51
|
Huang P, Zhang LY, Tan YY, Chen SD. Links between COVID-19 and Parkinson's disease/Alzheimer's disease: reciprocal impacts, medical care strategies and underlying mechanisms. Transl Neurodegener 2023; 12:5. [PMID: 36717892 PMCID: PMC9885419 DOI: 10.1186/s40035-023-00337-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The impact of coronavirus disease 2019 (COVID-19) pandemic on patients with neurodegenerative diseases and the specific neurological manifestations of COVID-19 have aroused great interest. However, there are still many issues of concern to be clarified. Therefore, we review the current literature on the complex relationship between COVID-19 and neurodegenerative diseases with an emphasis on Parkinson's disease (PD) and Alzheimer's disease (AD). We summarize the impact of COVID-19 infection on symptom severity, disease progression, and mortality rate of PD and AD, and discuss whether COVID-19 infection could trigger PD and AD. In addition, the susceptibility to and the prognosis of COVID-19 in PD patients and AD patients are also included. In order to achieve better management of PD and AD patients, modifications of care strategies, specific drug therapies, and vaccines during the pandemic are also listed. At last, mechanisms underlying the link of COVID-19 with PD and AD are reviewed.
Collapse
Affiliation(s)
- Pei Huang
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Lin-Yuan Zhang
- grid.412478.c0000 0004 1760 4628Department of Neurology, Shanghai General Hospital, Shanghai, 200080 China
| | - Yu-Yan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
52
|
Gauthier AG, Lin M, Zefi S, Kulkarni A, Thakur GA, Ashby CR, Mantell LL. GAT107-mediated α7 nicotinic acetylcholine receptor signaling attenuates inflammatory lung injury and mortality in a mouse model of ventilator-associated pneumonia by alleviating macrophage mitochondrial oxidative stress via reducing MnSOD-S-glutathionylation. Redox Biol 2023; 60:102614. [PMID: 36717349 PMCID: PMC9950665 DOI: 10.1016/j.redox.2023.102614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Supraphysiological concentrations of oxygen (hyperoxia) can compromise host defense and increase susceptibility to bacterial and viral infections, causing ventilator-associated pneumonia (VAP). Compromised host defense and inflammatory lung injury are mediated, in part, by high extracellular concentrations of HMGB1, which can be decreased by GTS-21, a partial agonist of α7 nicotinic acetylcholine receptor (α7nAChR). Here, we report that a novel α7nAChR agonistic positive allosteric modulator (ago-PAM), GAT107, at 3.3 mg/kg, i.p., significantly decreased animal mortality and markers of inflammatory injury in mice exposed to hyperoxia and subsequently infected with Pseudomonas aeruginosa. The incubation of macrophages with 3.3 μM of GAT107 significantly decreased hyperoxia-induced extracellular HMGB1 accumulation and HMGB1-induced macrophage phagocytic dysfunction. Hyperoxia-compromised macrophage function was correlated with impaired mitochondrial membrane integrity, increased superoxide levels, and decreased manganese superoxide dismutase (MnSOD) activity. This compromised MnSOD activity is due to a significant increase in its level of glutathionylation. The incubation of hyperoxic macrophages with 3.3 μM of GAT107 significantly decreases the levels of glutathionylated MnSOD, and restores MnSOD activity and mitochondrial membrane integrity. Thus, GAT107 restored hyperoxia-compromised phagocytic functions by decreasing HMGB1 release, most likely via a mitochondrial-directed pathway. Overall, our results suggest that GAT107 may be a potential treatment to decrease acute inflammatory lung injury by increasing host defense in patients with VAP.
Collapse
Affiliation(s)
- Alex G. Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sidorela Zefi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | | | | | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA,Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA,Corresponding author. Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 128 St. Albert Hall, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
53
|
Zhang N, Li Y, Feng Z. Inhibition effect of choline and parecoxib sodium on chronic constriction nerve injury-induced neuropathic pain in rats. BMC Anesthesiol 2023; 23:22. [PMID: 36639747 PMCID: PMC9837992 DOI: 10.1186/s12871-022-01913-0] [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: 02/22/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The simultaneous use of drugs with different mechanisms of analgesic action is a strategy for achieving effective pain control while minimizing dose-related side effects. Choline was described to potentiate the analgesic action of parecoxib sodium at small doses in several inflammatory pain models. However, these findings are still very limited, and more associated data are required to confirm the effectiveness of the combined choline and parecoxib sodium therapy against inflammatory pain. METHODS Adult rats were randomly divided into 9 groups (N = 6/group). The sham surgery group received an intraperitoneal (i.p.) injection of saline. Rats with chronic constriction injury (CCI) of the sciatic nerve received saline, choline (cho, 6, 12 and 24 mg/kg), parecoxib sodium (pare, 3, 6, and 12 mg/kg), or a combination of choline 6 mg/kg and parecoxib sodium 3 mg/kg. Mechanical and heat pain thresholds were measured at 30 min after drug treatment at Days 3, 5, 7, 10, and 14 after CCI. Another 30 rats were divided into 5 groups (N = 6/group): the sham, CCI + saline, CCI + cho-6 mg/kg, CCI + pare-3 mg/kg, and CCI + cho-6 mg/kg + pare-3 mg/kg groups. After repeated drug treatment for 7 days, five rats were randomly selected from each group, and the lumbar dorsal root ganglia (DRGs) (L4-6) were harvested for western blot analysis. RESULTS Choline significantly attenuated mechanical and heat hypersensitivity in CCI rats at 12 and 24 mg/kg doses (P < 0.05) but was not effective at the 6 mg/kg dose. Parecoxib sodium exerted significant pain inhibitory effects at the 6 and 12 mg/kg doses (P < 0.05) but not at the 3 mg/kg dose. Combining a low dose of choline (6 mg/kg) and parecoxib sodium (3 mg/kg) produced significant pain inhibition in CCI rats and reduced the expression of high mobility group protein 1 (HMGB1) and nuclear factor-kappa Bp65 (NF-κBp65) in L4-6 DRGs. CONCLUSION 1. In a rat model of chronic neuropathic pain (CCI), at a certain dose, choline or parecoxib sodium can alleviate mechanical pain and thermal hyperalgesia caused by CCI. 2. The combination of choline and parecoxib sodium in nonanalgesic doses can effectively relieve neuropathic pain, and its mechanism may be related to the inhibition of the high mobility group protein 1 (HMGB1)/Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway.
Collapse
Affiliation(s)
- Na Zhang
- grid.459327.eAnesthesiology Department, Civil Aviation General Hospital, Beijing, 100123 People’s Republic of China
| | - Yang Li
- grid.488137.10000 0001 2267 2324Chinese PLA Medical School, Beijing, 100853 China
| | - Zeguo Feng
- grid.414252.40000 0004 1761 8894Department of Pain Medicine, First Medical Center, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853 People’s Republic of China
| |
Collapse
|
54
|
Tao R, Mi B, Hu Y, Lin S, Xiong Y, Lu X, Panayi AC, Li G, Liu G. Hallmarks of peripheral nerve function in bone regeneration. Bone Res 2023; 11:6. [PMID: 36599828 PMCID: PMC9813170 DOI: 10.1038/s41413-022-00240-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/27/2022] [Accepted: 11/03/2022] [Indexed: 01/06/2023] Open
Abstract
Skeletal tissue is highly innervated. Although different types of nerves have been recently identified in the bone, the crosstalk between bone and nerves remains unclear. In this review, we outline the role of the peripheral nervous system (PNS) in bone regeneration following injury. We first introduce the conserved role of nerves in tissue regeneration in species ranging from amphibians to mammals. We then present the distribution of the PNS in the skeletal system under physiological conditions, fractures, or regeneration. Furthermore, we summarize the ways in which the PNS communicates with bone-lineage cells, the vasculature, and immune cells in the bone microenvironment. Based on this comprehensive and timely review, we conclude that the PNS regulates bone regeneration through neuropeptides or neurotransmitters and cells in the peripheral nerves. An in-depth understanding of the roles of peripheral nerves in bone regeneration will inform the development of new strategies based on bone-nerve crosstalk in promoting bone repair and regeneration.
Collapse
Affiliation(s)
- Ranyang Tao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Xuan Lu
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, 02215, MA, USA
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China.
| |
Collapse
|
55
|
Jin X, Wang X, Sun J, Tan W, Zhang G, Han J, Xie M, Zhou L, Yu Z, Xu T, Wang C, Wang Y, Zhou X, Jiang H. Subthreshold splenic nerve stimulation prevents myocardial Ischemia-Reperfusion injury via neuroimmunomodulation of proinflammatory factor levels. Int Immunopharmacol 2023; 114:109522. [PMID: 36502595 DOI: 10.1016/j.intimp.2022.109522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Clinical outcomes following myocardial ischemia-reperfusion (I/R) injury are strongly related to the intensity and duration of inflammation. The splenic nerve (SpN) is indispensable for the anti-inflammatory reflex. This study aimed to investigate whether splenic nerve stimulation (SpNS) plays a cardioprotective role in myocardial I/R injury and the potential underlying mechanism. METHODS Sprague-Dawley rats were randomly divided into four groups: sham group, I/R group, SpNS group, and I/R plus SpNS group. The highest SpNS intensity that did not influence heart rate was identified, and SpNS at this intensity was used as the subthreshold stimulus. Continuous subthreshold SpNS was applied for 1 h before ligation of the left coronary artery for 45 min. After 72 h of reperfusion, samples were collected for analysis. RESULTS SpN activity and splenic concentrations of cholinergic anti-inflammatory pathway (CAP)-related neurotransmitters were significantly increased by SpNS. The infarct size, oxidative stress, sympathetic tone, and the levels of proinflammatory cytokines, including TNF-α, IL-1β, and IL-6, were significantly reduced in rats subjected to subthreshold SpNS after myocardial I/R injury compared with those subjected to I/R injury alone. CONCLUSIONS Subthreshold SpNS ameliorates myocardial damage, the inflammatory response, and cardiac remodelling induced by myocardial I/R injury via neuroimmunomodulation of proinflammatory factor levels. SpNS is a potential therapeutic strategy for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Xiaoxing Jin
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Guocheng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Jiapeng Han
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Zhiyao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Tianyou Xu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Changyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| |
Collapse
|
56
|
Trifluoro-icaritin ameliorates spared nerve injury-induced neuropathic pain by inhibiting microglial activation through α7nAChR-mediated blockade of BDNF/TrkB/KCC2 signaling in the spinal cord of rats. Biomed Pharmacother 2023; 157:114001. [PMID: 36375307 DOI: 10.1016/j.biopha.2022.114001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain is still a serious and unsolved health problem. Activation of α7 nicotinic acetylcholine receptor (α7nAChR) is known to modulate neuropathic pain by inhibiting microglial activation and BDNF/TrkB/KCC2 signaling. We previously identified that trifluoro-icaritin (ICTF) has an attenuated effect on spared nerve injury (SNI)-induced neuropathic pain, but its potential mechanisms remain unknown. Here, the pain-related behaviors were determined by paw withdrawal threshold (PWT), CatWalk gait analysis, rotarod test, open field test and elevated plus maze test. The expression of pain-related signal molecules was evaluated by Western blot and immunofluorescence staining. The results showed that ICTF (5.0 mg/kg, i.p.) successfully relieved SNI-induced mechanical allodynia and anxiety-like behavior, we subsequently found there existed either positive or negative correlation between mechanical allodynia and gait parameters or rotating speed following ICTF treatment. Moreover, ICTF not only enhanced the expression of spinal α7nAChR, KCC2, CD206 and IL-10, but also decreased the levels of spinal BDNF, TrkB, CD11b, Iba-1, CD40 and IL-1β in SNI rats. Conversely, α7nAChR antagonist α-Bgtx (I.T.) effectively reversed the inhibitory effects of ICTF on SNI rats, resulting in a remarkable improvement of mechanical allodynia, activation of microglia. and suppression of α7nAChR-mediated BDNF/TrkB/KCC2 signaling. Additionally, exogenous BDNF (I.T.) dramatically abrogated both blockade of BDNF/TrkB/KCC2 cascade and alleviation of mechanical allodynia by ICTF treatment. Altogether, the study highlighted that ICTF could relieve SNI-induced neuropathic pain by suppressing microglial activation via α7nAChR-mediated inhibition of BDNF/TrkB/KCC2 signaling in the spinal cord, suggesting that ICTF may be served as a possible painkiller against neuropathic pain.
Collapse
|
57
|
Spinal Cord Stimulation Attenuates Neural Remodeling, Inflammation, and Fibrosis After Myocardial Infarction. Neuromodulation 2023; 26:57-67. [PMID: 35088742 DOI: 10.1016/j.neurom.2021.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2020] [Accepted: 09/28/2021] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is an established neuromodulation method that regulates the cardiac autonomic system. However, the biological mechanisms of the therapeutic effects of SCS after myocardial infarction (MI) remain unclear. MATERIALS AND METHODS Twenty-five rabbits were divided into five groups: SCS-MI (voltage: 0.5 v; pulse width: 0.2 ms; 50 Hz; ten minutes on and 30 minutes off; two weeks; n = 5), MI (n = 5), sham SCS-MI (voltage: 0 v; two weeks; n = 5), sham MI (n = 5), and blank control (n = 5) groups. MI was induced by permanent left anterior descending artery ligation. SCS-MI and sham SCS-MI rabbits received the corresponding interventions 24 hours after MI. Autonomic remodeling was evaluated using enzyme-linked immunosorbent assay and immunohistochemistry. Inflammation and myocardial fibrosis were assessed using immunohistochemistry, quantitative polymerase chain reaction, hematoxylin and eosin staining, Masson staining, and Western blot. RESULTS SCS improved the abnormal systemic autonomic activity. Cardiac norepinephrine decreased after MI (p < 0.01) and did not improve with SCS. Cardiac acetylcholine increased with SCS compared with the MI group (p < 0.05). However, no difference was observed between the MI and blank control groups. Growth-associated protein 43 (p < 0.001) and tyrosine hydroxylase (p < 0.001) increased whereas choline acetyltransferase (p < 0.05) decreased in the MI group compared with the blank control group. These changes were attenuated with SCS. SCS inhibited inflammation, decreased the ratio of phosphorylated-Erk to Erk (p < 0.001), and increased the ratio of phosphorylated-STAT3 to STAT3 (p < 0.001) compared with the MI group. Myocardial fibrosis was also attenuated by SCS. CONCLUSIONS SCS improved abnormal autonomic activity after MI, leading to reduced inflammation, reactivation of STAT3, and inhibition of Erk. Additionally, SCS attenuated myocardial fibrosis. Our results warrant future studies of biological mechanisms of the therapeutic effects of SCS after MI.
Collapse
|
58
|
Oshaghi M, Kourosh-Arami M, Roozbehkia M. Role of neurotransmitters in immune-mediated inflammatory disorders: a crosstalk between the nervous and immune systems. Neurol Sci 2023; 44:99-113. [PMID: 36169755 DOI: 10.1007/s10072-022-06413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a group of common heterogeneous disorders, characterized by an alteration of cellular homeostasis. Primarily, it has been shown that the release and diffusion of neurotransmitters from nervous tissue could result in signaling through lymphocyte cell-surface receptors and the modulation of immune function. This finding led to the idea that the neurotransmitters could serve as immunomodulators. It is now manifested that neurotransmitters can also be released from leukocytes and act as autocrine or paracrine modulators. Increasing data indicate that there is a crosstalk between inflammation and alterations in neurotransmission. The primary goal of this review is to demonstrate how these two pathways may converge at the level of the neuron and glia to involve in IMID. We review the role of neurotransmitters in IMID. The different effects that these compounds exert on a variety of immune cells are also reviewed. Current and future developments in understanding the cross-talk between the immune and nervous systems will undoubtedly identify new ways for treating immune-mediated diseases utilizing agonists or antagonists of neurotransmitter receptors.
Collapse
Affiliation(s)
- Mojgan Oshaghi
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Roozbehkia
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
59
|
Lv C, Sun M, Guo Y, Xia W, Qiao S, Tao Y, Fang Y, Zhang Q, Zhu Y, Yalikun Y, Xia Y, Wei Z, Dai Y. Cholinergic dysfunction-induced insufficient activation of alpha7 nicotinic acetylcholine receptor drives the development of rheumatoid arthritis through promoting protein citrullination via the SP3/PAD4 pathway. Acta Pharm Sin B 2023; 13:1600-1615. [PMID: 37139415 PMCID: PMC10150100 DOI: 10.1016/j.apsb.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/10/2022] [Accepted: 11/23/2022] [Indexed: 01/19/2023] Open
Abstract
Both cholinergic dysfunction and protein citrullination are the hallmarks of rheumatoid arthritis (RA), but the relationship between the two phenomena remains unclear. We explored whether and how cholinergic dysfunction accelerates protein citrullination and consequently drives the development of RA. Cholinergic function and protein citrullination levels in patients with RA and collagen-induced arthritis (CIA) mice were collected. In both neuron-macrophage coculture system and CIA mice, the effect of cholinergic dysfunction on protein citrullination and expression of peptidylarginine deiminases (PADs) was assessed by immunofluorescence. The key transcription factors for PAD4 expression were predicted and validated. Cholinergic dysfunction in the patients with RA and CIA mice negatively correlated with the degree of protein citrullination in synovial tissues. The cholinergic or alpha7 nicotinic acetylcholine receptor (α7nAChR) deactivation and activation resulted in the promotion and reduction of protein citrullination in vitro and in vivo, respectively. Especially, the activation deficiency of α7nAChR induced the earlier onset and aggravation of CIA. Furthermore, deactivation of α7nAChR increased the expression of PAD4 and specificity protein-3 (SP3) in vitro and in vivo. Our results suggest that cholinergic dysfunction-induced deficient α7nAChR activation, which induces the expression of SP3 and its downstream molecule PAD4, accelerating protein citrullination and the development of RA.
Collapse
Affiliation(s)
- Changjun Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minghui Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wenxin Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Simiao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yulai Fang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qin Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yusufu Yalikun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Corresponding authors.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Corresponding authors.
| |
Collapse
|
60
|
Yu LH, Jia GW, Liu YL, Wang SR, Ma JX. Vagus nerve stimulation is a potential treatment for ischemic stroke. Neural Regen Res 2023; 18:825-831. [DOI: 10.4103/1673-5374.350698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
61
|
Yasin S, Görücü Yılmaz Ş, Geyik S, Oğuzkan Balcı S. The holistic approach to the CHRNA7 gene, hsa-miR-3158-5p, and 15q13.3 hotspot CNVs in migraineurs. Mol Pain 2023; 19:17448069231152104. [PMID: 36604774 PMCID: PMC9850133 DOI: 10.1177/17448069231152104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/03/2022] [Accepted: 12/29/2022] [Indexed: 01/07/2023] Open
Abstract
Migraine is a neurological disease characterized by severe headache attacks. Combinations of different genetic variations such as copy number variation (CNV) in a gene and microRNA (miRNA) expression can provide a holistic approach to the disease as a pathophysiological, diagnostic, and therapeutic target. CNVs, the Cholinergic Receptor Nicotinic Alpha 7 Subunit (CHRNA7) gene, and expression of gene-targeting miRNAs (hsa-miR-548e-5p and hsa-miR-3158-5p) in migraineurs (n = 102; with aura, n = 43; without aura, n = 59) and non-migraines (n = 120) aged 15-60 years, comparative, case-control study was conducted. Genetic markers were compared with biochemical parameters (BMI, WBC, Urea, GFR, ESR, CRP, HBG). All analyzes were performed by quantitative Real-Time PCR (q-PCR) and fold change was calculated with the 2-ΔΔCT method. The diagnostic power of the CHRNA7 gene, CNV, and miRNAs were analyzed with the receiver operating curve (ROC). CHRNA7 gene and hsa-miR-3158-5p are down-regulated in migraineurs and the gene is controlled by this miRNA via CNVs (p < .05). Both deletion and duplication were detected in patients with migraine for CVN numbers (p = .05). The number of CNV deletions was higher than duplications. When CHRNA7-CNV-hsa-miR-3158-5p was modeled together in the ROC analysis, the area under the curve (AUC) was 0.805, and the diagnostic power was "good". In migraineurs, the CHRNA7 gene can be controlled by hsa-miR-3158-5p via CNVs to modulate the mechanism of pain. These three genetic markers have diagnostic potential and may be used in antimigraine treatments.
Collapse
Affiliation(s)
- Sedat Yasin
- Department of Neurology,
Gaziantep
University, Gaziantep, Turkey
| | - Şenay Görücü Yılmaz
- Department of Nutrition and
Dietetics, Gaziantep
University, Gaziantep, Turkey
| | - Sırma Geyik
- Department of Neurology,
Gaziantep
University, Gaziantep, Turkey
| | | |
Collapse
|
62
|
Gao S, Jiang Y, Chen Z, Zhao X, Gu J, Wu H, Liao Y, Sun H, Wang J, Chen W. Metabolic Reprogramming of Microglia in Sepsis-Associated Encephalopathy: Insights from Neuroinflammation. Curr Neuropharmacol 2023; 21:1992-2005. [PMID: 36529923 PMCID: PMC10514522 DOI: 10.2174/1570159x21666221216162606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction caused by sepsis that manifests as a range of brain dysfunctions from delirium to coma. It is a relatively common complication of sepsis associated with poor patient prognosis and mortality. The pathogenesis of SAE involves neuroinflammatory responses, neurotransmitter dysfunction, blood-brain barrier (BBB) disruption, abnormal blood flow regulation, etc. Neuroinflammation caused by hyperactivation of microglia is considered to be a key factor in disease development, which can cause a series of chain reactions, including BBB disruption and oxidative stress. Metabolic reprogramming has been found to play a central role in microglial activation and executive functions. In this review, we describe the pivotal role of energy metabolism in microglial activation and functional execution and demonstrate that the regulation of microglial metabolic reprogramming might be crucial in the development of clinical therapeutics for neuroinflammatory diseases like SAE.
Collapse
Affiliation(s)
- Shenjia Gao
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yi Jiang
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Jiahui Gu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Han Wu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yun Liao
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Hao Sun
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jun Wang
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Fudan Zhangjiang Institute, Shanghai, 201203, China
| |
Collapse
|
63
|
Kirkland LG, Garbe CG, Hadaya J, Benson PV, Wagener BM, Tankovic S, Hoover DB. Sympathetic innervation of human and porcine spleens: implications for between species variation in function. Bioelectron Med 2022; 8:20. [PMID: 36536461 PMCID: PMC9762010 DOI: 10.1186/s42234-022-00102-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The vagus nerve affects innate immune responses by activating spleen-projecting sympathetic neurons, which modulate leukocyte function. Recent basic and clinical research investigating vagus nerve stimulation to engage the cholinergic anti-inflammatory pathway (CAP) has shown promising therapeutic results for a variety of inflammatory diseases. Abundant sympathetic innervation occurs in rodent spleens, and use of these species has dominated mechanistic research investigating the CAP. However, previous neuroanatomical studies of human spleen found a more restricted pattern of innervation compared to rodents. Therefore, our primary goal was to establish the full extent of sympathetic innervation of human spleens using donor tissue with the shortest procurement to fixation time. Parallel studies of porcine spleen, a large animal model, were performed as a positive control and for comparison. METHODS Human and porcine spleen tissue were fixed immediately after harvest and prepared for immunohistochemistry. Human heart and porcine spleen were stained in conjunction as positive controls. Several immunohistochemical protocols were compared for best results. Tissue was stained for tyrosine hydroxylase (TH), a noradrenergic marker, using VIP purple chromogen. Consecutive tissue slices were stained for neuropeptide Y (NPY), which often co-localizes with TH, or double-labelled for TH and CD3, a T cell marker. High-magnification images and full scans of the tissue were obtained and analyzed for qualitative differences between species. RESULTS TH had dominant perivascular localization in human spleen, with negligible innervation of parenchyma, but such nerves were abundant throughout ventricular myocardium. In marked contrast, noradrenergic innervation was abundant in all regions of porcine spleen, with red pulp having more nerves than white pulp. NPY stain results were consistent with this pattern. In human spleen, noradrenergic nerves only ran close to T cells at the boundary of the periarterial lymphatic sheath and arteries. In porcine spleen, noradrenergic nerves were closely associated with T cells in both white and red pulp as well as other leukocytes in red pulp. CONCLUSION Sympathetic innervation of the spleen varies between species in both distribution and abundance, with humans and pigs being at opposite extremes. This has important implications for sympathetic regulation of neuroimmune interactions in the spleen of different species and focused targeting of the CAP in humans.
Collapse
Affiliation(s)
- Logan G. Kirkland
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Chloe G. Garbe
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Joseph Hadaya
- grid.19006.3e0000 0000 9632 6718UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA ,grid.19006.3e0000 0000 9632 6718Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA USA
| | - Paul V. Benson
- grid.265892.20000000106344187Department of Pathology, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Brant M. Wagener
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Sanjin Tankovic
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Donald B. Hoover
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA ,grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine and Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
64
|
Cirillo G, Negrete-Diaz F, Yucuma D, Virtuoso A, Korai SA, De Luca C, Kaniusas E, Papa M, Panetsos F. Vagus Nerve Stimulation: A Personalized Therapeutic Approach for Crohn's and Other Inflammatory Bowel Diseases. Cells 2022; 11:cells11244103. [PMID: 36552867 PMCID: PMC9776705 DOI: 10.3390/cells11244103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis, are incurable autoimmune diseases characterized by chronic inflammation of the gastrointestinal tract. There is increasing evidence that inappropriate interaction between the enteric nervous system and central nervous system and/or low activity of the vagus nerve, which connects the enteric and central nervous systems, could play a crucial role in their pathogenesis. Therefore, it has been suggested that appropriate neuroprosthetic stimulation of the vagus nerve could lead to the modulation of the inflammation of the gastrointestinal tract and consequent long-term control of these autoimmune diseases. In the present paper, we provide a comprehensive overview of (1) the cellular and molecular bases of the immune system, (2) the way central and enteric nervous systems interact and contribute to the immune responses, (3) the pathogenesis of the inflammatory bowel disease, and (4) the therapeutic use of vagus nerve stimulation, and in particular, the transcutaneous stimulation of the auricular branch of the vagus nerve. Then, we expose the working hypotheses for the modulation of the molecular processes that are responsible for intestinal inflammation in autoimmune diseases and the way we could develop personalized neuroprosthetic therapeutic devices and procedures in favor of the patients.
Collapse
Affiliation(s)
- Giovanni Cirillo
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Flor Negrete-Diaz
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias (IdISSC), Hospital Clinico San Carlos de Madrid, 28040 Madrid, Spain
| | - Daniela Yucuma
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Andalusian School of Public Health, University of Granada, 18011 Granada, Spain
| | - Assunta Virtuoso
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Sohaib Ali Korai
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Ciro De Luca
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | | | - Michele Papa
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy
- Correspondence: (M.P.); (F.P.)
| | - Fivos Panetsos
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias (IdISSC), Hospital Clinico San Carlos de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
- Correspondence: (M.P.); (F.P.)
| |
Collapse
|
65
|
Pooladgar P, Sakhabakhsh M, Taghva A, Soleiman-Meigooni S. Donepezil Beyond Alzheimer's Disease? A Narrative Review of Therapeutic Potentials of Donepezil in Different Diseases. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e128408. [PMID: 36942075 PMCID: PMC10024338 DOI: 10.5812/ijpr-128408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Donepezil hydrochloride is an acetylcholine esterase inhibitor studied and approved to treat Alzheimer's disease (AD). However, this drug can have positive therapeutic potential in treating different conditions, including various neurodegenerative disorders such as other types of dementia, multiple sclerosis, Parkinson's disease, psychiatric and mood disorders, and even infectious diseases. Hence, this study reviewed the therapeutic potential of this drug in treating Alzheimer's and other diseases by reviewing the articles from databases including Web of Science, Scopus, PubMed, Cochrane, and Science Direct. It was shown that donepezil could affect the pathophysiology of these diseases via mechanisms such as increasing the concentration of acetylcholine, modulating local and systemic inflammatory processes, affecting acetylcholine receptors like nicotinic and muscarinic receptors, and activating various cellular signaling via receptors like sigma-1 receptors. Despite many therapeutic potentials, this drug has not yet been approved for treating non-Alzheimer's diseases, and more comprehensive studies are needed.
Collapse
Affiliation(s)
- Parham Pooladgar
- Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Sakhabakhsh
- Head of Department of Neurology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Arsia Taghva
- Department of Psychiatry, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
66
|
McCamy KM, Rees KA, Winzer-Serhan UH. Peripheral immune challenges elicit differential up-regulation of hippocampal cytokine and chemokine mRNA expression in a mouse model of the 15q13.3 microdeletion syndrome. Cytokine 2022; 159:156005. [PMID: 36084604 DOI: 10.1016/j.cyto.2022.156005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/03/2022]
Abstract
The human heterozygous 15q13.3 microdeletion is associated with neuropathological disorders, most prominently with epilepsy and intellectual disability. The 1.5 Mb deletion encompasses six genes (FAN1 [MTMR15], MTMR10, TRPM1, KLF13, OTUD7A, and CHRNA7); all but one (TRPM1) are expressed in the brain. The 15q13.3 microdeletion causes highly variable neurological symptoms, and confounding factors may contribute to a more severe phenotype. CHRNA7 and KLF13 are involved in immune system regulation and altered immune responses may contribute to neurological deficits. We used the Df[h15q13]/+ transgenic mouse model with a heterozygous deletion of the orthologous region (Het) to test the hypothesis that the microdeletion increases innate immune responses compared to wild type (WT). Male and female mice were acutely challenged with the bacteriomimetic lipopolysaccharide (LPS, 0.1 mg/kg, i.p.) or the viral mimetic polyinosinic:polycytidylic acid (Poly(I:C), 5 mg/kg). Hippocampal mRNA expression of pro-inflammatory cytokines and chemokines were determined three hours after injection using quantitative PCR analysis. In controls, expression was not affected by sex or genotype. LPS and Poly(I:C) resulted in significantly increased hippocampal expression of cytokines, chemokines, and interferon-γ (IFNγ), with more robust increases for TNF-α, IL-6, IL-1β, CXCL1, and CCL2 by LPS, higher induction of IFNγ by Poly(I:C), and similar increases of CCL4 and CCL5 by both agents. Generally, Hets exhibited stronger responses than WT mice, and significant effects of genotype or genotype × treatment interactions were detected for CXCL1 and CCL5, and IL-6, IL-1β, and CCL4, respectively, after LPS. Sex differences were detected for some targets. LPS but not Poly(I:C), reduced overnight burrowing independent of sex or genotype, suggesting that LPS induced sickness behavior. Thus, mice carrying the microdeletion have an increased innate immune response following a LPS challenge, but further studies will have to determine the extent and mechanisms of altered immune activation and subsequent contributions to 15q13.3 microdeletion associated deficits.
Collapse
Affiliation(s)
- Kristin M McCamy
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Katherine A Rees
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Ursula H Winzer-Serhan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States.
| |
Collapse
|
67
|
Parasympathetic-macrophages-ductal epithelial cells axis promotes female rat submandibular gland regeneration after excretory duct ligation/deligation. Arch Oral Biol 2022; 145:105586. [DOI: 10.1016/j.archoralbio.2022.105586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
|
68
|
Roles of alpha-7 nicotinic acetylcholine receptors and spleen in the lung injury induced by a repeated saline lavage in rat. BMC Pulm Med 2022; 22:367. [PMID: 36167538 PMCID: PMC9513867 DOI: 10.1186/s12890-022-02151-3] [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: 05/06/2022] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The study aimed to determine whether or notα7 nicotinic acetylcholine receptors (α7nAChR) induce anti-inflammatory effects directly in the lung or through the spleen pathway in a sterile model of lung injury by saline lavage. METHODS Male Sprague Dawley rats were divided into seven groups; Sham, splenectomy (SPX), saline lavage (LAV), LAV treated with α7nAChR agonist nicotine (LAV + NIC), and LAV treated with NIC and a selective α7nAChR antagonist MLA (LAV+MLA+NIC), LAV and splenectomy (LAV+SPX), and LAV+SPX treated with nicotine (LAV+SPX+NIC). Tracheostomy and catheterization of the femoral artery were performed under deep anesthesia. Animals were subjected to volume-controlled ventilation and lung injury by 10 repeated saline lavages. Splenectomy was achieved one week before the induction of lung injury. The recovery phase lasted for 3 h, and drugs were injected 1 h after the last lavage. RESULTS Mean arterial blood pressure (MBP), heart rate (HR), PaO2, PaO2/FiO2 ratio, and pH decreased, whereas, maximal inspiratory (MIP) and expiratory (MEP) pressures, and PaCO2 increased 1 h after the saline lavage. Nicotine corrected entirely all the above parameters in the LAV + NIC group. MLA or SPX prevented the effects of nicotine on the above parameters, except that MLA had no extra effect on MIP or MEP. In addition, nicotine improved lung compliance in the LAV + NIC and LAV + SPX + NIC groups, though it was inhibited by MLA in the LAV + MLA + NIC group. The increases of plasma and lung tissue malondialdehyde (MDA) in the LAV group were diminished by nicotine, whereas, MLA and SPX prevented these reductions. Besides, nicotine could reduce plasma MDA in the LAV + SPX + NIC group. Total BAL cell count, protein BAL/protein plasma ratio, and lung histological scores were attenuated by nicotine in the LAV + NIC group, whereas, MLA reversed the mentioned alterations in the LAV + MLA + NIC group. However, splenectomy could not stop the decreasing effect of nicotine on the total BAL cell in the LAV + SPX + NIC group. CONCLUSIONS In this study, we indicated that α7nAChR and spleen play roles in cholinergic anti-inflammatory pathways in saline lavage-induced lung injury. However, our results are in favor of at least some direct effects of α 7nAChR in the lung.
Collapse
|
69
|
Wu F, Liu Z, Zhou L, Ye D, Zhu Y, Huang K, Weng Y, Xiong X, Zhan R, Shen J. Systemic immune responses after ischemic stroke: From the center to the periphery. Front Immunol 2022; 13:911661. [PMID: 36211352 PMCID: PMC9533176 DOI: 10.3389/fimmu.2022.911661] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/18/2022] [Indexed: 12/01/2022] Open
Abstract
Ischemic stroke is a leading cause of disability and death. It imposes a heavy economic burden on individuals, families and society. The mortality rate of ischemic stroke has decreased with the help of thrombolytic drug therapy and intravascular intervention. However, the nerve damage caused by ischemia-reperfusion is long-lasting and followed by multiple organ dysfunction. In this process, the immune responses manifested by systemic inflammatory responses play an important role. It begins with neuroinflammation following ischemic stroke. The large number of inflammatory cells released after activation of immune cells in the lesion area, along with the deactivated neuroendocrine and autonomic nervous systems, link the center with the periphery. With the activation of systemic immunity and the emergence of immunosuppression, peripheral organs become the second “battlefield” of the immune response after ischemic stroke and gradually become dysfunctional and lead to an adverse prognosis. The purpose of this review was to describe the systemic immune responses after ischemic stroke. We hope to provide new ideas for future research and clinical treatments to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Fan Wu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zongchi Liu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lihui Zhou
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Di Ye
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zhu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kaiyuan Huang
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yuxiang Weng
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxing Xiong
- Department of Clinical Laboratory, Renmin Hospital, Faculty of Medical Sciences, Wuhan University, Wuhan, China
| | - Renya Zhan
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Jian Shen, ; Renya Zhan,
| | - Jian Shen
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Jian Shen, ; Renya Zhan,
| |
Collapse
|
70
|
Espeter F, Künne D, Garczarek L, Kuhlmann H, Skarabis A, Zivkovic AR, Brenner T, Schmidt K. Critically Ill COVID-19 Patients Show Reduced Point of Care-Measured Butyrylcholinesterase Activity—A Prospective, Monocentric Observational Study. Diagnostics (Basel) 2022; 12:diagnostics12092150. [PMID: 36140551 PMCID: PMC9498245 DOI: 10.3390/diagnostics12092150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 01/08/2023] Open
Abstract
A biomarker for risk stratification and disease severity assessment in SARS-CoV-2 infections has not yet been established. Point of care testing (POCT) of butyrylcholinesterase (BChE) enables early detection of systemic inflammatory responses and correlates with disease severity in sepsis and burns. In acute care or resource-limited settings, POCT facilitates rapid clinical decision making, a particularly beneficial aspect in the management of pandemic situations. In this prospective observational study, POCT-measured BChE activity was assessed in 52 critically ill COVID-19 patients within 24 h of ICU admission and on the third and seventh day after ICU admission. Forty (77%) of these patients required venovenous extracorporeal membrane oxygenation (vvECMO). In critically ill COVID-19 patients, BChE activity is significantly decreased compared with healthy subjects, but also compared with other inflammatory conditions such as sepsis, burns, or trauma. POCT BChE activity reflects the severity of organ dysfunction and allows prediction of 28-day mortality in critically ill COVID-19 patients. Implementing early POCT BChE measurement could facilitate risk stratification and support admission and transfer decisions in resource-limited settings.
Collapse
Affiliation(s)
- Florian Espeter
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-84485
| | - David Künne
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Lena Garczarek
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Henning Kuhlmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Annabell Skarabis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | | | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Karsten Schmidt
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
71
|
Azam S, Kim YS, Jakaria M, Yu YJ, Ahn JY, Kim IS, Choi DK. Dioscorea nipponica Makino Rhizome Extract and Its Active Compound Dioscin Protect against Neuroinflammation and Scopolamine-Induced Memory Deficits. Int J Mol Sci 2022; 23:ijms23179923. [PMID: 36077321 PMCID: PMC9456145 DOI: 10.3390/ijms23179923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Activation of microglial cells by intrinsic or extrinsic insult causes neuroinflammation, a common phenomenon in neurodegenerative diseases. Prevention of neuroinflammation may ameliorate many neurodegenerative disease progressions. Dioscorea nipponica Makino (DN) extract can alleviate muscular atrophy and inflammatory diseases; however, the efficacy and mechanism of action in microglial cells remain unknown. The current study investigates the possible anti-inflammatory effects and mechanisms of Dioscorea nipponica Makino ethanol extract and its steroidal saponin dioscin. Our in vitro study shows that Dioscorea nipponica rhizome ethanol extract (DNRE) and dioscin protect against lipopolysaccharide (LPS)-activated inflammatory responses in BV-2 microglial cells by inhibiting phosphorylation and the nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), resulting in the downregulation of pro-inflammatory cytokines and enzymes. Consistent with our previous report of dioscin-mediated enhancement of neurotrophic factors in dopaminergic cells, here we found that dioscin upregulates brain-derived neurotrophic factor (BDNF) and cAMP-response element binding protein (CREB) phosphorylation (pCREB) in the cerebral cortex and hippocampus regions of the mouse brain. Scopolamine treatment increased pro-inflammatory enzyme levels and reduced the expression of BDNF and pCREB in the hippocampus and cortex regions, which led to impaired learning and referencing memory in mice. Pre-treatment of dioscin for 7 days substantially enhanced mice performances in maze studies, indicating amelioration in cognitive deficits. In conclusion, DNRE and its active compound dioscin protect against neurotoxicity most likely by suppressing NF-κB phosphorylation and upregulating neurotrophic factor BDNF.
Collapse
Affiliation(s)
- Shofiul Azam
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
| | - Yon-Suk Kim
- BKplus GLOCAL Education Program of Nutraceuticals Development, Konkuk University, Chungju 27478, Korea
| | - Md. Jakaria
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ye-Ji Yu
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
| | - Jae-Yong Ahn
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
| | - In-Su Kim
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
| | - Dong-Kug Choi
- BK21 Program, Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju 27478, Korea
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: ; Tel.: +82-43-840-3610; Fax: +82-43-840-3872
| |
Collapse
|
72
|
Yu J, Zhang RF, Mao YL. Cerebellar fastigial nucleus electrostimulation attenuates inflammation in a Post-Infarction rat model by activating cholinergic anti-inflammatory pathways. Neurosci Lett 2022; 788:136860. [PMID: 36041546 DOI: 10.1016/j.neulet.2022.136860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
There are negative correlations between indices of heart rate variability (HRV) and markers of inflammation. The inflammation plays an important role in myocardial damages after myocardial infarction (MI). Our previous study found that fastigial nucleus electrostimulation (FNS) improved abnormal HRV in a rat model of MI. Whether and how it can reduce inflammation and improve cardiac function after MI and the underlying mechanisms remain unknown. 66 Sprague Dawley rats were randomly divided into 4 groups as follows: i) Sham operation group (Sham); ii) Myocardial infarction group (MI); iii) FNS+MI group (FNS plus MI): left fastigial nucleus electrostimulation; iv) FNL+FNS+MI group (left fastigial nucleus lesion plus FNS plus MI). The serum expressions of acetylcholine (ACh), pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and anti-inflammatory cytokines IL-10 after FNS were measured using ELISA. Subsequently, the infarct size, the infiltration of inflammatory cells, the fibrotic area, and cardiac function were also evaluated. Additionally, the effects of FNS on the cholinergic anti-inflammatory pathway (CAP)-related proteins expression were determined by Western blot. We found that FNS significantly upregulated ACh and IL-10 expressions in serum, and decreased TNF-α and IL-6 levels. FNS significantly attenuated inflammatory cell infiltration and infarct size, decreased fibrosis, increased left ventricular ejection fraction (LVEF), and reduced mortality. Besides, the levels of p-STAT3/STAT3 and p-NF-κB/NF-κB significantly elevated after MI. FNS down-regulated the expression of p-STAT3/STAT3 and p-NF-κB/NF-κB. The protective effects of FNS were partially reversed by the fastigial nucleus lesion. Our data suggested that FNS can alleviate the inflammation after MI, and its cardiac neuroprotective mechanism may be achieved by increasing vagus tone, releasing ACh, and further activating the CAP via α7nAChR. The precise mechanism remains to be elucidated.
Collapse
Affiliation(s)
- Jiang Yu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China; Department of Cardiology, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang 621000, Sichuan, China
| | - Run-Feng Zhang
- Department of Cardiology, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang 621000, Sichuan, China.
| | - Yi-Li Mao
- Department of Cardiology, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang 621000, Sichuan, China
| |
Collapse
|
73
|
Sepsis-Induced Brain Dysfunction: Pathogenesis, Diagnosis, and Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1328729. [PMID: 36062193 PMCID: PMC9433216 DOI: 10.1155/2022/1328729] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/30/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Dysregulated host response to infection, which cause life-threatening organ dysfunction, was defined as sepsis. Sepsis can cause acute and long-term brain dysfunction, namely, sepsis-associated encephalopathy (SAE) and cognitive impairment. SAE refers to changes in consciousness without direct evidence of central nervous system infection. It is highly prevalent and may cause poor outcomes in sepsis patients. Cognitive impairment seriously affects the life quality of sepsis patients and increases the medical burden. The pathogenesis of sepsis-induced brain dysfunction is mainly characterized by the interaction of systemic inflammation, blood-brain barrier (BBB) dysfunction, neuroinflammation, microcirculation dysfunction, and brain dysfunction. Currently, the diagnosis of sepsis-induced brain dysfunction is based on clinical manifestation of altered consciousness along with neuropathological examination, and the treatment is mainly involves controlling sepsis. Although treatments for sepsis-induced brain dysfunction have been tested in animals, clinical treat sepsis-induced brain dysfunction is still difficult. Therefore, we review the underlying mechanisms of sepsis-induced brain injury, which mainly focus on the influence of systemic inflammation on BBB, neuroinflammation, brain microcirculation, and the brain function, which want to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating brain dysfunction.
Collapse
|
74
|
Effect of Vagus Nerve Stimulation on Blood Inflammatory Markers in Children with Drug-Resistant Epilepsy: A Pilot Study. CHILDREN 2022; 9:children9081133. [PMID: 36010024 PMCID: PMC9406968 DOI: 10.3390/children9081133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/20/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022]
Abstract
Background: Since one of the suggested mechanisms of action of VNS on epilepsy is the reduction of central inflammation, we carried out a comprehensive analysis of blood inflammatory markers in children considered for VNS surgery. Materials and methods: Five pediatric patients were studied. An extensive analysis of blood inflammatory markers was performed before surgery (T0) and six weeks after VNS implantation (T1). An epileptological outcome was obtained according to the McHugh score. Results: The variations of IgA, IgE, IgG, CD19, and PTX3 displayed a tendency toward a positive statistical correlation between T0 and T1. According to McHugh score, the patients were divided into Group 1 (i.e., Class I) and Group 2 (i.e., Classes II and III). IL-1β and PTX-3 tended to decrease more in Group 1, while TNF-α decreased in Group 2 (−56.65%) and slightly increased (+3.61%) in Group 1 at T1 without statistical correlation. Conclusions: The variation of IL-1β and PTX-3 seem to be related to a better outcome; thus, they do not reach statistical significance. A larger series of patients is needed to determine whether biochemical changes could relay with the clinical improvement of epilepsy.
Collapse
|
75
|
Czura CJ, Bikson M, Charvet L, Chen JDZ, Franke M, Fudim M, Grigsby E, Hamner S, Huston JM, Khodaparast N, Krames E, Simon BJ, Staats P, Vonck K. Neuromodulation Strategies to Reduce Inflammation and Improve Lung Complications in COVID-19 Patients. Front Neurol 2022; 13:897124. [PMID: 35911909 PMCID: PMC9329660 DOI: 10.3389/fneur.2022.897124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022] Open
Abstract
Since the outbreak of the COVID-19 pandemic, races across academia and industry have been initiated to identify and develop disease modifying or preventative therapeutic strategies has been initiated. The primary focus has been on pharmacological treatment of the immune and respiratory system and the development of a vaccine. The hyperinflammatory state ("cytokine storm") observed in many cases of COVID-19 indicates a prognostically negative disease progression that may lead to respiratory distress, multiple organ failure, shock, and death. Many critically ill patients continue to be at risk for significant, long-lasting morbidity or mortality. The human immune and respiratory systems are heavily regulated by the central nervous system, and intervention in the signaling of these neural pathways may permit targeted therapeutic control of excessive inflammation and pulmonary bronchoconstriction. Several technologies, both invasive and non-invasive, are available and approved for clinical use, but have not been extensively studied in treatment of the cytokine storm in COVID-19 patients. This manuscript provides an overview of the role of the nervous system in inflammation and respiration, the current understanding of neuromodulatory techniques from preclinical and clinical studies and provides a rationale for testing non-invasive neuromodulation to modulate acute systemic inflammation and respiratory dysfunction caused by SARS-CoV-2 and potentially other pathogens. The authors of this manuscript have co-founded the International Consortium on Neuromodulation for COVID-19 to advocate for and support studies of these technologies in the current coronavirus pandemic.
Collapse
Affiliation(s)
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, New York, NY, United States
| | - Leigh Charvet
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
| | - Jiande D. Z. Chen
- Division of Gastroenterology and Hepatology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | | | - Marat Fudim
- Division of Cardiology, Duke Clinical Research Institute, Duke University, Durham, NC, United States
| | | | - Sam Hamner
- Cala Health, Burlingame, CA, United States
| | - Jared M. Huston
- Departments of Surgery and Science Education, Zucker School of Medicine at Hofstra/Northwell, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | | | - Elliot Krames
- Pacific Pain Treatment Center, Napa, CA, United States
| | | | - Peter Staats
- National Spine and Pain, ElectroCore, Inc., Jacksonville, FL, United States
| | - Kristl Vonck
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
76
|
Special Issue: Cholinergic Control of Inflammation. Int J Mol Sci 2022; 23:ijms23147758. [PMID: 35887105 PMCID: PMC9319851 DOI: 10.3390/ijms23147758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Inflammation caused by infection, tissue trauma, and disease states such as arthritis and inflammatory bowel disease is perceived by the Central nervous System (CNS) through different routes that, by means of neural reflex circuits, regulate the immune system response [...]
Collapse
|
77
|
Ahmad F. Medicinal nicotine in COVID-19 acute respiratory distress syndrome, the new corticosteroid. World J Crit Care Med 2022; 11:228-235. [PMID: 36051943 PMCID: PMC9305679 DOI: 10.5492/wjccm.v11.i4.228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/23/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023] Open
Abstract
The cholinergic anti-inflammatory pathway (CAP) refers to the anti-inflammatory effects mediated by the parasympathetic nervous system. Existence of this pathway was first demonstrated when acetylcholinesterase inhibitors showed benefits in animal models of sepsis. CAP functions via the vagus nerve. The systemic anti-inflammatory effects of CAP converges on the α7 nicotinic acetylcholine receptor on splenic macrophages, leading to suppression of pro-inflammatory cytokines and simultaneous stimulation of anti-inflammatory cytokines, including interleukin 10. CAP offers a novel mechanism to mitigate inflammation. Electrical vagal nerve stimulation has shown benefits in patients suffering from rheumatoid arthritis. Direct agonists like nicotine and GTS-1 have also demonstrated anti-inflammatory properties in models of sepsis and acute respiratory distress syndrome, as have acetylcholinesterase inhibitors like Galantamine and Physostigmine. Experience with coronavirus disease 2019 (COVID-19) induced acute respiratory distress syndrome indicates that immunomodulators have a protective role in patient outcomes. Dexamethasone is the only medication currently in use that has shown to improve clinical outcomes. This is likely due to the suppression of what is referred to as a cytokine storm, which is implicated in the lethality of viral pneumonia. Nicotine transdermal patch activates CAP and harvests its anti-inflammatory potential by means of an easily administered depot delivery mechanism. It could prove to be a promising, safe and inexpensive additional tool in the currently limited armamentarium at our disposal for management of COVID-19 induced acute hypoxic respiratory failure.
Collapse
Affiliation(s)
- Farrukh Ahmad
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA 01608, United States
| |
Collapse
|
78
|
Zhu L, Huang L, Le A, Wang TJ, Zhang J, Chen X, Wang J, Wang J, Jiang C. Interactions between the Autonomic Nervous System and the Immune System after Stroke. Compr Physiol 2022; 12:3665-3704. [PMID: 35766834 DOI: 10.1002/cphy.c210047] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute stroke is one of the leading causes of morbidity and mortality worldwide. Stroke-induced immune-inflammatory response occurs in the perilesion areas and the periphery. Although stroke-induced immunosuppression may alleviate brain injury, it hinders brain repair as the immune-inflammatory response plays a bidirectional role after acute stroke. Furthermore, suppression of the systemic immune-inflammatory response increases the risk of life-threatening systemic bacterial infections after acute stroke. Therefore, it is essential to explore the mechanisms that underlie the stroke-induced immune-inflammatory response. Autonomic nervous system (ANS) activation is critical for regulating the local and systemic immune-inflammatory responses and may influence the prognosis of acute stroke. We review the changes in the sympathetic and parasympathetic nervous systems and their influence on the immune-inflammatory response after stroke. Importantly, this article summarizes the mechanisms on how ANS regulates the immune-inflammatory response through neurotransmitters and their receptors in immunocytes and immune organs after stroke. To facilitate translational research, we also discuss the promising therapeutic approaches modulating the activation of the ANS or the immune-inflammatory response to promote neurologic recovery after stroke. © 2022 American Physiological Society. Compr Physiol 12:3665-3704, 2022.
Collapse
Affiliation(s)
- Li Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Anh Le
- Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xuemei Chen
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.,Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
79
|
Luo Q, Ma H, Guo E, Yu L, Jia L, Zhang B, Feng G, Liu R. MicroRNAs Promote the Progression of Sepsis-Induced Cardiomyopathy and Neurovascular Dysfunction Through Upregulation of NF-kappaB Signaling Pathway-Associated HDAC7/ACTN4. Front Neurol 2022; 13:909828. [PMID: 35756932 PMCID: PMC9218607 DOI: 10.3389/fneur.2022.909828] [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: 03/31/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction The objective of this study was to determine the NF-kappaB pathway, hub genes, and transcription factors (TFs) in monocytes implicated in the progression of neurovascular-related sepsis-induced cardiomyopathy (SIC) as well as potential miRNAs with regulatory functions. Methods : Sepsis-induced cardiomyopathy—and heart failure (HF)-related differentially expressed genes (DEGs) between SIC and HF groups were identified separately by differential analysis. In addition, DEGs and differentially expressed miRNAs (DEmiRNAs) in monocytes between sepsis and the HC group were identified. Then, common DEGs in SIC, HF, and monocyte groups were identified by intersection analysis. Based on the functional pathways enriched by these DEGs, genes related to the NF-kB-inducing kinase (NIK)/NF-kappaB signaling pathway were selected for further intersection analysis to obtain hub genes. These common DEGs, together with sepsis-related DEmiRNAs, were used to construct a molecular interplay network and to identify core TFs in the network. Results : A total of 153 upregulated genes and 25 downregulated genes were obtained from SIC-, HF-, and monocyte-related DEGs. Functional pathway analysis revealed that the upregulated genes were enriched in NF-κB signaling pathway. A total of eight genes associated with NF-κB signaling pathway were then further identified from the 178 DEGs. In combination with sepsis-related DEmiRNAs, HDAC7/ACTN4 was identified as a key transcriptional regulatory pair in the progression of SIC and in monocyte regulation. hsa-miR-23a-3p, hsa-miR-3175, and hsa-miR-23b-3p can regulate the progression of SIC through the regulation of HDAC7/ACTN4. Finally, gene set enrichment analysis (GSEA) suggested that HDAC7/ACTN4 may be associated with apoptosis in addition to the inflammatory response. Conclusion : hsa-miR-23a-3p, hsa-miR-3175, and hsa-miR-23b-3p are involved in SIC progression by regulating NF-κB signaling signaling pathway-related HDAC7/ACTN4 in monocytes and cardiac tissue cells. These mechanisms may contribute to sepsis-induced neurovascular damage.
Collapse
Affiliation(s)
- Qiancheng Luo
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Hanning Ma
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Shanghai, China
| | - Enwei Guo
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Lin Yu
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Ling Jia
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Bingyu Zhang
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Gang Feng
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Rui Liu
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| |
Collapse
|
80
|
Chen Z, Fu L, Liu XA, Yang Z, Li W, Li F, Luo Q. Real-time effects of nicotine exposure and withdrawal on neurotransmitter metabolism of hippocampal neuronal cells by microfluidic chip-coupled LC-MS. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.09.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
81
|
Jankauskaite L, Malinauskas M, Mickeviciute GC. HMGB1: A Potential Target of Nervus Vagus Stimulation in Pediatric SARS-CoV-2-Induced ALI/ARDS. Front Pediatr 2022; 10:884539. [PMID: 35633962 PMCID: PMC9132499 DOI: 10.3389/fped.2022.884539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Abstract
From the start of pandemics, children were described as the ones who were less affected by SARS-Cov-2 or COVID-19, which was mild in most of the cases. However, with the growing vaccination rate of the adult population, children became more exposed to the virus and more cases of severe SARS-CoV-2-induced ARDS are being diagnosed with the disabling consequences or lethal outcomes associated with the cytokine storm. Thus, we do hypothesize that some of the children could benefit from nervus vagus stimulation during COVID-19 ARDS through the inhibition of HMGB1 release and interaction with the receptor, resulting in decreased neutrophil accumulation, oxidative stress, and coagulopathy as well as lung vascular permeability. Moreover, stimulation through alpha-7 nicotinic acetylcholine receptors could boost macrophage phagocytosis and increase the clearance of DAMPs and PAMPs. Further rise of FGF10 could contribute to lung stem cell proliferation and potential regeneration of the injured lung. However, this stimulation should be very specific, timely, and of proper duration, as it could lead to such adverse effects as increased viral spread and systemic infection, especially in small children or infants due to specific pediatric immunity state and anatomical features of the respiratory system.
Collapse
Affiliation(s)
- Lina Jankauskaite
- Lithuanian University of Health Sciences, Medical Academy, Pediatric Department, Kaunas, Lithuania
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
| | - Mantas Malinauskas
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
| | - Goda-Camille Mickeviciute
- Lithuanian University of Health Sciences, Medical Academy, Pediatric Department, Kaunas, Lithuania
- Lithuanian University of Health Sciences, Medical Academy, Institute of Physiology and Pharmacology, Kaunas, Lithuania
- Rehabilitation Center “Palangos Linas”, Palanga, Lithuania
| |
Collapse
|
82
|
Keasey MP, Lovins C, Jia C, Hagg T. Liver vitronectin release into the bloodstream increases due to reduced vagal muscarinic signaling after cerebral stroke in female mice. Physiol Rep 2022; 10:e15301. [PMID: 35531929 PMCID: PMC9082388 DOI: 10.14814/phy2.15301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022] Open
Abstract
Vitronectin (VTN) is a glycoprotein enriched in the blood and activates integrin receptors. VTN blood levels increase only in female mice 24 h after an ischemic stroke and exacerbate brain injury through IL-6-driven inflammation, but the VTN induction mechanism is unknown. Here, a 30 min middle cerebral artery occlusion (MCAO) in female mice induced VTN protein in the liver (normally the main source) in concert with plasma VTN. Male mice were excluded as VTN is not induced after stroke. MCAO also increased plasma VTN levels after de novo expression of VTN in the liver of VTN-/- female mice, using a hepatocyte-specific (SERPINA1) promoter. MCAO did not affect SERPINA1 or VTN mRNA in the liver, brain, or several peripheral organs, or platelet VTN, compared to sham mice. Thus, hepatocytes are the source of stroke-induced increases in plasma VTN, which is independent of transcription. The cholinergic innervation by the parasympathetic vagus nerve is a potential source of brain-liver signaling after stroke. Right-sided vagotomy at the cervical level led to increased plasma VTN levels, suggesting that VTN release is inhibited by vagal tone. Co-culture of hepatocytes with cholinergic neurons or treatment with acetylcholine, but not noradrenaline (sympathetic transmitter), suppressed VTN expression. Hepatocytes have muscarinic receptors and the M1/M3 agonist bethanechol decreased VTN mRNA and protein release in vitro via M1 receptors. Finally, systemic bethanechol treatment blocked stroke-induced plasma VTN. Thus, VTN translation and release are inhibited by muscarinic signaling from the vagus nerve and presents a novel target for lessening detrimental VTN expression.
Collapse
Affiliation(s)
- Matthew P. Keasey
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Chiharu Lovins
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Cuihong Jia
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Theo Hagg
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| |
Collapse
|
83
|
Long L, Zang Q, Jia G, Fan M, Zhang L, Qi Y, Liu Y, Yu L, Wang S. Transcutaneous Auricular Vagus Nerve Stimulation Promotes White Matter Repair and Improves Dysphagia Symptoms in Cerebral Ischemia Model Rats. Front Behav Neurosci 2022; 16:811419. [PMID: 35493949 PMCID: PMC9051615 DOI: 10.3389/fnbeh.2022.811419] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
Background Clinical and animal studies have shown that transcutaneous auricular vagus nerve stimulation (ta-VNS) exerts neuroprotection following cerebral ischemia. Studies have revealed that white matter damage after ischemia is related to swallowing defects, and the degree of white matter damage is related to the severity of dysphagia. However, the effect of ta-VNS on dysphagia symptoms and white matter damage in dysphagic animals after an ischemic stroke has not been investigated. Methods Middle cerebral artery occlusion (MCAO) rats were randomly divided into the sham, control and vagus nerve stimulation (VNS) group, which subsequently received ta-VNS for 3 weeks. The swallowing reflex was measured once weekly by electromyography (EMG). White matter remyelination, volume, angiogenesis and the inflammatory response in the white matter were assessed by electron microscopy, immunohistochemistry, stereology, enzyme-linked immunosorbent assay (ELISA) and Western blotting. Results ta-VNS significantly increased the number of swallows within 20 s and reduced the onset latency to the first swallow. ta-VNS significantly improved remyelination but did not alleviate white matter shrinkage after MCAO. Stereology revealed that ta-VNS significantly increased the density of capillaries and increased vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF2) expression in the white matter. ta-VNS significantly alleviated the increase inTLR4, MyD88, phosphorylated MAPK and NF-κB protein levels and suppressed the expression of the proinflammatory factors IL-1β and TNF-α. Conclusion These results indicated ta-VNS slightly improved dysphagia symptoms after ischemic stroke, possibly by increasing remyelination, inducing angiogenesis, and inhibiting the inflammatory response in the white matter of cerebral ischaemia model rats, implying that ta-VNS may be an effective therapeutic strategy for the treatment of dysphagia after ischemic stroke.
Collapse
Affiliation(s)
- Lu Long
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianwen Zang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gongwei Jia
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Fan
- Department of Traditional Chinese Medicine, Weinan Central Hospital, Weinan, China
| | - Liping Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingqiang Qi
- Center of Electron Microscope, Institute of Life Science of Chongqing Medical University, Chongqing, China
| | - Yilin Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sanrong Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Sanrong Wang
| |
Collapse
|
84
|
Burzynski H, Macht V, Woodruff J, Crawford J, Erichsen J, Piroli G, Grillo C, Fadel J, Reagan L. Pyridostigmine bromide elicits progressive and chronic impairments in the cholinergic anti-inflammatory pathway in the prefrontal cortex and hippocampus of male rats. Neurobiol Stress 2022; 18:100446. [PMID: 35573808 PMCID: PMC9095881 DOI: 10.1016/j.ynstr.2022.100446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Gulf War Illness (GWI) is a multi-symptom illness that continues to affect over 250,000 American Gulf War veterans. The causes of GWI remain equivocal; however, prophylactic use of the acetylcholinesterase inhibitor pyridostigmine bromide (PB), and the stress of combat have been identified as two potential causative factors. Both PB and stress alter acetylcholine (ACh), which mediates both cognition and anti-inflammatory responses. As inflammation has been proposed to contribute to the cognitive deficits and immune dysregulation in GWI, the goal of this study was to determine the long-term effects of PB and stress on the cholinergic anti-inflammatory pathway in the central nervous system and periphery. We used our previously established rat model of GWI and in vivo microdialysis to assess cholinergic neurochemistry in the prefrontal cortex (PFC) and hippocampus following a mild immune challenge (lipopolysaccharide; LPS). We then examined LPS-induced changes in inflammatory markers in PFC and hippocampal homogenates. We found that PB treatment produces a long-lasting potentiation of the cholinergic response to LPS in both the PFC and hippocampus. Interestingly, this prolonged effect of PB treatment enhancing cholinergic responses to LPS was accompanied by paradoxical increases in the release of pro-inflammatory cytokines in these brain regions. Collectively, these findings provide evidence that neuroinflammation resulting from dysregulation of the cholinergic anti-inflammatory pathway is a mechanistic mediator in the progression of the neurochemical and neurocognitive deficits in GWI and more broadly suggest that dysregulation of this pathway may contribute to neuroinflammatory processes in stress-related neurological disorders. Inflammation is thought to contribute to the progressive nature of GWI pathology. PB potentiates the central cholinergic response to LPS over time in model of GWI. PB progressively exacerbates the neuroinflammatory response to LPS. GWI may result from the dysregulation of the cholinergic anti-inflammatory pathway.
Collapse
|
85
|
Pattanaik B, Hammarlund M, Mjörnstedt F, Ulleryd MA, Zhong W, Uhlén M, Gummesson A, Bergström G, Johansson ME. Polymorphisms in alpha 7 nicotinic acetylcholine receptor gene, CHRNA7, and its partially duplicated gene, CHRFAM7A, associate with increased inflammatory response in human peripheral mononuclear cells. FASEB J 2022; 36:e22271. [PMID: 35344211 DOI: 10.1096/fj.202101898r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/14/2022] [Accepted: 03/11/2022] [Indexed: 01/16/2023]
Abstract
The vagus nerve can, via the alpha 7 nicotinic acetylcholine receptor (α7nAChR), regulate inflammation. The gene coding for the α7nAChR, CHRNA7, can be partially duplicated, that is, CHRFAM7A, which is reported to impair the anti-inflammatory effect mediated via the α7nAChR. Several single nucleotide polymorphisms (SNPs) have been described in both CHRNA7 and CHRFAM7A, however, the functional role of these SNPs for immune responses remains to be investigated. In the current study, we set out to investigate whether genetic variants of CHRNA7 and CHRFAM7A can influence immune responses. By investigating data available from the Swedish SciLifeLab SCAPIS Wellness Profiling (S3WP) study, in combination with droplet digital PCR and freshly isolated PBMCs from the S3WP participants, challenged with lipopolysaccharide (LPS), we show that CHRNA7 and CHRFAM7A are expressed in human PBMCs, with approximately four times higher expression of CHRFAM7A compared with CHRNA7. One SNP in CHRFAM7A, rs34007223, is positively associated with hsCRP in healthy individuals. Furthermore, gene ontology (GO)-terms analysis of plasma proteins associated with gene expression of CHRNA7 and CHRFAM7A demonstrated an involvement for these genes in immune responses. This was further supported by in vitro data showing that several SNPs in both CHRNA7 and CHRFAM7A are significantly associated with cytokine response. In conclusion, genetic variants of CHRNA7 and CHRFAM7A alters cytokine responses. Furthermore, given that CHRFAM7A SNP rs34007223 is associated with inflammatory marker hsCRP in healthy individuals suggests that CHRFAM7A may have a more pronounced role in regulating inflammatory processes in humans than previously been recognized.
Collapse
Affiliation(s)
- Bagmi Pattanaik
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria Hammarlund
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Filip Mjörnstedt
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marcus A Ulleryd
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, Sweden
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
86
|
Di Lascio S, Fornasari D, Benfante R. The Human-Restricted Isoform of the α7 nAChR, CHRFAM7A: A Double-Edged Sword in Neurological and Inflammatory Disorders. Int J Mol Sci 2022; 23:ijms23073463. [PMID: 35408823 PMCID: PMC8998457 DOI: 10.3390/ijms23073463] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
CHRFAM7A is a relatively recent and exclusively human gene arising from the partial duplication of exons 5 to 10 of the α7 neuronal nicotinic acetylcholine receptor subunit (α7 nAChR) encoding gene, CHRNA7. CHRNA7 is related to several disorders that involve cognitive deficits, including neuropsychiatric, neurodegenerative, and inflammatory disorders. In extra-neuronal tissues, α7nAChR plays an important role in proliferation, differentiation, migration, adhesion, cell contact, apoptosis, angiogenesis, and tumor progression, as well as in the modulation of the inflammatory response through the “cholinergic anti-inflammatory pathway”. CHRFAM7A translates the dupα7 protein in a multitude of cell lines and heterologous systems, while maintaining processing and trafficking that are very similar to the full-length form. It does not form functional ion channel receptors alone. In the presence of CHRNA7 gene products, dupα7 can assemble and form heteromeric receptors that, in order to be functional, should include at least two α7 subunits to form the agonist binding site. When incorporated into the receptor, in vitro and in vivo data showed that dupα7 negatively modulated α7 activity, probably due to a reduction in the number of ACh binding sites. Very recent data in the literature report that the presence of the duplicated gene may be responsible for the translational gap in several human diseases. Here, we will review the studies that have been conducted on CHRFAM7A in different pathologies, with the intent of providing evidence regarding when and how the expression of this duplicated gene may be beneficial or detrimental in the pathogenesis, and eventually in the therapeutic response, to CHRNA7-related neurological and non-neurological diseases.
Collapse
Affiliation(s)
- Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milan, Italy; (S.D.L.); (D.F.)
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milan, Italy; (S.D.L.); (D.F.)
- CNR Institute of Neuroscience, 20845 Vedano al Lambro, Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milan, Italy; (S.D.L.); (D.F.)
- CNR Institute of Neuroscience, 20845 Vedano al Lambro, Italy
- NeuroMi, Milan Center for Neuroscience, University of Milano Bicocca, 20126 Milan, Italy
- Correspondence:
| |
Collapse
|
87
|
Pan S, Wu YJ, Zhang SS, Cheng XP, Olatunji OJ, Yin Q, Zuo J. The Effect of α7nAChR Signaling on T Cells and Macrophages and Their Clinical Implication in the Treatment of Rheumatic Diseases. Neurochem Res 2022; 47:531-544. [PMID: 34783974 DOI: 10.1007/s11064-021-03480-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune disease and until now, the etiology and pathogenesis of RA is not fully understood, although dysregulation of immune cells is one of the leading cause of RA-related pathological changes. Based on current understanding, the priority of anti-rheumatic treatments is to restore immune homeostasis. There are several anti-rheumatic drugs with immunomodulatory effects available nowadays, but most of them have obvious safety or efficacy shortcomings. Therefore, the development of novel anti-rheumatic drugs is still in urgently needed. Cholinergic anti-inflammatory pathway (CAP) has been identified as an important aspect of the so-called neuro-immune regulation feedback, and the interaction between acetylcholine and alpha 7 nicotinic acetylcholine receptor (α7nAChR) serves as the foundation for this signaling. Consistent to its immunomodulatory functions, α7nAChR is extensively expressed by immune cells. Accordingly, CAP activation greatly affects the differentiation and function of α7nAChR-expressing immune cells. As a result, targeting α7nAChR will bring profound therapeutic impacts on the treatment of inflammatory diseases like RA. RA is widely recognized as a CD4+ T cells-driven disease. As a major component of innate immunity, macrophages also significantly contribute to RA-related immune abnormalities. Theoretically, manipulation of CAP in immune cells is a feasible way to treat RA. In this review, we summarized the roles of different T cells and macrophages subsets in the occurrence and progression of RA, and highlighted the immune consequences of CAP activation in these cells under RA circumstances. The in-depth discussion is supposed to inspire the development of novel cell-specific CAP-targeting anti-rheumatic regimens.
Collapse
Affiliation(s)
- Shu Pan
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, China
| | - Yi-Jin Wu
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, China
| | - Sa-Sa Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, China
| | - Xiu-Ping Cheng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, China
| | - Opeyemi Joshua Olatunji
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, 90110, Thailand
| | - Qin Yin
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China.
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China.
| | - Jian Zuo
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, China.
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241000, China.
| |
Collapse
|
88
|
Weijie W, Xiaonan Y, Yilin W, Hudan P, Liang L. Study on the compatibility principle of Wutou Decoction based on network pharmacology. DIGITAL CHINESE MEDICINE 2022. [DOI: 10.1016/j.dcmed.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
89
|
Trifluoro-icaritin alleviates chronic inflammatory pain through α7nAChR-mediated suppression of HMGB1/NF-κB signaling in the spinal cord of rats. Brain Res Bull 2022; 183:13-26. [PMID: 35202753 DOI: 10.1016/j.brainresbull.2022.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 01/13/2022] [Accepted: 02/20/2022] [Indexed: 01/02/2023]
Abstract
Inflammatory pain is a chronic, persistent and serious disease that greatly impacts public health, which is often accompanied by allodynia, hyperalgesia, and spontaneous pain. It is evident that α7 nicotinic acetylcholine receptor (α7nAChR) plays a key role in cholinergic anti-inflammatory pathway and exhibits the inhibition of neuroinflammation in chronic pain. Trifluoro-icaritin (ICTF), a derivative of icaritin from the extract of a genus of Epimedium plant, is identified to possess profound anti-inflammatory activity. However, whether ICTF has anti-nociceptive effect on inflammatory pain and its potential mechanisms remain poorly elucidated. Intraperitoneal injection (i.p.) of ICTF to complete Freund's adjuvant (CFA)-induced inflammatory pain rats once daily for 21 consecutive days. Pain-related behaviors were evaluated with paw withdrawal threshold (PWT), paw withdrawal latency (PWL), and CatWalk gait analysis. Expression of pain-related signaling molecules in the spinal cord were detected using qRT-PCR, western blot assay, and immunofluorescence staining. This results showed that ICTF (3.0mg/kg, i.p.) effectively alleviated mechanical allodynia and thermal hyperalgesia not 0.3 and 1.0mg/kg in CFA rats. Subsequently, we further observed that ICTF (3.0mg/kg) dramatically decreased the mRNA and protein levels of HMGB1, NF-κB p65, and IL-1β but markedly enhanced α7nAChR and IL-10 expression in the spinal cord of CFA rats, and Immunofluorescence staining also showed that ICTF (3.0mg/kg) significantly increased the expression of α7nAChR and reduced IBA1 in the spinal cord of CFA rats, along with suppressing the alterations of gait parameters induced by CFA. Moreover, Intrathecal injection (i.t.) of α7nAChR antagonist alpha-bungarotoxin (α-Bgtx, 1.0μg/kg) not only reversed the anti-nociceptive effect of ICTF on pain hypersensitivity, but also inhibited the down-regulation of HMGB1, NF-κB p65, and IL-1β as well as the up-regulation of α7nAChR and IL-10 protein expression induced by ICTF treatment. Altogether, our results illustrate that ICTF enables to ameliorate CFA-induced inflammatory pain through α7nAChR-mediated inhibition of HMGB1/NF-κB signaling pathway in the spinal cord of rats, suggesting that ICTF may be exploited as a potential painkiller against chronic inflammatory pain.
Collapse
|
90
|
Acrylamide Neurotoxicity as a Possible Factor Responsible for Inflammation in the Cholinergic Nervous System. Int J Mol Sci 2022; 23:ijms23042030. [PMID: 35216144 PMCID: PMC8880090 DOI: 10.3390/ijms23042030] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Acrylamide (ACR) is a chemical compound that exhibits neurotoxic and genotoxic effects. It causes neurological symptoms such as tremors, general weakness, numbness, tingling in the limbs or ataxia. Numerous scientific studies show the effect of ACR on nerve endings and its close connection with the cholinergic system. The cholinergic system is part of the autonomic nervous system that regulates higher cortical functions related to memory, learning, concentration and attention. Within the cholinergic system, there are cholinergic neurons, anatomical cholinergic structures, the neurotransmitter acetylcholine (ACh) and cholinergic receptors. Some scientific reports suggest a negative effect of ACR on the cholinergic system and inflammatory reactions within the body. The aim of the study was to review the current state of knowledge on the influence of acrylamide on the cholinergic system and to evaluate its possible effect on inflammatory processes. The cholinergic anti-inflammatory pathway (CAP) is a neuroimmunomodulatory pathway that is located in the blood and mucous membranes. The role of CAP is to stop the inflammatory response in the appropriate moment. It prevents the synthesis and the release of pro-inflammatory cytokines and ultimately regulates the local and systemic immune response. The cellular molecular mechanism for inhibiting cytokine synthesis is attributed to acetylcholine (ACh), the major vagal neurotransmitter, and the α7 nicotinic receptor (α7nAChR) subunit is a key receptor for the cholinergic anti-inflammatory pathway. The combination of ACh with α7nAChR results in inhibition of the synthesis and release of pro-inflammatory cytokines. The blood AChE is able to terminate the stimulation of the cholinergic anti-inflammatory pathway due to splitting ACh. Accordingly, cytokine production is essential for pathogen protection and tissue repair, but over-release of cytokines can lead to systemic inflammation, organ failure, and death. Inflammatory responses are precisely regulated to effectively protect against harmful stimuli. The central nervous system dynamically interacts with the immune system, modulating inflammation through the humoral and nervous pathways. The stress-induced rise in acetylcholine (ACh) level acts to ease the inflammatory response and restore homeostasis. This signaling process ends when ACh is hydrolyzed by acetylcholinesterase (AChE). There are many scientific reports indicating the harmful effects of ACR on AChE. Most of them indicate that ACR reduces the concentration and activity of AChE. Due to the neurotoxic effect of acrylamide, which is related to the disturbance of the secretion of neurotransmitters, and its influence on the disturbance of acetylcholinesterase activity, it can be concluded that it disturbs the normal inflammatory response.
Collapse
|
91
|
Vagus nerve stimulation alleviated cerebral ischemia and reperfusion injury in rats by inhibiting pyroptosis via α7 nicotinic acetylcholine receptor. Cell Death Dis 2022; 8:54. [PMID: 35136042 PMCID: PMC8825823 DOI: 10.1038/s41420-022-00852-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/06/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
Cumulative evidence suggests that pyroptosis, a new sort of programmed cell death, is closely related to cerebral ischemia/reperfusion (I/R) injury. Our previous studies have testified that vagus nerve stimulation (VNS) was involved in many different neuroprotective and neuroplasticity pathways via α7 nicotinic acetylcholine receptor (α7nAchR), a vital node of the cholinergic anti-inflammatory pathway during cerebral I/R injury. We aimed to determine the neuroprotective effects of VNS through α7nAchR-mediated inhibition of pyroptosis. Focal cerebral ischemic stroke rat models were obtained by middle cerebral artery occlusion for 120 min. Expression of the NLRP3 inflammasome was evaluated using western blotting and immunofluorescence (IF) staining. The neurological deficit score, infarct volume, TUNEL staining findings, transmission electron microscopy findings, and expression of inflammatory cytokines were assessed 3 days after I/R injury. Our findings suggested that the protein expression levels of NLRP3, GSDMD-N, cleaved caspase-1, and ASC gradually increased until they peaked on day 3 after I/R injury. VNS inhibited the expression of pyroptosis-related molecules and decreased the number of pyroptotic cells and membrane pores. Administration of α7nAchR-antagonist and agonist helped in further assessment of the role of α7nAchR in pyroptosis. α7nAchR-agonist mimicked VNS’s neuroprotective effects on the improvement of neurological deficits, the reduction of infarct volumes, and the inhibition of neuronal pyroptosis after cerebral I/R injury. Conversely, the neuroprotection provided by VNS could be reversed by the administration of α7nAchR-antagonist. In conclusion, VNS-induced neuroprotection via inhibition of neuronal pyroptosis was α7nAchR-dependent, highlighting the pivotal role of α7nAChR in suppressing cellular pyroptosis and neuroinflammation. These findings may allow a better understanding of treatment principles for cerebral I/R injury.
Collapse
|
92
|
Deng L, Wang S, Guo H, Liu X, Zou X, Zhang R, Lin Y, Tan W. (RS)-bambuterol and its enantiomers: Potential improvement of (R)-bambuterol in mice with colitis. Int Immunopharmacol 2022; 103:108501. [PMID: 34974400 DOI: 10.1016/j.intimp.2021.108501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022]
Abstract
Bambuterol (BMB) has been used clinically to treat asthma due to its bronchodilation activity. However, the effect of BMB on ulcerative colitis (UC) has not been examined. The present work focused on the effects of enantiomeric BMB on UC. Acute UC was induced in mice by 3% dextran sulfate sodium (DSS), and (R)-, (S) and (RS)-BMB were orally administered. Body weight loss and the disease activity index (DAI) were measured once a day. Inflammatory factors were detected by ELISA and qRT-PCR. Histological evaluations of colon samples were performed. IL-6, STAT3, and RORγt pathway-related proteins were analyzed by western blotting. The results verified that colitis severity was dramatically ameliorated by (R)-BMB, which was significantlybetter than the effect of (RS)-BMB or (S)-BMB, as evidenced by body weight loss, DAI, colon length, spleen/body weight ratio and histopathological manifestations. Furthermore, (R)-BMB treatment significantly diminished the levels of inflammatory cytokines and macrophages infiltration in mice with colitis. Besides, treated with (R)-BMB obviously elevated the level of β2AR. In addition, (R)-BMB decreased the expression of IL-6, IL-17, retinoic acid receptor-related orphan receptor-gamma t (RORt), and phosphorylated STAT3 (p-STAT3) in a dose-dependent manner in the colon tissues. The efficacy of (R)-BMB was more notable than aminosalicylic acid (5-ASA). (R)-BMB is either butyrilcholinesterase inhibitor or β2AR agonist which offers new treatment of colitis.
Collapse
Affiliation(s)
- Liangjun Deng
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Shanping Wang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Haihua Guo
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Xiaoming Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinfeng Zou
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Rui Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Yue Lin
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Wen Tan
- Post-Doctoral Innovation Site, Jinan University Affiliation, Yuanzhi Health Technology Co, Ltd, Hengqin New District, Zhuhai, Guangdong 519000, China; Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| |
Collapse
|
93
|
Canning BJ, Liu Q, Tao M, DeVita R, Perelman M, Hay DW, Dicpinigaitis PV, Liang J. Evidence for Alpha 7 Nicotinic Receptor Activation During the Cough Suppressing Effects Induced by Nicotine and Identification of ATA-101 as a Potential Novel Therapy for the Treatment of Chronic Cough. J Pharmacol Exp Ther 2022; 380:94-103. [PMID: 34782407 PMCID: PMC8969114 DOI: 10.1124/jpet.121.000641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 11/11/2021] [Indexed: 02/03/2023] Open
Abstract
Studies performed in healthy smokers have documented a diminished responsiveness to tussive challenges, and several lines of experimental evidence implicate nicotine as an antitussive component in both cigarette smoke and the vapors generated by electronic cigarettes (eCigs). We set out to identify the nicotinic receptor subtype involved in the antitussive actions of nicotine and to further evaluate the potential of nicotinic receptor-selective agonists as cough-suppressing therapeutics. We confirmed an antitussive effect of nicotine in guinea pigs. We additionally observed that the alpha-4 beta-2 (α 4 β 2)-selective agonist Tc-6683 was without effect on evoked cough responses in guinea pigs, while the α 7-selective agonist PHA 543613 dose-dependently inhibited evoked coughing. We subsequently describe the preclinical evidence in support of ATA-101, a potent and highly selective (α 7) selective nicotinic receptor agonist, as a potential candidate for antitussive therapy in humans. ATA-101, formerly known as Tc-5619, was orally bioavailable and moderately central nervous system (CNS) penetrant and dose-dependently inhibited coughing in guinea pigs evoked by citric acid and bradykinin. Comparing the effects of airway targeted administration versus systemic dosing and the effects of repeated dosing at various times prior to tussive challenge, our data suggest that the antitussive actions of ATA-101 require continued engagement of α 7 nicotinic receptors, likely in the CNS. Collectively, the data provide the preclinical rationale for α 7 nicotinic receptor engagement as a novel therapeutic strategy for cough suppression. The data also suggest that α 7 nicotinic acetylcholine receptor (nAChR) activation by nicotine may be permissive to nicotine delivery in a way that may promote addiction. SIGNIFICANCE STATEMENT: This study documents the antitussive actions of nicotine and identifies the α7 nicotinic receptor subtype as the target for nicotine during cough suppression described in humans. We additionally present evidence suggesting that ATA-101 and other α7 nicotinic receptor-selective agonists may be promising candidates for the treatment of chronic refractory cough.
Collapse
Affiliation(s)
- Brendan J Canning
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Qi Liu
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Mayuko Tao
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Robert DeVita
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Michael Perelman
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Douglas W Hay
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Peter V Dicpinigaitis
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| | - Jing Liang
- The Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland (B.J.C, Q.L.); Tokyo Medical and Dental University, Tokyo, Japan (M.T.); RJD Medicinal Chemistry Consulting LLC, Westfield, New Jersey (R.D.); Michael Perelman Consulting, Winter Park, Florida (M.P.); Hay Drug Discovery Consulting, Valley Forge, Pennsylvania (D.W.H.); Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York (P.V.D.); Apple Helix Bioventures, New York, New York (J.L.)
| |
Collapse
|
94
|
Mehranfard D, Speth RC. Cholinergic anti-inflammatory pathway and COVID-19. BIOIMPACTS : BI 2022; 12:171-174. [PMID: 35411295 PMCID: PMC8905591 DOI: 10.34172/bi.2022.23980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 01/09/2023]
Abstract
The cholinergic anti-inflammatory pathway (CAP) first described by Wang et al, 2003 has contemporary interest arising from the COVID-19 pandemic. While tobacco smoking has been considered an aggravating factor in the severity of COVID-19 infections, it has been suggested by some that the nicotine derived from tobacco could lessen the severity of COVID-19 infections. This spotlight briefly describes the CAP and its potential role as a therapeutic target for the treatment of COVID-19 infections using vagus nerve stimulation or selective alpha7 nicotinic acetylcholine receptor agonists.
Collapse
Affiliation(s)
- Danial Mehranfard
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Robert C. Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA
| |
Collapse
|
95
|
Apolipoprotein (a)/Lipoprotein(a)-Induced Oxidative-Inflammatory α7-nAChR/p38 MAPK/IL-6/RhoA-GTP Signaling Axis and M1 Macrophage Polarization Modulate Inflammation-Associated Development of Coronary Artery Spasm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9964689. [PMID: 35096275 PMCID: PMC8793348 DOI: 10.1155/2022/9964689] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/21/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Objective. Apolipoprotein (a)/lipoprotein(a) (Lp(a)), a major carrier of oxidized phospholipids, and α7-nicotinic acetylcholine receptor (α7-nAChR) may play an important role in the development of coronary artery spasm (CAS). In CAS, the association between Lp(a) and the α7-nAChR-modulated inflammatory macrophage polarization and activation and smooth muscle cell dysfunction remains unknown. Methods. We investigated the relevance of Lp(a)/α7-nAChR signaling in patient monocyte-derived macrophages and human coronary artery smooth muscle cells (HCASMCs) using expression profile correlation analyses, fluorescence-assisted cell sorting flow cytometry, immunoblotting, quantitative real-time polymerase chain reaction, and clinicopathological analyses. Results. There are increased serum Lp(a) levels (3.98-fold,
) and macrophage population (3.30-fold,
) in patients with CAS compared with patients without CAS. Serum Lp(a) level was positively correlated with high-sensitivity C-reactive protein (
,
), IL-6 (
,
), and α7-nAChR (
,
) in patients with CAS, but not in patients without CAS. Compared with untreated or low-density lipoprotein- (LDL-) treated macrophages, Lp(a)-treated macrophages exhibited markedly enhanced α7-nAChR mRNA expression (
) and activity (
), in vitro and ex vivo. Lp(a) but not LDL preferentially induced CD80+ macrophage (M1) polarization and reduced the inducible nitric oxide synthase expression and the subsequent NO production. While shRNA-mediated loss of α7-nAChR function reduced the Lp(a)-induced CD80+ macrophage pool, both shRNA and anti-IL-6 receptor tocilizumab suppressed Lp(a)-upregulated α7-nAChR, p-p38 MAPK, IL-6, and RhoA-GTP protein expression levels in cultures of patient monocyte-derived macrophages and HCASMCs. Conclusions. Elevated Lp(a) levels upregulate α7-nAChR/IL-6/p38 MAPK signaling in macrophages of CAS patients and HCASMC, suggesting that Lp(a)-triggered inflammation mediates CAS through α7-nAChR/p38 MAPK/IL-6/RhoA-GTP signaling induction, macrophage M1 polarization, and HCASMC activation.
Collapse
|
96
|
Cholinergic blockade of neuroinflammation – from tissue to RNA regulators. Neuronal Signal 2022; 6:NS20210035. [PMID: 35211331 PMCID: PMC8837817 DOI: 10.1042/ns20210035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory stimuli and consequent pro-inflammatory immune responses may facilitate neurodegeneration and threaten survival following pathogen infection or trauma, but potential controllers preventing these risks are incompletely understood. Here, we argue that small RNA regulators of acetylcholine (ACh) signaling, including microRNAs (miRs) and transfer RNA fragments (tRFs) may tilt the balance between innate and adaptive immunity, avoid chronic inflammation and prevent the neuroinflammation-mediated exacerbation of many neurological diseases. While the restrictive permeability of the blood–brain barrier (BBB) protects the brain from peripheral immune events, this barrier can be disrupted by inflammation and is weakened with age. The consequently dysregulated balance between pro- and anti-inflammatory processes may modify the immune activities of brain microglia, astrocytes, perivascular macrophages, oligodendrocytes and dendritic cells, leading to neuronal damage. Notably, the vagus nerve mediates the peripheral cholinergic anti-inflammatory reflex and underlines the consistent control of body–brain inflammation by pro-inflammatory cytokines, which affect cholinergic functions; therefore, the disruption of this reflex can exacerbate cognitive impairments such as attention deficits and delirium. RNA regulators can contribute to re-balancing the cholinergic network and avoiding its chronic deterioration, and their activities may differ between men and women and/or wear off with age. This can lead to hypersensitivity of aged patients to inflammation and higher risks of neuroinflammation-driven cholinergic impairments such as delirium and dementia following COVID-19 infection. The age- and sex-driven differences in post-transcriptional RNA regulators of cholinergic elements may hence indicate new personalized therapeutic options for neuroinflammatory diseases.
Collapse
|
97
|
Activation of α-7 Nicotinic Acetylcholine Receptor Attenuates Cardiac Inflammation Through NLRP3/Caspase-1/IL-18 Pathway. Biochem Genet 2022; 60:1333-1345. [PMID: 34988776 DOI: 10.1007/s10528-021-10162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 11/04/2021] [Indexed: 11/02/2022]
Abstract
Activation of α-7 nicotinic acetylcholine receptor (α7nAChR) receptor might induce cardiac inflammation, cardiac remodeling, and dysfunction. In this regard, this study aims to clarify the role and mechanism of α7nAChR in the process of cardiac inflammation and damage. Normal male C57BL/6J and NLRP3-knockout mice were used to evaluate the effect of PHA-543613, a selective agonist of α7nAChR, on cardiac inflammation and possible involvement of NLRP3/Caspase-1/IL-18 using western blotting and ELISA. Activation of α7nAChR using PHA-543613 (NE), at the doses of 0.5 mg/kg and 1 mg/kg, induced cardiac inflammation. In addition, both in vivo and in vitro studies showed higher expression of NLRP3 and higher activation of Caspase-1 and IL-18 after treating animals with NE. On the other hand, we did not observe any significant changes in inflammatory cytokines and cardiac inflammation after administration of NE in NLRP3-knockout mice. It could be concluded that blocking the NLRP3/Caspase-1/IL-18 pathway can simultaneously inhibit the inflammatory response mediated by α7nAChR and it would a novel target for inhibiting cardiac inflammation and remodeling.
Collapse
|
98
|
Villa C, Rivellini E, Lavitrano M, Combi R. Can SARS-CoV-2 Infection Exacerbate Alzheimer's Disease? An Overview of Shared Risk Factors and Pathogenetic Mechanisms. J Pers Med 2022; 12:29. [PMID: 35055344 PMCID: PMC8780286 DOI: 10.3390/jpm12010029] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
The current coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2, is affecting every aspect of global society, including public healthcare systems, medical care access, and the economy. Although the respiratory tract is primarily affected by SARS-CoV-2, emerging evidence suggests that the virus may also reach the central nervous system (CNS), leading to several neurological issues. In particular, people with a diagnosis of Alzheimer's disease (AD) are a vulnerable group at high risk of contracting COVID-19, and develop more severe forms and worse outcomes, including death. Therefore, understanding shared links between COVID-19 and AD could aid the development of therapeutic strategies against both. Herein, we reviewed common risk factors and potential pathogenetic mechanisms that might contribute to the acceleration of neurodegenerative processes in AD patients infected by SARS-CoV-2.
Collapse
Affiliation(s)
- Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Eleonora Rivellini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
99
|
Xu XJ, Yang MS, Zhang B, Ge QQ, Niu F, Dong JQ, Zhuang Y, Liu BY. Genome-wide interrogation of transfer RNA-derived small RNAs in a mouse model of traumatic brain injury. Neural Regen Res 2022; 17:386-394. [PMID: 34269214 PMCID: PMC8463968 DOI: 10.4103/1673-5374.314315] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are a recently established family of regulatory small non-coding RNAs that modulate diverse biological processes. Growing evidence indicates that tsRNAs are involved in neurological disorders and play a role in the pathogenesis of neurodegenerative disease. However, whether tsRNAs are involved in traumatic brain injury-induced secondary injury remains poorly understood. In this study, a mouse controlled cortical impact model of traumatic brain injury was established, and integrated tsRNA and messenger RNA (mRNA) transcriptome sequencing were used. The results revealed that 103 tsRNAs were differentially expressed in the mouse model of traumatic brain injury at 72 hours, of which 56 tsRNAs were upregulated and 47 tsRNAs were downregulated. Based on microRNA-like seed matching and Pearson correlation analysis, 57 differentially expressed tsRNA-mRNA interaction pairs were identified, including 29 tsRNAs and 26 mRNAs. Moreover, Gene Ontology annotation of target genes revealed that the significantly enriched terms were primarily associated with inflammation and synaptic function. Collectively, our findings suggest that tsRNAs may be associated with traumatic brain injury-induced secondary brain injury, and are thus a potential therapeutic target for traumatic brain injury. The study was approved by the Beijing Neurosurgical Institute Animal Care and Use Committee (approval No. 20190411) on April 11, 2019.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian-Qian Ge
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University; Nerve Injury and Repair Center of Beijing Institute for Brain Disorders; China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
100
|
The Mechanisms Mediated by α7 Acetylcholine Nicotinic Receptors May Contribute to Peripheral Nerve Regeneration. Molecules 2021; 26:molecules26247668. [PMID: 34946750 PMCID: PMC8709212 DOI: 10.3390/molecules26247668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/25/2023] Open
Abstract
Due to the microenvironment created by Schwann cell (SC) activity, peripheral nerve fibers are able to regenerate. Inflammation is the first response to nerve damage and the removal of cellular and myelin debris is essential in preventing the persistence of the local inflammation that may negatively affect nerve regeneration. Acetylcholine (ACh) is one of the neurotransmitters involved in the modulation of inflammation through the activity of its receptors, belonging to both the muscarinic and nicotinic classes. In this report, we evaluated the expression of α7 nicotinic acetylcholine receptors (nAChRs) in rat sciatic nerve, particularly in SCs, after peripheral nerve injury. α7 nAChRs are absent in sciatic nerve immediately after dissection, but their expression is significantly enhanced in SCs after 24 h in cultured sciatic nerve segments or in the presence of the proinflammatory neuropeptide Bradykinin (BK). Moreover, we found that activation of α7 nAChRs with the selective partial agonist ICH3 causes a decreased expression of c-Jun and an upregulation of uPA, MMP2 and MMP9 activity. In addition, ICH3 treatment inhibits IL-6 transcript level expression as well as the cytokine release. These results suggest that ACh, probably released from regenerating axons or by SC themselves, may actively promote through α7 nAChRs activation an anti-inflammatory microenvironment that contributes to better improving the peripheral nerve regeneration.
Collapse
|