mediated antiapoptotic effect of protecting cardiomyocytes from CVB3-induced apoptosis in vivo and in vitro. This protection correlated with the PI3K/Akt pathway and the inducement of the antiapoptotic protein survivin. A combination of these mechanisms serves as a novel protective response to treat viral myocarditis.
Collapse
|
16
|
Liu T, Zhang M, Niu H, Liu J, Ruilian M, Wang Y, Xiao Y, Xiao Z, Sun J, Dong Y, Liu X. Astragalus polysaccharide from Astragalus Melittin ameliorates inflammation via suppressing the activation of TLR-4/NF-κB p65 signal pathway and protects mice from CVB3-induced virus myocarditis. Int J Biol Macromol 2018; 126:179-186. [PMID: 30586589 DOI: 10.1016/j.ijbiomac.2018.12.207] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
Inflammation plays a crucial role in regulating cardiomyopathy and injuries of coxsackievirus B3 (CVB3)-induced viral myocarditis (VM). It has been reported that Astragalus polysaccharide (AP) from Astragalus Melittin could inhabit inflammatory gene expression under a variety of pathological conditions. However, the functional roles of AP in CVB3-induced VM still remain unknown. Here, we found that AP significantly enhanced survival for CVB3-induced mice. AP protected the mice against CVB3-induced myocardial injuries characterized by the increased body weight and depressed serum level of creatine kinase-MB (CK-MB), aspartate transaminases (AST) and lactate dehydrogenase (LDH), enhanced left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS). At the pathological level, AP ameliorated the mice against CVB3-induced myocardial damage, dilated cardiomyopathy and chronic myocardial fibrosis. We subsequently found that AP significantly suppressed CVB3-induced expression of inflammation marker (IL-1β, IL-6, TNF-α, INF-γ and MCP-1) in heart. Furthermore, we confirmed that AP suppressed the CVB3-induced expression of TLR-4 and phosphorylated NF-κB p65 in heart. Taken together, the data suggest that AP protects against CVB3-induced myocardial damage and inflammation, which may partly attribute to the regulation of TLR-4/NF-κB p65 signal pathway, moreover, suppressive effect of AP on CVB3-induced activation of TLR-4/NF-κB p65 signal was TNF-α-independent.
Collapse
Affiliation(s)
- Tianlong Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Mingjie Zhang
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Haiyan Niu
- Department of Pharmacology, College of Pharmacy, Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Jing Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Ma Ruilian
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Yi Wang
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Yunfeng Xiao
- Department of Pharmacology, College of Pharmacy, Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Zhibin Xiao
- Department of Pharmacology, College of Pharmacy, Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Jianjun Sun
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, 010059 Hohhot, PR China
| | - Yu Dong
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, PR China.
| | - Xiaolei Liu
- Department of Pharmacology, College of Pharmacy, Inner Mongolia Medical University, 010059 Hohhot, PR China.
| |
Collapse
|
17
|
Sang Y, Gu X, Pan L, Zhang C, Rong X, Wu T, Xia T, Li Y, Ge L, Zhang Y, Chu M. Melatonin Ameliorates Coxsackievirus B3-Induced Myocarditis by Regulating Apoptosis and Autophagy. Front Pharmacol 2018; 9:1384. [PMID: 30564119 PMCID: PMC6288359 DOI: 10.3389/fphar.2018.01384] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 11/12/2018] [Indexed: 01/29/2023] Open
Abstract
Current therapeutics options for viral myocarditis are unsatisfactory. Melatonin (MLT), a hormone secreted by the pineal gland and other organs, has protective effects on ischemic heart injury. However, the potential therapeutic effect of MLT on viral myocarditis is unknown. In this study, we investigated the protective effect of MLT on viral myocarditis in a mouse model of myocarditis infected with coxsackievirus B3 (CVB3) and explored the probable mechanisms. Mice with CVB3-induced myocarditis displayed inflammatory cell infiltration and interstitial edema. MLT treatment significantly ameliorated the myocardial injuries. In addition, the rate of autophagy changed, although apoptosis was inhibited in mouse hearts following treatment with MLT. These results suggest that MLT has a strong therapeutic effect on acute viral myocarditis, which is associated with changes in autophagy and apoptosis in the heart. Thus, MLT could be a promising novel therapeutic approach against viral myocarditis.
Collapse
Affiliation(s)
- Yimiao Sang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Pediatrics, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xiaohong Gu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lulu Pan
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China.,Child Health Manage Department, Maternal and Child Health Care Institution, Wenzhou, China
| | - Chunxiang Zhang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xing Rong
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| | - Tianhe Xia
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yuechun Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lisha Ge
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuanhai Zhang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| | - Maoping Chu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Kong W, Kang K, Gao Y, Liu H, Meng X, Cao Y, Yang S, Liu W, Zhang J, Yu K, Zhao M. GTS-21 Protected Against LPS-Induced Sepsis Myocardial Injury in Mice Through α7nAChR. Inflammation 2018; 41:1073-1083. [PMID: 29680908 DOI: 10.1007/s10753-018-0759-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis-induced myocardial injury is a well-known cause of mortality. The cholinergic anti-inflammatory pathway (CHAIP) is a physiological mechanism by which the central nervous system regulates immune response through the vagus nerve and acetylcholine; the α7-nicotinic acetylcholine receptor (α7nAChR) is the main component of CHAIP; GTS-21, a synthetic α7nAChR selective agonist, has repeatedly shown its powerful anti-inflammatory effect. However, little is known about its effect on LPS-induced myocardial injury. We investigated the protective effects of GTS-21 on lipopolysaccharide (LPS)-induced cardiomyopathy via the cholinergic anti-inflammatory pathway in a mouse sepsis model. We constructed the model of myocardial injury in sepsis mice by C57BL/6 using LPS and determined the time of LPS treatment by hematoxylin-eosin (HE) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). C57BL/6 mice were randomized into five groups: blank control group, model group, α-bungarotoxin + LPS group, GTS-21 + LPS group, and α-bungarotoxin + GTS-21 + LPS group. The pathological results of myocardial tissue were detected by the HE method; the apoptosis rate was detected by the TUNEL method; the relative expressions of NF-κB p65, Caspase-3, Caspase-8, Bcl-2, Bax, p53, and a7nAChR were detected by real-time quantitative PCR (RT-PCR); and the protein expressions of IL-6, IL-1 β, TNF-α, and pSTAT3 were detected by western blot. The results showed that LPS-induced myocardial pathological and apoptosis changes were significant compared with the blank group, which was reversed by GTS-21; however, pretreatment with α-bungarotoxin obviously blocked the protective effect of GTS-21. NF-κB p65, Caspase-3, Caspase-8, Bax, p53, IL-6, IL-1β, TNF-α, and pSTAT3 were significantly increased in the model group, while a7nAChR and Bcl-2 were significantly decreased; GTS-21 treatment reversed that result, while pretreatment with α-bungarotoxin strengthened the result in the model. And pretreatment with α-bungarotoxin blocked the protective effect of GTS-21. GTS-21 can alleviate the LPS-induced damage in the heart via a7nAChR, and pretreatment with α-bungarotoxin obviously blocked the protective effect of GTS-21 on sepsis in mice.
Collapse
Affiliation(s)
- Weilan Kong
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Kai Kang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Yang Gao
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, China
| | - Haitao Liu
- Department of Critical Care Medicine, the Cancer Hospital of Harbin Medical University, 150 Haping Road, Harbin, 150081, China
| | - Xianglin Meng
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Yanhui Cao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Songliu Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Wen Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Jiannan Zhang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, the Cancer Hospital of Harbin Medical University, 150 Haping Road, Harbin, 150081, China. .,Institute of Critical Care Medicine in Sino Russian Medical Research Center of Harbin Medical University, 150 Haping Road, Harbin, 150081, China.
| | - Mingyan Zhao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China.
| |
Collapse
|
19
|
Zhang Z, Dai X, Qi J, Ao Y, Yang C, Li Y. Astragalus mongholicus (Fisch.) Bge Improves Peripheral Treg Cell Immunity Imbalance in the Children With Viral Myocarditis by Reducing the Levels of miR-146b and miR-155. Front Pediatr 2018; 6:139. [PMID: 29977885 PMCID: PMC6021496 DOI: 10.3389/fped.2018.00139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/25/2018] [Indexed: 12/03/2022] Open
Abstract
Viral myocarditis (VMC) is a common cardiac disease, however, there still lacks an effective therapeutic strategy for VMC. Astragalus mongholicus (Fisch.) Bge (AB), a Chinese herb with some functional metabolites, may have some pharmacological effects on VMC. AB ingredients were measured by a full-scan LCQ mass spectrum. We aimed to explore the effects of AB on the VMC children by investigating peripheral Treg cell homeostasis. A total of 68 VMC children were random and evenly assigned into an AG group (received 10-mL AB oral liquid daily), and a CG group (received placebo daily). Peripheral blood mononuclear cells (PBMC) were obtained from peripheral blood and Treg cells were isolated. The levels of miR-146b, miR-155, Treg immunity activity and myocarditis biomarkers were measured in Treg cells. There were four main components (sucrose, calycosin, Astragaloside IV and calycosin-7-glucoside) in AB. The cases sinus tachycardia, frequent premature ventricular contractions, and supraventricular tachycardia were significantly reduced in the AG group (P < 0.05). Meanwhile, the myocardial enzymes and cardiac function indexes were improved in the AG group when compared with the CG group (P < 0.05). The time of electrocardiogram recovery, symptom duration and hospital stay was shorter in the AG group than in the CG group (P < 0.05). The levels of miR-146b and miR-155 were higher in the CG group than in the AG group (P < 0.05). The levels of ROR-γt (retinoic acid receptor-related orphan nuclear receptor gamma), FoxP3 (forkhead transcription factor), IL-10 (interleukin-11) and TGF-β (transforming growth factor beta) were lower in the CG group than in the AG group (P < 0.05). In contrast, the levels of IL-17, IL-21, CK-MB (creatine kinase-MB), cTnI (cardiac troponin I), GrB (granzyme B), sFasL (soluble fas ligand) and caspase-3 were higher in the CG group than in the AG group (P < 0.05). Furthermore, the levels of ROR-γt, FoxP3, IL-10, and TGF-β were positively, whereas the levels of IL-17, IL-21, CK-MB, cTnI, GrB, sFasL and caspase-3 were negatively, associated with the levels of miR-146b and miR-155 (P < 0.05). AB treatment improved cardiac functions, peripheral Treg cell immunity imbalance in the children with VMC by reducing the levels of miR-146b and miR-155.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of PICU, The First Hospital of Jilin University, Changchun, China
| | - Xinlun Dai
- Clinical Medical College, Jilin University, Changchun, China
| | - Ji Qi
- Department of PICU, The First Hospital of Jilin University, Changchun, China
| | - Yu Ao
- Department of PICU, The First Hospital of Jilin University, Changchun, China
| | - Chunfeng Yang
- Department of PICU, The First Hospital of Jilin University, Changchun, China
| | - Yumei Li
- Department of PICU, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|