1
|
Lu J, Chen K, Cen Z, Huang Y, Li Y, Chen L, Wu W. α7nAChR on B cells directs T cell differentiation to prevent viral myocarditis. JCI Insight 2025; 10:e189323. [PMID: 40337863 DOI: 10.1172/jci.insight.189323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Patients with viral myocarditis (VMC) exhibit evident autonomic nervous system imbalance, and adverse cardiac remodeling is involved in impaired cholinergic function. The α7 nicotinic acetylcholine receptor (α7nAChR), which is a neurotransmitter receptor, exerts immunoregulatory effects. Recent advances have illuminated the evolution and functions of peripheral and cardiac B cells in heart disease. However, the role of α7nAChR expressed by B cells in the progression of VMC has not been established. We revealed the neuroimmune communication landscape in the heart and found that the phenotypes of cardiac and splenic B cells and their α7nAChR expression changed dynamically during the progression of VMC to dilated cardiomyopathy. α7nAChR on B cells serves as a negative regulator by inhibiting their proinflammatory functions and signaling pathways. B cell-specific α7nAChR deficiency exacerbated myocardial inflammation, fibrosis, and cardiac dysfunction. However, these effects were abrogated in non-B cells from mice with IL-17A knockdown. Enhanced degradation of acetylcholine leads to an imbalance in cholinergic signaling, resulting in impaired neurotransmission. The acetylcholinesterase inhibitor pyridostigmine bromide could improve cardiac remodeling and prevent the progression of VMC to the chronic phase, which was partly dependent on the α7nAChR on B cells. Our findings provide notable insights into cardiac-neural-immune communication during myocardial injury.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cardiology, and
| | | | | | | | - Yong Li
- Emergency Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | | | - Weifeng Wu
- Department of Cardiology, and
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co. constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
2
|
Yang Z, Gao Y, Zhao L, Lv X, Du Y. Molecular mechanisms of Sepsis attacking the immune system and solid organs. Front Med (Lausanne) 2024; 11:1429370. [PMID: 39267971 PMCID: PMC11390691 DOI: 10.3389/fmed.2024.1429370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Remarkable progress has been achieved in sepsis treatment in recent times, the mortality rate of sepsis has experienced a gradual decline as a result of the prompt administration of antibiotics, fluid resuscitation, and the implementation of various therapies aimed at supporting multiple organ functions. However, there is still significant mortality and room for improvement. The mortality rate for septic patients, 22.5%, is still unacceptably high, accounting for 19.7% of all global deaths. Therefore, it is crucial to thoroughly comprehend the pathogenesis of sepsis in order to enhance clinical diagnosis and treatment methods. Here, we summarized classic mechanisms of sepsis progression, activation of signal pathways, mitochondrial quality control, imbalance of pro-and anti- inflammation response, diseminated intravascular coagulation (DIC), cell death, presented the latest research findings for each mechanism and identify potential therapeutic targets within each mechanism.
Collapse
Affiliation(s)
- Zhaoyun Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yan Gao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanwei Du
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
3
|
Zhang Q, Zhang L, Lin G, Luo F. The protective role of vagus nerve stimulation in ischemia-reperfusion injury. Heliyon 2024; 10:e30952. [PMID: 38770302 PMCID: PMC11103530 DOI: 10.1016/j.heliyon.2024.e30952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) encompasses the damage resulting from the restoration of blood supply following tissue ischemia. This phenomenon commonly occurs in clinical scenarios such as hemorrhagic shock, severe trauma, organ transplantation, and thrombolytic therapy. Despite its prevalence, existing treatments exhibit limited efficacy against IRI. Vagus nerve stimulation (VNS) is a widely utilized technique for modulating the autonomic nervous system. Numerous studies have demonstrated that VNS significantly reduces IRI in various organs, including the heart, brain, and liver. This article reviews the pathological processes during IRI and summarizes the role and possible mechanisms of VNS in IRI of different organs. Furthermore, this review addresses the current challenges of VNS clinical applications, providing a novel perspective on IRI treatment.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Guoqiang Lin
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Fanyan Luo
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
4
|
Kelly MJ, Breathnach C, Tracey KJ, Donnelly SC. Manipulation of the inflammatory reflex as a therapeutic strategy. Cell Rep Med 2022; 3:100696. [PMID: 35858588 PMCID: PMC9381415 DOI: 10.1016/j.xcrm.2022.100696] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 06/20/2021] [Accepted: 06/23/2022] [Indexed: 02/07/2023]
Abstract
The cholinergic anti-inflammatory pathway is the efferent arm of the inflammatory reflex, a neural circuit through which the CNS can modulate peripheral immune responses. Signals communicated via the vagus and splenic nerves use acetylcholine, produced by Choline acetyltransferase (ChAT)+ T cells, to downregulate the inflammatory actions of macrophages expressing α7 nicotinic receptors. Pre-clinical studies using transgenic animals, cholinergic agonists, vagotomy, and vagus nerve stimulation have demonstrated this pathway's role and therapeutic potential in numerous inflammatory diseases. In this review, we summarize what is understood about the inflammatory reflex. We also demonstrate how pre-clinical findings are being translated into promising clinical trials, and we draw particular attention to innovative bioelectronic methods of harnessing the cholinergic anti-inflammatory pathway for clinical use.
Collapse
Affiliation(s)
- Mark J Kelly
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland; Tallaght University Hospital, Dublin, Ireland
| | | | - Kevin J Tracey
- Center for Biomedical Science and Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Seamas C Donnelly
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland; Tallaght University Hospital, Dublin, Ireland.
| |
Collapse
|
5
|
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
|
6
|
The cholinergic anti-inflammatory pathway in humans: State-of-the-art review and future directions. Neurosci Biobehav Rev 2022; 136:104622. [PMID: 35300992 DOI: 10.1016/j.neubiorev.2022.104622] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
Abstract
The parasympathetic nervous system modulates inflammation through efferent vagus nerve signaling. Tracey (2002) termed this process as the cholinergic anti-inflammatory pathway (CAP). Interest in the potential practical use of this immune-modulatory process is increasing alongside increasing appreciation for the role of systemic inflammation in the etiology of somatic and psychological disease. A diverse literature exists providing expansive correlational evidence and some preliminary experimental evidence of the CAP in humans. However, so far this literature has not been well integrated and critically evaluated. This review describes the current state-of-the-art of research into vagus nerve driven parasympathetic control of inflammation in humans. Substantial limitations and gaps in the literature are identified, and promising directions for future research are highlighted.
Collapse
|
7
|
Costantini TW, Coimbra R, Weaver JL, Eliceiri BP. Precision targeting of the vagal anti-inflammatory pathway attenuates the systemic inflammatory response to burn injury. J Trauma Acute Care Surg 2022; 92:323-329. [PMID: 34789702 PMCID: PMC8792272 DOI: 10.1097/ta.0000000000003470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND The systemic inflammatory response (SIRS) drives late morbidity and mortality after injury. The α7 nicotinic acetylcholine receptor (α7nAchR) expressed on immune cells regulates the vagal anti-inflammatory pathway that prevents an overwhelming SIRS response to injury. Nonspecific pharmacologic stimulation of the vagus nerve has been evaluated as a potential therapeutic to limit SIRS. Unfortunately, the results of clinical trials have been underwhelming. We hypothesized that directly targeting the α7nAchR would more precisely stimulate the vagal anti-inflammatory pathway on immune cells and decrease gut and lung injury after severe burn. METHODS C57BL/6 mice underwent 30% total body surface area steam burn. Mice were treated with an intraperitoneal injection of a selective agonist of the α7nAchR (AR-R17779) at 30 minutes postburn. Intestinal permeability to 4 kDa FITC-dextran was measured at multiple time points postinjury. Lung vascular permeability was measured 6 hours after burn injury. Serial behavioral assessments were performed to quantify activity levels. RESULTS Intestinal permeability peaked at 6 hours postburn. AR-R17779 decreased burn-induced intestinal permeability in a dose-dependent fashion (p < 0.001). There was no difference in gut permeability to 4 kDa FITC-dextran between sham and burn-injured animals treated with 5 mg/kg of AR-R17779. While burn injury increased lung permeability 10-fold, AR-R17779 prevented burn-induced lung permeability with no difference compared with sham (p < 0.01). Postinjury activity levels were significantly improved in burned animals treated with AR-R17779. CONCLUSION Directly stimulating the α7nAchR prevents burn-induced gut and lung injury. Directly targeting the α7nAChR that mediates the cholinergic anti-inflammatory response may be an improved strategy compared with nonspecific vagal agonists.
Collapse
Affiliation(s)
- Todd W. Costantini
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| | - Raul Coimbra
- Comparative Effectiveness and Clinical Outcomes Research Center, Riverside University Health System, Loma Linda University School of Medicine, Riverside, CA
| | - Jessica L. Weaver
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| | - Brian P. Eliceiri
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| |
Collapse
|
8
|
Marine Origin Ligands of Nicotinic Receptors: Low Molecular Compounds, Peptides and Proteins for Fundamental Research and Practical Applications. Biomolecules 2022; 12:biom12020189. [PMID: 35204690 PMCID: PMC8961598 DOI: 10.3390/biom12020189] [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: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
The purpose of our review is to briefly show what different compounds of marine origin, from low molecular weight ones to peptides and proteins, offer for understanding the structure and mechanism of action of nicotinic acetylcholine receptors (nAChRs) and for finding novel drugs to combat the diseases where nAChRs may be involved. The importance of the mentioned classes of ligands has changed with time; a protein from the marine snake venom was the first excellent tool to characterize the muscle-type nAChRs from the electric ray, while at present, muscle and α7 receptors are labeled with the radioactive or fluorescent derivatives prepared from α-bungarotoxin isolated from the many-banded krait. The most sophisticated instruments to distinguish muscle from neuronal nAChRs, and especially distinct subtypes within the latter, are α-conotoxins. Such information is crucial for fundamental studies on the nAChR revealing the properties of their orthosteric and allosteric binding sites and mechanisms of the channel opening and closure. Similar data are provided by low-molecular weight compounds of marine origin, but here the main purpose is drug design. In our review we tried to show what has been obtained in the last decade when the listed classes of compounds were used in the nAChR research, applying computer modeling, synthetic analogues and receptor mutants, X-ray and electron-microscopy analyses of complexes with the nAChRs, and their models which are acetylcholine-binding proteins and heterologously-expressed ligand-binding domains.
Collapse
|
9
|
Transcutaneous vagal nerve simulation to reduce a systemic inflammatory response syndrome and the associated intestinal failure: study protocol of a prospective, two-armed, sham-controlled, double-blinded trial in healthy subjects (the NeuroSIRS-Study). Int J Colorectal Dis 2022; 37:259-270. [PMID: 34599686 PMCID: PMC8760201 DOI: 10.1007/s00384-021-04034-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE Surgery initiates pro-inflammatory mediator cascades leading to a variably pronounced sterile inflammation (SIRS). SIRS is associated with intestinal paralysis and breakdown of intestinal barrier and might result in abdominal sepsis. Technological progress led to the development of a neurostimulator for transcutaneous auricular vagal nerve stimulation (taVNS), which is associated with a decline in inflammatory parameters and peristalsis improvement in rodents and healthy subjects via activation of the cholinergic anti-inflammatory pathway. Therefore, taVNS might be a strategy for SIRS prophylaxis. METHODS The NeuroSIRS-Study is a prospective, randomized two-armed, sham-controlled, double-blind clinical trial. The study is registered at DRKS00016892 (09.07.2020). A controlled endotoxemia is used as a SIRS-mimicking model. 2 ng/kg bodyweight lipopolysaccharide (LPS) will be administered after taVNS or sham stimulation. The primary objective is a reduction of clinical symptoms of SIRS after taVNS compared to sham stimulation. Effects of taVNS on release of inflammatory cytokines, intestinal function, and vital parameters will be analyzed. DISCUSSION TaVNS is well-tolerated, with little to no side effects. Despite not fully mimicking postoperative inflammation, LPS challenge is the most used experimental tool to imitate SIRS and offers standardization and reproducibility. The restriction to healthy male volunteers exerts a certain bias limiting generalizability to the surgical population. Still, this pilot study aims to give first insights into taVNS as a prophylactic treatment in postoperative inflammation to pave the way for further clinical trials in patients at risk for SIRS. This would have major implications for future therapeutic approaches.
Collapse
|
10
|
The Regulation Effect of α7nAChRs and M1AChRs on Inflammation and Immunity in Sepsis. Mediators Inflamm 2021; 2021:9059601. [PMID: 34776789 PMCID: PMC8580654 DOI: 10.1155/2021/9059601] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/14/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
The inflammatory storm in the early stage and immunosuppression in the late stage are responsible for the high mortality rates and multiple organ dysfunction in sepsis. In recent years, studies have found that the body's cholinergic system can spontaneously and dynamically regulate inflammation and immunity in sepsis according to the needs of the body. Firstly, the vagus nerve senses and regulates local or systemic inflammation by means of the Cholinergic Anti-inflammatory Pathway (CAP) and activation of α7-nicotinic acetylcholine receptors (α7nAChRs); thus, α7nAChRs play important roles for the central nervous system (CNS) to modulate peripheral inflammation; secondly, the activation of muscarinic acetylcholine receptors 1 (M1AChRs) in the forebrain can affect the neurons of the Medullary Visceral Zone (MVZ), the core of CAP, to regulate systemic inflammation and immunity. Based on the critical role of these two cholinergic receptor systems in sepsis, it is necessary to collect and analyze the related findings in recent years to provide ideas for further research studies and clinical applications. By consulting the related literature, we draw some conclusions: MVZ is the primary center for the nervous system to regulate inflammation and immunity. It coordinates not only the sympathetic system and vagus system but also the autonomic nervous system and neuroendocrine system to regulate inflammation and immunity; α7nAChRs are widely expressed in immune cells, neurons, and muscle cells; the activation of α7nAChRs can suppress local and systemic inflammation; the expression of α7nAChRs represents the acute or chronic inflammatory state to a certain extent; M1AChRs are mainly expressed in the advanced centers of the brain and regulate systemic inflammation; neuroinflammation of the MVZ, hypothalamus, and forebrain induced by sepsis not only leads to their dysfunctions but also underlies the regulatory dysfunction on systemic inflammation and immunity. Correcting the neuroinflammation of these regulatory centers and adjusting the function of α7nAChRs and M1AChRs may be two key strategies for the treatment of sepsis in the future.
Collapse
|
11
|
Abstract
Sepsis is expected to have a substantial impact on public health and cost as its prevalence increases. Factors contributing to increased prevalence include a progressively aging population, advances in the use of immunomodulatory agents to treat a rising number of diseases, and immune-suppressing therapies in organ transplant recipients and cancer patients. It is now recognized that sepsis is associated with profound and sustained immunosuppression, which has been implicated as a predisposing factor in the increased susceptibility of patients to secondary infections and mortality. In this review, we discuss mechanisms of sepsis-induced immunosuppression and biomarkers that identify a state of impaired immunity. We also highlight immune-enhancing strategies that have been evaluated in patients with sepsis, as well as therapeutics under current investigation. Finally, we describe future challenges and the need for a new treatment paradigm, integrating predictive enrichment with patient factors that may guide the future selection of tailored immunotherapy. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lisa K Torres
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, New York, NY, USA;
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands;
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands;
| |
Collapse
|
12
|
Lauwers M, Courties A, Sellam J, Wen C. The cholinergic system in joint health and osteoarthritis: a narrative-review. Osteoarthritis Cartilage 2021; 29:643-653. [PMID: 33609692 DOI: 10.1016/j.joca.2021.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) poses a major health and economic burden worldwide due to an increasing number of patients and the unavailability of disease-modifying drugs. In this review, the latest understanding of the involvement of the cholinergic system in joint homeostasis and OA will be outlined. First of all, the current evidence on the presence of the cholinergic system in the normal and OA joint will be described. Cholinergic innervation as well as the non-neuronal cholinergic system are detected. In a variety of inflammatory diseases, the classic cholinergic anti-inflammatory pathway lately received a lot of attention as via this pathway cholinergic agonists can reduce inflammation. The role of this cholinergic anti-inflammatory pathway in the context of OA will be discussed. Activation of this pathway improved the progression of the disease. Secondly, chondrocyte hypertrophy plays a pivotal role in osteophyte formation and OA development; the impact of the cholinergic system on hypertrophic chondroblasts and endochondral ossification will be evaluated. Cholinergic stimulation increased chondrocyte proliferation, delayed chondrocyte differentiation and caused early mineralisation. Moreover, acetylcholinesterase and butyrylcholinesterase affect the endochondral ossification via an acetylcholine-independent pathway. Thirdly, subchondral bone is critical for cartilage homeostasis and metabolism; the cholinergic system in subchondral bone homeostasis and disorders will be explored. An increase in osteoblast proliferation and osteoclast apoptosis is observed. Lastly, current therapeutic strategies for OA are limited to symptom relief; here the impact of smoking on disease progression and the potential of acetylcholinesterase inhibitors as candidate disease-modifying drug for OA will be discussed.
Collapse
Affiliation(s)
- M Lauwers
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| | - A Courties
- Department of Rheumatology, Assistance Publique - Hôpitaux de Paris (AP-HP), Inserm UMRS_938, Sorbonne Université, Saint-Antoine Hospital, Paris, France.
| | - J Sellam
- Department of Rheumatology, Assistance Publique - Hôpitaux de Paris (AP-HP), Inserm UMRS_938, Sorbonne Université, Saint-Antoine Hospital, Paris, France.
| | - C Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
13
|
Halder N, Lal G. Cholinergic System and Its Therapeutic Importance in Inflammation and Autoimmunity. Front Immunol 2021; 12:660342. [PMID: 33936095 PMCID: PMC8082108 DOI: 10.3389/fimmu.2021.660342] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurological and immunological signals constitute an extensive regulatory network in our body that maintains physiology and homeostasis. The cholinergic system plays a significant role in neuroimmune communication, transmitting information regarding the peripheral immune status to the central nervous system (CNS) and vice versa. The cholinergic system includes the neurotransmitter\ molecule, acetylcholine (ACh), cholinergic receptors (AChRs), choline acetyltransferase (ChAT) enzyme, and acetylcholinesterase (AChE) enzyme. These molecules are involved in regulating immune response and playing a crucial role in maintaining homeostasis. Most innate and adaptive immune cells respond to neuronal inputs by releasing or expressing these molecules on their surfaces. Dysregulation of this neuroimmune communication may lead to several inflammatory and autoimmune diseases. Several agonists, antagonists, and inhibitors have been developed to target the cholinergic system to control inflammation in different tissues. This review discusses how various molecules of the neuronal and non-neuronal cholinergic system (NNCS) interact with the immune cells. What are the agonists and antagonists that alter the cholinergic system, and how are these molecules modulate inflammation and immunity. Understanding the various functions of pharmacological molecules could help in designing better strategies to control inflammation and autoimmunity.
Collapse
Affiliation(s)
- Namrita Halder
- Laboratory of Autoimmunity and Tolerance, National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Girdhari Lal
- Laboratory of Autoimmunity and Tolerance, National Centre for Cell Science, Ganeshkhind, Pune, India
| |
Collapse
|
14
|
Possible Therapeutic Role of Cholinergic Agonists on COVID-19 related inflammatory response. JOURNAL OF BASIC AND CLINICAL HEALTH SCIENCES 2021. [DOI: 10.30621/jbachs.869857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
15
|
Nicotinic Acetylcholine Receptor Involvement in Inflammatory Bowel Disease and Interactions with Gut Microbiota. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031189. [PMID: 33572734 PMCID: PMC7908252 DOI: 10.3390/ijerph18031189] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
The gut-brain axis describes a complex interplay between the central nervous system and organs of the gastrointestinal tract. Sensory neurons of dorsal root and nodose ganglia, neurons of the autonomic nervous system, and immune cells collect and relay information about the status of the gut to the brain. A critical component in this bi-directional communication system is the vagus nerve which is essential for coordinating the immune system’s response to the activities of commensal bacteria in the gut and to pathogenic strains and their toxins. Local control of gut function is provided by networks of neurons in the enteric nervous system also called the ‘gut-brain’. One element common to all of these gut-brain systems is the expression of nicotinic acetylcholine receptors. These ligand-gated ion channels serve myriad roles in the gut-brain axis including mediating fast synaptic transmission between autonomic pre- and postganglionic neurons, modulation of neurotransmitter release from peripheral sensory and enteric neurons, and modulation of cytokine release from immune cells. Here we review the role of nicotinic receptors in the gut-brain axis with a focus on the interplay of these receptors with the gut microbiome and their involvement in dysregulation of gut function and inflammatory bowel diseases.
Collapse
|
16
|
Wang H, Cai D, Chen Z, Wang Y. GTS-21 Promotes α7 nAChR to Alleviate Intestinal Ischemia-Reperfusion-Induced Apoptosis and Inflammation of Enterocytes. Med Sci Monit 2020; 26:e921618. [PMID: 32417847 PMCID: PMC7251968 DOI: 10.12659/msm.921618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Intestinal ischemia-reperfusion injury is a serious intestinal disease, with main symptoms of inflammatory reaction and severe oxidative damage. In addition, GTS-21-induced α7 nAChR has been shown to exert anti-inflammatory effects and anti-oxidation effects in various organs. However, whether α7 nAChR can alleviate ischemia-reperfusion-induced intestinal injury is unclear. Material/Methods We used intestinal epithelial cells (IEC-6) to perform the experiments. Oxygen glucose deprivation/reoxygenation (OGD/R) was used to simulate the physiological environment of ischemia-reperfusion. First, the expression of α7 nAChR was determined in these cells which was cultured under OGD/R conditions. After that, the GTS-21 was used to treat these cells and the levels of inflammatory factors (TNF-α, IL-1β, IL-6, and IL-10) were assessed by ELISA. Next, the levels of ROS, SOD, and MDA were determined in IEC-6 cells. Finally, the apoptosis rates of IEC-6 cells were measured by flow cytometry. Results Results showed that the expression of TNF-α, IL-1β, and IL-6 was enhanced when the IEC-6 cells were cultured under OGD/R conditions. However, after treatment with GTS-21, the levels of these proinflammatory factors were suppressed. In addition, the levels of ROS and MDA were also inhibited and the expression of SOD was promoted after GTS-21 treatment. We also found that the ratios of apoptotic cells declined after GTS-21 treatment. Conclusions GTS-21-induced α7 nAChR decreased the OGD/R-induced inflammatory response, oxidative damage, and apoptosis of intestinal epithelial cells.
Collapse
Affiliation(s)
- Haisong Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Dongmiao Cai
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Zhenyi Chen
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
17
|
Sinagra E, Utzeri E, Morreale GC, Fabbri C, Pace F, Anderloni A. Microbiota-gut-brain axis and its affect inflammatory bowel disease: Pathophysiological concepts and insights for clinicians. World J Clin Cases 2020; 8:1013-1025. [PMID: 32258072 PMCID: PMC7103973 DOI: 10.12998/wjcc.v8.i6.1013] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/14/2020] [Accepted: 03/05/2020] [Indexed: 02/05/2023] Open
Abstract
Despite the bi-directional interaction between gut microbiota and the brain not being fully understood, there is increasing evidence arising from animal and human studies that show how this intricate relationship may facilitate inflammatory bowel disease (IBD) pathogenesis, with consequent important implications on the possibility to improve the clinical outcomes of the diseases themselves, by acting on the different components of this system, mainly by modifying the microbiota. With the emergence of precision medicine, strategies in which patients with IBD might be categorized other than for standard gut symptom complexes could offer the opportunity to tailor therapies to individual patients. The aim of this narrative review is to elaborate on the concept of the gut-brain-microbiota axis and its clinical significance regarding IBD on the basis of recent scientific literature, and finally to focus on pharmacological therapies that could allow us to favorably modify the function of this complex system.
Collapse
Affiliation(s)
- Emanuele Sinagra
- Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, Cefalù 90015, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo 90100, Italy
| | - Erika Utzeri
- Nuova Casa di Cura di Decimomannu, Cagliari 09100, Italy
| | | | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena, Azienda USL Romagna, Forlì 47121, Italy
| | - Fabio Pace
- Unit of Gastroenterology, Bolognini Hospital, Bergamo 24100, Italy
| | - Andrea Anderloni
- Digestive Endoscopy Unit, Division of Gastroenterology, Humanitas Research Hospital, Rozzano 20089, Italy
| |
Collapse
|
18
|
Mogilevski T, Burgell R, Aziz Q, Gibson PR. Review article: the role of the autonomic nervous system in the pathogenesis and therapy of IBD. Aliment Pharmacol Ther 2019; 50:720-737. [PMID: 31418887 DOI: 10.1111/apt.15433] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/25/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND There is a growing body of evidence implicating a role for the brain-gut axis in the pathogenesis of inflammation in patients with IBD. AIMS To perform a narrative review of published literature regarding the association of the autonomic nervous system and intestinal inflammation and to describe the rationale for and emerging use of autonomic manipulation as a therapeutic agent METHODS: Current relevant literature was summarised and critically examined. RESULTS There is substantial pre-clinical and clinical evidence for a multifaceted anti-inflammatory effect of the vagus at both systemic and local intestinal levels. It acts via acetylcholine-mediated activation of α-7-acetylcholine receptors involving multiple cell types in innate and adaptive immunity and the enteric nervous system with subsequent protective influences on the intestinal barrier, inflammatory mechanisms and the microbiome. In patients with IBD, there is evidence for a sympatho-vagal imbalance, functional enteric neuronal depletion and hyporeactivity of the hypothalamic-pituitary-adrenal axis. Direct or transcutaneous vagal neuromodulation up-regulates the cholinergic anti-inflammatory pathway in pre-clinical and clinical models with down-regulation of systemic and local intestinal inflammation. This is supported by two small studies in Crohn's disease although remains to be investigated in ulcerative colitis. CONCLUSIONS Modulating the cholinergic anti-inflammatory pathway influences inflammation both systemically and at a local intestinal level. It represents a potentially underutilised anti-inflammatory therapeutic strategy. Given the likely pathogenic role of the autonomic nervous system in patients with IBD, vagal neuromodulation, an apparently safe and successful means of increasing vagal tone, warrants further clinical exploration.
Collapse
Affiliation(s)
- Tamara Mogilevski
- Centre for Neuroscience, Surgery and Trauma, Barts and the London School of Medicine and Dentistry, Blizard Institute, Wingate Institute of Neurogastroenterology, London, UK.,Barts Health NHS Trust, London, UK.,Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Rebecca Burgell
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Qasim Aziz
- Centre for Neuroscience, Surgery and Trauma, Barts and the London School of Medicine and Dentistry, Blizard Institute, Wingate Institute of Neurogastroenterology, London, UK.,Barts Health NHS Trust, London, UK
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| |
Collapse
|
19
|
Kiers D, Leijte GP, Gerretsen J, Zwaag J, Kox M, Pickkers P. Comparison of different lots of endotoxin and evaluation of in vivo potency over time in the experimental human endotoxemia model. Innate Immun 2019; 25:34-45. [PMID: 30782041 PMCID: PMC6830888 DOI: 10.1177/1753425918819754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022] Open
Abstract
The experimental human endotoxemia model is used to study the systemic inflammatory response in vivo. The previously used lot of endotoxin, which was used for over a decade, is no longer approved for human use and a new Good Manufacturing Practices-grade batch has become available. We compared the inflammatory response induced by either bolus or continuous administration of either the previously used lot #1188844 or new lots of endotoxin (#94332B1 and #94332B4). Compared with lot #1188844, bolus administration of lot #94332B1 induced a more pronounced systemic inflammatory response including higher plasma levels of pro-inflammatory cytokines and more pronounced clinical signs of inflammation. In contrast, continuous infusion of lot #94332B4 resulted in a slightly less pronounced inflammatory response compared with lot #1188844. Furthermore, we evaluated whether lot #1188844 displayed in vivo potency loss by reviewing inflammatory parameters obtained from 17 endotoxemia studies performed in our centre between 2007 and 2016. Despite inter-study variability in endotoxemia-induced effects on temperature, heart rate, symptoms, and leukocyte counts, the magnitude of these effects did not decrease over time. In conclusion, although all lots of endotoxin induce a pronounced inflammatory response, the magnitude differs between lots. We observed no potency loss of endotoxin over time.
Collapse
Affiliation(s)
- Dorien Kiers
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
- Both authors contributed equally to this work (shared first
authorship)
| | - Guus P. Leijte
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
- Both authors contributed equally to this work (shared first
authorship)
| | - Jelle Gerretsen
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
| | - Jelle Zwaag
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
| | - Matthijs Kox
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
| | - Peter Pickkers
- Dept. of Intensive Care Medicine, Radboud University Medical
Center, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
20
|
Zhao L, Xiong Q, Stary CM, Mahgoub OK, Ye Y, Gu L, Xiong X, Zhu S. Bidirectional gut-brain-microbiota axis as a potential link between inflammatory bowel disease and ischemic stroke. J Neuroinflammation 2018; 15:339. [PMID: 30537997 PMCID: PMC6290529 DOI: 10.1186/s12974-018-1382-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence suggests that gut-brain-microbiota axis (GBMAx) may play a pivotal role linking gastrointestinal and neuronal disease. In this review, we summarize the latest advances in studies of GBMAx in inflammatory bowel disease (IBD) and ischemic stroke. A more thorough understanding of the GBMAx could advance our knowledge about the pathophysiology of IBD and ischemic stroke and help to identify novel therapeutic targets via modulation of the GBMAx.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiutang Xiong
- Diabetes Research Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Creed M. Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305 USA
| | | | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Rd, Wuhan, 430060 Hubei China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000 Zhejiang China
| |
Collapse
|
21
|
Kong W, Kang K, Gao Y, Liu H, Meng X, Cao Y, Yang S, Liu W, Zhang J, Yu K, Zhao M. GTS-21 Protected Against LPS-Induced Sepsis Myocardial Injury in Mice Through α7nAChR. Inflammation 2018; 41:1073-1083. [PMID: 29680908 DOI: 10.1007/s10753-018-0759-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis-induced myocardial injury is a well-known cause of mortality. The cholinergic anti-inflammatory pathway (CHAIP) is a physiological mechanism by which the central nervous system regulates immune response through the vagus nerve and acetylcholine; the α7-nicotinic acetylcholine receptor (α7nAChR) is the main component of CHAIP; GTS-21, a synthetic α7nAChR selective agonist, has repeatedly shown its powerful anti-inflammatory effect. However, little is known about its effect on LPS-induced myocardial injury. We investigated the protective effects of GTS-21 on lipopolysaccharide (LPS)-induced cardiomyopathy via the cholinergic anti-inflammatory pathway in a mouse sepsis model. We constructed the model of myocardial injury in sepsis mice by C57BL/6 using LPS and determined the time of LPS treatment by hematoxylin-eosin (HE) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). C57BL/6 mice were randomized into five groups: blank control group, model group, α-bungarotoxin + LPS group, GTS-21 + LPS group, and α-bungarotoxin + GTS-21 + LPS group. The pathological results of myocardial tissue were detected by the HE method; the apoptosis rate was detected by the TUNEL method; the relative expressions of NF-κB p65, Caspase-3, Caspase-8, Bcl-2, Bax, p53, and a7nAChR were detected by real-time quantitative PCR (RT-PCR); and the protein expressions of IL-6, IL-1 β, TNF-α, and pSTAT3 were detected by western blot. The results showed that LPS-induced myocardial pathological and apoptosis changes were significant compared with the blank group, which was reversed by GTS-21; however, pretreatment with α-bungarotoxin obviously blocked the protective effect of GTS-21. NF-κB p65, Caspase-3, Caspase-8, Bax, p53, IL-6, IL-1β, TNF-α, and pSTAT3 were significantly increased in the model group, while a7nAChR and Bcl-2 were significantly decreased; GTS-21 treatment reversed that result, while pretreatment with α-bungarotoxin strengthened the result in the model. And pretreatment with α-bungarotoxin blocked the protective effect of GTS-21. GTS-21 can alleviate the LPS-induced damage in the heart via a7nAChR, and pretreatment with α-bungarotoxin obviously blocked the protective effect of GTS-21 on sepsis in mice.
Collapse
Affiliation(s)
- Weilan Kong
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Kai Kang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Yang Gao
- Department of Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086, China
| | - Haitao Liu
- Department of Critical Care Medicine, the Cancer Hospital of Harbin Medical University, 150 Haping Road, Harbin, 150081, China
| | - Xianglin Meng
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Yanhui Cao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Songliu Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Wen Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Jiannan Zhang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, the Cancer Hospital of Harbin Medical University, 150 Haping Road, Harbin, 150081, China. .,Institute of Critical Care Medicine in Sino Russian Medical Research Center of Harbin Medical University, 150 Haping Road, Harbin, 150081, China.
| | - Mingyan Zhao
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Road, Harbin, 150001, China.
| |
Collapse
|
22
|
Schaller SJ, Nagashima M, Schönfelder M, Sasakawa T, Schulz F, Khan MAS, Kem WR, Schneider G, Schlegel J, Lewald H, Blobner M, Jeevendra Martyn JA. GTS-21 attenuates loss of body mass, muscle mass, and function in rats having systemic inflammation with and without disuse atrophy. Pflugers Arch 2018; 470:1647-1657. [PMID: 30006848 DOI: 10.1007/s00424-018-2180-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/19/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022]
Abstract
Muscle changes of critical illness are attributed to systemic inflammatory responses and disuse atrophy. GTS-21 (3-(2,4-dimethoxy-benzylidene)anabaseine), also known as DMBX-A) is a synthetic derivative of the natural product anabaseine that acts as an agonist at α7-acetylcholine receptors (α7nAChRs). Hypothesis tested was that modulation of inflammation by agonist GTS-21 (10 mg/kg b.i.d. intraperitoneally) will attenuate body weight (BW) and muscle changes. Systemic sham inflammation was produced in 125 rats by Cornyebacterium parvum (C.p.) or saline injection on days 0/4/8. Seventy-four rats had one immobilized-limb producing disuse atrophy. GTS-21 effects on BW, tibialis muscle mass (TMM), and function were assessed on day 12. Systemically, methemoglobin levels increased 26-fold with C.p. (p < 0.001) and decreased significantly (p < 0.033) with GTS-21. Control BW increased (+ 30 ± 9 g, mean ± SD) at day 12, but decreased with C.p. and superimposed disuse (p = 0.005). GTS-21 attenuated BW loss in C.p. (p = 0.005). Compared to controls, TMM decreased with C.p. (0.43 ± 0.06 g to 0.26 ± 0.03 g) and with superimposed disuse (0.18 ± 0.04 g); GTS-21 ameliorated TMM loss to 0.32 ± 0.04 (no disuse, p = 0.028) and to 0.22 ± 0.03 (with disuse, p = 0.004). Tetanic tensions decreased with C.p. or disuse and GTS-21 attenuated tension decrease in animals with disuse (p = 0.006) and in animals with C.p. and disuse (p = 0.029). C.p.-induced 11-fold increased muscle α7nAChR expression was decreased by > 60% with GTS-21 treatment. In conclusion, GTS-21 modulates systemic inflammation, evidenced by both decreased methemoglobin levels and decrease of α7nAChR expression, and mitigates inflammation-mediated loss of BW, TMM, fiber size, and function.
Collapse
Affiliation(s)
- Stefan J Schaller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA.
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany.
| | - Michio Nagashima
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
- Department of Intensive Care Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Martin Schönfelder
- Institute of Exercise Biology, Technische Universität München, Georg-Brauchle-Ring 60/62, 80992, Munich, Germany
| | - Tomoki Sasakawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
- Department of Anesthesiology and Critical Care Medicine, Asahikawa Medical University, 1 Chome-1-1 Midorigaoka Higashi 2 Jō, Asahikawa-shi, Hokkaidō, 078-8802, Japan
| | - Fabian Schulz
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Mohammed A S Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
| | - William R Kem
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, P.O. Box 100267, Gainesville, FL, 32610-0267, USA
| | - Gerhard Schneider
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Jürgen Schlegel
- Institute of Pathology, Technische Universität München, Ismaningerstr. 22, 81675, Munich, Germany
| | - Heidrun Lewald
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Manfred Blobner
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - J A Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
| |
Collapse
|
23
|
An ALPHA7 Nicotinic Acetylcholine Receptor Agonist (GTS-21) Promotes C2C12 Myonuclear Accretion in Association with Release of Interleukin-6 (IL-6) and Improves Survival in Burned Mice. Shock 2018; 48:227-235. [PMID: 28282360 DOI: 10.1097/shk.0000000000000849] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of interleukin-6 (IL-6) in physiological processes and disease is poorly understood. The hypothesis tested in this study was that selective alpha7 acetylcholine receptor (α7AChR) agonist, GTS-21, releases IL-6 in association with myonuclear accretion and enhances insulin signaling in muscle cells, and improves survival of burn injured (BI) mice. The in vitro effects of GTS-21 were determined in C2C12 myoblasts and 7-day differentiated myotubes (myotubes). The in vivo effects of GTS-21 were tested in BI wild-type (WT) and IL-6 knockout (IL6KO) mice. GTS-21 dose-dependently (0 μM, 100 μM, and 200 μM) significantly increased IL-6 levels in myoblasts and myotubes at 6 and 9 h. GTS-21-induced IL-6 release in myotubes was attenuated by methyllycaconitine (α7AChR antagonist), and by Stat-3 or Stat-5 inhibitors. GTS-21 increased MyoD and Pax7 protein expression, myonuclear accretion, and insulin-induced phosphorylation of Akt, GSK-3β, and Glut4 in myotubes. The glucose levels of burned IL6KO mice receiving GTS-21 decreased significantly compared with sham-burn IL6KO mice. Superimposition of BI on IL6KO mice caused 100% mortality; GTS-21 reduced mortality to 75% in the IL6KO mice. The 75% mortality in burned WT mice was reduced to 0% with GTS-21. The in vitro findings suggest that GTS-21-induced IL-6 release from muscle is mediated via α7AChRs upstream of Stat-3 and -5 pathways and is associated with myonuclear accretion, possibly via MyoD and Pax7 expression. GTS-21 in vivo improves survival in burned WT mice and IL6KO mice, suggesting a potential therapeutic application of α7AChR agonists in the treatment of BI.
Collapse
|
24
|
Prevention of Burn-Induced Inflammatory Responses and Muscle Wasting by GTS-21, a Specific Agonist for α7 Nicotinic Acetylcholine Receptors. Shock 2018; 47:61-69. [PMID: 27529131 DOI: 10.1097/shk.0000000000000729] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Muscle wasting (MW) in catabolic conditions (e.g., burn injury [BI]) is a major risk factor affecting prognosis. Activation of interleukin-1β (IL-1β)/nuclear factor-kappa B (NF-κB), interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3), and/or forkhead box O transcriptional factor (FOXO)-mediated gene transcription pathways is the pivotal trigger of inflammatory response-induced protein catabolic processes in muscle. The α7 acetylcholine receptors (α7AChRs) are upregulated in macrophages and peripheral tissues including skeletal muscle during MW conditions. Stimulation of α7AChRs mitigates inflammatory responses. Hypothesis tested is that attenuation of inflammation by α7AChR stimulation with specific α7AChR agonist, GTS-21, will reverse BI-induced body mass and MW by modulating inflammatory and proteolytic signals. METHODS Body surface area (30%) BI or sham BI mice were treated with GTS-21 or saline. Tibialis anterior (TA) muscle was harvested at 6 h, day 1 or 3 to examine inflammatory and proteolytic signals. RESULTS GTS-21 significantly ameliorated the BI-induced increased expression of inflammatory cytokines IL-6, IL-1β, C-X-C motif chemokine ligand 2 (6 h), phosphorylated STAT3, and NF-κB (day 1) in TA muscle. GTS-21 also significantly inhibited BI-induced increase of MuRF1 and FOXO1 (day 1). Consistent with the cytokine and inflammatory mediator changes, BI-induced body weight and TA muscle mass loss at day 3 were mitigated by GTS-21 treatment. The beneficial effect of GTS-21 on BI changes was absent in methyllycaconitine (α7AChR antagonist)-treated wild-type and α7AChR knockout mice. CONCLUSION GTS-21 stimulation of α7AChRs, by modulating multiple molecular signals related to inflammation and proteolysis, attenuates protein wasting, evidenced by maintenance of body weight and attenuation of distant muscle mass loss after BI. GTS-21 can be a novel, potent therapeutic option for reversal of BI-induced MW.
Collapse
|
25
|
Seyedabadi M, Rahimian R, Ghia JE. The role of alpha7 nicotinic acetylcholine receptors in inflammatory bowel disease: involvement of different cellular pathways. Expert Opin Ther Targets 2018; 22:161-176. [PMID: 29298542 DOI: 10.1080/14728222.2018.1420166] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Autonomic imbalance plays a pivotal role in the pathophysiology of inflammatory bowel diseases (IBD). The central nervous system (CNS) cooperates dynamically with the immune system to regulate inflammation through humoral and neural pathways. In particular, acetylcholine (Ach), the main neurotransmitter in the vagus nerve, decreases the production of pro-inflammatory cytokines through a mechanism dependent on the α7 nicotinic Ach receptors (α7nAChRs). Areas covered: Here, we review the evidence for involvement of the cholinergic anti-inflammatory pathway (CAP) in IBD. We also elaborate the role of α7nAChRs and subsequent cellular pathways in CAP. Finally, we review potential therapeutic implications of modulators of these receptors. Expert opinion: Alpha7nAChR modulators possess both cognitive improving and anti-inflammatory properties. Although, these agents demonstrated therapeutic benefits in experimental models, their efficacy has not always been translated in clinical trials. Thus, development of more specific α7nAChR ligands as well as more experimental studies and better controlled trials, especially in the field of IBD, are encouraged for a progress in this field.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- a Department of Pharmacology, School of Medicine , Bushehr University of Medical Sciences , Bushehr , Iran.,b The Persian Gulf Biomedical Sciences Research Institute , Bushehr University of Medical Sciences , Bushehr , Iran.,c Education Development Center , Bushehr University of Medical Sciences , Bushehr , Iran
| | - Reza Rahimian
- d Department of Psychiatry and Neuroscience, Faculty of Medicine , CERVO Brain Research Center, Laval University , Quebec , Quebec , Canada
| | - Jean-Eric Ghia
- e Department of Immunology , University of Manitoba , Winnipeg , Manitoba , Canada.,f Department of Internal Medicine Section of Gastroenterology, and Inflammatory Bowel Disease Clinical & Research Center , University of Manitoba , Winnipeg , Manitoba , Canada
| |
Collapse
|
26
|
Schmidt K, Bhakdisongkhram S, Uhle F, Philipsenburg C, Zivkovic AR, Brenner T, Motsch J, Weigand MA, Hofer S. GTS-21 reduces microvascular permeability during experimental endotoxemia. Microvasc Res 2017; 115:75-82. [PMID: 28818494 DOI: 10.1016/j.mvr.2017.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/03/2017] [Accepted: 08/12/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION No effective pharmacological therapy is currently available to attenuate tissue edema formation due to increased microvascular permeability in sepsis. Cholinergic mediators have been demonstrated to exert anti-inflammatory effects via the α7 nicotinic acetylcholine receptor (α7nAChR) during inflammation. GTS-21, a partial α7nAChR agonist, is an appealing therapeutic substance for sepsis-induced microvascular inflammation due to its demonstrated cholinergic anti-inflammatory properties and its favorable safety profile in clinical trials. This study evaluated the effect of GTS-21 on microvascular permeability and leukocyte adhesion during experimental endotoxemia. METHODS Male Wistar rats (n=60) were anesthetized and prepared for intravital microscopy (IVM). Sevoflurane inhalation combined with propofol (10mg/kg) and fentanyl (5μg/kg) was used for anesthesia induction, followed by continuous intravenous anesthesia with propofol (10-40mg/kg/h) and fentanyl (10μg/kg/h). The rat mesentery was prepared for evaluation of macromolecular leakage, leukocyte adhesion and venular wall shear rate in postcapillary venules using IVM. Following baseline IVM recording, GTS-21 (1mg/kg) was applied simultaneously with, 1h prior to and 1h after administration of lipopolysaccharide (LPS, 5mg/kg). Test substances (crystalloid solution, LPS, GTS-21) were administered as volume equivalent intravenous infusions over 5min in the respective treatment groups. The consecutive IVMs were performed at 60, 120 and 180min after the baseline IVM. The systemic inflammatory response was evaluated by measuring TNF-α levels after the 180min IVM. RESULTS Microvascular permeability was significantly reduced in animals treated with GTS-21 simultaneously and 1h after induction of endotoxemia. Leukocyte adhesion, venular wall shear rate and TNF-α levels were not affected by GTS-21 treatment compared to the untreated endotoxemic animals. CONCLUSION GTS-21 has a protective effect on microvascular barrier function during endotoxemia. Considering its anti-inflammatory efficacy and safety profile, its clinical use might prove beneficial for the treatment of capillary leakage in sepsis therapy.
Collapse
Affiliation(s)
- Karsten Schmidt
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Sukanya Bhakdisongkhram
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Christoph Philipsenburg
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Aleksandar R Zivkovic
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Johann Motsch
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Stefan Hofer
- Clinic for Anesthesiology, Intensive Care and Emergency Medicine I, Westpfalz Hospital, Hellmut-Hartert-Str. 1, 67655 Kaiserslautern, Germany.
| |
Collapse
|
27
|
Hoover DB. Cholinergic modulation of the immune system presents new approaches for treating inflammation. Pharmacol Ther 2017; 179:1-16. [PMID: 28529069 DOI: 10.1016/j.pharmthera.2017.05.002] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The nervous system and immune system have broad and overlapping distributions in the body, and interactions of these ubiquitous systems are central to the field of neuroimmunology. Over the past two decades, there has been explosive growth in our understanding of neuroanatomical, cellular, and molecular mechanisms that mediate central modulation of immune functions through the autonomic nervous system. A major catalyst for growth in this field was the discovery that vagal nerve stimulation (VNS) caused a prominent attenuation of the systemic inflammatory response evoked by endotoxin in experimental animals. This effect was mediated by acetylcholine (ACh) stimulation of nicotinic receptors on splenic macrophages. Hence, the circuit was dubbed the "cholinergic anti-inflammatory pathway". Subsequent work identified the α7 nicotinic ACh receptor (α7nAChR) as the crucial target for attenuation of pro-inflammatory cytokine release from macrophages and dendritic cells. Further investigation made the important discovery that cholinergic T cells within the spleen and not cholinergic nerve cells were the source of ACh that stimulated α7 receptors on splenic macrophages. Given the important role that inflammation plays in numerous disease processes, cholinergic anti-inflammatory mechanisms are under intensive investigation from a basic science perspective and in translational studies of animal models of diseases such as inflammatory bowel disease and rheumatoid arthritis. This basic work has already fostered several clinical trials examining the efficacy of VNS and cholinergic therapeutics in human inflammatory diseases. This review provides an overview of basic and translational aspects of the cholinergic anti-inflammatory response and relevant pharmacology of drugs acting at the α7nAChR.
Collapse
Affiliation(s)
- Donald B Hoover
- Department of Biomedical Sciences and Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.
| |
Collapse
|
28
|
Ren C, Tong YL, Li JC, Lu ZQ, Yao YM. The Protective Effect of Alpha 7 Nicotinic Acetylcholine Receptor Activation on Critical Illness and Its Mechanism. Int J Biol Sci 2017; 13:46-56. [PMID: 28123345 PMCID: PMC5264260 DOI: 10.7150/ijbs.16404] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022] Open
Abstract
Critical illnesses and injuries are recognized as major threats to human health, and they are usually accompanied by uncontrolled inflammation and dysfunction of immune response. The alpha 7 nicotinic acetylcholine receptor (α7nAchR), which is a primary receptor of cholinergic anti-inflammatory pathway (CAP), exhibits great benefits for critical ill conditions. It is composed of 5 identical α7 subunits that form a central pore with high permeability for calcium. This putative structure is closely associated with its functional states. Activated α7nAChR exhibits extensive anti-inflammatory and immune modulatory reactions, including lowered pro-inflammatory cytokines levels, decreased expressions of chemokines as well as adhesion molecules, and altered differentiation and activation of immune cells, which are important in maintaining immune homeostasis. Well understanding of the effects and mechanisms of α7nAChR will be of great value in exploring effective targets for treating critical diseases.
Collapse
Affiliation(s)
- Chao Ren
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Ya-Lin Tong
- Department of Burns and Plastic Surgery, the 181st Hospital of Chinese PLA, Guilin 541002, People's Republic of China
| | - Jun-Cong Li
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Zhong-Qiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.; State Key Laboratory of Kidney Disease, the Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| |
Collapse
|
29
|
Georgin-Lavialle S, Moura DS, Salvador A, Chauvet-Gelinier JC, Launay JM, Damaj G, Côté F, Soucié E, Chandesris MO, Barète S, Grandpeix-Guyodo C, Bachmeyer C, Alyanakian MA, Aouba A, Lortholary O, Dubreuil P, Teyssier JR, Trojak B, Haffen E, Vandel P, Bonin B, Hermine O, Gaillard R. Mast cells' involvement in inflammation pathways linked to depression: evidence in mastocytosis. Mol Psychiatry 2016; 21:1511-1516. [PMID: 26809839 DOI: 10.1038/mp.2015.216] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/24/2015] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
Converging sources of evidence point to a role for inflammation in the development of depression, fatigue and cognitive dysfunction. More precisely, the tryptophan (TRP) catabolism is thought to play a major role in inflammation-induced depression. Mastocytosis is a rare disease in which chronic symptoms, including depression, are related to mast cell accumulation and activation. Our objectives were to study the correlations between neuropsychiatric features and the TRP catabolism pathway in mastocytosis in order to demonstrate mast cells' potential involvement in inflammation-induced depression. Fifty-four patients with mastocytosis and a mean age of 50.1 years were enrolled in the study and compared healthy age-matched controls. Depression and stress were evaluated with the Beck Depression Inventory revised and the Perceived Stress Scale. All patients had measurements of TRP, serotonin (5-HT), kynurenine (KYN), indoleamine 2,3-dioxygenase 1 (IDO1) activity (ratio KYN/TRP), kynurenic acid (KA) and quinolinic acid (QA). Patients displayed significantly lower levels of TRP and 5-HT without hypoalbuminemia or malabsorption, higher IDO1 activity, and higher levels of KA and QA, with an imbalance towards the latter. High perceived stress and high depression scores were associated with low TRP and high IDO1 activity. In conclusion, TRP metabolism is altered in mastocytosis and correlates with perceived stress and depression, demonstrating mast cells' involvement in inflammation pathways linked to depression.
Collapse
Affiliation(s)
- S Georgin-Lavialle
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - D S Moura
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Laboratoire de Psychopathologie et Processus de Santé, EA 4057, IUPDP, Institut de Psychologie, Université Paris Descartes, Paris, France
| | - A Salvador
- Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France
| | - J-C Chauvet-Gelinier
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | - J-M Launay
- Laboratoire de biochimie et biologie moléculaire, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - G Damaj
- Service des Maladies du Sang, Centre Hospitalier Universitaire, Hôpital Sud, Amiens, France
| | - F Côté
- INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - E Soucié
- INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - M-O Chandesris
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - S Barète
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - C Grandpeix-Guyodo
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - C Bachmeyer
- Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M-A Alyanakian
- Laboratoire d'immunologie, Hôpital Necker, Paris, France
| | - A Aouba
- Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - O Lortholary
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Service des infectieuses et tropicales, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants malades, Paris, France
| | - P Dubreuil
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - J-R Teyssier
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France
| | - B Trojak
- Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Service de Psychiatrie, Département de Neurosciences, Dijon, France
| | - E Haffen
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France.,Service de Psychiatrie, CHU de Besançon, Besançon, France.,Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France
| | - P Vandel
- Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France.,Centre d'Investigation Clinique CIC-IT 808 INSERM, CHU de Besaçon, Besançon, France
| | - B Bonin
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | | | - O Hermine
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - R Gaillard
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Human Histopathology and Animal Models, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| |
Collapse
|
30
|
Galle-Treger L, Suzuki Y, Patel N, Sankaranarayanan I, Aron JL, Maazi H, Chen L, Akbari O. Nicotinic acetylcholine receptor agonist attenuates ILC2-dependent airway hyperreactivity. Nat Commun 2016; 7:13202. [PMID: 27752043 PMCID: PMC5071851 DOI: 10.1038/ncomms13202] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
Allergic asthma is a complex and chronic inflammatory disorder that is associated with airway hyperreactivity (AHR) and driven by Th2 cytokine secretion. Type 2 innate lymphoid cells (ILC2s) produce large amounts of Th2 cytokines and contribute to the development of AHR. Here, we show that ILC2s express the α7-nicotinic acetylcholine receptor (α7nAChR), which is thought to have an anti-inflammatory role in several inflammatory diseases. We show that engagement of a specific agonist with α7nAChR on ILC2s reduces ILC2 effector function and represses ILC2-dependent AHR, while decreasing expression of ILC2 key transcription factor GATA-3 and critical inflammatory modulator NF-κB, and reducing phosphorylation of upstream kinase IKKα/β. Additionally, the specific α7nAChR agonist reduces cytokine production and AHR in a humanized ILC2 mouse model. Collectively, our data suggest that α7nAChR expressed by ILC2s is a potential therapeutic target for the treatment of ILC2-mediated asthma. Airway hyperreactivity is driven by type 2 cytokines produced by ILC2 and Th2 cells. Here the authors show that an α7-nicotinic receptor agonist (GTS-21) inhibits ILC2 responses and is therapeutic against Alternaria-induced airway hyperreactivity in a humanized mouse model.
Collapse
Affiliation(s)
- Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Yuzo Suzuki
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Nisheel Patel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Ishwarya Sankaranarayanan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Jennifer L Aron
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Lin Chen
- Departments of Biological Science and Chemistry, University of Southern California, 1050 Childs Way RIH 201, Los Angeles, California 90089, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| |
Collapse
|
31
|
Abstract
Research during the last decade has significantly advanced our understanding of the molecular mechanisms at the interface between the nervous system and the immune system. Insight into bidirectional neuro-immune communication has characterized the nervous system as an important partner of the immune system in the regulation of inflammation. Neuronal pathways, including the vagus nerve-based inflammatory reflex, are physiological regulators of immune function and inflammation. In parallel, neuronal function is altered in conditions characterized by immune dysregulation and inflammation. Here, we review these regulatory mechanisms and describe the neural circuitry modulating immunity. Understanding these mechanisms reveals possibilities to use targeted neuromodulation as a therapeutic approach for inflammatory and autoimmune disorders. These findings and current clinical exploration of neuromodulation in the treatment of inflammatory diseases define the emerging field of Bioelectronic Medicine.
Collapse
Affiliation(s)
- Valentin A Pavlov
- Center for Biomedical Science, The Feinstein Institute for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
| | - Kevin J Tracey
- Center for Biomedical Science, The Feinstein Institute for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
| |
Collapse
|
32
|
Abstract
Inflammation and immunity are regulated by neural reflexes. Recent basic science research has demonstrated that a neural reflex, termed the inflammatory reflex, modulates systemic and regional inflammation in a multiplicity of clinical conditions encountered in perioperative medicine and critical care. In this review, the authors describe the anatomic and physiologic basis of the inflammatory reflex and review the evidence implicating this pathway in the modulation of sepsis, ventilator-induced lung injury, postoperative cognitive dysfunction, myocardial ischemia-reperfusion injury, and traumatic hemorrhage. The authors conclude with a discussion of how these new insights might spawn novel therapeutic strategies for the treatment of inflammatory diseases in the context of perioperative and critical care medicine.
Collapse
|
33
|
Georgin-Lavialle S, Gaillard R, Moura D, Hermine O. Mastocytosis in adulthood and neuropsychiatric disorders. Transl Res 2016; 174:77-85.e1. [PMID: 27063957 DOI: 10.1016/j.trsl.2016.03.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/04/2016] [Accepted: 03/15/2016] [Indexed: 12/18/2022]
Abstract
Patients with mastocytosis can display various disabling general and neuropsychological symptoms among one third of them, including general signs such as fatigue and musculoskeletal pain, which can have a major impact on quality of life. Neurological symptoms are less frequent and mainly consist of acute or chronic headache (35%), rarely syncopes (5%), acute onset back pain (4%), and in a few cases, clinical and radiological symptoms resembling or allowing the diagnosis of multiple sclerosis (1.3%). Headaches are associated with symptoms related to mast cell activation syndrome (flushes, prurit, and so forth) and more frequently present as migraine (37.5%), with often aura (66%). Depression-anxiety like symptoms can occur in 40% to 60% of the patients and cognitive impairment is not rare (38.6%). The pathophysiology of these symptoms could be linked to tissular mast cell infiltration or to mast cell mediators release or both. The tryptophan metabolism could be involved in mast cell-induced neuroinflammation through indoleamine-2,3-dioxygenase activation. Treatments targeting mast cell may be useful to target neuropsychological features associated with mastocytosis, including tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Sophie Georgin-Lavialle
- Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Raphaël Gaillard
- Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France; Human Histopathology and Animal Models, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - Daniela Moura
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Paris, France
| | - Olivier Hermine
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Paris, France; INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders Hôpital Necker-Enfants malades, Institut Imagine, Paris, France; Service d'hématologie adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France.
| |
Collapse
|
34
|
Goverse G, Stakenborg M, Matteoli G. The intestinal cholinergic anti-inflammatory pathway. J Physiol 2016; 594:5771-5780. [PMID: 26959627 DOI: 10.1113/jp271537] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/02/2016] [Indexed: 01/10/2023] Open
Abstract
The main task of the immune system is to distinguish and respond accordingly to 'danger' or 'non-danger' signals. This is of critical importance in the gastrointestinal tract in which immune cells are constantly in contact with food antigens, symbiotic microflora and potential pathogens. This complex mixture of food antigens and symbionts are essential for providing vital nutrients, so they must be tolerated by the intestinal immune system to prevent aberrant inflammation. Therefore, in the gut the balance between immune activation and tolerance should be tightly regulated to maintain intestinal homeostasis and to prevent hypersensitivity to harmless luminal antigens. Loss of this delicate equilibrium can lead to abnormal activation of the intestinal immune system resulting in devastating gastrointestinal disorders such as inflammatory bowel disease (IBD). Recent evidence supports the idea that the central nervous system interacts dynamically via the vagus nerve with the intestinal immune system to modulate inflammation through humoral and neural pathways, using a mechanism also referred to as the intestinal cholinergic anti-inflammatory pathway. In this review, we will focus on the current understanding of the mechanisms and neuronal circuits involved in the intestinal cholinergic anti-inflammatory pathway. Further investigation on the crosstalk between the nervous and intestinal immune system will hopefully provide new insights leading to the identification of innovative therapeutic approaches to treat intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Gera Goverse
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Michelle Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.
| |
Collapse
|
35
|
Abstract
The vagus nerve can sense peripheral inflammation and transmit action potentials from the periphery to the brainstem. Vagal afferent signaling is integrated in the brainstem, and efferent vagus nerves carry outbound signals that terminate in spleen and other organs. Stimulation of efferent vagus nerve leads to the release of acetylcholine in these organs. In turn, acetylcholine interacts with members of the nicotinic acetylcholine receptor (nAChR) family, particularly with the alpha7 nicotinic acetylcholine receptor (α7nAChR), which is expressed by macrophages and other cytokine-producing cells. Ultimately, the production of proinflammatory cytokines is markedly inhibited. This neuroimmune communication is termed "the inflammatory reflex". The uncontrolled inflammation as a result from sepsis can lead to multiple organ failure, and even death. Experimental data show that regulation of the inflammatory reflex appears to be a useful interventional strategy for septic response. Herein, we review recent advances in the understanding of the inflammatory reflex and discuss potential therapeutics that vagal modulation of the immune system for the treatment of severe sepsis and septic shock.
Collapse
Affiliation(s)
- Da-Wei Wang
- a Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital , Beijing , China.,b Department of ICU , Weihai Municipal Hospital , Weihai , China
| | - Yi-Mei Yin
- b Department of ICU , Weihai Municipal Hospital , Weihai , China
| | - Yong-Ming Yao
- a Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital , Beijing , China
| |
Collapse
|
36
|
Anti-inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection. Biochem Pharmacol 2015; 97:463-472. [DOI: 10.1016/j.bcp.2015.07.032] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/27/2015] [Indexed: 12/15/2022]
|
37
|
Kox M, van Eijk LT, Verhaak T, Frenzel T, Kiers HD, Gerretsen J, van der Hoeven JG, Kornet L, Scheiner A, Pickkers P. Transvenous vagus nerve stimulation does not modulate the innate immune response during experimental human endotoxemia: a randomized controlled study. Arthritis Res Ther 2015; 17:150. [PMID: 26049730 PMCID: PMC4480894 DOI: 10.1186/s13075-015-0667-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/29/2015] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Vagus nerve stimulation (VNS) exerts beneficial anti-inflammatory effects in various animal models of inflammation, including collagen-induced arthritis, and is implicated in representing a novel therapy for rheumatoid arthritis. However, evidence of anti-inflammatory effects of VNS in humans is very scarce. Transvenous VNS (tVNS) is a newly developed and less invasive method to stimulate the vagus nerve. In the present study, we determined whether tVNS is a feasible and safe procedure and investigated its putative anti-inflammatory effects during experimental human endotoxemia. METHODS We performed a randomized double-blind sham-controlled study in healthy male volunteers. A stimulation catheter was inserted in the left internal jugular vein at spinal level C5-C7, adjacent to the vagus nerve. In the tVNS group (n = 10), stimulation was continuously performed for 30 minutes (0-10 V, 1 ms, 20 Hz), starting 10 minutes before intravenous administration of 2 ng kg(-1) Escherichia coli lipopolysaccharide (LPS). Sham-instrumented subjects (n = 10) received no electrical stimulation. RESULTS No serious adverse events occurred throughout the study. In the tVNS group, stimulation of the vagus nerve was achieved as indicated by laryngeal vibration. Endotoxemia resulted in fever, flu-like symptoms, and hemodynamic changes that were unaffected by tVNS. Furthermore, plasma levels of inflammatory cytokines increased sharply during endotoxemia, but responses were similar between groups. Finally, cytokine production by leukocytes stimulated with LPS ex vivo, as well as neutrophil phagocytosis capacity, were not influenced by tVNS. CONCLUSIONS tVNS is feasible and safe, but does not modulate the innate immune response in humans in vivo during experimental human endotoxemia. TRIAL REGISTRATION Clinicaltrials.gov NCT01944228. Registered 12 September 2013.
Collapse
Affiliation(s)
- Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lucas T van Eijk
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Verhaak
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Tim Frenzel
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Harmke D Kiers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| | - Lilian Kornet
- Medtronic Inc., Endepolsdomein 5, 6229, GW, Maastricht, The Netherlands.
| | - Avram Scheiner
- Medtronic Inc., 8200 Coral Sea St NE, Mounds View, MN, 55112, USA.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Geert Grooteplein 10, 6500, HB, Nijmegen, The Netherlands.
| |
Collapse
|
38
|
Findlay JS, Ulaeto D, D'Elia RV. Cytokines and viral hemorrhagic fever: potential for therapeutic intervention. Future Virol 2015. [DOI: 10.2217/fvl.15.5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT The recent Ebola outbreak in West Africa highlights the need to improve our understanding of why viral hemorrhagic fevers (VHFs) are so devastating. There is a requirement to generate effective prophylactics, such as vaccines, and therapies, especially those that are effective postsymptomatically. For a range of pathogens, it appears that overstimulation of pro-inflammatory cytokines, the ‘cytokine storm’, causes serious immunopathology in patients. In this review, we will focus on the cytokine response following infection by representatives of the viruses which can cause VHF: Ebola virus and Marburg virus, Crimean–Congo hemorrhagic fever virus, Dengue virus, Junin and Lassa virus. Specifically, the role of the cytokine storm in causing VHF and the use of therapeutic immunomodulatory compounds to help treat these fatal and debilitating diseases will be explored.
Collapse
Affiliation(s)
- James S Findlay
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| | - David Ulaeto
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| | - Riccardo V D'Elia
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| |
Collapse
|
39
|
Li MJ, Niu JK, Miao YL. Relationship between brain-gut axis and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2015; 23:1097-1103. [DOI: 10.11569/wcjd.v23.i7.1097] [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] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is a chronic relapsing inflammatory disease affecting the gastrointestinal tract. The incidence of IBD has increased dramatically year by year in China. Currently, the IBD research is focused on genetically predisposed factors, immune response, environmental triggers and infections. However, the etiology of IBD is still unclear. Recently, more attention has been paid to the research of neural regulation affecting the progression of IBD. Previous research has revealed that psycho-neuro-endocrine-immune modulation through the brain-gut axis plays a crucial role in the pathogenesis of IBD. It is important to explore other psychotherapies applied to adjutant therapy in IBD. This review reviews the recent advances in understanding the relationship between the brain-gut axis and inflammatory bowel disease.
Collapse
|
40
|
Cedillo JL, Arnalich F, Martín-Sánchez C, Quesada A, Rios JJ, Maldifassi MC, Atienza G, Renart J, Fernández-Capitán C, García-Rio F, López-Collazo E, Montiel C. Usefulness of α7 nicotinic receptor messenger RNA levels in peripheral blood mononuclear cells as a marker for cholinergic antiinflammatory pathway activity in septic patients: results of a pilot study. J Infect Dis 2014; 211:146-55. [PMID: 25092899 DOI: 10.1093/infdis/jiu425] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Stimulation of the vagus nerve in the so-called cholinergic antiinflammatory pathway (CAP) attenuates systemic inflammation, improving survival in animal sepsis models via α7 nicotinic acetylcholine receptors on immunocompetent cells. Because the relevance of this regulatory pathway is unknown in human sepsis, this pilot study assessed whether the α7 gene expression level in septic patients' peripheral blood mononuclear cells (PBMC) might be used to assess CAP activity and clinical outcome. METHODS The PBMCs α7 messenger RNA levels were determined by real-time quantitative reverse-transcription polymerase chain reaction in 33 controls and 33 patients at enrollment and after their hospital discharge. Data were analyzed to find significant associations between α7 level, vagally mediated heart rate variability as an indirect reflection of CAP activity, serum concentrations of different inflammation markers, and clinical course. RESULTS Septic patients' α7 levels were significantly increased and returned to control values after recovery. These α7 levels correlated directly with the vagal heart input and inversely with the magnitude of the patient's inflammatory state, disease severity, and clinical outcome. CONCLUSIONS This study reveals that the PBMC α7 gene expression level is a clinically relevant marker for CAP activity in sepsis: the higher the α7 expression, the better the inflammation control and the prognosis.
Collapse
Affiliation(s)
- José L Cedillo
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma
| | | | | | | | | | - María C Maldifassi
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma
| | - Gema Atienza
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma
| | - Jaime Renart
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma, Instituto de Investigacion Sanitaria IdiPAZ, Madrid, Spain
| | | | | | - Eduardo López-Collazo
- Laboratory of Tumor Immunology, Unidad de Investigación, Hospital Universitario La Paz
| | - Carmen Montiel
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma
| |
Collapse
|
41
|
Dal-Pizzol F, Tomasi CD, Ritter C. Septic encephalopathy: does inflammation drive the brain crazy? REVISTA BRASILEIRA DE PSIQUIATRIA 2014; 36:251-8. [DOI: 10.1590/1516-4446-2013-1233] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/11/2013] [Indexed: 11/21/2022]
|
42
|
Clark RB, Lamppu D, Libertine L, McDonough A, Kumar A, LaRosa G, Rush R, Elbaum D. Discovery of novel 2-((pyridin-3-yloxy)methyl)piperazines as α7 nicotinic acetylcholine receptor modulators for the treatment of inflammatory disorders. J Med Chem 2014; 57:3966-83. [PMID: 24814197 DOI: 10.1021/jm5004599] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Herein we report the design, synthesis, and structure-activity relationships for a new class of α7 nicotinic acetylcholine receptor (nAChR) modulators based on the 2-((pyridin-3-yloxy)methyl)piperazine scaffold. The oxazolo[4,5-b]pyridine, (R)-18, and 4-methoxyphenylurea, (R)-47, were identified as potent and selective modulators of the α7 nAChR with favorable in vitro safety profiles and good oral bioavailability in mouse. Both compounds were shown to significantly inhibit cellular infiltration in a murine model of allergic lung inflammation. Despite the structural and in vivo functional similarities in the compounds, only (R)-18 was shown to be an agonist. Compound (R)-47 demonstrated silent agonist activity. These data support the hypothesis that the anti-inflammatory activity of the α7 nAChR is mediated by a signal transduction pathway that is independent of ion current.
Collapse
Affiliation(s)
- Roger B Clark
- Critical Therapeutics, Inc. , 60 Westview Street, Lexington, Massachusetts 02421, United States
| | | | | | | | | | | | | | | |
Collapse
|
43
|
The role of α7 nicotinic acetylcholine receptor in modulation of heart rate dynamics in endotoxemic rats. PLoS One 2013; 8:e82251. [PMID: 24340009 PMCID: PMC3858293 DOI: 10.1371/journal.pone.0082251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/22/2013] [Indexed: 01/10/2023] Open
Abstract
Previous reports have indicated that artificial stimulation of the vagus nerve reduces systemic inflammation in experimental models of sepsis. This phenomenon is a part of a broader cholinergic anti-inflammatory pathway which activates the vagus nerve to modulate inflammation through activation of alpha7 nicotinic acetylcholine receptors (α7nACHR). Heart rate variability represents the complex interplay between autonomic nervous system and cardiac pacemaker cells. Reduced heart rate variability and increased cardiac cycle regularity is a hallmark of clinical conditions that are associated with systemic inflammation (e.g. endotoxemia and sepsis). The present study was aimed to assess the role of α7nACHR in modulation of heart rate dynamics during systemic inflammation. Systemic inflammation was induced by injection of endotoxin (lipopolysaccharide) in rats. Electrocardiogram and body temperature were recorded in conscious animals using a telemetric system. Linear and non-linear indices of heart rate variability (e.g. sample entropy and fractal-like temporal structure) were assessed. RT-PCR and immunohistochemistry studies showed that α7nACHR is expressed in rat atrium and is mainly localized at the endothelial layer. Systemic administration of an α7nACHR antagonist (methyllycaconitine) did not show a significant effect on body temperature or heart rate dynamics in naïve rats. However, α7nACHR blockade could further reduce heart rate variability and elicit a febrile response in endotoxemic rats. Pre-treatment of endotoxemic animals with an α7nACHR agonist (PHA-543613) was unable to modulate heart rate dynamics in endotoxemic rats but could prevent the effect of endotoxin on body temperature within 24 h experiment. Neither methyllycaconitine nor PHA-543613 could affect cardiac beating variability of isolated perfused hearts taken from control or endotoxemic rats. Based on our observations we suggest a tonic role for nicotinic acetylcholine receptors in modulation of heart rate dynamics during systemic inflammation.
Collapse
|
44
|
Sussman ES, Kellner CP, McDowell MM, Bruce SS, Heuts SG, Zhuang Z, Bruce RA, Claassen J, Connolly ES. Alpha-7 nicotinic acetylcholine receptor agonists in intracerebral hemorrhage: an evaluation of the current evidence for a novel therapeutic agent. Neurosurg Focus 2013; 34:E10. [PMID: 23634914 DOI: 10.3171/2013.2.focus1315] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intracerebral hemorrhage (ICH) is the most deadly and least treatable subtype of stroke, and at the present time there are no evidence-based therapeutic interventions for patients with this disease. Secondary injury mechanisms are known to cause substantial rates of morbidity and mortality following ICH, and the inflammatory cascade is a major contributor to this post-ICH secondary injury. The alpha-7 nicotinic acetylcholine receptor (α7-nAChR) agonists have a well-established antiinflammatory effect and have been shown to attenuate perihematomal edema volume and to improve functional outcome in experimental ICH. The authors evaluate the current evidence for the use of an α7-nAChR agonist as a novel therapeutic agent in patients with ICH.
Collapse
Affiliation(s)
- Eric S Sussman
- Department of Neurological Surgery, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Loram LC, Taylor FR, Strand KA, Maier SF, Speake JD, Jordan KG, James JW, Wene SP, Pritchard RC, Green H, Van Dyke K, Mazarov A, Letchworth SR, Watkins LR. Systemic administration of an alpha-7 nicotinic acetylcholine agonist reverses neuropathic pain in male Sprague Dawley rats. THE JOURNAL OF PAIN 2013. [PMID: 23182225 DOI: 10.1016/j.jpain.2012.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
UNLABELLED Alpha-7 nicotinic acetylcholine receptor (α7 nAChR) agonists attenuate pain and inflammation in preclinical models. This study tested whether systemic delivery of an α7 nAChR agonist attenuates neuropathic pain and associated immune-mediated pro-inflammation. Hind paw response thresholds to mechanical stimuli in male Sprague Dawley rats were assessed before and after sciatic chronic constriction injury (CCI) or sham surgery. Osmotic mini-pumps containing TC-7020, an α7 nAChR selective agonist, were implanted 10 to 14 days after surgery. TC-7020 (1, 3, and 10 mg/kg/d; s.c.) significantly attenuated CCI-induced allodynia, which lasted through 2 weeks of test compound administration. Spinal cords were collected after 2 weeks and processed for microglial and astrocyte activation markers within the ipsilateral L4-L6 dorsal horn. In addition, ipsilateral L4-5 dorsal root ganglia (DRGs) were processed for neuronal injury and satellite cell activation markers. CCI-induced central glial cell activation markers were not suppressed by TC-7020, even though TC-7020 is mildly blood-brain barrier permeable. However, TC-7020 downregulated the integrated density of activation transcription factor 3 (ATF3) but not the number of ATF positive cells. TC-7020 also downregulated phosphorylated extracellular signal kinase (p-ERK) and satellite cell activation in the CCI-affected DRGs. Therefore, systemic α7 nAChR agonist may be effective in treating neuropathic pain via reducing neuronal injury and immune cells activation occurring in the periphery. PERSPECTIVE These studies demonstrated that TC-7020, an alpha7 nicotinic acetylcholine receptor agonist with partial blood-brain barrier permeability, reversed neuropathic pain in rats, likely via attenuation of inflammation in the DRG and/or the site of sciatic injury.
Collapse
Affiliation(s)
- Lisa C Loram
- Department of Psychology & Neuroscience, and Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The central nervous system interacts dynamically with the immune system to modulate inflammation through humoral and neural pathways. Recently, in animal models of sepsis, the vagus nerve (VN) has been proposed to play a crucial role in the regulation of the immune response, also referred to as the cholinergic anti-inflammatory pathway. The VN, through release of acetylcholine, dampens immune cell activation by interacting with α-7 nicotinic acetylcholine receptors. Recent evidence suggests that the vagal innervation of the gastrointestinal tract also plays a major role controlling intestinal immune activation. Indeed, VN electrical stimulation potently reduces intestinal inflammation restoring intestinal homeostasis, whereas vagotomy has the reverse effect. In this review, we will discuss the current understanding concerning the mechanisms and effects involved in the cholinergic anti-inflammatory pathway in the gastrointestinal tract. Deeper investigation on this counter-regulatory neuroimmune mechanism will provide new insights in the cross-talk between the nervous and immune system leading to the identification of new therapeutic targets to treat intestinal immune disease.
Collapse
Affiliation(s)
- Gianluca Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Herestraat 49, Leuven 3000, Belgium.
| | - Guy E Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium,Department of Clinical and Experimental Medicine, University Hospital Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
47
|
D'Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC. Targeting the "cytokine storm" for therapeutic benefit. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:319-27. [PMID: 23283640 PMCID: PMC3592351 DOI: 10.1128/cvi.00636-12] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation is the body's first line of defense against infection or injury, responding to challenges by activating innate and adaptive responses. Microbes have evolved a diverse range of strategies to avoid triggering inflammatory responses. However, some pathogens, such as the influenza virus and the Gram-negative bacterium Francisella tularensis, do trigger life-threatening "cytokine storms" in the host which can result in significant pathology and ultimately death. For these diseases, it has been proposed that downregulating inflammatory immune responses may improve outcome. We review some of the current candidates for treatment of cytokine storms which may prove useful in the clinic in the future and compare them to more traditional therapeutic candidates that target the pathogen rather than the host response.
Collapse
|
48
|
Cui WY, Zhao S, Polanowska-Grabowska R, Wang J, Wei J, Dash B, Chang SL, Saucerman JJ, Gu J, Li MD. Identification and characterization of poly(I:C)-induced molecular responses attenuated by nicotine in mouse macrophages. Mol Pharmacol 2013; 83:61-72. [PMID: 23028093 PMCID: PMC3533466 DOI: 10.1124/mol.112.081497] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/01/2012] [Indexed: 12/15/2022] Open
Abstract
To further our understanding of the effects of nicotine on the molecular responses of macrophages during virus or virus-like infections, poly(I:C)-stimulated macrophage-like RAW264.2 cells or mouse primary peritoneal macrophages were challenged with nicotine; and their molecular responses were evaluated using a qRT-PCR array, antibody array, ELISA, Western blotting, and Ca(2+) imaging. Of 51 genes expressed in the Toll-like receptor (TLR) and RIG-I-like receptor (RLR) pathways, mRNA expression of 15 genes in RAW264.7 cells was attenuated by nicotine, of which mRNA expression of IL-6, TNF-α, and IL-1β was confirmed to be attenuated in peritoneal macrophages. Concurrently, nicotine treatment attenuated the release of IL-6 and TNF-α from poly(I:C)-stimulated macrophages. However, when poly(I:C)-stimulated macrophages were challenged with nicotine plus α-bungarotoxin (α-BTX), secretion of IL-6 and TNF-α was found to be in a level seen with poly(I:C) stimulation only, indicating that α7-nAChR, a highly Ca(2+) permeable ion channel sensitive to blockade by α-BTX, is involved in this process. Furthermore, results from an antibody array indicated that nicotine treatment attenuated the phosphorylation of 82 sites, including Thr286 on CaMKIIα, from poly(I:C)-stimulated RAW264.7 cells, of which 28 are expressed in the downstream cascade of Ca(2+) signaling. Coincidentally, poly(I:C)-stimulated macrophages showed attenuated expression of phosphorylated CaMKIIα when pretreated with nicotine. In addition, nicotine attenuated intracellular Ca(2+) signal from poly(I:C)-stimulated RAW264.7 cells. Collectively, these results indicate that poly(I:C)-induced molecular responses of macrophages could be significantly attenuated by nicotine.
Collapse
Affiliation(s)
- Wen-Yan Cui
- Department of Psychiatry and Neurobehavioral Sciences University of Virginia 1670 Discovery Drive, Suite 110, Charlottesville, VA 22911, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bonaz BL, Bernstein CN. Brain-gut interactions in inflammatory bowel disease. Gastroenterology 2013; 144:36-49. [PMID: 23063970 DOI: 10.1053/j.gastro.2012.10.003] [Citation(s) in RCA: 481] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/23/2012] [Accepted: 10/01/2012] [Indexed: 12/13/2022]
Abstract
Psycho-neuro-endocrine-immune modulation through the brain-gut axis likely has a key role in the pathogenesis of inflammatory bowel disease (IBD). The brain-gut axis involves interactions among the neural components, including (1) the autonomic nervous system, (2) the central nervous system, (3) the stress system (hypothalamic-pituitary-adrenal axis), (4) the (gastrointestinal) corticotropin-releasing factor system, and (5) the intestinal response (including the intestinal barrier, the luminal microbiota, and the intestinal immune response). Animal models suggest that the cholinergic anti-inflammatory pathway through an anti-tumor necrosis factor effect of the efferent vagus nerve could be a therapeutic target in IBD through a pharmacologic, nutritional, or neurostimulation approach. In addition, the psychophysiological vulnerability of patients with IBD, secondary to the potential presence of any mood disorders, distress, increased perceived stress, or maladaptive coping strategies, underscores the psychological needs of patients with IBD. Clinicians need to address these issues with patients because there is emerging evidence that stress or other negative psychological attributes may have an effect on the disease course. Future research may include exploration of markers of brain-gut interactions, including serum/salivary cortisol (as a marker of the hypothalamic-pituitary-adrenal axis), heart rate variability (as a marker of the sympathovagal balance), or brain imaging studies. The widespread use and potential impact of complementary and alternative medicine and the positive response to placebo (in clinical trials) is further evidence that exploring other psycho-interventions may be important therapeutic adjuncts to the conventional therapeutic approach in IBD.
Collapse
Affiliation(s)
- Bruno L Bonaz
- Stress et Interactions Neuro-Digestives, Grenoble Institut des Neurosciences, Centre de Recherche INSERM 836 UJF-CEA-CHU, Grenoble, France.
| | | |
Collapse
|
50
|
Boeckxstaens G. The clinical importance of the anti-inflammatory vagovagal reflex. HANDBOOK OF CLINICAL NEUROLOGY 2013; 117:119-34. [PMID: 24095121 DOI: 10.1016/b978-0-444-53491-0.00011-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Excessive activation of the immune system is prevented by anti-inflammatory mediators such as corticosteroids and anti-inflammatory cytokines. Recently, it became clear that the brain not only senses peripheral inflammation through vagal afferent nerve fibers, but also provides an integrated response dampening the immune system through vagal efferents. This so-called anti-inflammatory pathway has been introduced as a third system by which the immune system is modulated. In sepsis, the anti-inflammatory effect is mediated by modulation of splenic macrophages, whereas in the gut, vagal nerve fibers synapse with enteric cholinergic neurons interacting with resident intestinal macrophages. In this chapter, the preclinical data underscoring the importance of this pathway are summarized, and its clinical significance is reviewed. Finally, the current data supporting its relevance to human disease and its therapeutic potential will be discussed. Insight in the mechanisms underlying these crucial properties will lead to better understanding of immune-mediated diseases and ultimately to improved anti-inflammatory therapies.
Collapse
Affiliation(s)
- G Boeckxstaens
- Department of Gastroenterology, University Hospital Leuven, University of Leuven, Leuven, Belgium.
| |
Collapse
|