1
|
Gao F, Du W, Guo C, Geng P, Liu W, Jin X. α7nACh receptor, a promising target to reduce BBB damage by regulating inflammation and autophagy after ischemic stroke. Biomed Pharmacother 2024; 179:117337. [PMID: 39191022 DOI: 10.1016/j.biopha.2024.117337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Increased blood-brain barrier (BBB) permeability can lead to cerebral vasogenic edema and hemorrhagic transformation (HT) after reperfusion with tissue plasminogen activator (tPA), the only United States Food and Drug Administration (FDA)-approved treatment for acute ischemia stroke (AIS). The therapeutic benefits of tPA after AIS are partially outweighed by a more than a six-fold increase in the risk of symptomatic intracerebral hemorrhage. Therefore, strategies to protect the integrity of BBB are urgently needed to reduce HT and vasogenic edema after tPA thrombolysis or endovascular thrombectomy. Interestingly, an NIH study showed that smokers treated with tPA had a significantly lower prevalence of brain hemorrhage than nonsmokers, suggesting that cigarette smoking may protect patients treated with tPA from the side effects of cerebral hemorrhage. Importantly, we recently showed that treatment with nicotine reduces AIS-induced BBB damage and that modulating α7nAChR by modulation could reduce ischemia/reperfusion-induced BBB damage, suggesting that α7nAChR could be a potential target to reduce BBB after AIS. In this review, we first provide an overview of stroke and the impact of α7nAChR activation on BBB damage. Next, we discuss the features and mechanism of BBB destruction after AIS. We then discuss the effect of nicotine effect on BBB integrity as well as the mechanism underlying those effects. Finally, we discuss the side effects and potential strategies for modulating α7nAChR to reduce AIS-induced BBB damage.
Collapse
Affiliation(s)
- Fengying Gao
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Wencao Liu
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
2
|
Busceti CL, Bucci D, Damato A, De Lucia M, Venturini E, Ferrucci M, Lazzeri G, Puglisi-Allegra S, Scioli M, Carrizzo A, Nicoletti F, Vecchione C, Fornai F. Methamphetamine-Induced Blood Pressure Sensitization Correlates with Morphological Alterations within A1/C1 Catecholamine Neurons. Int J Mol Sci 2024; 25:10282. [PMID: 39408612 PMCID: PMC11476956 DOI: 10.3390/ijms251910282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Methamphetamine (METH) is a drug of abuse, which induces behavioral sensitization following repeated doses. Since METH alters blood pressure, in the present study we assessed whether systolic and diastolic blood pressure (SBP and DBP, respectively) are sensitized as well. In this context, we investigated whether alterations develop within A1/C1 neurons in the vasomotor center. C57Bl/6J male mice were administered METH (5 mg/kg, daily for 5 consecutive days). Blood pressure was measured by tail-cuff plethysmography. We found a sensitized response both to SBP and DBP, along with a significant decrease of catecholamine neurons within A1/C1 (both in the rostral and caudal ventrolateral medulla), while no changes were detected in glutamic acid decarboxylase. The decrease of catecholamine neurons was neither associated with the appearance of degeneration-related marker Fluoro-Jade B nor with altered expression of α-synuclein. Rather, it was associated with reduced free radicals and phospho-cJun and increased heat shock protein-70 and p62/sequestosome within A1/C1 cells. Blood pressure sensitization was not associated with altered arterial reactivity. These data indicate that reiterated METH administration may increase blood pressure persistently and may predispose to an increased cardiovascular response to METH. These data may be relevant to explain cardiovascular events following METH administration and stressful conditions.
Collapse
Affiliation(s)
- Carla Letizia Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Domenico Bucci
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Antonio Damato
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Massimiliano De Lucia
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Eleonora Venturini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| | - Stefano Puglisi-Allegra
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Mariarosaria Scioli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
| | - Albino Carrizzo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana” University of Salerno, 84081 Baronissi, Italy
| | - Ferdinando Nicoletti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Physiology and Pharmacology, University Sapienza, 00185 Roma, Italy
| | - Carmine Vecchione
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana” University of Salerno, 84081 Baronissi, Italy
| | - Francesco Fornai
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy; (C.L.B.); (D.B.); (A.D.); (M.D.L.); (E.V.); (S.P.-A.); (M.S.); (A.C.); (F.N.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.F.); (G.L.)
| |
Collapse
|
3
|
Costa PCT, de Luna Freire MO, de Oliveira Coutinho D, Godet M, Magnani M, Antunes VR, de Souza EL, Vidal H, de Brito Alves JL. Nutraceuticals in the management of autonomic function and related disorders: A comprehensive review. Pharmacol Res 2024; 208:107368. [PMID: 39191337 DOI: 10.1016/j.phrs.2024.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
Nutraceuticals have been described as phytocomplexes when derived from foods of plant origin or a pool of secondary metabolites when derived from foods of animal origin, which are concentrated and administered in an appropriate form and can promote beneficial health effects in the prevention/treatment of diseases. Considering that pharmaceutical medications can cause side effects, there is a growing interest in using nutraceuticals as an adjuvant therapeutic tool for several disorders involving autonomic dysfunction, such as obesity, atherosclerosis and other cardiometabolic diseases. This review summarizes and discusses the evidence from the literature on the effects of various nutraceuticals on autonomic control, addressing the gut microbiota modulation, production of secondary metabolites from bioactive compounds, and improvement of physical and chemical properties of cell membranes. Additionally, the safety of nutraceuticals and prospects are discussed. Probiotics, resveratrol, quercetin, curcumin, nitrate, inositol, L-carnosine, and n-3 polyunsaturated fatty acids (n-3 PUFAs) are among the nutraceuticals most studied to improve autonomic dysfunction in experimental animal models and clinical trials. Further human studies are needed to elucidate the effects of nutraceuticals formulated of multitarget compounds and their underlying mechanisms of action, which could benefit conditions involving autonomic dysfunction.
Collapse
Affiliation(s)
- Paulo César Trindade Costa
- Department of Nutrition, Federal University of Paraíba, João Pessoa, PB, Brazil; Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | | | | | - Murielle Godet
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Marciane Magnani
- Department of Food Engineering, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Vagner Roberto Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Hubert Vidal
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | | |
Collapse
|
4
|
Natarajan C, Le LHD, Gunasekaran M, Tracey KJ, Chernoff D, Levine YA. Electrical stimulation of the vagus nerve ameliorates inflammation and disease activity in a rat EAE model of multiple sclerosis. Proc Natl Acad Sci U S A 2024; 121:e2322577121. [PMID: 38968104 PMCID: PMC11252997 DOI: 10.1073/pnas.2322577121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Multiple sclerosis (MS) is a demyelinating central nervous system (CNS) disorder that is associated with functional impairment and accruing disability. There are multiple U.S. Food and Drug Administration (FDA)-approved drugs that effectively dampen inflammation and slow disability progression. However, these agents do not work well for all patients and are associated with side effects that may limit their use. The vagus nerve (VN) provides a direct communication conduit between the CNS and the periphery, and modulation of the inflammatory reflex via electrical stimulation of the VN (VNS) shows efficacy in ameliorating pathology in several CNS and autoimmune disorders. We therefore investigated the impact of VNS in a rat experimental autoimmune encephalomyelitis (EAE) model of MS. In this study, VNS-mediated neuroimmune modulation is demonstrated to effectively decrease EAE disease severity and duration, infiltration of neutrophils and pathogenic lymphocytes, myelin damage, blood-brain barrier disruption, fibrinogen deposition, and proinflammatory microglial activation. VNS modulates expression of genes that are implicated in MS pathogenesis, as well as those encoding myelin proteins and transcription factors regulating new myelin synthesis. Together, these data indicate that neuroimmune modulation via VNS may be a promising approach to treat MS, that not only ameliorates symptoms but potentially also promotes myelin repair (remyelination).
Collapse
Affiliation(s)
| | | | | | - Kevin J. Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY11030
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
| | | | - Yaakov A. Levine
- SetPoint Medical, Valencia, CA91355
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY11549
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm171 76, Sweden
| |
Collapse
|
5
|
Zhang Z, Zhang D, Lin Q, Cui X. Therapeutically Fine-Tuning Autonomic Nervous System to Treat Sepsis: A New Perspective on the Immunomodulatory Effects of Acupuncture. J Inflamm Res 2024; 17:4373-4387. [PMID: 38988505 PMCID: PMC11233988 DOI: 10.2147/jir.s477181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Recent studies have highlighted the immunomodulatory effects of acupuncture on sepsis and proposed novel non-pharmacological or bioelectronic approaches to managing inflammatory illnesses. Establishing rules for selectively activating sympathetic or vagal nerve-mediated anti-inflammatory pathways using acupuncture has valuable clinical applications. Over the years, studies have revealed the segmental modulatory role of acupuncture in regulating visceral function by targeting the autonomic nervous system (ANS). In this review, we aim to summarize recent findings on acupuncture in treating sepsis, focusing on the underlying ANS mechanism, as well as the rules of acupoint specificity, intensity, frequency, and other parameters utilized in these studies. Mechanistically, the immunomodulatory properties of the sympathetic nervous system have been highlighted. Furthermore, we explore the immunotherapeutic benefits of acupuncture in treating sepsis. A better understanding of the immunoregulatory mechanism of sympathetic nervous system may offer novel approaches for the development of therapeutics to treat or prevent a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Dingdan Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, 21287, USA
| | - Xiang Cui
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| |
Collapse
|
6
|
Luo S, Lin H, Wu C, Zhu L, Hua Q, Weng Y, Wang L, Fan X, Zhao KB, Liu G, Wang Y, Chen HT, Xu L, Zheng L. Cholinergic macrophages promote the resolution of peritoneal inflammation. Proc Natl Acad Sci U S A 2024; 121:e2402143121. [PMID: 38923993 PMCID: PMC11228479 DOI: 10.1073/pnas.2402143121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The non-neural cholinergic system plays a critical role in regulating immune equilibrium and tissue homeostasis. While the expression of choline acetyltransferase (ChAT), the enzyme catalyzing acetylcholine biosynthesis, has been well documented in lymphocytes, its role in the myeloid compartment is less understood. Here, we identify a significant population of macrophages (Mϕs) expressing ChAT and synthesizing acetylcholine in the resolution phase of acute peritonitis. Using Chat-GFP reporter mice, we observed marked upregulation of ChAT in monocyte-derived small peritoneal Mϕs (SmPMs) in response to Toll-like receptor agonists and bacterial infections. These SmPMs, phenotypically and transcriptionally distinct from tissue-resident large peritoneal macrophages, up-regulated ChAT expression through a MyD88-dependent pathway involving MAPK signaling. Notably, this process was attenuated by the TRIF-dependent TLR signaling pathway, and our tests with a range of neurotransmitters and cytokines failed to induce a similar response. Functionally, Chat deficiency in Mϕs led to significantly decreased peritoneal acetylcholine levels, reduced efferocytosis of apoptotic neutrophils, and a delayed resolution of peritonitis, which were reversible with exogenous ACh supplementation. Intriguingly, despite B lymphocytes being a notable ChAT-expressing population within the peritoneal cavity, Chat deletion in B cells did not significantly alter the resolution process. Collectively, these findings underscore the crucial role of Mϕ-derived acetylcholine in the resolution of inflammation and highlight the importance of the non-neuronal cholinergic system in immune regulation.
Collapse
Affiliation(s)
- Shufeng Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, People's Republic of China
| | - Huiling Lin
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Chong Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Lan Zhu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Qiaomin Hua
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, People's Republic of China
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Yulan Weng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Lu Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Xiaoli Fan
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Kai-Bo Zhao
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Gaoteng Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Yuting Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Hai-Tian Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Li Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, People's Republic of China
| | - Limin Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, People's Republic of China
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Ministry of Education Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| |
Collapse
|
7
|
Lam KHS, Su DCJ, Wu YT, Varrassi G, Suryadi T, Reeves KD. A Novel Ultrasound-Guided Bilateral Vagal Nerve Hydrodissection With 5% Dextrose Without Local Anesthetic for Recalcitrant Chronic Multisite Pain and Autonomic Dysfunction. Cureus 2024; 16:e63609. [PMID: 38957517 PMCID: PMC11218924 DOI: 10.7759/cureus.63609] [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: 06/09/2024] [Accepted: 06/30/2024] [Indexed: 07/04/2024] Open
Abstract
Chronic pain is a complex condition that often poses diagnostic and management challenges due to its multifactorial etiology. This case report describes a 49-year-old pastor who presented with a three-year history of chronic pain affecting multiple sites, including the neck, bilateral shoulders, thoracic region, lower back, and bilateral knees. Additionally, he experienced shortness of breath on mild exertion, which adversely affected his ability to converse and speak publicly. The patient had a rapid resting heart rate of 100-120 beats per minute, occasional palpitations, and a 24-hour electrocardiogram that confirmed 15% premature ventricular complexes with bigeminy and trigeminy. He complained of limited appetite with early satiety, intermittent nausea, and regurgitation. Despite consultations with multiple specialists, no underlying causes were identified in the cardiac, respiratory, gastrointestinal, or psychological domains. Ultrasound-guided bilateral vagus nerve hydrodissection using 5% dextrose without local anesthetics was administered three times at monthly intervals, resulting in remarkable pain relief within three months and the effects persisted at the nine-month follow-up. Tachycardia was no longer perceived, resting heart rate slowed to 70-80 beats per minute, shortness of breath improved, and public speaking ability was restored. The patient's early satiety, nausea, and reflux complaints were resolved. This case report highlights the potential effectiveness of this novel intervention for chronic pain. Further research is warranted to validate these findings and explore the mechanism of action.
Collapse
Affiliation(s)
- King Hei Stanley Lam
- Board of Clinical Research, The Hong Kong Institute of Musculoskeletal Medicine, Kowloon, HKG
- Faculty of Medicine, The Chinese University of Hong Kong, New Territories, HKG
- Faculty of Medicine, The University of Hong Kong, Hong Kong, HKG
| | | | - Yung-Tsan Wu
- Physical Medicine and Rehabilitation, Tri-Service General Hospital, Taipei, TWN
| | | | - Teinny Suryadi
- Physical Medicine and Rehabilitation, Synergy Clinic, Jakarta, IDN
- Physical Medicine and Rehabilitation, Hermina Podomoro Hospital, Jakarta, IDN
| | - K Dean Reeves
- Rehabilitation Medicine, Private Practice, Kansas City, USA
| |
Collapse
|
8
|
Maldonado-García JL, García-Mena LH, Mendieta-Cabrera D, Pérez-Sánchez G, Becerril-Villanueva E, Alvarez-Herrera S, Homberg T, Vallejo-Castillo L, Pérez-Tapia SM, Moreno-Lafont MC, Ortuño-Sahagún D, Pavón L. Use of Extracellular Monomeric Ubiquitin as a Therapeutic Option for Major Depressive Disorder. Pharmaceuticals (Basel) 2024; 17:841. [PMID: 39065692 PMCID: PMC11279398 DOI: 10.3390/ph17070841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Major depressive disorder (MDD) is a mood disorder that has become a global health emergency according to the World Health Organization (WHO). It affects 280 million people worldwide and is a leading cause of disability and financial loss. Patients with MDD present immunoendocrine alterations like cortisol resistance and inflammation, which are associated with alterations in neurotransmitter metabolism. There are currently numerous therapeutic options for patients with MDD; however, some studies suggest a high rate of therapeutic failure. There are multiple hypotheses explaining the pathophysiological mechanisms of MDD, in which several systems are involved, including the neuroendocrine and immune systems. In recent years, inflammation has become an important target for the development of new therapeutic options. Extracellular monomeric ubiquitin (emUb) is a molecule that has been shown to have immunomodulatory properties through several mechanisms including cholinergic modulation and the generation of regulatory T cells. In this perspective article, we highlight the influence of the inflammatory response in MDD. In addition, we review and discuss the evidence for the use of emUb contained in Transferon as a concomitant treatment with selective serotonin reuptake inhibitors (SSRIs).
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Lissette Haydee García-Mena
- Departamento de Salud Digital, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico;
| | - Danelia Mendieta-Cabrera
- Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 14370, Mexico;
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| | - Toni Homberg
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Luis Vallejo-Castillo
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Sonia Mayra Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (T.H.); (L.V.-C.)
- Laboratorio Nacional Para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) Para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
| | - Martha C. Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (S.M.P.-T.)
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas (IICB), CUCS, Universidad de Guadalajara, Jalisco 44340, Mexico;
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (G.P.-S.); (E.B.-V.); (S.A.-H.)
| |
Collapse
|
9
|
Shu LZ, Ding YD, Zhang JY, He RS, Xiao L, Pan BX, Deng H. Interactions between MDSCs and the Autonomic Nervous System: Opportunities and Challenges in Cancer Neuroscience. Cancer Immunol Res 2024; 12:652-662. [PMID: 38568775 DOI: 10.1158/2326-6066.cir-23-0976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/11/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Myeloid-derived suppressor cells (MDSC) are a population of heterogeneous immune cells that are involved in precancerous conditions and neoplasms. The autonomic nervous system (ANS), which is composed of the sympathetic nervous system and the parasympathetic nervous system, is an important component of the tumor microenvironment that responds to changes in the internal and external environment mainly through adrenergic and cholinergic signaling. An abnormal increase of autonomic nerve density has been associated with cancer progression. As we discuss in this review, growing evidence indicates that sympathetic and parasympathetic signals directly affect the expansion, mobilization, and redistribution of MDSCs. Dysregulated autonomic signaling recruits MDSCs to form an immunosuppressive microenvironment in chronically inflamed tissues, resulting in abnormal proliferation and differentiation of adult stem cells. The two components of the ANS may also be responsible for the seemingly contradictory behaviors of MDSCs. Elucidating the underlying mechanisms has the potential to provide more insights into the complex roles of MDSCs in tumor development and lay the foundation for the development of novel MDSC-targeted anticancer strategies.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yi-Dan Ding
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jin-Yao Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Rui-Shan He
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Li Xiao
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huan Deng
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Rommel FR, Tumala S, Urban AL, Siebenhaar F, Kruse J, Gieler U, Peters EMJ. Stress Affects Mast Cell Proteases in Murine Skin in a Model of Atopic Dermatitis-like Allergic Inflammation. Int J Mol Sci 2024; 25:5738. [PMID: 38891925 PMCID: PMC11171663 DOI: 10.3390/ijms25115738] [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: 04/29/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Stress exposure worsens allergic inflammatory diseases substantially. Mast cells (MCs) play a key role in peripheral immune responses to neuroendocrine stress mediators such as nerve growth factor (NGF) and substance P (SP). Mast cell proteases (MCPs) and cholinergic factors (Chrna7, SLURP1) were recently described to modulate MC stress response. We studied MCPs and Chrna7/SLURP1 and their interplay in a mouse model for noise induced stress (NiS) and atopic dermatitis-like allergic inflammation (AlD) and in cultured MC lacking Chrna7. We found that the cholinergic stress axis interacts with neuroendocrine stress mediators and stress-mediator cleaving enzymes in AlD. SP-cleaving mMCP4+ MC were upregulated in AlD and further upregulated by stress in NiS+AlD. Anti-NGF neutralizing antibody treatment blocked the stress-induced upregulation in vivo, and mMCP4+ MCs correlated with measures of AlD disease activity. Finally, high mMCP4 production in response to SP depended on Chrna7/SLURP1 in cultured MCs. In conclusion, mMCP4 and its upstream regulation by Chrna7/SLURP1 are interesting novel targets for the treatment of allergic inflammation and its aggravation by stress.
Collapse
Affiliation(s)
- Frank R. Rommel
- Psychoneuroimmunology Laboratory, Department of Psychosomatic Medicine and Psychotherapy, Justus Liebig University Giessen, 35390 Giessen, Germany
| | - Susanne Tumala
- Psychoneuroimmunology Laboratory, Department of Psychosomatic Medicine and Psychotherapy, Justus Liebig University Giessen, 35390 Giessen, Germany
| | - Anna-Lena Urban
- Psychoneuroimmunology Laboratory, Department of Psychosomatic Medicine and Psychotherapy, Justus Liebig University Giessen, 35390 Giessen, Germany
| | - Frank Siebenhaar
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, 12203 Berlin, Germany
| | - Johannes Kruse
- Department of Psychosomatic Medicine and Psychotherapy, Justus Liebig University Giessen, 35390 Giessen, Germany
| | - Uwe Gieler
- Department of Dermatology, University Hospital Giessen, 35392 Giessen, Germany
| | - Eva M. J. Peters
- Psychoneuroimmunology Laboratory, Department of Psychosomatic Medicine and Psychotherapy, Justus Liebig University Giessen, 35390 Giessen, Germany
- Charité Center 12 for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
11
|
Lataro RM, Brognara F, Iturriaga R, Paton JFR. Inflammation of some visceral sensory systems and autonomic dysfunction in cardiovascular disease. Auton Neurosci 2024; 251:103137. [PMID: 38104365 DOI: 10.1016/j.autneu.2023.103137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
The sensitization and hypertonicity of visceral afferents are highly relevant to the development and progression of cardiovascular and respiratory disease states. In this review, we described the evidence that the inflammatory process regulates visceral afferent sensitivity and tonicity, affecting the control of the cardiovascular and respiratory system. Some inflammatory mediators like nitric oxide, angiotensin II, endothelin-1, and arginine vasopressin may inhibit baroreceptor afferents and contribute to the baroreflex impairment observed in cardiovascular diseases. Cytokines may act directly on peripheral afferent terminals that transmit information to the central nervous system (CNS). TLR-4 receptors, which recognize lipopolysaccharide, were identified in the nodose and petrosal ganglion and have been implicated in disrupting the blood-brain barrier, which can potentiate the inflammatory process. For example, cytokines may cross the blood-brain barrier to access the CNS. Additionally, pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and some of their receptors have been identified in the nodose ganglion and carotid body. These pro-inflammatory cytokines also sensitize the dorsal root ganglion or are released in the nucleus of the solitary tract. In cardiovascular disease, pro-inflammatory mediators increase in the brain, heart, vessels, and plasma and may act locally or systemically to activate/sensitize afferent nervous terminals. Recent evidence demonstrated that the carotid body chemoreceptor cells might sense systemic pro-inflammatory molecules, supporting the novel proposal that the carotid body is part of the afferent pathway in the central anti-inflammatory reflexes. The exact mechanisms of how pro-inflammatory mediators affects visceral afferent signals and contribute to the pathophysiology of cardiovascular diseases awaits future research.
Collapse
Affiliation(s)
- R M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - F Brognara
- Department of Nursing, General and Specialized, Nursing School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Iturriaga
- Facultad de Ciencias Biológicas, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Investigación en Fisiología y Medicina en Altura - FIMEDALT, Universidad de Antofagasta, Antofagasta, Chile
| | - J F R Paton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
12
|
Zhang J, Chen S, Hu X, Huang L, Loh P, Yuan X, Liu Z, Lian J, Geng L, Chen Z, Guo Y, Chen B. The role of the peripheral system dysfunction in the pathogenesis of sepsis-associated encephalopathy. Front Microbiol 2024; 15:1337994. [PMID: 38298892 PMCID: PMC10828041 DOI: 10.3389/fmicb.2024.1337994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Sepsis is a condition that greatly impacts the brain, leading to neurological dysfunction and heightened mortality rates, making it one of the primary organs affected. Injury to the central nervous system can be attributed to dysfunction of various organs throughout the entire body and imbalances within the peripheral immune system. Furthermore, central nervous system injury can create a vicious circle with infection-induced peripheral immune disorders. We collate the pathogenesis of septic encephalopathy, which involves microglial activation, programmed cell death, mitochondrial dysfunction, endoplasmic reticulum stress, neurotransmitter imbalance, and blood-brain barrier disruption. We also spotlight the effects of intestinal flora and its metabolites, enterocyte-derived exosomes, cholinergic anti-inflammatory pathway, peripheral T cells and their cytokines on septic encephalopathy.
Collapse
Affiliation(s)
- Jingyu Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuangli Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiyou Hu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lihong Huang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - PeiYong Loh
- School of International Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinru Yuan
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinyu Lian
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lianqi Geng
- Binhai New Area Hospital of TCM, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture and Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Binhai New Area Hospital of TCM, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture and Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
13
|
D’Haens G, Eberhardson M, Cabrijan Z, Danese S, van den Berg R, Löwenberg M, Fiorino G, Schuurman PR, Lind G, Almqvist P, Olofsson PS, Tracey KJ, Hanauer SB, Zitnik R, Chernoff D, Levine YA. Neuroimmune Modulation Through Vagus Nerve Stimulation Reduces Inflammatory Activity in Crohn's Disease Patients: A Prospective Open-label Study. J Crohns Colitis 2023; 17:1897-1909. [PMID: 37738465 PMCID: PMC10798868 DOI: 10.1093/ecco-jcc/jjad151] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] is a debilitating, inflammatory condition affecting the gastrointestinal tract. There is no cure and sustained clinical and endoscopic remission is achieved by fewer than half of patients with current therapies. The immunoregulatory function of the vagus nerve, the 'inflammatory reflex', has been established in patients with rheumatoid arthritis and biologic-naive CD. The aim of this study was to explore the safety and efficacy of vagus nerve stimulation in patients with treatment-refractory CD, in a 16-week, open-label, multicentre, clinical trial. METHODS A vagus nerve stimulator was implanted in 17 biologic drug-refractory patients with moderately to severely active CD. One patient exited the study pre-treatment, and 16 patients were treated with vagus nerve stimulation [4/16 receiving concomitant biologics] during 16 weeks of induction and 24 months of maintenance treatment. Endpoints included clinical improvement, patient-reported outcomes, objective measures of inflammation [endoscopic/molecular], and safety. RESULTS There was a statistically significant and clinically meaningful decrease in CD Activity Index at Week 16 [mean ± SD: -86.2 ± 92.8, p = 0.003], a significant decrease in faecal calprotectin [-2923 ± 4104, p = 0.015], a decrease in mucosal inflammation in 11/15 patients with paired endoscopies [-2.1 ± 1.7, p = 0.23], and a decrease in serum tumour necrosis factor and interferon-γ [46-52%]. Two quality-of-life indices improved in 7/11 patients treated without biologics. There was one study-related severe adverse event: a postoperative infection requiring device explantation. CONCLUSIONS Neuroimmune modulation via vagus nerve stimulation was generally safe and well tolerated, with a clinically meaningful reduction in clinical disease activity associated with endoscopic improvement, reduced levels of faecal calprotectin and serum cytokines, and improved quality of life.
Collapse
Affiliation(s)
- Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michael Eberhardson
- Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Zeljko Cabrijan
- Division of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, Zagreb, Croatia
- Division of Gastroenterology, University of Applied Health Sciences, Zagreb, Croatia
- Josip Juraj Strossmayer University of Osijek School of Medicine, Osijek, Croatia
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Italy
- Department of Gastroenterology and Endoscopy, University Vita-Salute San Raffaele, Milano, Italy
| | - Remco van den Berg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gionata Fiorino
- Department of Gastroenterology and Digestive Endoscopy, VIta-Salute San Raffaele Hospital, Milan, Italy
- IBD Unit, Department of Gastroenterology and Digestive Endoscopy, San Camillo-Forlanini Hospital, Rome, Italy
| | | | - Göran Lind
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Per Almqvist
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
- Neurosurgery Stockholm AB, Stockholm, Sweden
| | - Peder S Olofsson
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Feinstein Institutes for Medical Research, Manhasset, New York
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Stephen B Hanauer
- Division of Gastroenterology and Hepatology, Northwestern University–Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ralph Zitnik
- SetPoint Medical, Valencia, California, USA
- Valerio Consulting, Santa Barbara, California, USA
| | | | - Yaakov A Levine
- Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- SetPoint Medical, Valencia, California, USA
| |
Collapse
|
14
|
Apaydin O, Altaikyzy A, Filosa A, Sawamiphak S. Alpha-1 adrenergic signaling drives cardiac regeneration via extracellular matrix remodeling transcriptional program in zebrafish macrophages. Dev Cell 2023; 58:2460-2476.e7. [PMID: 37875117 DOI: 10.1016/j.devcel.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 06/24/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
The autonomic nervous system plays a pivotal role in cardiac repair. Here, we describe the mechanistic underpinning of adrenergic signaling in fibrotic and regenerative response of the heart to be dependent on immunomodulation. A pharmacological approach identified adrenergic receptor alpha-1 as a key regulator of macrophage phenotypic diversification following myocardial damage in zebrafish. Genetic manipulation and single-cell transcriptomics showed that the receptor signals activation of an "extracellular matrix remodeling" transcriptional program in a macrophage subset, which serves as a key regulator of matrix composition and turnover. Mechanistically, adrenergic receptor alpha-1-activated macrophages determine activation of collagen-12-expressing fibroblasts, a cellular determinant of cardiac regenerative niche, through midkine-mediated paracrine crosstalk, allowing lymphatic and blood vessel growth and cardiomyocyte proliferation at the lesion site. These findings identify the mechanism of adrenergic signaling in macrophage phenotypic and functional determination and highlight the potential of neural modulation for regulation of fibrosis and coordination of myocardial regenerative response.
Collapse
Affiliation(s)
- Onur Apaydin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Akerke Altaikyzy
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alessandro Filosa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suphansa Sawamiphak
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
15
|
Refisch A, Walter M. [The importance of the human microbiome for mental health]. DER NERVENARZT 2023; 94:1001-1009. [PMID: 37847418 PMCID: PMC10620288 DOI: 10.1007/s00115-023-01552-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/06/2023] [Indexed: 10/18/2023]
Abstract
Many common diseases including psychiatric disorders show characteristic alterations in the microbiome. Preclinical studies have uncovered important mechanisms by which the microbiome interacts bidirectionally with neural functions. Dysregulation of the complex interplay between the microbiome, immune system, stress response, and energy homeostasis, particularly in the early stages of life, can predispose to the development of psychiatric symptoms later in life. Although few clinical studies are available to date, the broad influence of the microbiome on neural and mental functions as well as its high plasticity, have generated great interest in its therapeutic potential for common psychiatric disorders.
Collapse
Affiliation(s)
- Alexander Refisch
- Klinik für Psychiatrie und Psychotherapie, Universitätsklinikum Jena, Philosophenweg 3, 07743, Jena, Deutschland.
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena, Deutschland.
| | - Martin Walter
- Klinik für Psychiatrie und Psychotherapie, Universitätsklinikum Jena, Philosophenweg 3, 07743, Jena, Deutschland
- Deutsches Zentrum für psychische Gesundheit (DZP), Jena, Deutschland
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena, Deutschland
| |
Collapse
|
16
|
Kaplan A, Lakkis B, El-Samadi L, Karaayvaz EB, Booz GW, Zouein FA. Cooling Down Inflammation in the Cardiovascular System via the Nicotinic Acetylcholine Receptor. J Cardiovasc Pharmacol 2023; 82:241-265. [PMID: 37539950 DOI: 10.1097/fjc.0000000000001455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023]
Abstract
ABSTRACT Inflammation is a major player in many cardiovascular diseases including hypertension, atherosclerosis, myocardial infarction, and heart failure. In many individuals, these conditions coexist and mutually exacerbate each other's progression. The pathophysiology of these diseases entails the active involvement of both innate and adaptive immune cells. Immune cells that possess the α7 subunit of the nicotinic acetylcholine receptor on their surface have the potential to be targeted through both pharmacological and electrical stimulation of the cholinergic system. The cholinergic system regulates the inflammatory response to various stressors in different organ systems by systematically suppressing spleen-derived monocytes and chemokines and locally improving immune cell function. Research on the cardiovascular system has demonstrated the potential for atheroma plaque stabilization and regression as favorable outcomes. Smaller infarct size and reduced fibrosis have been associated with improved cardiac function and a decrease in adverse cardiac remodeling. Furthermore, enhanced electrical stability of the myocardium can lead to a reduction in the incidence of ventricular tachyarrhythmia. In addition, improving mitochondrial dysfunction and decreasing oxidative stress can result in less myocardial tissue damage caused by reperfusion injury. Restoring baroreflex activity and reduction in renal damage can promote blood pressure regulation and help counteract hypertension. Thus, the present review highlights the potential of nicotinic acetylcholine receptor activation as a natural approach to alleviate the adverse consequences of inflammation in the cardiovascular system.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- Department of Cardiology, Kemer Public Hospital, Kemer, Antalya, Turkey
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Bachir Lakkis
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Lana El-Samadi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
| | - Ekrem Bilal Karaayvaz
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS; and
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
| |
Collapse
|
17
|
Viscarra F, Chrestia JF, Sanchez Y, Pérez EG, Biggin PC, Bouzat C, Bermudez I, López JJ. Side Groups Convert the α7 Nicotinic Receptor Agonist Ether Quinuclidine into a Type I Positive Allosteric Modulator. ACS Chem Neurosci 2023; 14:2876-2887. [PMID: 37535446 DOI: 10.1021/acschemneuro.3c00225] [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] [Indexed: 08/05/2023] Open
Abstract
The quinuclidine scaffold has been extensively used for the development of nicotinic acetylcholine receptor (nAChR) agonists, with hydrophobic substituents at position 3 of the quinuclidine framework providing selectivity for α7 nAChRs. In this study, six new ligands (4-9) containing a 3-(pyridin-3-yloxy)quinuclidine moiety (ether quinuclidine) were synthesized to gain a better understanding of the structural-functional properties of ether quinuclidines. To evaluate the pharmacological activity of these ligands, two-electrode voltage-clamp and single-channel recordings were performed. Only ligand 4 activated α7 nAChR. Ligands 5 and 7 had no effects on α7 nAChR, but ligands 6, 8, and 9 potentiated the currents evoked by ACh. Ligand 6 was the most potent and efficacious of the potentiating ligands, with an estimated EC50 for potentiation of 12.6 ± 3.32 μM and a maximal potentiation of EC20 ACh responses of 850 ± 120%. Ligand 6 increased the maximal ACh responses without changing the kinetics of the current responses. At the single-channel level, the potentiation exerted by ligand 6 was evidenced in the low micromolar concentration range by the appearance of prolonged bursts of channel openings. Furthermore, computational studies revealed the preference of ligand 6 for an intersubunit site in the transmembrane domain and highlighted some putative key interactions that explain the different profiles of the synthesized ligands. Notably, Met276 in the 15' position of the transmembrane domain 2 almost abolished the effects of ligand 6 when mutated to Leu. We conclude that ligand 6 is a novel type I positive allosteric modulator (PAM-I) of α7 nAChR.
Collapse
Affiliation(s)
- Franco Viscarra
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, U.K
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, Oxford University, South Parks Road, Oxford OX1 3QU, U.K
| | - Juan Facundo Chrestia
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km 7, Bahía Blanca 8000, Argentina
| | - Yaima Sanchez
- Department of Organic Chemistry, Faculty of Chemistry, Universidad de Concepción, Edmundo Larenas 129, Concepción 4070371, Chile
| | - Edwin G Pérez
- Department of Organic Chemistry, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago 7820436, Chile
| | - Philip C Biggin
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, Oxford University, South Parks Road, Oxford OX1 3QU, U.K
| | - Cecilia Bouzat
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km 7, Bahía Blanca 8000, Argentina
| | - Isabel Bermudez
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, U.K
| | - Jhon J López
- Department of Organic Chemistry, Faculty of Chemistry, Universidad de Concepción, Edmundo Larenas 129, Concepción 4070371, Chile
| |
Collapse
|
18
|
Petsakou A, Liu Y, Liu Y, Comjean A, Hu Y, Perrimon N. Epithelial Ca 2+ waves triggered by enteric neurons heal the gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553227. [PMID: 37645990 PMCID: PMC10461974 DOI: 10.1101/2023.08.14.553227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the etiology of chronic disorders such as inflammatory bowel diseases and cancer. We used the Drosophila midgut to investigate this question and discovered that during regeneration a subpopulation of cholinergic enteric neurons triggers Ca2+ currents among enterocytes to promote return of the epithelium to homeostasis. Specifically, we found that down-regulation of the cholinergic enzyme Acetylcholinesterase in the epithelium enables acetylcholine from defined enteric neurons, referred as ARCENs, to activate nicotinic receptors in enterocytes found near ARCEN-innervations. This activation triggers high Ca2+ influx that spreads in the epithelium through Inx2/Inx7 gap junctions promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki activation and increase of inflammatory cytokines together with hyperplasia, reminiscent of inflammatory bowel diseases. Altogether, we found that during gut regeneration the conserved cholinergic pathway facilitates epithelial Ca2+ waves that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric-dependent intestinal regeneration which advance the current understanding of how a tissue returns to its homeostatic state after injury and could ultimately help existing therapeutics.
Collapse
Affiliation(s)
| | - Yifang Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Aram Comjean
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Boston, USA
| |
Collapse
|
19
|
Karkoska KA, Gollamudi J, Hyacinth HI. Molecular and environmental contributors to neurological complications in sickle cell disease. Exp Biol Med (Maywood) 2023; 248:1319-1332. [PMID: 37688519 PMCID: PMC10625341 DOI: 10.1177/15353702231187646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobinopathy in which affected hemoglobin polymerizes under hypoxic conditions resulting in red cell distortion and chronic hemolytic anemia. SCD affects millions of people worldwide, primarily in Sub-Saharan Africa and the Indian subcontinent. Due to vaso-occlusion of sickled red cells within the microvasculature, SCD affects virtually every organ system and causes significant morbidity and early mortality. The neurological complications of SCD are particularly devastating and diverse, ranging from overt stroke to covert cerebral injury, including silent cerebral infarctions and blood vessel tortuosity. However, even individuals without evidence of neuroanatomical changes in brain imaging have evidence of cognitive deficits compared to matched healthy controls likely due to chronic cerebral hypoxemia and neuroinflammation. In this review, we first examined the biological contributors to SCD-related neurological complications and then discussed the equally important socioenvironmental contributors. We then discuss the evidence for neuroprotection from the two primary disease-modifying therapies, chronic monthly blood transfusions and hydroxyurea, and end with several experimental therapies designed to specifically target these complications.
Collapse
Affiliation(s)
- Kristine A Karkoska
- Division of Hematology & Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45219-0525, USA
| | - Jahnavi Gollamudi
- Division of Hematology & Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45219-0525, USA
| | - Hyacinth I Hyacinth
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0525, USA
| |
Collapse
|
20
|
Frasch MG. Heart Rate Variability Code: Does It Exist and Can We Hack It? Bioengineering (Basel) 2023; 10:822. [PMID: 37508849 PMCID: PMC10375964 DOI: 10.3390/bioengineering10070822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
A code is generally defined as a system of signals or symbols for communication. Experimental evidence is synthesized for the presence and utility of such communication in heart rate variability (HRV) with particular attention to fetal HRV: HRV contains signatures of information flow between the organs and of response to physiological or pathophysiological stimuli as signatures of states (or syndromes). HRV exhibits features of time structure, phase space structure, specificity with respect to (organ) target and pathophysiological syndromes, and universality with respect to species independence. Together, these features form a spatiotemporal structure, a phase space, that can be conceived of as a manifold of a yet-to-be-fully understood dynamic complexity. The objective of this article is to synthesize physiological evidence supporting the existence of HRV code: hereby, the process-specific subsets of HRV measures indirectly map the phase space traversal reflecting the specific information contained in the code required for the body to regulate the physiological responses to those processes. The following physiological examples of HRV code are reviewed, which are reflected in specific changes to HRV properties across the signal-analytical domains and across physiological states and conditions: the fetal systemic inflammatory response, organ-specific inflammatory responses (brain and gut), chronic hypoxia and intrinsic (heart) HRV (iHRV), allostatic load (physiological stress due to surgery), and vagotomy (bilateral cervical denervation). Future studies are proposed to test these observations in more depth, and the author refers the interested reader to the referenced publications for a detailed study of the HRV measures involved. While being exemplified mostly in the studies of fetal HRV, the presented framework promises more specific fetal, postnatal, and adult HRV biomarkers of health and disease, which can be obtained non-invasively and continuously.
Collapse
Affiliation(s)
- Martin Gerbert Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
21
|
Salm DC, Horewicz VV, Tanaka F, Ferreira JK, de Oliveira BH, Maio JMB, Donatello NN, Ludtke DD, Mazzardo-Martins L, Dutra AR, Mack JM, de C H Kunzler D, Cargnin-Ferreira E, Salgado ASI, Bittencourt EB, Bianco G, Piovezan AP, Bobinski F, Moré AOO, Martins DF. Electrical Stimulation of the Auricular Branch Vagus Nerve Using Random and Alternating Frequencies Triggers a Rapid Onset and Pronounced Antihyperalgesia via Peripheral Annexin A1-Formyl Peptide Receptor 2/ALX Pathway in a Mouse Model of Persistent Inflammatory Pain. Mol Neurobiol 2023; 60:2889-2909. [PMID: 36745336 DOI: 10.1007/s12035-023-03237-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 01/13/2023] [Indexed: 02/07/2023]
Abstract
This study evaluated the antihyperalgesic and anti-inflammatory effects of percutaneous vagus nerve electrical stimulation (pVNS) by comparing the effects of alternating and random frequencies in an animal model of persistent inflammatory hyperalgesia. The model was induced by Freund's complete adjuvant (CFA) intraplantar (i.pl.) injection. Mice were treated with different protocols of time (10, 20, or 30 min), ear laterality (right, left or both), and frequency (alternating or random). Mechanical hyperalgesia was evaluated, and some groups received i.pl. WRW4 (FPR2/ALX antagonist) to determine the involvement. Edema, paw surface temperature, and spontaneous locomotor activity were evaluated. Interleukin-1β, IL-6, IL-10, and IL4 levels were verified by enzyme-linked immunosorbent assay. AnxA1, FPR2/ALX, neutrophil, M1 and M2 phenotype macrophage, and apoptotic cells markers were identified using western blotting. The antihyperalgesic effect pVNS with alternating and random frequency effect is depending on the type of frequency, time, and ear treated. The pVNS random frequency in the left ear for 10 min had a longer lasting antihyperalgesic effect, superior to classical stimulation using alternating frequency and the FPR2/ALX receptor was involved in this effect. There was a reduction in the levels of pro-inflammatory cytokines and an increase in the immunocontent of AnxA1 and CD86 in mice paw. pVNS with a random frequency in the left ear for 10 min showed to be optimal for inducing an antihyperalgesic effect. Thus, the random frequency was more effective than the alternating frequency. Therefore, pVNS may be an important adjunctive treatment for persistent inflammatory pain.
Collapse
Affiliation(s)
- Daiana C Salm
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Verônica V Horewicz
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Fernanda Tanaka
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Júlia K Ferreira
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Bruna H de Oliveira
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Julia Maria Batista Maio
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Nathalia N Donatello
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Daniela D Ludtke
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Leidiane Mazzardo-Martins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Aline R Dutra
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Josiel M Mack
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Deborah de C H Kunzler
- Department of Physiotherapy, State University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | | | | | - Gianluca Bianco
- Research Laboratory of Posturology and Neuromodulation RELPON, Department of Human Neuroscience, Sapienza University, Rome, Italy
- Istituto Di Formazione in Agopuntura E Neuromodulazione IFAN, Rome, Italy
| | - Anna Paula Piovezan
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Ari O O Moré
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Integrative Medicine and Acupuncture Division, University Hospital, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel F Martins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil.
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil.
| |
Collapse
|
22
|
Liu T, Fu Y, Shi J, He S, Chen D, Li W, Chen Y, Zhang L, Lv Q, Yang Y, Jin Q, Wang J, Xie M. Noninvasive ultrasound stimulation to treat myocarditis through splenic neuro-immune regulation. J Neuroinflammation 2023; 20:94. [PMID: 37069636 PMCID: PMC10108488 DOI: 10.1186/s12974-023-02773-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway (CAP) has been widely studied to modulate the immune response. Current stimulating strategies are invasive or imprecise. Noninvasive low-intensity pulsed ultrasound (LIPUS) has become increasingly appreciated for targeted neuronal modulation. However, its mechanisms and physiological role on myocarditis remain poorly defined. METHODS The mouse model of experimental autoimmune myocarditis was established. Low-intensity pulsed ultrasound was targeted at the spleen to stimulate the spleen nerve. Under different ultrasound parameters, histological tests and molecular biology were performed to observe inflammatory lesions and changes in immune cell subsets in the spleen and heart. In addition, we evaluated the dependence of the spleen nerve and cholinergic anti-inflammatory pathway of low-intensity pulsed ultrasound in treating autoimmune myocarditis in mice through different control groups. RESULTS The echocardiography and flow cytometry of splenic or heart infiltrating immune cells revealed that splenic ultrasound could alleviate the immune response, regulate the proportion and function of CD4+ Treg and macrophages by activating cholinergic anti-inflammatory pathway, and finally reduce heart inflammatory injury and improve cardiac remodeling, which is as effective as an acetylcholine receptor agonists GTS-21. Transcriptome sequencing showed significant differential expressed genes due to ultrasound modulation. CONCLUSIONS It is worth noting that the ultrasound therapeutic efficacy depends greatly on acoustic pressure and exposure duration, and the effective targeting organ was the spleen but not the heart. This study provides novel insight into the therapeutic potentials of LIPUS, which are essential for its future application.
Collapse
Affiliation(s)
- Tianshu Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanan Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shukun He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dandan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
23
|
Ahmed O, Caravaca AS, Crespo M, Dai W, Liu T, Guo Q, Leiva M, Sabio G, Shavva VS, Malin SG, Olofsson PS. Hepatic stellate cell activation markers are regulated by the vagus nerve in systemic inflammation. Bioelectron Med 2023; 9:6. [PMID: 36997988 PMCID: PMC10064698 DOI: 10.1186/s42234-023-00108-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND The liver is an important immunological organ and liver inflammation is part of the pathophysiology of non-alcoholic steatohepatitis, a condition that may promote cirrhosis, liver cancer, liver failure, and cardiovascular disease. Despite dense innervation of the liver parenchyma, little is known about neural regulation of liver function in inflammation. Here, we study vagus nerve control of the liver response to acute inflammation. METHODS Male C57BL/6 J mice were subjected to either sham surgery, surgical vagotomy, or electrical vagus nerve stimulation followed by intraperitoneal injection of the TLR2 agonist zymosan. Animals were euthanized and tissues collected 12 h after injection. Samples were analyzed by qPCR, RNAseq, flow cytometry, or ELISA. RESULTS Hepatic mRNA levels of pro-inflammatory mediators Ccl2, Il-1β, and Tnf-α were significantly higher in vagotomized mice compared with mice subjected to sham surgery. Differences in liver Ccl2 levels between treatment groups were largely reflected in the plasma chemokine (C-C motif) ligand 2 (CCL2) concentration. In line with this, we observed a higher number of macrophages in the livers of vagotomized mice compared with sham as measured by flow cytometry. In mice subjected to electrical vagus nerve stimulation, hepatic mRNA levels of Ccl2, Il1β, and Tnf-α, and plasma CCL2 levels, were significantly lower compared with sham. Interestingly, RNAseq revealed that a key activation marker for hepatic stellate cells (HSC), Pnpla3, was the most significantly differentially expressed gene between vagotomized and sham mice. Of note, several HSC-activation associated transcripts were higher in vagotomized mice, suggesting that signals in the vagus nerve contribute to HSC activation. In support of this, we observed significantly higher number of activated HSCs in vagotomized mice as compared with sham as measured by flow cytometry. CONCLUSIONS Signals in the cervical vagus nerve controlled hepatic inflammation and markers of HSC activation in zymosan-induced peritonitis.
Collapse
Affiliation(s)
- Osman Ahmed
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry, Faculty of Medicine, Khartoum University, Khartoum, Sudan
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - April S Caravaca
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Crespo
- Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain
| | - Wanmin Dai
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ting Liu
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Qi Guo
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Magdalena Leiva
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Guadalupe Sabio
- Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain
| | - Vladimir S Shavva
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Stephen G Malin
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peder S Olofsson
- Department of Medicine Solna, Laboratory of Immunobiology, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Medicine Solna, Stockholm Center for Bioelectronic Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
24
|
Elgoyhen AB. The α9α10 acetylcholine receptor: a non-neuronal nicotinic receptor. Pharmacol Res 2023; 190:106735. [PMID: 36931539 DOI: 10.1016/j.phrs.2023.106735] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Within the superfamily of pentameric ligand-gated ion channels, cholinergic nicotinic receptors (nAChRs) were classically identified to mediate synaptic transmission in the nervous system and the neuromuscular junction. The α9 and α10 nAChR subunits were the last ones to be identified. Surprisingly, they do not fall into the dichotomic neuronal/muscle classification of nAChRs. They assemble into heteropentamers with a well-established function as canonical ion channels in inner ear hair cells, where they mediate central nervous system control of auditory and vestibular sensory processing. The present review includes expression, pharmacological, structure-function, molecular evolution and pathophysiological studies, that define receptors composed from α9 and α10 subunits as distant and distinct members within the nAChR family. Thus, although α9 and α10 were initially included within the neuronal subdivision of nAChR subunits, they form a distinct clade within the phylogeny of nAChRs. Following the classification of nAChR subunits based on their main synaptic site of action, α9 and α10 should receive a name in their own right.
Collapse
Affiliation(s)
- Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina.
| |
Collapse
|
25
|
Tate WP, Walker MOM, Peppercorn K, Blair ALH, Edgar CD. Towards a Better Understanding of the Complexities of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID. Int J Mol Sci 2023; 24:ijms24065124. [PMID: 36982194 PMCID: PMC10048882 DOI: 10.3390/ijms24065124] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex condition arising in susceptible people, predominantly following viral infection, but also other stressful events. The susceptibility factors discussed here are both genetic and environmental although not well understood. While the dysfunctional physiology in ME/CFS is becoming clearer, understanding has been hampered by different combinations of symptoms in each affected person. A common core set of mainly neurological symptoms forms the modern clinical case definition, in the absence of an accessible molecular diagnostic test. This landscape has prompted interest in whether ME/CFS patients can be classified into a particular phenotype/subtype that might assist better management of their illness and suggest preferred therapeutic options. Currently, the same promising drugs, nutraceuticals, or behavioral therapies available can be beneficial, have no effect, or be detrimental to each individual patient. We have shown that individuals with the same disease profile exhibit unique molecular changes and physiological responses to stress, exercise and even vaccination. Key features of ME/CFS discussed here are the possible mechanisms determining the shift of an immune/inflammatory response from transient to chronic in ME/CFS, and how the brain and CNS manifests the neurological symptoms, likely with activation of its specific immune system and resulting neuroinflammation. The many cases of the post viral ME/CFS-like condition, Long COVID, following SARS-CoV-2 infection, and the intense research interest and investment in understanding this condition, provide exciting opportunities for the development of new therapeutics that will benefit ME/CFS patients.
Collapse
Affiliation(s)
- Warren P Tate
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Max O M Walker
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Anna L H Blair
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Christina D Edgar
- Department of Biochemistry, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
26
|
Adang EAMC, Strous MTA, van den Bergh JP, Gach D, van Kampen VEM, van Zeeland REP, Barten DG, van Osch FHM. Association of Heart Rate Variability with Pulmonary Function Impairment and Symptomatology Post-COVID-19 Hospitalization. SENSORS (BASEL, SWITZERLAND) 2023; 23:s23052473. [PMID: 36904676 PMCID: PMC10007596 DOI: 10.3390/s23052473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 06/12/2023]
Abstract
The persistence of symptoms beyond three months after COVID-19 infection, often referred to as post-COVID-19 condition (PCC), is commonly experienced. It is hypothesized that PCC results from autonomic dysfunction with decreased vagal nerve activity, which can be indexed by low heart rate variability (HRV). The aim of this study was to assess the association of HRV upon admission with pulmonary function impairment and the number of reported symptoms beyond three months after initial hospitalization for COVID-19 between February and December 2020. Follow-up took place three to five months after discharge and included pulmonary function tests and the assessment of persistent symptoms. HRV analysis was performed on one 10 s electrocardiogram obtained upon admission. Analyses were performed using multivariable and multinomial logistic regression models. Among 171 patients who received follow-up, and with an electrocardiogram at admission, decreased diffusion capacity of the lung for carbon monoxide (DLCO) (41%) was most frequently found. After a median of 119 days (IQR 101-141), 81% of the participants reported at least one symptom. HRV was not associated with pulmonary function impairment or persistent symptoms three to five months after hospitalization for COVID-19.
Collapse
Affiliation(s)
- Estelle A. M. C. Adang
- Department of Emergency Medicine, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
| | - Maud T. A. Strous
- Department of Internal Medicine, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
| | - Joop P. van den Bergh
- Department of Internal Medicine, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Debbie Gach
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Clinical Epidemiology, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
| | | | | | - Dennis G. Barten
- Department of Emergency Medicine, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
| | - Frits H. M. van Osch
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Clinical Epidemiology, VieCuri Medical Centre, 5912 BL Venlo, The Netherlands
- Department of Epidemiology, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
27
|
Mohanta SK, Yin C, Weber C, Habenicht AJR. Neuroimmune cardiovascular interfaces in atherosclerosis. Front Cell Dev Biol 2023; 11:1117368. [PMID: 36793445 PMCID: PMC9923102 DOI: 10.3389/fcell.2023.1117368] [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: 12/06/2022] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Two pairs of biological systems acting over long distances have recently been defined as major participants in the regulation of physiological and pathological tissue reactions: i) the nervous and vascular systems form various blood-brain barriers and control axon growth and angiogenesis; and ii) the nervous and immune systems emerge as key players to direct immune responses and maintain blood vessel integrity. The two pairs have been explored by investigators in relatively independent research areas giving rise to the concepts of the rapidly expanding topics of the neurovascular link and neuroimmunology, respectively. Our recent studies on atherosclerosis led us to consider a more inclusive approach by conceptualizing and combining principles of the neurovascular link and neuroimmunology: we propose that the nervous system, the immune system and the cardiovascular system undergo complex crosstalks in tripartite rather than bipartite interactions to form neuroimmune cardiovascular interfaces (NICIs).
Collapse
Affiliation(s)
- Sarajo K. Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Andreas J. R. Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
28
|
A role of gut-microbiota-brain axis via subdiaphragmatic vagus nerve in depression-like phenotypes in Chrna7 knock-out mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110652. [PMID: 36191806 DOI: 10.1016/j.pnpbp.2022.110652] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/21/2022]
Abstract
The α7 subtype of the nicotinic acetylcholine receptor (α7 nAChR: coded by Chrna7) is known to regulate the cholinergic ascending anti-inflammatory pathway. We previously reported that Chrna7 knock-out (KO) mice show depression-like behaviors through abnormal composition of gut microbiota and systemic inflammation. Given the role of subdiaphragmatic vagus nerve in gut-microbiota-brain axis, we investigated whether subdiaphragmatic vagotomy (SDV) could affect depression-like behaviors, abnormal composition of gut microbiota, and microbes-derived metabolites in Chrna7 KO mice. SDV blocked depression-like behaviors and reduced expression of synaptic proteins in the medial prefrontal cortex (mPFC) of Chrna7 KO mice. LEfSe (linear discriminant analysis effect size) analysis revealed that the species Lactobacillus sp. BL302, the species Lactobacillus hominis, and the species Lactobacillus reuteri, were identified as potential microbial markers in the KO + SDV group. There were several genus and species altered among the three groups [wild-type (WT) + sham group, KO + sham group, KO + SDV group]. Furthermore, there were several plasma metabolites altered among the three groups. Moreover, there were correlations between relative abundance of several microbiome and behavioral data (or synaptic proteins). Network analysis showed correlations between relative abundance of several microbiome and plasma metabolites (or behavioral data). These data suggest that Chrna7 KO mice produce depression-like behaviors and reduced expression of synaptic proteins in the mPFC through gut-microbiota-brain axis via subdiaphragmatic vagus nerve.
Collapse
|
29
|
PSpice modeling of cervical and site-focused vagus nerve ultrasonic stimulation for reduced tumor necrosis factor-α production. Sci Rep 2022; 12:21470. [PMID: 36509814 PMCID: PMC9744857 DOI: 10.1038/s41598-022-25944-2] [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: 06/13/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Clinical ultrasound is widely used as a diagnostic and therapeutic tool. Recently, it has been used to perform neuromodulation to treat diverse effects, including inflammation reduction through the vagus nerve. Although the mechanism by which ultrasound propagates through tissue for diagnostic purposes has been established, there is not a complete understanding of how it interacts with neurons to elicit excitation and inhibit inflammation. This work presents a novel technique based on a well-established electrical engineering tool, PSpice, to model cervical and site-focused vagus nerve ultrasonic stimulation to understand its capability in reducing tumor necrosis factor-α (TNF-α) production in the spleen. Transmission line theory is utilized as the basis for the different tissue layers. The models supported the hypothesis that site-focused stimulation has the advantage to decrease undesired efferent effects that would otherwise occur with cervical stimulation. Two different acoustic pressures, 0.25 and 0.83 MPa, were simulated for theoretical efficacy and safety based on previous experimental work conducted by others. The 0.25 MPa simulation was ideal for neurostimulation and reduction of TNF-α, while 0.83 MPa resulted in much higher intensity levels that will most likely induce additional inflammation.
Collapse
|
30
|
Cherifi F, Lefevre Arbogast S, Font J, Abdeddaim C, Becourt S, Penel N, Coquan E, Lequesne J, Gidron Y, Joly F. The promising prognostic value of vagal nerve activity at the initial management of ovarian cancer. Front Oncol 2022; 12:1049970. [PMID: 36523968 PMCID: PMC9745166 DOI: 10.3389/fonc.2022.1049970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2023] Open
Abstract
OBJECTIVE Identifying new modifiable prognostic markers is important for ovarian cancer (OC). Low parasympathic activity is associated with inflammation, oxidative stress and sympathetic nervous system activation. Previous studies reported that low vagal nerve activity, measured by low heart rate variability (HRV), may predict poor cancer prognosis. We aimed to examine the prognostic value of HRV in OC. METHODS This bicentric retrospective study included patients diagnosed with serous OC FIGO stage ≥IIB, between January 2015 and August 2019, with electrocardiograms (ECG) available around diagnosis. HRV was measured from ECG using the time domain parameter of standard deviation of all normal-to-normal heartbeat intervals (SDNN). Optimal SDNN cut-off was determined using the Youden index criteria of time-dependent ROC curves. We used multivariate cox proportional hazard models to investigate the association between HRV and overall survival (OS), while adjusting for well-known OC prognostic factors. RESULTS The 202 patients included were 65.7 years-old on average, 93% had stage FIGO IIIC/IV, 56% had complete surgical resection. Median OS was 38.6 months [95%CI:34.4-47.4]. The median SDNN was 11.1ms, with an optimal cut-off of 10ms to predict OS. OS was shorter for patients with low HRV compared to high HRV (26.4 vs 45.1 months; p<0.001). In multivariate analysis, HRV remained an independent prognostic factor with a two-fold higher risk of death among patients with low SDNN compared to those with high SDNN (HR=2.03, 95%CI=1.35-3.06, p<0.001). CONCLUSION Low HRV, was associated with worse OS in OC patients, supporting previous studies on the prognostic role of HRV in cancer. If replicated in prospective studies, vagal nerve activity may be a new therapeutic target in OC.
Collapse
Affiliation(s)
- François Cherifi
- Department of Medical Oncology, Centre François Baclesse, Caen, France
- Department of Clinical Research, Centre Francois Baclesse, Caen, France
| | - Sophie Lefevre Arbogast
- Department of Clinical Research, Centre Francois Baclesse, Caen, France
- Normandie Université, UNICAEN, Institut National de la Santé et de la Recherche Médicale (Inserm) U1086, ANTICIPE, Caen, France
| | - Jonaz Font
- Department of Cardiology, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Cyril Abdeddaim
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | | | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Elodie Coquan
- Department of Medical Oncology, Centre François Baclesse, Caen, France
- Department of Clinical Research, Centre Francois Baclesse, Caen, France
| | - Justine Lequesne
- Department of Clinical Research, Centre Francois Baclesse, Caen, France
| | - Yori Gidron
- Department of Nursing, Faculty of Health Sciences, Haifa University, Haifa, Israel
| | - Florence Joly
- Department of Medical Oncology, Centre François Baclesse, Caen, France
- Normandie Université, UNICAEN, Institut National de la Santé et de la Recherche Médicale (Inserm) U1086, ANTICIPE, Caen, France
| |
Collapse
|
31
|
Muñoz AI, Maldonado-García JL, Fragozo A, Vallejo-Castillo L, Lucas-Gonzalez A, Trejo-Martínez I, Pavón L, Pérez-Sánchez G, Cobos-Marin L, Pérez-Tapia SM. Altered neutrophil-to-lymphocyte ratio in sepsis secondary to canine parvoviral enteritis treated with and without an immunomodulator in puppies. Front Vet Sci 2022; 9:995443. [PMID: 36425123 PMCID: PMC9679511 DOI: 10.3389/fvets.2022.995443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/20/2022] [Indexed: 08/29/2023] Open
Abstract
Neutrophil-to-lymphocyte ratio (NLR) is a cheap and easy-to-obtain biomarker that mirrors the balance between innate and adaptive immunity. Cortisol and catecholamines have been identified as major drivers of NLR. High cortisol levels increase neutrophils while simultaneously decreasing lymphocyte counts. Likewise, endogenous catecholamines may cause leukocytosis and lymphopenia. Thus, NLR allows us to monitor patient severity in conditions such as sepsis. Twenty-six puppies with sepsis secondary to canine parvoviral enteritis were treated with and without an immunomodulator. Our group determined the NLR and the plasmatic cortisol levels by chemiluminescence, and norepinephrine (NE) and epinephrine (E) by HPLC during the first 72 h of clinical follow-up. Our results showed that at admission puppies presented an NLR value of 1.8, cortisol of 314.9 nmol/L, NE 3.7, and E 3.3 pmol/mL. Both treatments decreased admission NLR values after 24 h of treatment. However, only the puppies treated with the immunomodulator (I) remained without significant changes in NLR (0.7-1.4) compared to the CT group, and that showed a significant difference (P < 0.01) in their NLR value (0.4-4.6). In addition, we found significant differences in the slope values between the admission and final values of NLR (P < 0.005), cortisol (P < 0.02), and E (P < 0.05) between treatments. Then, our data suggest that the immunomodulator positively affects the number of lymphocytes and neutrophils involved in NLR as well as major drivers like cortisol and epinephrine, which is reflected in clinical parameters and survival.
Collapse
Affiliation(s)
- Adriana I. Muñoz
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - José Luis Maldonado-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Ana Fragozo
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I + D + i) para Farmoquímicos y Biotecnológicos (LANSEIDI-FarBiotec-CONACyT), Mexico City, Mexico
| | - Luis Vallejo-Castillo
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I + D + i) para Farmoquímicos y Biotecnológicos (LANSEIDI-FarBiotec-CONACyT), Mexico City, Mexico
| | - Amellalli Lucas-Gonzalez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I + D + i) para Farmoquímicos y Biotecnológicos (LANSEIDI-FarBiotec-CONACyT), Mexico City, Mexico
| | - Ismael Trejo-Martínez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I + D + i) para Farmoquímicos y Biotecnológicos (LANSEIDI-FarBiotec-CONACyT), Mexico City, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Laura Cobos-Marin
- Laboratorio de Virología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I + D + i) para Farmoquímicos y Biotecnológicos (LANSEIDI-FarBiotec-CONACyT), Mexico City, Mexico
| |
Collapse
|
32
|
Stapelberg NJC, Branjerdporn G, Adhikary S, Johnson S, Ashton K, Headrick J. Environmental Stressors and the PINE Network: Can Physical Environmental Stressors Drive Long-Term Physical and Mental Health Risks? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13226. [PMID: 36293807 PMCID: PMC9603079 DOI: 10.3390/ijerph192013226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Both psychosocial and physical environmental stressors have been linked to chronic mental health and chronic medical conditions. The psycho-immune-neuroendocrine (PINE) network details metabolomic pathways which are responsive to varied stressors and link chronic medical conditions with mental disorders, such as major depressive disorder via a network of pathophysiological pathways. The primary objective of this review is to explore evidence of relationships between airborne particulate matter (PM, as a concrete example of a physical environmental stressor), the PINE network and chronic non-communicable diseases (NCDs), including mental health sequelae, with a view to supporting the assertion that physical environmental stressors (not only psychosocial stressors) disrupt the PINE network, leading to NCDs. Biological links have been established between PM exposure, key sub-networks of the PINE model and mental health sequelae, suggesting that in theory, long-term mental health impacts of PM exposure may exist, driven by the disruption of these biological networks. This disruption could trans-generationally influence health; however, long-term studies and information on chronic outcomes following acute exposure event are still lacking, limiting what is currently known beyond the acute exposure and all-cause mortality. More empirical evidence is needed, especially to link long-term mental health sequelae to PM exposure, arising from PINE pathophysiology. Relationships between physical and psychosocial stressors, and especially the concept of such stressors acting together to impact on PINE network function, leading to linked NCDs, evokes the concept of syndemics, and these are discussed in the context of the PINE network.
Collapse
Affiliation(s)
- Nicolas J. C. Stapelberg
- Gold Coast Hospital and Health Service, Gold Coast, QLD 4215, Australia
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4226, Australia
| | - Grace Branjerdporn
- Gold Coast Hospital and Health Service, Gold Coast, QLD 4215, Australia
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4226, Australia
| | - Sam Adhikary
- Mater Young Adult Health Centre, Mater Hospital, Brisbane, QID 4101, Australia
| | - Susannah Johnson
- Gold Coast Hospital and Health Service, Gold Coast, QLD 4215, Australia
| | - Kevin Ashton
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4226, Australia
| | - John Headrick
- School of Medical Science, Griffith University, Gold Coast, QID 4215, Australia
| |
Collapse
|
33
|
McKinley MJ, Martelli D, Trevizan-Baú P, McAllen RM. Divergent splanchnic sympathetic efferent nerve pathways regulate interleukin-10 and tumour necrosis factor-α responses to endotoxaemia. J Physiol 2022; 600:4521-4536. [PMID: 36056471 DOI: 10.1113/jp283217] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/18/2022] [Indexed: 01/05/2023] Open
Abstract
The efferent branches of the splanchnic sympathetic nerves that enhance interleukin-10 (IL-10) and suppress tumour necrosis factor-α (TNF) levels in the reflex response to systemic immune challenge were investigated in anaesthetized, ventilated rats. Plasma levels of TNF and IL-10 were measured 90 min after intravenous lipopolysaccharide (LPS, 60 µg/kg). Splanchnic nerve section, ganglionic blockade with pentolinium tartrate or β2 adrenoreceptor antagonism with ICI 118551 all blocked IL-10 responses. Restoring plasma adrenaline after splanchnic denervation rescued IL-10 responses. TNF responses were disinhibited by splanchnic denervation or pentolinium treatment, but not by ICI 118551. Splanchnic nerve branches were cut individually or in combination in vagotomized rats, ruling out any vagal influence on results. Distal splanchnic denervation, sparing the adrenal nerves, disinhibited TNF but did not reduce IL-10 responses. Selective adrenal denervation depressed IL-10 but did not disinhibit TNF responses. Selective denervation of either spleen or liver did not affect IL-10 or TNF responses, but combined splenic and adrenal denervation did so. Finally, combined section of the cervical and lumbar sympathetic nerves did not affect cytokine responses to LPS. Together, these results show that the endogenous anti-inflammatory reflex is mediated by sympathetic efferent fibres that run in the splanchnic, but not other sympathetic nerves, nor the vagus. Within the splanchnic nerves, divergent pathways control these two cytokine responses: neurally driven adrenaline, acting via β2 adrenoreceptors, regulates IL-10, while TNF is restrained by sympathetic nerves to abdominal organs including the spleen, where non-β2 adrenoreceptor mechanisms are dominant. KEY POINTS: An endogenous neural reflex, mediated by the splanchnic, but not other sympathetic nerves, moderates the cytokine response to systemic inflammatory challenge. This reflex suppresses the pro-inflammatory cytokine tumour necrosis factor-α (TNF), while enhancing levels of the anti-inflammatory cytokine interleukin-10 (IL-10). The reflex enhancement of IL-10 depends on the splanchnic nerve supply to the adrenal gland and on β2 adrenoreceptors, consistent with mediation by circulating adrenaline. After splanchnic nerve section it can be rescued by restoring circulating adrenaline. The reflex suppression of TNF depends on splanchnic nerve branches that innervate abdominal tissues including, but not restricted to, spleen: it is not blocked by adrenal denervation or β2 adrenoreceptor antagonism. Distinct sympathetic efferent pathways are thus responsible for pro- and anti-inflammatory cytokine components of the reflex regulating inflammation.
Collapse
Affiliation(s)
- Michael J McKinley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia.,Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Davide Martelli
- Department of Biomedical and Neuromotor Sciences, Physiology Division, University of Bologna, Bologna, Italy
| | - Pedro Trevizan-Baú
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Robin M McAllen
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Asiedu K. Role of ocular surface neurobiology in neuronal-mediated inflammation in dry eye disease. Neuropeptides 2022; 95:102266. [PMID: 35728484 DOI: 10.1016/j.npep.2022.102266] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/18/2023]
Abstract
Inflammation is the consequence of dry eye disease regardless of its etiology. Several injurious or harmless processes to the ocular surface neurons promote ocular surface neurogenic inflammation, leading to the vicious cycle of dry eye disease. These processes include the regular release of neuromediators during the conduction of ocular surface sensations, hyperosmolarity-induced ocular surface neuronal damage, neuro-regenerative activities, and neuronal-mediated dendritic cell activities. Neurogenic inflammation appears to be the main culprit, instigating the self-perpetuating inflammation observed in patients with dry eye disease.
Collapse
Affiliation(s)
- Kofi Asiedu
- School of Optometry & Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
35
|
Irwin MR. Sleep disruption induces activation of inflammation and heightens risk for infectious disease: Role of impairments in thermoregulation and elevated ambient temperature. Temperature (Austin) 2022; 10:198-234. [PMID: 37332305 PMCID: PMC10274531 DOI: 10.1080/23328940.2022.2109932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/25/2022] [Accepted: 07/30/2022] [Indexed: 10/15/2022] Open
Abstract
Thermoregulation and sleep are tightly coordinated, with evidence that impairments in thermoregulation as well as increases in ambient temperature increase the risk of sleep disturbance. As a period of rest and low demand for metabolic resources, sleep functions to support host responses to prior immunological challenges. In addition by priming the innate immune response, sleep prepares the body for injury or infection which might occur the following day. However when sleep is disrupted, this phasic organization between nocturnal sleep and the immune system becomes misaligned, cellular and genomic markers of inflammation are activated, and increases of proinflammatory cytokines shift from the nighttime to the day. Moreover, when sleep disturbance is perpetuated due to thermal factors such as elevated ambient temperature, the beneficial crosstalk between sleep and immune system becomes further imbalanced. Elevations in proinflammatory cytokines have reciprocal effects and induce sleep fragmentation with decreases in sleep efficiency, decreases in deep sleep, and increases in rapid eye movement sleep, further fomenting inflammation and inflammatory disease risk. Under these conditions, sleep disturbance has additional potent effects to decrease adaptive immune response, impair vaccine responses, and increase vulnerability to infectious disease. Behavioral interventions effectively treat insomnia and reverse systemic and cellular inflammation. Further, insomnia treatment redirects the misaligned inflammatory- and adaptive immune transcriptional profiles with the potential to mitigate risk of inflammation-related cardiovascular, neurodegenerative, and mental health diseases, as well as susceptibility to infectious disease.
Collapse
Affiliation(s)
- Michael R. Irwin
- University of California, Los Angeles – Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
36
|
Olsen LK, Moore RJ, Bechmann NA, Ethridge VT, Gargas NM, Cunningham SD, Kuang Z, Whicker JK, Rohan JG, Hatcher-Solis CN. Vagus nerve stimulation-induced cognitive enhancement: Hippocampal neuroplasticity in healthy male rats. Brain Stimul 2022; 15:1101-1110. [PMID: 35970317 DOI: 10.1016/j.brs.2022.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) improves cognition in humans and rodents, but the effects of a single session of VNS on performance and plasticity are not well understood. OBJECTIVE Behavioral performance and hippocampal (HC) electrophysiology/neurotrophin expression were measured in healthy adult rats after VNS paired training to investigate changes in cognition and synaptic plasticity. METHODS Platinum/iridium electrodes were surgically implanted around the left cervical branch of the VN of anesthetized male Sprague-Dawley rats (N = 47). VNS (100 μs biphasic pulses, 30 Hz, 0.8 mA) paired Novel Object Recognition (NOR)/Passive Avoidance Task (PAT) were assessed 24 h after training and post-mortem tissue was collected 48 h after VNS (N = 28). Electrophysiology recordings were collected using a microelectrode array system to assess functional effects on HC slices 90 min after VNS (N = 19). Sham received the same treatment without VNS and experimenters were blinded. RESULTS Stimulated rats exhibited improved performance in NOR (p < 0.05, n = 12) and PAT (p < 0.05, n = 14). VNS enhanced long-term potentiation (p < 0.05, n = 7-12), and spontaneous spike amplitude (p < 0.05, n = 7-12) and frequency (p < 0.05, n = 7-12) in the CA1. Immunohistochemical analysis found increased brain-derived neurotrophic factor expression in the CA1 (p < 0.05, n = 8-9) and CA2 (p < 0.01, n = 7-8). CONCLUSION These findings suggest that our VNS parameters promote synaptic plasticity and target the CA1, which may mediate the positive cognitive effects of VNS. This study significantly contributes to a better understanding of VNS mediated HC synaptic plasticity, which may improve clinical utilization of VNS for cognitive enhancement.
Collapse
Affiliation(s)
- Laura K Olsen
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Raquel J Moore
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA; Infoscitex, Inc., Dayton, OH, USA
| | - Naomi A Bechmann
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA; Infoscitex, Inc., Dayton, OH, USA
| | - Victoria T Ethridge
- Naval Medical Research Unit Dayton, Wright-Patterson AFB, OH, USA; Odyssey Systems Consulting Group, Wakefield, MA, USA
| | - Nathan M Gargas
- Naval Medical Research Unit Dayton, Wright-Patterson AFB, OH, USA; Odyssey Systems Consulting Group, Wakefield, MA, USA
| | - Sylvia D Cunningham
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Zhanpeng Kuang
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA
| | - Joshua K Whicker
- Cognitive Neuroscience, 711th HPW, AFRL, Wright-Patterson AFB, OH, USA
| | - Joyce G Rohan
- Naval Medical Research Unit Dayton, Wright-Patterson AFB, OH, USA
| | | |
Collapse
|
37
|
Akoolo L, Djokic V, Rocha SC, Ulloa L, Parveen N. Sciatic-Vagal Nerve Stimulation by Electroacupuncture Alleviates Inflammatory Arthritis in Lyme Disease-Susceptible C3H Mice. Front Immunol 2022; 13:930287. [PMID: 35924250 PMCID: PMC9342905 DOI: 10.3389/fimmu.2022.930287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022] Open
Abstract
Lyme disease is caused by Borrelia burgdorferi, and the pathogenesis of the disease is complex with both bacterial and host factors contributing to inflammatory responses. Lyme disease affects different organs including joints and results in arthritis. Immune responses stimulated by B. burgdorferi through toll-like receptors cause infiltration of leukocytes, which produce inflammatory cytokines and facilitate spirochete clearance. However, arthritic manifestations and chronic fatigue syndrome-like symptoms persist long after completion of antibiotic treatment regimens in a significant number of patients. To counter the effects of inflammation, treatment by non-steroidal anti-inflammatory drugs, hydroxychloroquine, or synovectomy to eradicate inflammatory arthritis in the involved joint could be employed; however, they often have long-term consequences. Acupuncture has been used for a long time in Asian medicine to diminish pain during various ailments, but the effects and its mechanism are just beginning to be explored. Control of inflammation by neuronal stimulation has been exploited as a systemic therapeutic intervention to arrest inflammatory processes. Our objective was to determine whether activation of the sciatic-vagal network by electroacupuncture on ST36 acupoint, which is used to control systemic inflammation in experimental models of infectious disorders such as endotoxemia, can also alleviate Lyme arthritis symptoms in mice. This aim was further strengthened by the reports that sciatic-vagal neuronal network stimulation can lead to dopamine production in the adrenal medulla and moderate the production of inflammatory factors. We first assessed whether electroacupuncture affects spirochete colonization to attenuate Lyme arthritis. Interestingly, bioluminescent B. burgdorferi burden detected by live imaging and qPCR were similar in electroacupuncture- and mock-treated mice, while electroacupuncture induced a lasting anti-inflammatory effect on mice. Despite the discontinuation of treatment at 2 weeks, the simultaneous decrease in neutrophils in the joints and inflammatory cytokine levels throughout the body at 4 weeks suggests a systemic and persistent effect of electroacupuncture that attenuates Lyme arthritis. Our results suggest that electroacupuncture-mediated anti-inflammatory responses could offer promising healthcare benefits in patients suffering from long-term Lyme disease manifestations.
Collapse
Affiliation(s)
- Lavoisier Akoolo
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Vitomir Djokic
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sandra C. Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Luis Ulloa
- Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
38
|
Czura CJ, Bikson M, Charvet L, Chen JDZ, Franke M, Fudim M, Grigsby E, Hamner S, Huston JM, Khodaparast N, Krames E, Simon BJ, Staats P, Vonck K. Neuromodulation Strategies to Reduce Inflammation and Improve Lung Complications in COVID-19 Patients. Front Neurol 2022; 13:897124. [PMID: 35911909 PMCID: PMC9329660 DOI: 10.3389/fneur.2022.897124] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022] Open
Abstract
Since the outbreak of the COVID-19 pandemic, races across academia and industry have been initiated to identify and develop disease modifying or preventative therapeutic strategies has been initiated. The primary focus has been on pharmacological treatment of the immune and respiratory system and the development of a vaccine. The hyperinflammatory state ("cytokine storm") observed in many cases of COVID-19 indicates a prognostically negative disease progression that may lead to respiratory distress, multiple organ failure, shock, and death. Many critically ill patients continue to be at risk for significant, long-lasting morbidity or mortality. The human immune and respiratory systems are heavily regulated by the central nervous system, and intervention in the signaling of these neural pathways may permit targeted therapeutic control of excessive inflammation and pulmonary bronchoconstriction. Several technologies, both invasive and non-invasive, are available and approved for clinical use, but have not been extensively studied in treatment of the cytokine storm in COVID-19 patients. This manuscript provides an overview of the role of the nervous system in inflammation and respiration, the current understanding of neuromodulatory techniques from preclinical and clinical studies and provides a rationale for testing non-invasive neuromodulation to modulate acute systemic inflammation and respiratory dysfunction caused by SARS-CoV-2 and potentially other pathogens. The authors of this manuscript have co-founded the International Consortium on Neuromodulation for COVID-19 to advocate for and support studies of these technologies in the current coronavirus pandemic.
Collapse
Affiliation(s)
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, New York, NY, United States
| | - Leigh Charvet
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
| | - Jiande D. Z. Chen
- Division of Gastroenterology and Hepatology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | | | - Marat Fudim
- Division of Cardiology, Duke Clinical Research Institute, Duke University, Durham, NC, United States
| | | | - Sam Hamner
- Cala Health, Burlingame, CA, United States
| | - Jared M. Huston
- Departments of Surgery and Science Education, Zucker School of Medicine at Hofstra/Northwell, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | | | - Elliot Krames
- Pacific Pain Treatment Center, Napa, CA, United States
| | | | - Peter Staats
- National Spine and Pain, ElectroCore, Inc., Jacksonville, FL, United States
| | - Kristl Vonck
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
39
|
Chaudhari S, Pham GS, Brooks CD, Dinh VQ, Young-Stubbs CM, Shimoura CG, Mathis KW. Should Renal Inflammation Be Targeted While Treating Hypertension? Front Physiol 2022; 13:886779. [PMID: 35770194 PMCID: PMC9236225 DOI: 10.3389/fphys.2022.886779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/17/2022] [Indexed: 11/28/2022] Open
Abstract
Despite extensive research and a plethora of therapeutic options, hypertension continues to be a global burden. Understanding of the pathological roles of known and underexplored cellular and molecular pathways in the development and maintenance of hypertension is critical to advance the field. Immune system overactivation and inflammation in the kidneys are proposed alternative mechanisms of hypertension, and resistant hypertension. Consideration of the pathophysiology of hypertension in chronic inflammatory conditions such as autoimmune diseases, in which patients present with autoimmune-mediated kidney inflammation as well as hypertension, may reveal possible contributors and novel therapeutic targets. In this review, we 1) summarize current therapies used to control blood pressure and their known effects on inflammation; 2) provide evidence on the need to target renal inflammation, specifically, and especially when first-line and combinatory treatment efforts fail; and 3) discuss the efficacy of therapies used to treat autoimmune diseases with a hypertension/renal component. We aim to elucidate the potential of targeting renal inflammation in certain subsets of patients resistant to current therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Keisa W. Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
40
|
Yeater TD, Cruz CJ, Cruz-Almeida Y, Allen KD. Autonomic Nervous System Dysregulation and Osteoarthritis Pain: Mechanisms, Measurement, and Future Outlook. Curr Rheumatol Rep 2022; 24:175-183. [PMID: 35420372 PMCID: PMC9189055 DOI: 10.1007/s11926-022-01071-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW The autonomic nervous system is an important regulator of stress responses and exhibits functional changes in chronic pain states. This review discusses potential overlap among autonomic dysregulation, osteoarthritis (OA) progression, and chronic pain. From this foundation, we then discuss preclinical to clinical research opportunities to close gaps in our knowledge of autonomic dysregulation and OA. Finally, we consider the potential to generate new therapies for OA pain via modulation of the autonomic nervous system. RECENT FINDINGS Recent reviews provide a framework for the autonomic nervous system in OA progression; however, research is still limited on the topic. In other chronic pain states, functional overlaps between the central autonomic network and pain processing centers in the brain suggest relationships between concomitant dysregulation of the two systems. Non-pharmacological therapeutics, such as vagus nerve stimulation, mindfulness-based meditation, and exercise, have shown promise in alleviating painful symptoms of joint diseases, and these interventions may be partially mediated through the autonomic nervous system. The autonomic nervous system appears to be dysregulated in OA progression, and further research on rebalancing autonomic function may lead to novel therapeutic strategies for treating OA pain.
Collapse
Affiliation(s)
- Taylor D. Yeater
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.,Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA
| | - Carlos J. Cruz
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.,Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA
| | - Yenisel Cruz-Almeida
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA.,Department of Community Dentistry & Behavioral Sciences, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Kyle D. Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.,Department of Orthopedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, FL, USA.,Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA
| |
Collapse
|
41
|
Caravaca AS, Gallina AL, Tarnawski L, Shavva VS, Colas RA, Dalli J, Malin SG, Hult H, Arnardottir H, Olofsson PS. Vagus nerve stimulation promotes resolution of inflammation by a mechanism that involves Alox15 and requires the α7nAChR subunit. Proc Natl Acad Sci U S A 2022; 119:e2023285119. [PMID: 35622894 PMCID: PMC9295760 DOI: 10.1073/pnas.2023285119] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/09/2022] [Indexed: 12/31/2022] Open
Abstract
Nonresolving inflammation underlies a range of chronic inflammatory diseases, and therapeutic acceleration of resolution of inflammation may improve outcomes. Neural reflexes regulate the intensity of inflammation (for example, through signals in the vagus nerve), but whether activation of the vagus nerve promotes the resolution of inflammation in vivo has been unknown. To investigate this, mice were subjected to electrical vagus nerve stimulation (VNS) or sham surgery at the cervical level followed by zymosan-induced peritonitis. The duration of inflammation resolution was significantly reduced and efferocytosis was significantly increased in mice treated with VNS as compared with sham. Lipid mediator (LM) metabololipidomics revealed that mice treated with VNS had higher levels of specialized proresolving mediators (SPMs), particularly from the omega-3 docosahexaenoic (DHA) and docosapentaenoic (n-3 DPA) metabolomes, in peritoneal exudates. VNS also shifted the ratio between proinflammatory and proresolving LMs toward a proresolving profile, but this effect by VNS was inverted in mice deficient in 12/15-lipoxgenase (Alox15), a key enzyme in this SPM biosynthesis. The significant VNS-mediated reduction of neutrophil numbers in peritoneal exudates was absent in mice deficient in the cholinergic α7-nicotinic acetylcholine receptor subunit (α7nAChR), an essential component of the inflammatory reflex. Thus, VNS increased local levels of SPM and accelerated resolution of inflammation in zymosan-induced peritonitis by a mechanism that involves Alox15 and requires the α7nAChR.
Collapse
Affiliation(s)
- April S. Caravaca
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Alessandro L. Gallina
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Laura Tarnawski
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Vladimir S. Shavva
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Romain A. Colas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Stephen G. Malin
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Henrik Hult
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
- Department of Mathematics, KTH Royal Institute of Technology, Stockholm, 114 28, Sweden
| | - Hildur Arnardottir
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Peder S. Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, 11030
| |
Collapse
|
42
|
Tate W, Walker M, Sweetman E, Helliwell A, Peppercorn K, Edgar C, Blair A, Chatterjee A. Molecular Mechanisms of Neuroinflammation in ME/CFS and Long COVID to Sustain Disease and Promote Relapses. Front Neurol 2022; 13:877772. [PMID: 35693009 PMCID: PMC9174654 DOI: 10.3389/fneur.2022.877772] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a disease now well-documented as having arisen commonly from a viral infection, but also from other external stressors, like exposure to agricultural chemicals, other types of infection, surgery, or other severe stress events. Research has shown these events produce a systemic molecular inflammatory response and chronic immune activation and dysregulation. What has been more difficult to establish is the hierarchy of the physiological responses that give rise to the myriad of symptoms that ME/CFS patients experience, and why they do not resolve and are generally life-long. The severity of the symptoms frequently fluctuates through relapse recovery periods, with brain-centered symptoms of neuroinflammation, loss of homeostatic control, "brain fog" affecting cognitive ability, lack of refreshing sleep, and poor response to even small stresses. How these brain effects develop with ME/CFS from the initiating external effector, whether virus or other cause, is poorly understood and that is what our paper aims to address. We propose the hypothesis that following the initial stressor event, the subsequent systemic pathology moves to the brain via neurovascular pathways or through a dysfunctional blood-brain barrier (BBB), resulting in chronic neuroinflammation and leading to a sustained illness with chronic relapse recovery cycles. Signaling through recognized pathways from the brain back to body physiology is likely part of the process by which the illness cycle in the peripheral system is sustained and why healing does not occur. By contrast, Long COVID (Post-COVID-19 condition) is a very recent ME/CFS-like illness arising from the single pandemic virus, SARS-CoV-2. We believe the ME/CFS-like ongoing effects of Long COVID are arising by very similar mechanisms involving neuroinflammation, but likely with some unique signaling, resulting from the pathology of the initial SARS-CoV-2 infection. The fact that there are very similar symptoms in both ongoing diseases, despite the diversity in the nature of the initial stressors, supports the concept of a similar dysfunctional CNS component common to both.
Collapse
Affiliation(s)
- Warren Tate
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Max Walker
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Eiren Sweetman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Amber Helliwell
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Christina Edgar
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Anna Blair
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
43
|
Yang H, George SJ, Thompson DA, Silverman HA, Tsaava T, Tynan A, Pavlov VA, Chang EH, Andersson U, Brines M, Chavan SS, Tracey KJ. Famotidine activates the vagus nerve inflammatory reflex to attenuate cytokine storm. Mol Med 2022; 28:57. [PMID: 35578169 PMCID: PMC9109205 DOI: 10.1186/s10020-022-00483-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/25/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Severe COVID-19 is characterized by pro-inflammatory cytokine release syndrome (cytokine storm) which causes high morbidity and mortality. Recent observational and clinical studies suggest famotidine, a histamine 2 receptor (H2R) antagonist widely used to treat gastroesophageal reflux disease, attenuates the clinical course of COVID-19. Because evidence is lacking for a direct antiviral activity of famotidine, a proposed mechanism of action is blocking the effects of histamine released by mast cells. Here we hypothesized that famotidine activates the inflammatory reflex, a brain-integrated vagus nerve mechanism which inhibits inflammation via alpha 7 nicotinic acetylcholine receptor (α7nAChR) signal transduction, to prevent cytokine storm. METHODS The potential anti-inflammatory effects of famotidine and other H2R antagonists were assessed in mice exposed to lipopolysaccharide (LPS)-induced cytokine storm. As the inflammatory reflex is integrated and can be stimulated in the brain, and H2R antagonists penetrate the blood brain barrier poorly, famotidine was administered by intracerebroventricular (ICV) or intraperitoneal (IP) routes. RESULTS Famotidine administered IP significantly reduced serum and splenic LPS-stimulated tumor necrosis factor (TNF) and IL-6 concentrations, significantly improving survival. The effects of ICV famotidine were significantly more potent as compared to the peripheral route. Mice lacking mast cells by genetic deletion also responded to famotidine, indicating the anti-inflammatory effects are not mast cell-dependent. Either bilateral sub-diaphragmatic vagotomy or genetic knock-out of α7nAChR abolished the anti-inflammatory effects of famotidine, indicating the inflammatory reflex as famotidine's mechanism of action. While the structurally similar H2R antagonist tiotidine displayed equivalent anti-inflammatory activity, the H2R antagonists cimetidine or ranitidine were ineffective even at very high dosages. CONCLUSIONS These observations reveal a previously unidentified vagus nerve-dependent anti-inflammatory effect of famotidine in the setting of cytokine storm which is not replicated by high dosages of other H2R antagonists in clinical use. Because famotidine is more potent when administered intrathecally, these findings are also consistent with a primarily central nervous system mechanism of action.
Collapse
Affiliation(s)
- Huan Yang
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Sam J George
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Dane A Thompson
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Harold A Silverman
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Téa Tsaava
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Aisling Tynan
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Valentin A Pavlov
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Eric H Chang
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Michael Brines
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sangeeta S Chavan
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| | - Kevin J Tracey
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
44
|
Chan A, Gao J, Houston M, Willett T, Farhadian B, Silverman M, Tran P, Jaradeh S, Thienemann M, Frankovich J. Children With PANS May Manifest POTS. Front Neurol 2022; 13:819636. [PMID: 35557616 PMCID: PMC9086964 DOI: 10.3389/fneur.2022.819636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/09/2022] [Indexed: 11/22/2022] Open
Abstract
Objectives Pediatric acute-onset neuropsychiatric syndrome (PANS) is characterized by an abrupt-onset of severe psychiatric symptoms including OCD, anxiety, cognitive difficulties, and sleep issues which is thought to be a post-infection brain inflammatory disorder. We observed postural orthostatic tachycardia syndrome (POTS) which resolved with immunomodulation in a patient with Pediatric acute-onset neuropsychiatric syndrome (PANS). Here, we aim to present a case of POTS and to examine the prevalence of (POTS) in our PANS cohort, and compare the clinical characteristics of patients with and without POTS. Study Design We conducted this cohort study of patients meeting PANS criteria who had at least three clinic visits during the study period. We included data from prospectively collected questionnaires and medical record review. We present a case followed by statistical comparisons within our cohort and a Kaplan-Meier analysis to determine the time-dependent risk of a POTS diagnosis. Results Our study included 204 patients: mean age of PANS onset was 8.6 years, male sex (60%), non-Hispanic White (78%). Evidence of POTS was observed in 19/204 patients (9%) with 5/19 having persistent POTS defined as persistent abnormal orthostatic vitals, persistent POTS symptoms, and/or continued need for pharmacotherapy for POTS symptoms for at least 6 months). In this PANS cohort, patients with POTS were more likely to have comorbid joint hypermobility (63 vs 37%, p = 0.04), chronic fatigue (42 vs 18%, p = 0.03), and a family history of chronic fatigue, POTS, palpitations and syncope. An unadjusted logistic regression model showed that a PANS flare (abrupt neuropsychiatric deterioration) was significantly associated with an exacerbation of POTS symptoms (OR 3.3, 95% CI 1.4–7.6, p < 0.01). Conclusions Our study describes a high prevalence of POTS in patients with PANS (compared to the general population) and supports an association between POTS presentation and PANS flare within our cohort.
Collapse
Affiliation(s)
- Avis Chan
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States.,Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | - Jaynelle Gao
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States.,Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | - Madison Houston
- Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States.,Department of Human Biology, Stanford University School of Humanities and Sciences, Stanford, CA, United States
| | - Theresa Willett
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States.,Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | - Bahare Farhadian
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States.,Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | - Melissa Silverman
- Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States.,Division of Child and Adolescent Psychiatry and Child Development, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Paula Tran
- Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States.,Division of Child and Adolescent Psychiatry and Child Development, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Safwan Jaradeh
- Autonomic Disorders Program, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Margo Thienemann
- Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States.,Division of Child and Adolescent Psychiatry and Child Development, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Jennifer Frankovich
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States.,Stanford PANS/Immune Behavioral Health Clinic and PANS Research Program at Lucile Packard Children's Hospital, Palo Alto, CA, United States
| |
Collapse
|
45
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
46
|
Jung H, Lee H, Kim D, Cheong E, Hyun YM, Yu JW, Um JW. Differential Regional Vulnerability of the Brain to Mild Neuroinflammation Induced by Systemic LPS Treatment in Mice. J Inflamm Res 2022; 15:3053-3063. [PMID: 35645573 PMCID: PMC9140139 DOI: 10.2147/jir.s362006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Hyeji Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Hyojeong Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Dongwook Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Young-Min Hyun
- Department of Anatomy and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Correspondence: Ji Won Um, Email
| |
Collapse
|
47
|
Gradek-Kwinta E, Slowik A, Dziedzic T. The use of anticholinergic medication is associated with an increased risk of stroke-associated pneumonia. Aging Clin Exp Res 2022; 34:1935-1938. [PMID: 35416612 DOI: 10.1007/s40520-022-02123-x] [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: 02/02/2022] [Accepted: 03/20/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Pneumonia is a frequent medical complication after stroke. A few studies showed that the use of anticholinergic medication is associated with a higher risk of community acquired pneumonia in the elderly. We aimed to determine if there is any association between anticholinergic medication used before stroke and stroke-associated pneumonia (SAP). METHODS We analysed prospectively collected data of 675 patients with acute stroke (mean age 71.4 ± 13.3; 53.1% female). We used the Anticholinergic Drug Scale to assess anticholinergic exposure during a month preceding stroke onset. RESULTS We diagnosed SAP in 14.7% of patients. The use of anticholinergic medication was associated with an elevated risk of SAP (OR 2.56, 95% CI 1.59-4.11, P < 0.01) in univariate analysis. This association remained significant in multivariable analysis adjusted for age, stroke severity, atrial fibrillation, previous myocardial infarction and respiratory tract diseases (OR 2.06, 95% CI 1.01-4.22, P = 0.04). CONCLUSIONS The use of anticholinergic medication before stroke is associated with an increased risk of SAP.
Collapse
Affiliation(s)
- Elżbieta Gradek-Kwinta
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503, Kraków, Poland
| | - Agnieszka Slowik
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503, Kraków, Poland
| | - Tomasz Dziedzic
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503, Kraków, Poland.
| |
Collapse
|
48
|
Yang H, George SJ, Thompson D, Silverman HA, Tsaava T, Tynan A, Pavlov VA, Chang E, Andersson U, Brines M, Chavan SS, Tracey KJ. Famotidine activates the vagus nerve inflammatory reflex to attenuate cytokine storm. RESEARCH SQUARE 2022:rs.3.rs-1493296. [PMID: 35441176 PMCID: PMC9016653 DOI: 10.21203/rs.3.rs-1493296/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background. Severe COVID-19 is characterized by pro-inflammatory cytokine release syndrome (cytokine storm) which causes high morbidity and mortality. Recent observational and clinical studies suggest famotidine, a histamine 2 receptor (H2R) antagonist widely used to treat gastroesophageal reflux disease , attenuates the clinical course of COVID-19. Because evidence is lacking for a direct antiviral activity of famotidine, a proposed mechanism of action is blocking the effects of histamine released by mast cells. Here we hypothesized that famotidine activates the inflammatory reflex, a brain-integrated vagus nerve mechanism which inhibits inflammation via alpha 7 nicotinic acetylcholine receptor ( α7nAChR ) signal transduction, to prevent cytokine storm. Methods. The potential anti-inflammatory effects of famotidine and other H2R antagonists was assessed in mice exposed to lipopolysaccharide (LPS)-induced cytokine storm. As the inflammatory reflex is integrated and can be stimulated in the brain, and H2R antagonists penetrate the blood brain barrier poorly, famotidine was administered by intracerebroventricular (ICV) or intraperitoneal (IP) routes. Results. Famotidine administered IP significantly reduced serum and splenic LPS-stimulated tumor necrosis factor α and interleukin-6 concentrations, significantly improving survival. The effects of ICV famotidine were significantly more potent as compared to the peripheral route. Mice lacking mast cells by genetic deletion also responded to famotidine, indicating the anti-inflammatory effects are not mast cell dependent. Either bilateral sub-diaphragmatic vagotomy or genetic knock-out of α7nAChR abolished the anti-inflammatory effects of famotidine, indicating the inflammatory reflex as famotidine's mechanism of action. While the structurally similar H2R antagonist tiotidine displayed equivalent anti-inflammatory activity, the H2R antagonists cimetidine or ranitidine were ineffective even at very high dosages. Conclusions. These observations reveal a previously unidentified vagus nerve-dependent anti-inflammatory effect of famotidine in the setting of cytokine storm which is not replicated by high dosages of other H2R antagonists in clinical use. Because famotidine is more potent when administered intrathecally, these findings are also consistent with a primarily central nervous system mechanism of action.
Collapse
Affiliation(s)
- Huan Yang
- Feinstein Institute for Medical Research
| | - Sam J George
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Dane Thompson
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Harold A Silverman
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Tea Tsaava
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Aisling Tynan
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Valentin A Pavlov
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Eric Chang
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | | | - Michael Brines
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Sangeeta S Chavan
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| | - Kevin J Tracey
- Feinstein Institute for Medical Research Fertility Research Laboratory: Northwell Health Feinstein Institutes for Medical Research
| |
Collapse
|
49
|
Burzynski H, Macht V, Woodruff J, Crawford J, Erichsen J, Piroli G, Grillo C, Fadel J, Reagan L. Pyridostigmine bromide elicits progressive and chronic impairments in the cholinergic anti-inflammatory pathway in the prefrontal cortex and hippocampus of male rats. Neurobiol Stress 2022; 18:100446. [PMID: 35573808 PMCID: PMC9095881 DOI: 10.1016/j.ynstr.2022.100446] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/18/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Gulf War Illness (GWI) is a multi-symptom illness that continues to affect over 250,000 American Gulf War veterans. The causes of GWI remain equivocal; however, prophylactic use of the acetylcholinesterase inhibitor pyridostigmine bromide (PB), and the stress of combat have been identified as two potential causative factors. Both PB and stress alter acetylcholine (ACh), which mediates both cognition and anti-inflammatory responses. As inflammation has been proposed to contribute to the cognitive deficits and immune dysregulation in GWI, the goal of this study was to determine the long-term effects of PB and stress on the cholinergic anti-inflammatory pathway in the central nervous system and periphery. We used our previously established rat model of GWI and in vivo microdialysis to assess cholinergic neurochemistry in the prefrontal cortex (PFC) and hippocampus following a mild immune challenge (lipopolysaccharide; LPS). We then examined LPS-induced changes in inflammatory markers in PFC and hippocampal homogenates. We found that PB treatment produces a long-lasting potentiation of the cholinergic response to LPS in both the PFC and hippocampus. Interestingly, this prolonged effect of PB treatment enhancing cholinergic responses to LPS was accompanied by paradoxical increases in the release of pro-inflammatory cytokines in these brain regions. Collectively, these findings provide evidence that neuroinflammation resulting from dysregulation of the cholinergic anti-inflammatory pathway is a mechanistic mediator in the progression of the neurochemical and neurocognitive deficits in GWI and more broadly suggest that dysregulation of this pathway may contribute to neuroinflammatory processes in stress-related neurological disorders. Inflammation is thought to contribute to the progressive nature of GWI pathology. PB potentiates the central cholinergic response to LPS over time in model of GWI. PB progressively exacerbates the neuroinflammatory response to LPS. GWI may result from the dysregulation of the cholinergic anti-inflammatory pathway.
Collapse
|
50
|
Deng L, Wang S, Guo H, Liu X, Zou X, Zhang R, Lin Y, Tan W. (RS)-bambuterol and its enantiomers: Potential improvement of (R)-bambuterol in mice with colitis. Int Immunopharmacol 2022; 103:108501. [PMID: 34974400 DOI: 10.1016/j.intimp.2021.108501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022]
Abstract
Bambuterol (BMB) has been used clinically to treat asthma due to its bronchodilation activity. However, the effect of BMB on ulcerative colitis (UC) has not been examined. The present work focused on the effects of enantiomeric BMB on UC. Acute UC was induced in mice by 3% dextran sulfate sodium (DSS), and (R)-, (S) and (RS)-BMB were orally administered. Body weight loss and the disease activity index (DAI) were measured once a day. Inflammatory factors were detected by ELISA and qRT-PCR. Histological evaluations of colon samples were performed. IL-6, STAT3, and RORγt pathway-related proteins were analyzed by western blotting. The results verified that colitis severity was dramatically ameliorated by (R)-BMB, which was significantlybetter than the effect of (RS)-BMB or (S)-BMB, as evidenced by body weight loss, DAI, colon length, spleen/body weight ratio and histopathological manifestations. Furthermore, (R)-BMB treatment significantly diminished the levels of inflammatory cytokines and macrophages infiltration in mice with colitis. Besides, treated with (R)-BMB obviously elevated the level of β2AR. In addition, (R)-BMB decreased the expression of IL-6, IL-17, retinoic acid receptor-related orphan receptor-gamma t (RORt), and phosphorylated STAT3 (p-STAT3) in a dose-dependent manner in the colon tissues. The efficacy of (R)-BMB was more notable than aminosalicylic acid (5-ASA). (R)-BMB is either butyrilcholinesterase inhibitor or β2AR agonist which offers new treatment of colitis.
Collapse
Affiliation(s)
- Liangjun Deng
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Shanping Wang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Haihua Guo
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Xiaoming Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinfeng Zou
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Rui Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Yue Lin
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Wen Tan
- Post-Doctoral Innovation Site, Jinan University Affiliation, Yuanzhi Health Technology Co, Ltd, Hengqin New District, Zhuhai, Guangdong 519000, China; Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| |
Collapse
|