1
|
Baker MC, Nagy D, Tamang S, Horváth-Puhó E, Sørensen HT. Vagotomy and the incidence of rheumatoid arthritis and osteoarthritis: a Danish register-based study. Arthritis Res Ther 2025; 27:106. [PMID: 40380285 DOI: 10.1186/s13075-025-03567-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
OBJECTIVES Given the potential role of vagus nerve stimulation in treating rheumatoid arthritis (RA), we examined the incidence of RA and osteoarthritis (OA) in patients who underwent different forms of vagotomy that disparately affect the inflammatory reflex. METHODS Using nationwide health registries, we constructed cohorts of patients in Denmark who underwent truncal or superselective vagotomy between 1977 and 1995 and comparison members from the general population matched 10:1 on birth year, sex, and calendar year. We identified incident RA or OA and used Cox proportional hazards models to compute adjusted hazard ratios (aHRs) and corresponding 95% CI. RESULTS Our cohorts consisted of 2,260 truncal vagotomy patients matched with 22,610 comparators, and 3,810 superselective vagotomy patients matched with 38,090 comparators. The incidence rate (IR) of RA per 1,000 person-years (95% CI) in the truncal vagotomy cohort was 10.2 (6.5-15.3) versus 7.2 (6.1-8.4) in the matched comparison cohort. The aHR (95% CI) for RA development was 2.62 (1.47-4.67) in the truncal vagotomy cohort and 1.05 (0.51-2.17) in the superselective vagotomy cohort, with respect to comparison cohorts. The risk of developing OA was not significantly different for either vagotomy cohort compared with comparison cohorts. CONCLUSION Truncal vagotomy was associated with an increased incidence of RA; this association was not observed with superselective vagotomy. No association with either form of vagotomy was seen with OA. These findings support the hypothesis that disruption of vagus nerve signaling impacts the inflammatory reflex and contributes to the development of RA.
Collapse
Affiliation(s)
- Matthew C Baker
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, (MCB, ST), Stanford, CA, USA.
| | - Dávid Nagy
- Department of Clinical Epidemiology, Department of Clinical Medicine and Center for Population Medicine, Aarhus University Hospital, Aarhus University, (DN, EHP, HTS), Aarhus, Denmark
| | - Suzanne Tamang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, (MCB, ST), Stanford, CA, USA
| | - Erzsébet Horváth-Puhó
- Department of Clinical Epidemiology, Department of Clinical Medicine and Center for Population Medicine, Aarhus University Hospital, Aarhus University, (DN, EHP, HTS), Aarhus, Denmark
| | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Department of Clinical Medicine and Center for Population Medicine, Aarhus University Hospital, Aarhus University, (DN, EHP, HTS), Aarhus, Denmark
- Clinical Excellence Science Center, Stanford University, (HTS), Stanford, CA, USA
| |
Collapse
|
2
|
Liu C, Liao Y, Jiang H, Tang Q, He C, Wang Y, Ren M, Wang C, Chen S, Tan L, Wan X, Chen D. Theabrownin: The 'rich hue' of Chinese dark tea, its extraction, and role in regulating inflammation and immune response. Food Res Int 2025; 209:116185. [PMID: 40253125 DOI: 10.1016/j.foodres.2025.116185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Theabrownin (TB) is one of the most representative bioactive components in Chinese dark tea, often referred to as the "gold in dark tea." The complex macromolecular structure of TB is influenced by its source (tea materials), extraction, separation, and purification methods, which affect its final structure and bioactivity. In recent years, research on TB has surged, becoming a hotspot in the field of tea functional components and health research. Extensive studies on its health benefits indicate that TB is a crucial active ingredient in dark tea with substantial potential for application in food, health care, industry, and medical fields. This review summarizes the formation of TB during dark tea manufacturing, especially the "piling" stage, extraction methods, various purification techniques, and the physicochemical properties of TB. Additionally, it comprehensively reviews recent research on TB's role in typical inflammation and immune imbalance-induced diseases such as colitis, atherosclerosis, non-alcoholic fatty liver disease, and innate immune diseases. The review concludes with a comparative summary of the biological activities of TB from the five major types of Chinese dark tea in terms of anti-inflammatory and immune regulatory effects.
Collapse
Affiliation(s)
- Chen Liu
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China; Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Yihong Liao
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Hanrui Jiang
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Qian Tang
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Chunlei He
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Yijun Wang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, PR China
| | - Mengyi Ren
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Chenbo Wang
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Shengxiang Chen
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Liqiang Tan
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, PR China.
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan, PR China.
| |
Collapse
|
3
|
Liu S, Zheng Y, Chen H, Li X, Yan Q, Mu W, Fu Y, Chen H, Hou H, Liu L, Tian C. Structural basis for allosteric agonism of human α7 nicotinic acetylcholine receptors. Cell Discov 2025; 11:35. [PMID: 40195322 PMCID: PMC11977206 DOI: 10.1038/s41421-025-00788-y] [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: 11/02/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
The α7 nicotinic acetylcholine receptor (nAChR), a pentameric ligand-gated ion channel, plays important roles in cognition, neuroprotection, and anti-inflammation. As a potential drug target, α7 nAChR has different binding sites for different ligands, particularly agonists and positive allosteric modulators (PAMs). Ago-PAMs can both directly activate and allosterically modulate α7 nAChR. However, the mechanism underlying α7 nAChR modulation by ago-PAM has yet to be fully elucidated. Here, we present cryo-EM structures of α7 nAChR in complex with the ago-PAM GAT107 and Ca2+ in the open and desensitized states, respectively. Our results from both structural comparisons and functional assays suggest an allosteric mechanism underlying GAT107 modulation and calcium potentiation of α7 nAChR, involving local conformational changes in the ECD-TMD coupling region and a global structural rearrangement in the transmembrane domain. This work provides a new mechanism of α7 nAChR gating distinct from that of conventional agonist binding. These findings would aid in drug design and enrich our biophysical understanding of pentameric ligand-gated ion channels.
Collapse
Affiliation(s)
- Sanling Liu
- Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, University of Science and Technology of China, Hefei, Anhui, China.
- Beijing Life Science Academy, Beijing, China.
| | - Yining Zheng
- Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, University of Science and Technology of China, Hefei, Anhui, China
| | - Haopeng Chen
- Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, University of Science and Technology of China, Hefei, Anhui, China
| | - Xin Li
- Beijing Life Science Academy, Beijing, China
| | - Qipeng Yan
- Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenjun Mu
- Beijing Life Science Academy, Beijing, China
| | - Yaning Fu
- Beijing Life Science Academy, Beijing, China
| | - Huan Chen
- Beijing Life Science Academy, Beijing, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing, China.
| | - Lei Liu
- Department of Chemistry, Tsinghua University, Beijing, China.
| | - Changlin Tian
- Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, University of Science and Technology of China, Hefei, Anhui, China.
- Beijing Life Science Academy, Beijing, China.
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China.
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
de Souza AA, Goularte KCM, Piccoli RC, Custódio SV, de Mello JE, Victor MG, Domingues WB, de Souza LP, Dos Santos Gonçalves L, Campos VF, Cunico W, Oses JP, Stefanello FM, de Aguiar MSS, Spanevello RM. 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one Attenuates Scopolamine-induced Cognitive Impairment in Rats: Insights Into Neuroprotective Effects. Mol Neurobiol 2025:10.1007/s12035-025-04887-5. [PMID: 40164887 DOI: 10.1007/s12035-025-04887-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's Disease (AD) is characterized by memory decline, dysregulation in cholinergic and purinergic signaling, neuroinflammation, and oxidative stress. Current treatments are limited, highlighting the need for new compounds to prevent or delay AD progression. Thiazolidinones have emerged as promising candidates due to their antioxidant, anti-inflammatory, and anticholinesterase properties. The aim of this study was to evaluate the effects of 3-(3-(diethylamino)propyl)-2-(4-(methylthio)phenyl)thiazolidin-4-one (DS27) in a rat model of scopolamine-induced memory deficits. Male rats were divided into groups: I - Control, II - Scopolamine (SCO) (1 mg/kg), III - SCO and DS27 (5 mg/kg), IV - SCO and DS27 (10 mg/kg), V - SCO and donepezil (5 mg/kg). The animals were treated orally with DS27 or donepezil for seven days. On the 8th day, they underwent the open field test and inhibitory avoidance training, followed by intraperitoneal administration SCO. Twenty-four hours later, an inhibitory avoidance test was conducted. Acetylcholinesterase (AChE) activity, oxidative stress, and inflammatory and purinergic parameters were analyzed in the cerebral cortex, hippocampus, cerebrospinal fluid, serum, lymphocytes, and liver. DS27 prevented memory deficits, alterations in AChE activity, and oxidative damage induced by SCO in brain structures. Additionally, DS27 prevented SCO-induced decrease in IL-10 levels, and increase in IL-6, and TNF-α expression in the cerebral cortex, and normalized ATP and ADP hydrolysis in cerebrospinal fluid and lymphocytes. DS27 did not induce oxidative stress in the liver or alter serum biochemical parameters. These findings suggest that DS27 has significant neuroprotective properties and could be a promising alternative for treating neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Anita Avila de Souza
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Kelen Cristiane Machado Goularte
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Raphaela Cassol Piccoli
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Solange Vega Custódio
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Julia Eisenhardt de Mello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Melinda Gomes Victor
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - William Borges Domingues
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Lucas Petitemberte de Souza
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Laís Dos Santos Gonçalves
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Vinicius Farias Campos
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
- Graduate Program in Biotechnology - Laboratory of Structural Genomics - Technological Development Center, Federal University of Pelotas, Capão Do Leão Campus, S/N, Pelotas-RS, RS, CEP 96010‑900, Brazil
| | - Wilson Cunico
- Graduate Program in Biochemistry and Bioprospecting - Laboratory of Chemistry Applied to Bioactive - Center the Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Jean Pierre Oses
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Francieli Moro Stefanello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Department of Clinical Medicine, Faculty of Medicine - Laboratory of Neuroscience and Behavior - Drug Research and Development Center, Federal University of Ceará, Fortaleza-CE, CEP 60020-181, Brazil
| | - Roselia Maria Spanevello
- Graduate Program in Biochemistry and Bioprospection - Laboratory of Neurochemistry, Inflammation, and Cancer - Center the Chemical, Pharmaceutical, and Food Sciences, Federal University of Pelotas, Capão Do Leão Campus S/N, Pelotas-RS, CEP 96010‑900, Brazil.
| |
Collapse
|
5
|
Mac CH, Nguyen GLT, Nguyen DTM, Huang SM, Peng HH, Chang Y, Lo SK, Chiang HHK, Yang YZ, Song HL, Chia WT, Lin YJ, Sung HW. Noninvasive Vagus Nerve Electrical Stimulation for Immune Modulation in Sepsis Therapy. J Am Chem Soc 2025; 147:8406-8421. [PMID: 40033812 PMCID: PMC11912339 DOI: 10.1021/jacs.4c16367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
Sepsis presents a significant medical challenge due to its intense inflammatory response to infection, often resulting in high mortality rates. A promising therapeutic strategy targets the cholinergic anti-inflammatory pathway (CAIP), which regulates immune responses. This study investigates the ingestion of piezoelectric particles that adhere to the stomach lining, specifically targeting TRPV1 receptors. In a mouse model of sepsis, these particles, when activated by low-intensity pulsed ultrasound, generate mild electrical pulses. These pulses stimulate vagal afferent fibers, transmitting signals to the brain and modulating the neural-immune network via the CAIP. Consequently, this leads to a reduction in systemic inflammation, mitigating weight loss, alleviating multiple tissue injuries, and preventing death by modulating immune cells in the spleen. This approach addresses the critical need for noninvasive sepsis therapies, potentially improving patient outcomes. Utilizing portable ultrasound equipment with minimal thermal effects, this technique offers a safe and convenient treatment option, even for home use.
Collapse
Affiliation(s)
- Cam-Hoa Mac
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Giang Le Thi Nguyen
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Dien Thi My Nguyen
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Sheng-Min Huang
- Department
of Pharmacology, College of Medicine, National
Cheng Kung University, Tainan 701401, Taiwan
| | - Hsu-Hsia Peng
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Yen Chang
- Taipei
Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and School of
Medicine, Tzu Chi University, Hualien 970473, Taiwan
| | - Shih-Kai Lo
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Hui-Hua Kenny Chiang
- Institute
of Biomedical Engineering, National Yang-Ming
Chiao Tung University, Taipei 112304, Taiwan
| | - Yuan-Zhen Yang
- Institute
of Biomedical Engineering, National Yang-Ming
Chiao Tung University, Taipei 112304, Taiwan
| | - Hsiang-Lin Song
- Department
of Pathology, National Taiwan University
Hospital, Hsinchu Branch, Hsinchu 302058, Taiwan
| | - Wei-Tso Chia
- Department
of Orthopedics, National Taiwan University
Hospital, Hsinchu Branch, Hsinchu 302058, Taiwan
| | - Yu-Jung Lin
- Research
Center for Applied Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Hsing-Wen Sung
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
6
|
Lenka A, Vernino S. Carbidopa: beyond Parkinson's disease. Clin Auton Res 2025:10.1007/s10286-025-01122-y. [PMID: 40056295 DOI: 10.1007/s10286-025-01122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
PURPOSE To revisit the pharmacology and real-world use of carbidopa in the management of autonomic disorders. METHODS To identify articles suitable for this review, a search of the PubMed database was conducted in January 2025 using the keywords "Carbidopa," "MK-486," and "L-alpha-methyldopa hydrazine." RESULTS The pharmacotherapeutic role of carbidopa extends beyond the management of Parkinson's disease. Our literature search revealed the use of carbidopa in three primary autonomic diseases to treat either nausea or symptoms of sympathetic hyperactivity: (1) familial dysautonomia, (2) hyperadrenergic postural orthostatic tachycardia syndrome (POTS), and (3) afferent baroreflex failure (familial or acquired). Even at a dose as high as 600 mg/day, carbidopa was not associated with bothersome side effects in some of the clinical trials on familial dysautonomia. Pre-clinical evidence also suggests in vitro and in vivo inhibition of T-cell activation by carbidopa and a potential therapeutic use in cytokine release syndrome. CONCLUSION Current evidence, although limited, suggests that carbidopa has a favorable safety profile. While large, well-designed studies are warranted, observations from case series and small studies suggest that carbidopa could have utility in treating nausea in familial dysautonomia and symptoms of sympathetic hyperactivity in hyperadrenergic postural orthostatic tachycardia syndrome (POTS) and afferent baroreflex failure.
Collapse
Affiliation(s)
- Abhishek Lenka
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Steven Vernino
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
7
|
Wu S, Xie Y, Jiang Y, Zhang X, Zhou Y, Zuo X, Li T. GTS-21 modulates rheumatoid arthritis Th17 and Th2 lymphocyte subset differentiation through the ɑ7nAch receptor. Clin Rheumatol 2025; 44:989-998. [PMID: 39812970 DOI: 10.1007/s10067-025-07320-3] [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: 05/17/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Previous research has demonstrated ɑ7nAch receptor (ɑ7nAchR) agonists to provide benefit for rheumatoid arthritis (RA) patients. However, the immunological mechanism of action for these ɑ7nAchR agonists has not been elucidated. Herein, the effect of GTS-21, a selective ɑ7nAchR agonist, on the differentiation of Th17 and Th2 cells was assessed. CD4 + T cells were obtained from the peripheral blood mononuclear cells (PBMCs) of RA patients and healthy donors. CD4 + T cells were separately differentiated into Th2 or Th17 cells with or without GTS-21 and with or without alpha-bungarotoxin (ɑBgt) (a ɑ7nAchR antagonist). The proportions of Th17 and Th2 cells were assessed by flow cytometry. Levels of the T cell cytokines, IL-17A and IL-4, were assessed by ELISA. Specific transcription factors, retinoic orphan receptor c (RORc), and GATA Binding Protein 3 (GATA-3) were detected by western blot. GTS-21 reduced IL-17A and increased IL-4 production by RA PBMCs. GTS-21 reduced the percentage of Th17 cells and increased the percentage of Th2 cells during Th17 and Th2 differentiation, respectively. GTS-21 downregulated RA CD4 + T cells RORc levels and reduced the secretion of IL-17A during Th17 differentiation. GTS-21 upregulated RA CD4 + T cells GATA3 and promoted IL-4 production during Th2 differentiation. ɑ-Bgt blocked the effects of GTS-21 during Th17 and Th2 differentiation. These results demonstrated that GTS-21 suppressed RA Th17 differentiation and promoted Th2 differentiation. As such, the use of GTS-21 may be a new therapeutic approach by which to treat RA patients. Key Points • GTS-21 suppressed RA Th17 differentiation and promoted Th2 differentiation via acting on ɑ7nAchR. • The protective effect of GTS-21 on RA may be related to its regulation of Th cell subsets.
Collapse
Affiliation(s)
- Shiyao Wu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanli Xie
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Jiang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoli Zhang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaou Zhou
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zuo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tong Li
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Xuan X, Huang Z, Kong Z, Li R, Li J, Huang H. GENETIC INSIGHTS INTO SEPSIS: MENDELIAN RANDOMIZATION ANALYSIS OF CEREBROSPINAL FLUID METABOLITES. Shock 2025; 63:379-384. [PMID: 39454631 DOI: 10.1097/shk.0000000000002494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
ABSTRACT Background: Sepsis, a life-threatening response to infection leading to systemic inflammation and organ dysfunction, has been hypothesized to be influenced by metabolic alterations in cerebrospinal fluid (CSF). Despite extensive research, the specific metabolic pathways contributing to sepsis remain unclear. This study aims to elucidate the causal relationships between CSF metabolites and sepsis risk using Mendelian randomization (MR), offering insights that could lead to novel therapeutic strategies. Methods: We conducted a two-sample MR analysis using genetic variants as instrumental variables (IVs) to investigate 338 CSF metabolites identified through a genome-wide association study. Data on sepsis-related outcomes were extracted from the genome-wide association study catalog encompassing 486,484 individuals of European descent. IVs were rigorously selected based on stringent genetic association and linkage disequilibrium criteria. Statistical analyses, including inverse variance weighting (IVW) and weighted median methods, were performed using the "TwoSampleMR" package in R software, supplemented by comprehensive sensitivity analyses to ensure the robustness of our findings. Results: Our analysis identified 19 CSF metabolites causally associated with sepsis risk. Notably, metabolites such as 1-palmitoyl-2-stearoyl-gpc (16:0/18:0) and 2-hydroxyglutarate showed significant negative correlations with sepsis risk. The reverse MR analysis further revealed that sepsis could negatively impact certain CSF metabolite levels, particularly ribonate, suggesting a bidirectional relationship. These relationships were substantiated by rigorous statistical testing and sensitivity analyses confirming the absence of horizontal pleiotropy and the stability of our results across various MR methods. Conclusions: This study demonstrates significant causal associations between specific CSF metabolites and the risk of developing sepsis, highlighting the potential for these metabolites to serve as biomarkers or therapeutic targets. The bidirectional nature of these findings also suggests that sepsis itself may alter metabolic profiles, offering further avenues for intervention.
Collapse
Affiliation(s)
- Xin Xuan
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhihao Huang
- Department of Otorhinolaryngology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhiqian Kong
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Ruoyu Li
- Department of Intensive Care Unit, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Jianfeng Li
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Haiyan Huang
- Department of Internal Medicine of Chinese Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| |
Collapse
|
9
|
Zoccali C, Mallamaci F, Kanbay M, Dounousi E, Kotanko P, Jankowski J, Chertow GM. Autonomic Dysfunction and Inflammation in CKD: A High-Risk Pathway. J Am Soc Nephrol 2025; 36:529-532. [PMID: 39621839 PMCID: PMC11888946 DOI: 10.1681/asn.0000000591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 11/26/2024] [Indexed: 01/15/2025] Open
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, New York
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), Reggio Calabria, Italy
| | - Francesca Mallamaci
- Divisione di Nefrologia e Trapianto Renale, Grande Ospedale Metropolitano, Reggio Calabria, Italy
- CNR-IFC, Institute of Clinical Physiology, Research Unit of Clinical Epidemiology, Reggio Calabria, Italy
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Peter Kotanko
- Renal Research Institute, New York, New York
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Glenn M. Chertow
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
10
|
Wong ML, Widerström-Noga E, Bolanos JL, Gonzalez G, Penedo FJ, Hosein PJ, Tovin MM, Gonzalez JP, McTeague LM. Feasibility of trancutaneous auricular vagus nerve stimulation in Black and Hispanic/Latino people with peripheral neuropathy. FRONTIERS IN PAIN RESEARCH 2025; 5:1516196. [PMID: 39896735 PMCID: PMC11782131 DOI: 10.3389/fpain.2024.1516196] [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: 10/24/2024] [Accepted: 12/31/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction Peripheral neuropathy (PN) is the most common neurodegenerative disorder, and the primary causes are chemotherapy-induced peripheral neuropathy (CIPN) and diabetic neuropathy (DN). Transcutaneous auricular vagus nerve stimulation (taVNS) is a promising non-pharmacological and non-invasive intervention that targets key pathways involved with PN. However, research is needed to determine the feasibility, acceptability, and effects of taVNS in people with PN. It is also critical that this research on taVNS include the perspectives of Black and Hispanic/Latino patients, who are often underrepresented in research. Methods This research was comprised of two consecutive studies: a survey and a pilot randomized sham-controlled trial (RCT). The survey assessed symptom burden, management strategies, and interest in taVNS among CIPN patients. The pilot RCT evaluated the feasibility, acceptability, and preliminary effects of taVNS in Black and Hispanic/Latino patients with CIPN or diabetic neuropathy. Participants were recruited from the University of Miami medical system, with culturally sensitive approaches to enhance minority participation. Results The survey included 62 respondents, 78% Black or Hispanic/Latino, revealing high symptom burden and significant interest in taVNS (82% expressed moderate to high interest). The pilot RCT enrolled 28 participants, achieving a 42% recruitment rate and 86% retention. taVNS was well tolerated, with no significant adverse effects. Preliminary data indicated a decrease in neuropathic symptoms and an increased heart rate variability (HRV) during active taVNS, suggesting autonomic modulation. Tingling sensation and pain decreased by median values of 2.0 and 1.5, respectively. Additionally, the median values for standard deviation of the RR interval increased from 34.9 (CI = 21.6-44.8) at baseline to 44.8 (CI = 26.5-50.3) during intervention. Exit interviews highlighted positive participant experiences and identified potential barriers, such as protocol length and distrust in medical research. Conclusion The findings underscore the need for novel CIPN treatments and demonstrate the feasibility of conducting taVNS research in historically underrepresented populations. High interest in taVNS and successful recruitment and retention rates suggest that culturally sensitive approaches can enhance minority participation in clinical trials. These findings will be used to develop a large clinical trial to determine the efficacy of repeated taVNS in a diverse cohort. Clinical Trial Registration https://clinicaltrials.gov, identifier (NCT05896202).
Collapse
Affiliation(s)
- Marlon L. Wong
- Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Eva Widerström-Noga
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
| | - Jessica L. Bolanos
- Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Gabriel Gonzalez
- Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Frank J. Penedo
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Peter J. Hosein
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Melissa M. Tovin
- Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Juan P. Gonzalez
- Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lisa M. McTeague
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
11
|
Gargus M, Ben-Azu B, Landwehr A, Dunn J, Errico JP, Tremblay MÈ. Mechanisms of vagus nerve stimulation for the treatment of neurodevelopmental disorders: a focus on microglia and neuroinflammation. Front Neurosci 2025; 18:1527842. [PMID: 39881804 PMCID: PMC11774973 DOI: 10.3389/fnins.2024.1527842] [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: 11/13/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
The vagus nerve (VN) is the primary parasympathetic nerve, providing two-way communication between the body and brain through a network of afferent and efferent fibers. Evidence suggests that altered VN signaling is linked to changes in the neuroimmune system, including microglia. Dysfunction of microglia, the resident innate immune cells of the brain, is associated with various neurodevelopmental disorders, including schizophrenia, attention deficit hyperactive disorder (ADHD), autism spectrum disorder (ASD), and epilepsy. While the mechanistic understanding linking the VN, microglia, and neurodevelopmental disorders remains incomplete, vagus nerve stimulation (VNS) may provide a better understanding of the VN's mechanisms and act as a possible treatment modality. In this review we examine the VN's important role in modulating the immune system through the inflammatory reflex, which involves the cholinergic anti-inflammatory pathway, which releases acetylcholine. Within the central nervous system (CNS), the direct release of acetylcholine can also be triggered by VNS. Homeostatic balance in the CNS is notably maintained by microglia. Microglia facilitate neurogenesis, oligodendrogenesis, and astrogenesis, and promote neuronal survival via trophic factor release. These cells also monitor the CNS microenvironment through a complex sensome, including groups of receptors and proteins enabling microglia to modify neuroimmune health and CNS neurochemistry. Given the limitations of pharmacological interventions for the treatment of neurodevelopmental disorders, this review seeks to explore the application of VNS as an intervention for neurodevelopmental conditions. Accordingly, we review the established mechanisms of VNS action, e.g., modulation of microglia and various neurotransmitter pathways, as well as emerging preclinical and clinical evidence supporting VNS's impact on symptoms associated with neurodevelopmental disorders, such as those related to CNS inflammation induced by infections. We also discuss the potential of adapting non-invasive VNS for the prevention and treatment of these conditions. Overall, this review is intended to increase the understanding of VN's potential for alleviating microglial dysfunction involved in schizophrenia, ADHD, ASD, and epilepsy. Additionally, we aim to reveal new concepts in the field of CNS inflammation and microglia, which could serve to understand the mechanisms of VNS in the development of new therapies for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Antonia Landwehr
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jaclyn Dunn
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Yelisyeyeva O, Kaminskyy D, Semen M, Chelpanova I, Semen KO. Redox Metabolism and Autonomic Regulation During Aging: Can Heart Rate Variability Be Used to Monitor Healthy Longevity? Biomedicines 2025; 13:161. [PMID: 39857745 PMCID: PMC11761282 DOI: 10.3390/biomedicines13010161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The functionality of redox metabolism is frequently named as an important contributor to the processes of aging and anti-aging. Excessive activation of free radical reactions accompanied by the inability of the antioxidant defense (AOD) mechanisms to control the flow of the reactive oxygen species (ROS) leads to the persistence of oxidative stress, hypoxia, impaired mitochondrial energy function and reduced ATP potential. From a long-term perspective, such changes contribute to the development of chronic diseases and facilitate aging. In turn, preconditioning of a biosystem with small doses of stressful stimuli might cause mobilization of the mechanisms of AOD and control an excessive flow of ROS, which supports optimal functioning of the redox reactions. Those mechanisms are of crucial importance for anti-aging and are also known as a eustress or hormetic response. To ensure continuous support of mild pro-oxidant activity in a metabolic system, close monitoring and timely corrections preventing the development of excessive ROS production are required. The paper introduces the potential of heart rate variability (HRV) as a biomarker of functional and metabolic reserves and a tool to measure stress resilience during aging. The practical approaches to interpretation of HRV are provided based on total power, changes in total power in response to an orthostatic test and activities of all spectral components. It is suggested that the complex of those parameters can reflect the depth of oxidative stress and may be used to guide lifestyle interventions and promote active longevity.
Collapse
Affiliation(s)
- Olha Yelisyeyeva
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (O.Y.); (I.C.)
| | - Danylo Kaminskyy
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine;
| | - Marta Semen
- Department of Propaedeutics of Pediatrics and Medical Genetics, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine;
| | - Ilona Chelpanova
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (O.Y.); (I.C.)
| | | |
Collapse
|
13
|
Heusch G, Kleinbongard P. The spleen in ischaemic heart disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01114-x. [PMID: 39743566 DOI: 10.1038/s41569-024-01114-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/04/2025]
Abstract
Ischaemic heart disease is a consequence of coronary atherosclerosis, and atherosclerosis is a systemic inflammatory disease. The spleen releases various immune cells in temporally distinct patterns. Neutrophils, monocytes, macrophages, B cells and T cells execute innate and adaptive immune processes in the coronary atherosclerotic plaque and in the ischaemic myocardium. Prolonged inflammation contributes to ischaemic heart failure. The spleen is also a target of neuromodulation through vagal, sympathetic and sensory nerve activation. Efferent vagal activation and subsequent activation of the noradrenergic splenic nerve activate β2-adrenergic receptors on splenic T cells, which release acetylcholine that ultimately results in attenuation of cytokine secretion from splenic macrophages. Coeliac vagal nerve activation increases splenic sympathetic nerve activity and drives the release of T cells, a process that depends on placental growth factor. Activation of the vagosplenic axis protects acutely from ischaemia-reperfusion injury during auricular tragus vagal stimulation and remote ischaemic conditioning. Splenectomy abrogates all these deleterious and beneficial actions on the cardiovascular system. The aggregate effect of splenectomy in humans is a long-term increase in mortality from ischaemic heart disease. The spleen has been appreciated as an important immune organ for inflammatory processes in atherosclerosis, myocardial infarction and heart failure, whereas its complex interaction with circulating blood factors and with the autonomic and somatic nervous systems, as well as its role in cardioprotection, have emerged only in the past decade. In this Review, we describe this newly identified cardioprotective function of the spleen and highlight the potential for translating the findings to patients with ischaemic heart disease.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Xingjun F, Ruijiao Z, Peihua Y, Shiyin W, Liqin C, Liangchao Q, Qinghua P. Left T7 paravertebral nerve blockade activate the α7nAChR-Dependent CAP in patients undergoing thoracoscopic lobectomy: a prospective controlled study. BMC Anesthesiol 2024; 24:475. [PMID: 39722047 DOI: 10.1186/s12871-024-02857-3] [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: 08/05/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVE This study aimed to observe the impact of Tthoracic paravertebral nerve blockade(TPVB) at left T7 level on the α7nAChR-dependent cholinergic anti-inflammatory pathway in patients undergoing thoracoscopic lobectomy. METHODS Scheduled thoracoscopic lung surgery patients at the First Affiliated Hospital of Nanchang University from August to September 2023 were divided into two groups according to the surgical site. The experimental group underwent left T7 paravertebral nerve blockade (LTPVB group), while the control group underwent right T7 paravertebral nerve blockade (RTPVB group). Relevant clinical data were collected, and Doppler ultrasound was used to measure the resistive index (RI) of the splenic artery before and after blockade. Additionally, perioperative α7nAChR levels and the expression levels of the inflammatory factors IL-1β, IL-6, and TNF-α were determined. RESULTS There were no significant differences in general conditions, perioperative blood pressure, heart rate, or pain VAS scores between the two groups (p > 0.05). Splenic Doppler ultrasound showed that compared to before blockade, the RI of the splenic artery in the LTPVB group significantly decreased (p < 0.05). The α7nAChR levels at 12 h and 24 h postoperatively were significantly increased (p < 0.05) in both groups, and the levels of IL-1β, IL-6, and TNF-α gradually increased over time in both groups. However, the levels were significantly lower in the LTPVB group compared to the RTPVB group at 12 h and 24 h postoperatively (p < 0.05). CONCLUSION TPVB at left T7 can activate the α7nAChR-dependent cholinergic anti-inflammatory pathway, thereby alleviating the postoperative inflammatory response in patients undergoing thoracoscopic lobectomy.
Collapse
Affiliation(s)
- Fang Xingjun
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China
- People's Hospital of Chizhou, Chizhou, 247000, Anhui, China
| | - Zhang Ruijiao
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China
| | - Yuan Peihua
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China
| | - Wu Shiyin
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China
| | - Cheng Liqin
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China
| | - Qu Liangchao
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China.
- Ganjiang New Area People's Hospital, 330029, Nanchang, Jiangxi, China.
| | - Peng Qinghua
- Department of Anesthesiology and Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, China.
| |
Collapse
|
15
|
Brem S. Vagus nerve stimulation: Novel concept for the treatment of glioblastoma and solid cancers by cytokine (interleukin-6) reduction, attenuating the SASP, enhancing tumor immunity. Brain Behav Immun Health 2024; 42:100859. [PMID: 39512605 PMCID: PMC11541944 DOI: 10.1016/j.bbih.2024.100859] [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: 03/27/2024] [Revised: 07/31/2024] [Accepted: 09/07/2024] [Indexed: 11/15/2024] Open
Abstract
Immuno-oncology, specifically immune checkpoint inhibitors (ICIs), has revolutionized cancer care with dramatic, long-term responses and increased survival, including patients with metastatic cancer to the brain. Glioblastomas, and other primary brain tumors, are refractory to ICIs as monotherapy or in combination with standard therapy. The tumor microenvironment (TME) poses multiple biological hurdles: blood-brain barrier, immune suppression, heterogeneity, and tumor infiltration. Genomic analysis of the senescence-associated secretory phenotype (SASP) and preclinical models of glioma suggest that an exciting approach would entail reprogramming of the glioma microenvironment, attenuating the pro-inflammatory, pro-tumorigenic cytokines of the SASP, especially interleukin-6 (IL-6). A testable hypothesis now proposed is to modulate the immune system by harnessing the body's 'inflammatory reflex' to reduce cytokines. Vagus nerve stimulation can activate T cell immunity by the cholinergic, α7nicotinic acetylcholine receptor agonist (α7nAchR), and suppress IL-6 systemically, as well as other pro-inflammatory cytokines of the SASP, interleukin -1β (IL-1β) and tumor necrosis factor-alpha (TNF-α). The hypothesis predicts that electrical activation of the vagus nerve, with cytokine reduction, in combination with ICIs, would convert an immune resistant ("cold") tumor to an immune responsive ("hot") tumor, and halt glioma progression. The hypothesis also envisions cancer as an immune "dysautonomia" whereby a therapeutic intervention, vagus nerve stimulation (VNS), resets the systemic and local cytokine levels. A prospective, randomized, phase II clinical trial, to confirm the hypothesis, is a logical, exigent, next step. Cytokine reduction by VNS could also be useful for other forms of human cancer, e.g., breast, colorectal, head and neck, lung, melanoma, ovarian, pancreatic, and prostate cancer, as the emerging field of "cancer neuroscience" shows a role for neural regulation of multiple tumor types. Because IL-6, and companion pro-inflammatory cytokines, participate in the initiation, progression, spread and recurrence of cancer, minimally invasive VNS could be employed to suppress glioma or cancer progression, while also mitigating depression and/or seizures, thereby enhancing quality of life. The current hypothesis reimagines glioma pathophysiology as a dysautonomia with the therapeutic objective to reset the autonomic nervous system and form an immune responsive state to halt tumor progression and prevent recurrence. VNS, as a novel method to control cancer, can be administered with ICIs, standard therapy, or in clinical trials, combined with emerging immunotherapy: dendritic cell, mRNA, or chimeric antigen receptor (CAR) T cell vaccines.
Collapse
Affiliation(s)
- Steven Brem
- University of Pennsylvania, Department of Neurosurgery, Perelman Center for Advanced Medicine, 15-141, 3400 Civic Center Blvd., Philadelphia, PA, 19104, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, United States
| |
Collapse
|
16
|
Biscola NP, Bartmeyer PM, Beshay Y, Stern E, Mihaylov PV, Powley TL, Ward MP, Havton LA. Laterality, sexual dimorphism, and human vagal projectome heterogeneity shape neuromodulation to vagus nerve stimulation. Commun Biol 2024; 7:1536. [PMID: 39562711 PMCID: PMC11576867 DOI: 10.1038/s42003-024-07222-1] [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: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Neuromodulation by vagus nerve stimulation (VNS) provides therapeutic benefits in multiple medical conditions, including epilepsy and clinical depression, but underlying mechanisms of action are not well understood. Cervical vagus nerve biopsies were procured from transplant organ donors for high resolution light microscopy (LM) and transmission electron microscopy (TEM) to map the human fascicular and sub-fascicular organization. Cervical vagal segments show laterality with right sided dominance in fascicle numbers and cross-sectional areas as well as sexual dimorphism with female dominance in fascicle numbers. The novel and unprecedented detection of numerous small fascicles by high resolution LM and TEM expand the known fascicle size range and morphological diversity of the human vagus nerve. Ground truth TEM quantification of all myelinated and unmyelinated axons within individual nerve fascicles show marked sub-fascicular heterogeneity of nerve fiber numbers, size, and myelination. A heuristic action potential interpreter (HAPI) tool predicts VNS-evoked compound nerve action potentials (CNAPs) generated by myelinated and unmyelinated nerve fibers and validates functional dissimilarity between fascicles. Our findings of laterality, sexual dimorphism, and an expanded range of fascicle size heterogeneity provide mechanistic insights into the varied therapeutic responses and off-target effects to VNS and may guide new refinement strategies for neuromodulation.
Collapse
Affiliation(s)
- Natalia P Biscola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Petra M Bartmeyer
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Youssef Beshay
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Esther Stern
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Plamen V Mihaylov
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthew P Ward
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leif A Havton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA.
| |
Collapse
|
17
|
Liu FJ, Wu J, Gong LJ, Yang HS, Chen H. Non-invasive vagus nerve stimulation in anti-inflammatory therapy: mechanistic insights and future perspectives. Front Neurosci 2024; 18:1490300. [PMID: 39605787 PMCID: PMC11599236 DOI: 10.3389/fnins.2024.1490300] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Non-invasive vagus nerve stimulation (VNS) represents a transformative approach for managing a broad spectrum of inflammatory and autoimmune conditions, including rheumatoid arthritis and inflammatory bowel disease. This comprehensive review delineates the mechanisms underlying VNS, emphasizing the cholinergic anti-inflammatory pathway, and explores interactions within the neuro-immune and vagus-gut axes based on both clinical outcomes and pre-clinical models. Clinical applications have confirmed the efficacy of VNS in managing specific autoimmune diseases, such as rheumatoid arthritis, and chronic inflammatory conditions like inflammatory bowel disease, showcasing the variability in stimulation parameters and patient responses. Concurrently, pre-clinical studies have provided insights into the potential of VNS in modulating cardiovascular and broader inflammatory responses, paving the way for its translational application in clinical settings. Innovations in non-invasive VNS technology and precision neuromodulation are enhancing its therapeutic potential, making it a viable option for patients who are unresponsive to conventional treatments. Nonetheless, the widespread adoption of this promising therapy is impeded by regulatory challenges, patient compliance issues, and the need for extensive studies on long-term efficacy and safety. Future research directions will focus on refining VNS technology, optimizing treatment parameters, and exploring synergistic effects with other therapeutic modalities, which could revolutionize the management of chronic inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Fu-Jun Liu
- Neurology Medical Center II, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Jing Wu
- Department of Medical Imaging, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Li-Jun Gong
- Center of Surgical Anesthesia, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong-Shuai Yang
- Central Operating Room, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Huan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Hou Z, Yang F, Zhang Q, Wang Y, Liu J, Liang F. Targeting the PI3K/AKT signaling pathway with PNU120596 protects against LPS-induced acute lung injury. J Pharm Pharmacol 2024; 76:1508-1520. [PMID: 39288376 DOI: 10.1093/jpp/rgae076] [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: 11/13/2023] [Accepted: 06/04/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES This study investigated the potential therapeutic benefits of PNU120596, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (α7nAChR), in mitigating acute lung injury (ALI) induced by lipopolysaccharide (LPS) in a mouse model. Specifically, we sought to examine the impact of PNU120596 on the PI3K/AKT signaling pathway in the context of ALI. METHODS ALI was induced in mice by LPS administration, and the protective effects of PNU120596 were assessed. Lung injury, lung function, and the inflammatory response were evaluated. Additionally, the activation of the PI3K/AKT signaling pathway was examined, along with the levels of inflammatory factors and oxidative stress markers. KEY FINDINGS PNU120596 significantly ameliorated LPS-induced lung injury, improved lung function, and reduced the inflammatory response in the mouse model of ALI. Furthermore, we observed that PNU120596 inhibited the activation of the PI3K/AKT signaling pathway, which was associated with decreased levels of inflammatory factors and oxidative stress markers. CONCLUSIONS PNU120596 exhibits promising therapeutic potential for the treatment of acute lung injury, potentially by targeting the PI3K/AKT signaling pathway. These findings suggest that modulation of the α7 nicotinic acetylcholine receptor with PNU120596 may offer a viable strategy for the management of ALI, warranting further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Zixin Hou
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
- Department of Anesthesiology, General Hospital, Hunan University of Medicine, Huaihua 418000, PR China
| | - Qiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Yuxia Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Junwen Liu
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| |
Collapse
|
19
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
20
|
Kuhn AM, Bosis KE, Wohleb ES. Looking Back to Move Forward: Research in Stress, Behavior, and Immune Function. Neuroimmunomodulation 2024; 31:211-229. [PMID: 39369707 DOI: 10.1159/000541592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND From the original studies investigating the effects of adrenal gland secretion to modern high-throughput multidimensional analyses, stress research has been a topic of scientific interest spanning just over a century. SUMMARY The objective of this review was to provide historical context for influential discoveries, surprising findings, and preclinical models in stress-related neuroimmune research. Furthermore, we summarize this work and present a current understanding of the stress pathways and their effects on the immune system and behavior. We focus on recent work demonstrating stress-induced immune changes within the brain and highlight studies investigating stress effects on microglia. Lastly, we conclude with potential areas for future investigation concerning microglia heterogeneity, bone marrow niches, and sex differences. KEY MESSAGES Stress is a phenomenon that ties together not only the central and peripheral nervous system, but the immune system as well. The cumulative effects of stress can enhance or suppress immune function, based on the intensity and duration of the stressor. These stress-induced immune alterations are associated with neurobiological changes, including structural remodeling of neurons and decreased neurogenesis, and these contribute to the development of behavioral and cognitive deficits. As such, research in this field has revealed important insights into neuroimmune communication as well as molecular and cellular mediators of complex behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- Alexander M Kuhn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelly E Bosis
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
21
|
Okdahl T, Kufaishi H, Kornum D, Bertoli D, Krogh K, K Knop F, Hansen CS, Størling J, Rossing P, Brock B, Drewes AM, Brock C. Transcutaneous vagus nerve stimulation has no anti-inflammatory effect in diabetes. Sci Rep 2024; 14:21042. [PMID: 39251831 PMCID: PMC11385211 DOI: 10.1038/s41598-024-72139-y] [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: 03/18/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
Chronic inflammation is associated with diabetes and contributes to the development and progression of micro- and macrovascular complications. Transcutaneous vagus nerve stimulation (tVNS) has been proposed to reduce levels of circulating inflammatory cytokines in non-diabetics by activating the cholinergic anti-inflammatory pathway. We investigated the anti-inflammatory potential of tVNS as a secondary endpoint of a randomized controlled trial in people with diabetes (NCT04143269). 131 people with diabetes (type 1: n = 63; type 2: n = 68), gastrointestinal symptoms and various degrees of autonomic neuropathy were included and randomly assigned to self-administer active (n = 63) or sham (n = 68) tVNS over two successive study periods: (1) Seven days with four daily administrations and, (2) 56 days with two daily administrations. Levels of systemic inflammatory cytokines (IL-6, IL-8, IL-10, TNF-α, IFN-γ) were quantified from blood samples by multiplex technology. Information regarding age, sex, diabetes type, and the presence of cardiac autonomic neuropathy (CAN) was included in the analysis as possible confounders. No differences in either cytokine were seen after study period 1 and 2 between active and sham tVNS (all p-values > 0.08). Age, sex, diabetes type, presence of CAN, and baseline levels of inflammatory cytokines were not associated with changes after treatment (all p-values > 0.07). A tendency towards slight reductions in TNF-α levels after active treatment was observed in those with no CAN compared to those with early or manifest CAN (p = 0.052). In conclusion, tVNS did not influence the level of systemic inflammation in people with diabetes.
Collapse
Affiliation(s)
- Tina Okdahl
- Mech-Sense, Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Health, Aalborg University Hospital, Aalborg, Denmark
| | - Huda Kufaishi
- Steno Diabetes Center Copenhagen, University of Copenhagen, Herlev, Denmark
| | - Ditte Kornum
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Davide Bertoli
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Klaus Krogh
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Filip K Knop
- Steno Diabetes Center Copenhagen, University of Copenhagen, Herlev, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Joachim Størling
- Steno Diabetes Center Copenhagen, University of Copenhagen, Herlev, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Herlev, Denmark
| | - Birgitte Brock
- Steno Diabetes Center Copenhagen, University of Copenhagen, Herlev, Denmark
| | - Asbjørn M Drewes
- Mech-Sense, Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Health, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetes Center North Jutland, Aalborg University Hospital, Aalborg, Denmark
| | - Christina Brock
- Mech-Sense, Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark.
- Department of Clinical Medicine, Faculty of Health, Aalborg University Hospital, Aalborg, Denmark.
- Steno Diabetes Center North Jutland, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
22
|
Honwad HH, Najibi M, Koscso B, Bogunovic M, Irazoqui JE. TFEB-Mediated Pro-inflammatory Response in Murine Macrophages Induced by Acute Alpha7 Nicotinic Receptor Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577408. [PMID: 39211236 PMCID: PMC11361017 DOI: 10.1101/2024.01.26.577408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Transcription factors TFEB and TFE3 are crucial for regulating autophagy, lysosomal biogenesis, and lipid metabolism, and have significant roles in macrophage function and innate immunity. The alpha7 nicotinic acetylcholine receptor (α7nAChR), a ligand-gated Ca 2+ channel known for its therapeutic potential in neurological and inflammatory disorders, has been implicated in modulating immune responses by modulating macrophage function. Stimulation of α7nAChR with chemical agonists has been claimed to activate TFEB in pancreatic acinar cells and neurons. However, the impact of α7nAChR activation on TFEB and TFE3 in macrophages remained unknown, posing an important question due to the potential implications for inflammation regulation. This study investigates the effects of acute α7nAChR activation on TFEB-mediated responses in murine macrophages using the specific agonist PNU-282987. We demonstrate that α7nAChR stimulation triggers TFEB nuclear translocation and lysosomal expansion. Surprisingly, PNU-282987 induces a broad pro-inflammatory gene signature without concomitant cytokine secretion, suggesting an uncoupling of gene expression from cytokine release. Mechanistically, TFEB activation requires the lysosomal Ca 2+ exporter MCOLN1 and the Ca 2+ -dependent phosphatase PPP3/calcineurin. Additionally, PNU-282987 elevates reactive oxygen species (ROS) levels, and ROS are involved in TFEB activation by PNU-282987. Notably, even with α7nAChR deletion, compensatory ROS-mediated TFEB activation persists, suggesting the involvement of additional nicotinic receptors. Our findings reveal a novel α7nAChR-TFEB signaling axis in macrophages, offer new insights into the cholinergic regulation of immune responses, establish a baseline for comparison with disease states, and identify potential therapeutic targets for modulating inflammation.
Collapse
|
23
|
Crews FT, Coleman LG, Macht VA, Vetreno RP. Alcohol, HMGB1, and Innate Immune Signaling in the Brain. Alcohol Res 2024; 44:04. [PMID: 39135668 PMCID: PMC11318841 DOI: 10.35946/arcr.v44.1.04] [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] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Binge drinking (i.e., consuming enough alcohol to achieve a blood ethanol concentration of 80 mg/dL, approximately 4-5 drinks within 2 hours), particularly in early adolescence, can promote progressive increases in alcohol drinking and alcohol-related problems that develop into compulsive use in the chronic relapsing disease, alcohol use disorder (AUD). Over the past decade, neuroimmune signaling has been discovered to contribute to alcohol-induced changes in drinking, mood, and neurodegeneration. This review presents a mechanistic hypothesis supporting high mobility group box protein 1 (HMGB1) and Toll-like receptor (TLR) signaling as key elements of alcohol-induced neuroimmune signaling across glia and neurons, which shifts gene transcription and synapses, altering neuronal networks that contribute to the development of AUD. This hypothesis may help guide further research on prevention and treatment. SEARCH METHODS The authors used the search terms "HMGB1 protein," "alcohol," and "brain" across PubMed, Scopus, and Embase to find articles published between 1991 and 2023. SEARCH RESULTS The database search found 54 references in PubMed, 47 in Scopus, and 105 in Embase. A total of about 100 articles were included. DISCUSSION AND CONCLUSIONS In the brain, immune signaling molecules play a role in normal development that differs from their functions in inflammation and the immune response, although cellular receptors and signaling are shared. In adults, pro-inflammatory signals have emerged as contributing to brain adaptation in stress, depression, AUD, and neurodegenerative diseases. HMGB1, a cytokine-like signaling protein released from activated cells, including neurons, is hypothesized to activate pro-inflammatory signals through TLRs that contribute to adaptations to binge and chronic heavy drinking. HMGB1 alone and in heteromers with other molecules activates TLRs and other immune receptors that spread signaling across neurons and glia. Both blood and brain levels of HMGB1 increase with ethanol exposure. In rats, an adolescent intermittent ethanol (AIE) binge drinking model persistently increases brain HMGB1 and its receptors; alters microglia, forebrain cholinergic neurons, and neuronal networks; and increases alcohol drinking and anxiety while disrupting cognition. Studies of human postmortem AUD brain have found elevated levels of HMGB1 and TLRs. These signals reduce cholinergic neurons, whereas microglia, the brain's immune cells, are activated by binge drinking. Microglia regulate synapses through complement proteins that can change networks affected by AIE that increase drinking, contributing to risks for AUD. Anti-inflammatory drugs, exercise, cholinesterase inhibitors, and histone deacetylase epigenetic inhibitors prevent and reverse the AIE-induced pathology. Further, HMGB1 antagonists and other anti-inflammatory treatments may provide new therapies for alcohol misuse and AUD. Collectively, these findings suggest that restoring the innate immune signaling balance is central to recovering from alcohol-related pathology.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Leon G. Coleman
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Victoria A. Macht
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Parada-Kusz M, Clatworthy AE, Goering ER, Blackwood SM, Shigeta JY, Mashin E, Salm EJ, Choi C, Combs S, Lee JSW, Rodriguez-Osorio C, Clish C, Tomita S, Hung DT. 3-Hydroxykynurenine targets kainate receptors to promote defense against infection. Nat Chem Biol 2024:10.1038/s41589-024-01635-z. [PMID: 38898166 DOI: 10.1038/s41589-024-01635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/07/2024] [Indexed: 06/21/2024]
Abstract
Bacterial infection involves a complex interaction between the pathogen and host where the outcome of infection is not solely determined by pathogen eradication. To identify small molecules that promote host survival by altering the host-pathogen dynamic, we conducted an in vivo chemical screen using zebrafish embryos and found that treatment with 3-hydroxykynurenine (3-HK) protects from lethal bacterial infection. 3-HK, a metabolite produced through host tryptophan metabolism, has no direct antibacterial activity but enhances host survival by restricting bacterial expansion in macrophages through a systemic mechanism that targets kainate-sensitive glutamate receptors. These findings reveal a new pathway by which tryptophan metabolism and kainate-sensitive glutamate receptors function and interact to modulate immunity, with important implications for the coordination between the immune and nervous systems in pathological conditions.
Collapse
Affiliation(s)
- Margarita Parada-Kusz
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Clatworthy
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Emily R Goering
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie M Blackwood
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jack Y Shigeta
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Elizabeth J Salm
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Catherine Choi
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Senya Combs
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenny S W Lee
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos Rodriguez-Osorio
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
25
|
Garbizu M, Aguado L, Martín A. Alpha7 nicotinic receptors as potential theranostic targets for experimental stroke. Neural Regen Res 2024; 19:939-940. [PMID: 37862178 PMCID: PMC10749633 DOI: 10.4103/1673-5374.385294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 10/22/2023] Open
Affiliation(s)
- Maider Garbizu
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Laura Aguado
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance, San Sebastian, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
26
|
Wang Z, Han B, Qi J, Cao X, Gu H, Sun J. Chuanzhitongluo capsule improves cognitive impairment in mice with chronic cerebral hypoperfusion via the cholinergic anti-inflammatory pathway. Exp Gerontol 2024; 189:112407. [PMID: 38522309 DOI: 10.1016/j.exger.2024.112407] [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: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Vascular cognitive impairment (VCI) has become a common disease-causing cognitive deficit in humans, second only to Alzheimer's Disease (AD). Chuanzhitongluo capsule (CZTL) is a Traditional Chinese Medicine (TCM) preparation known for its effective protection against cerebral ischemia. However, its potential to ameliorate VCI remains unclear. This study aimed to investigate the cognitive improvement effects of CZTL in a mouse model of VCI. Chronic cerebral hypoperfusion (CCH) was induced in mice by bilateral common carotid artery stenosis (BCAS) to simulate the pathological changes associated with VCI. Spatial learning and memory abilities were assessed using the Morris Water Maze (MWM). RNA sequencing (RNA-Seq) was employed to identify differentially expressed genes (DEGs) in the hippocampus. Levels of inflammatory factors were measured through enzyme-linked immunosorbent assay (ELISA), while immunofluorescence (IF) determined the expression intensity of target proteins. Western Blot (WB) confirmed the final action pathway. Results indicated that CZTL significantly improved the spatial learning and memory abilities of CCH mice, along with alterations in gene expression profiles in the hippocampus. It also reduced neuroinflammation in the hippocampus and upregulated the choline acetyltransferase (ChAT) and α7 subunit-containing nicotinic acetylcholine receptor (α7nAChR), which are in synaptic plasticity and neuronal development. Moreover, CZTL inhibited the NF-κB signaling pathway. In conclusion, CZTL may alleviate neuroinflammation induced by CCH and improve cognitive impairment in CCH mice by regulating the cholinergic anti-inflammatory pathway (CAIP) involving ChAT/α7nAChR/NF-κB.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Institute of Integrative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianjiao Qi
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuelei Cao
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huali Gu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
27
|
Bricher Choque PN, Porter MH, Teixeira MS, Dellê H, Elias RM, Durante B, Dutra MRH, Metz CN, Pavlov VA, Consolim Colombo FM. Cholinergic Stimulation Exerts Cardioprotective Effects and Alleviates Renal Inflammatory Responses after Acute Myocardial Infarction in Spontaneous Hypertensive Rats (SHRs). Pharmaceuticals (Basel) 2024; 17:547. [PMID: 38794117 PMCID: PMC11124479 DOI: 10.3390/ph17050547] [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: 03/07/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND In this investigation, we explored the effects of pharmacological cholinergic stimulation on cardiac function and renal inflammation following acute myocardial infarction (AMI) in spontaneously hypertensive rats (SHRs). METHODS Adult male SHRs were randomized into three experimental groups: sham-operated; AMI + Veh (infarcted, treated with vehicle); and AMI + PY (infarcted, treated with the cholinesterase inhibitor, pyridostigmine bromide (PY)-40 mg/kg, once daily for seven days). Rats were euthanized 7 or 30 days post-surgery. The clinical parameters were assessed on the day before euthanasia. Subsequent to euthanasia, blood samples were collected and renal tissues were harvested for histological and gene expression analyses aimed to evaluate inflammation and injury. RESULTS Seven days post-surgery, the AMI + PY group demonstrated improvements in left ventricular diastolic function and autonomic regulation, and a reduction in renal macrophage infiltration compared to the AMI + Veh group. Furthermore, there was a notable downregulation in pro-inflammatory gene expression and an upregulation in anti-inflammatory gene expression. Analysis 30 days post-surgery showed that PY treatment had a sustained positive effect on renal gene expression, correlated with a decrease in biomarkers, indicative of subclinical kidney injury. CONCLUSIONS Short-term cholinergic stimulation with PY provides both cardiac and renal protection by mitigating the inflammatory response after AMI.
Collapse
Affiliation(s)
- Pamela Nithzi Bricher Choque
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Maria Helena Porter
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Manuella S. Teixeira
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| | - Humberto Dellê
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Rosilene Motta Elias
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Bruno Durante
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| | - Marina Rascio Henriques Dutra
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Christine N. Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (C.N.M.); (V.A.P.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (C.N.M.); (V.A.P.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA
| | - Fernanda M. Consolim Colombo
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| |
Collapse
|
28
|
Ihnatovych I, Saddler RA, Sule N, Szigeti K. Translational implications of CHRFAM7A, an elusive human-restricted fusion gene. Mol Psychiatry 2024; 29:1020-1032. [PMID: 38200291 PMCID: PMC11176066 DOI: 10.1038/s41380-023-02389-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Genes restricted to humans may contribute to human-specific traits and provide a different context for diseases. CHRFAM7A is a uniquely human fusion gene and a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR). The α7 nAChR has been a promising target for diseases affecting cognition and higher cortical functions, however, the treatment effect observed in animal models failed to translate into human clinical trials. As CHRFAM7A was not accounted for in preclinical drug screens it may have contributed to the translational gap. Understanding the complex genetic architecture of the locus, deciphering the functional impact of CHRFAM7A on α7 nAChR neurobiology and utilizing human-relevant models may offer novel approaches to explore α7 nAChR as a drug target.
Collapse
Affiliation(s)
- Ivanna Ihnatovych
- Department of Neurology, State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ruth-Ann Saddler
- Department of Neurology, State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Norbert Sule
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY, 14203, USA
| | - Kinga Szigeti
- Department of Neurology, State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| |
Collapse
|
29
|
López-Otín C, Kroemer G. The missing hallmark of health: psychosocial adaptation. Cell Stress 2024; 8:21-50. [PMID: 38476764 PMCID: PMC10928495 DOI: 10.15698/cst2024.03.294] [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: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
The eight biological hallmarks of health that we initially postulated (Cell. 2021 Jan 7;184(1):33-63) include features of spatial compartmentalization (integrity of barriers, containment of local perturbations), maintenance of homeostasis over time (recycling & turnover, integration of circuitries, rhythmic oscillations) and an array of adequate responses to stress (homeostatic resilience, hormetic regulation, repair & regeneration). These hallmarks affect all eight somatic strata of the human body (molecules, organelles, cells, supracellular units, organs, organ systems, systemic circuitries and meta-organism). Here we postulate that mental and socioeconomic factors must be added to this 8×8 matrix as an additional hallmark of health ("psychosocial adaptation") and as an additional stratum ("psychosocial interactions"), hence building a 9×9 matrix. Potentially, perturbation of each of the somatic hallmarks and strata affects psychosocial factors and vice versa. Finally, we discuss the (patho)physiological bases of these interactions and their implications for mental health improvement.
Collapse
Affiliation(s)
- Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
30
|
Hesampour F, Bernstein CN, Ghia JE. Brain-Gut Axis: Invasive and Noninvasive Vagus Nerve Stimulation, Limitations, and Potential Therapeutic Approaches. Inflamm Bowel Dis 2024; 30:482-495. [PMID: 37738641 DOI: 10.1093/ibd/izad211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Indexed: 09/24/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing condition with no known etiology and is characterized by disrupted gut homeostasis, chronic inflammation, and ulcerative lesions. Although current treatments can reduce disease activity, IBD frequently recurs once treatments are discontinued, indicating that treatments are ineffective in providing long-term remission. The lack of responsiveness and reluctance of some affected persons to take medications because of potential adverse effects has enhanced the need for novel therapeutic approaches. The vagus nerve (VN) is likely important in the pathogenesis of IBD, considering the decreased activity of the parasympathetic nervous system, especially the VN, and the impaired interaction between the enteric nervous system and central nervous system in patients with IBD. Vagus nerve stimulation (VNS) has demonstrated anti-inflammatory effects in various inflammatory disorders, including IBD, by inhibiting the production of inflammatory cytokines by immune cells. It has been suggested that stimulating the vagus nerve to induce its anti-inflammatory effects may be a potential therapeutic approach for IBD. Noninvasive techniques for VNS have been developed. Considering the importance of VN function in the brain-gut axis, VNS is a promising treatment option for IBD. This review discusses the potential therapeutic advantages and drawbacks of VNS, particularly the use of noninvasive transcutaneous auricular vagus nerve stimulation.
Collapse
Affiliation(s)
| | - Charles N Bernstein
- Internal Medicine, University of Manitoba, Winnipeg, Canada
- Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, Canada
| | - Jean-Eric Ghia
- Immunology, University of Manitoba, Winnipeg, Canada
- Internal Medicine, University of Manitoba, Winnipeg, Canada
- Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| |
Collapse
|
31
|
Parente J, Carolyna Gianlorenco A, Rebello-Sanchez I, Kim M, Mario Prati J, Kyung Kim C, Choi H, Song JJ, Fregni F. Neural, Anti-Inflammatory, and Clinical Effects of Transauricular Vagus Nerve Stimulation in Major Depressive Disorder: A Systematic Review. Int J Neuropsychopharmacol 2024; 27:pyad058. [PMID: 37870480 PMCID: PMC10972554 DOI: 10.1093/ijnp/pyad058] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/22/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The discovery of effective treatments for major depressive disorder (MDD) may help target different brain pathways. Invasive vagus nerve stimulation (VNS) is an effective neuromodulation technique for the treatment of MDD; however, the effectiveness of the noninvasive technique, transauricular VNS (taVNS), remains unknown. Moreover, a mechanistic understanding of the neural effects behind its biological and therapeutic effects is lacking. This review aimed to evaluate the clinical evidence and the neural and anti-inflammatory effects of taVNS in MDD. METHODS Two searches were conducted using a systematic search strategy reviewed the clinical efficacy and neural connectivity of taVNS in MDD in humans and evaluated the changes in inflammatory markers after taVNS in humans or animal models of depression. A risk of bias assessment was performed in all human studies. RESULTS Only 5 studies evaluated the effects of taVNS in patients with depression. Although the studies demonstrated the efficacy of taVNS in treating depression, they used heterogeneous methodologies and limited data, thus preventing the conduct of pooled quantitative analyses. Pooled analysis could not be performed for studies that investigated the modulation of connectivity between brain areas; of the 6 publications, 5 were based on the same experiment. The animal studies that analyzed the presence of inflammatory markers showed a reduction in the level of pro-inflammatory cytokines or receptor expression. CONCLUSIONS Data on the clinical efficacy of taVNS in the treatment of MDD are limited. Although these studies showed positive results, no conclusions can be drawn regarding this topic considering the heterogeneity of these studies, as in the case of functional connectivity studies. Based on animal studies, the application of taVNS causes a decrease in the level of inflammatory factors in different parts of the brain, which also regulate the immune system. Therefore, further studies are needed to understand the effects of taVNS in patients with MDD.
Collapse
Affiliation(s)
- Joao Parente
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | | | | | - Minkyung Kim
- Department of Neurology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jose Mario Prati
- Department of Physical Therapy, Federal University of Sao Carlos, Sao Paulo, Brazil
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Hyuk Choi
- Department of Medical Sciences, Graduate School of Medicine, Korea University, Seoul, Republic of Korea
- Neurive Co., Ltd., Gimhae, Republic of Korea
| | - Jae-Jun Song
- Neurive Co., Ltd., Gimhae, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Medical Center, Seoul, Republic of Korea
| | - Felipe Fregni
- Neuromodulation Center and Center for Clinical Research Learning, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
González-González MA, Conde SV, Latorre R, Thébault SC, Pratelli M, Spitzer NC, Verkhratsky A, Tremblay MÈ, Akcora CG, Hernández-Reynoso AG, Ecker M, Coates J, Vincent KL, Ma B. Bioelectronic Medicine: a multidisciplinary roadmap from biophysics to precision therapies. Front Integr Neurosci 2024; 18:1321872. [PMID: 38440417 PMCID: PMC10911101 DOI: 10.3389/fnint.2024.1321872] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/10/2024] [Indexed: 03/06/2024] Open
Abstract
Bioelectronic Medicine stands as an emerging field that rapidly evolves and offers distinctive clinical benefits, alongside unique challenges. It consists of the modulation of the nervous system by precise delivery of electrical current for the treatment of clinical conditions, such as post-stroke movement recovery or drug-resistant disorders. The unquestionable clinical impact of Bioelectronic Medicine is underscored by the successful translation to humans in the last decades, and the long list of preclinical studies. Given the emergency of accelerating the progress in new neuromodulation treatments (i.e., drug-resistant hypertension, autoimmune and degenerative diseases), collaboration between multiple fields is imperative. This work intends to foster multidisciplinary work and bring together different fields to provide the fundamental basis underlying Bioelectronic Medicine. In this review we will go from the biophysics of the cell membrane, which we consider the inner core of neuromodulation, to patient care. We will discuss the recently discovered mechanism of neurotransmission switching and how it will impact neuromodulation design, and we will provide an update on neuronal and glial basis in health and disease. The advances in biomedical technology have facilitated the collection of large amounts of data, thereby introducing new challenges in data analysis. We will discuss the current approaches and challenges in high throughput data analysis, encompassing big data, networks, artificial intelligence, and internet of things. Emphasis will be placed on understanding the electrochemical properties of neural interfaces, along with the integration of biocompatible and reliable materials and compliance with biomedical regulations for translational applications. Preclinical validation is foundational to the translational process, and we will discuss the critical aspects of such animal studies. Finally, we will focus on the patient point-of-care and challenges in neuromodulation as the ultimate goal of bioelectronic medicine. This review is a call to scientists from different fields to work together with a common endeavor: accelerate the decoding and modulation of the nervous system in a new era of therapeutic possibilities.
Collapse
Affiliation(s)
- María Alejandra González-González
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatric Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NOVA University, Lisbon, Portugal
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Stéphanie C. Thébault
- Laboratorio de Investigación Traslacional en salud visual (D-13), Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Marta Pratelli
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Nicholas C. Spitzer
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Collaborative Center on Big Science Plan for Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Cuneyt G. Akcora
- Department of Computer Science, University of Central Florida, Orlando, FL, United States
| | | | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | | | - Kathleen L. Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, United States
| | - Brandy Ma
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
33
|
Abraham MN, Nedeljkovic-Kurepa A, Fernandes TD, Yaipen O, Brewer MR, Leisman DE, Taylor MD, Deutschman CS. M1 cholinergic signaling in the brain modulates cytokine levels and splenic cell sub-phenotypes following cecal ligation and puncture. Mol Med 2024; 30:22. [PMID: 38317082 PMCID: PMC10845657 DOI: 10.1186/s10020-024-00787-x] [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: 09/13/2023] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND The contribution of the central nervous system to sepsis pathobiology is incompletely understood. In previous studies, administration of endotoxin to mice decreased activity of the vagus anti-inflammatory reflex. Treatment with the centrally-acting M1 muscarinic acetylcholine (ACh) receptor (M1AChR) attenuated this endotoxin-mediated change. We hypothesize that decreased M1AChR-mediated activity contributes to inflammation following cecal ligation and puncture (CLP), a mouse model of sepsis. METHODS In male C57Bl/6 mice, we quantified basal forebrain cholinergic activity (immunostaining), hippocampal neuronal activity, serum cytokine/chemokine levels (ELISA) and splenic cell subtypes (flow cytometry) at baseline, following CLP and following CLP in mice also treated with the M1AChR agonist xanomeline. RESULTS At 48 h. post-CLP, activity in basal forebrain cells expressing choline acetyltransferase (ChAT) was half of that observed at baseline. Lower activity was also noted in the hippocampus, which contains projections from ChAT-expressing basal forebrain neurons. Serum levels of TNFα, IL-1β, MIP-1α, IL-6, KC and G-CSF were higher post-CLP than at baseline. Post-CLP numbers of splenic macrophages and inflammatory monocytes, TNFα+ and ILβ+ neutrophils and ILβ+ monocytes were higher than baseline while numbers of central Dendritic Cells (cDCs), CD4+ and CD8+ T cells were lower. When, following CLP, mice were treated with xanomeline activity in basal forebrain ChAT-expressing neurons and in the hippocampus was significantly higher than in untreated animals. Post-CLP serum concentrations of TNFα, IL-1β, and MIP-1α, but not of IL-6, KC and G-CSF, were significantly lower in xanomeline-treated mice than in untreated mice. Post-CLP numbers of splenic neutrophils, macrophages, inflammatory monocytes and TNFα+ neutrophils also were lower in xanomeline-treated mice than in untreated animals. Percentages of IL-1β+ neutrophils, IL-1β+ monocytes, cDCs, CD4+ T cells and CD8+ T cells were similar in xanomeline-treated and untreated post-CLP mice. CONCLUSION Our findings indicate that M1AChR-mediated responses modulate CLP-induced alterations in serum levels of some, but not all, cytokines/chemokines and affected splenic immune response phenotypes.
Collapse
Affiliation(s)
- Mabel N Abraham
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Ana Nedeljkovic-Kurepa
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Tiago D Fernandes
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Omar Yaipen
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Mariana R Brewer
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Daniel E Leisman
- Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - Matthew D Taylor
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Clifford S Deutschman
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York, USA.
- Sepsis Research Laboratories, The Feinstein Institutes for Medical Research, Northwell Health, Room 3140, 350 Community Drive, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
34
|
Giunta S, Xia S, Pelliccioni G, Olivieri F. Autonomic nervous system imbalance during aging contributes to impair endogenous anti-inflammaging strategies. GeroScience 2024; 46:113-127. [PMID: 37821752 PMCID: PMC10828245 DOI: 10.1007/s11357-023-00947-7] [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] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
Inflammaging refers to the age-related low grade, sterile, chronic, systemic, and long-lasting subclinical, proinflammatory status, currently recognized as the main risk factor for development and progression of the most common age-related diseases (ARDs). Extensive investigations were focused on a plethora of proinflammatory stimuli that can fuel inflammaging, underestimating and partly neglecting important endogenous anti-inflammaging mechanisms that could play a crucial role in such age-related proinflammatory state. Studies on autonomic nervous system (ANS) functions during aging highlighted an imbalance toward an overactive sympathetic nervous system (SNS) tone, promoting proinflammatory conditions, and a diminished parasympathetic nervous system (PNS) activity, playing anti-inflammatory effects mediated by the so called cholinergic anti-inflammatory pathway (CAP). At the molecular level, CAP is characterized by signals communicated via the vagus nerve (with the possible involvement of the splenic nerves) through acetylcholine release to downregulate the inflammatory actions of macrophages, key players of inflammaging. Notably, decreased vagal function and increased burden of activated/senescent macrophages (macrophaging) probably precede the development of several age-related risk factors and diseases, while increased vagal function and reduced macrophaging could be associated with relevant reduction of risk profiles. Hypothalamic-pituitary-adrenal axis (HPA axis) is another pathway related to ANS promoting some anti-inflammatory response mainly through increased cortisol levels. In this perspective review, we highlighted that CAP and HPA, representing broadly "anti-inflammaging" mechanisms, have a reduced efficacy and lose effectiveness in aged people, a phenomenon that could contribute to fuel inflammaging. In this framework, strategies aimed to re-balance PNS/SNS activities could be explored to modulate systemic inflammaging especially at an early subclinical stage, thus increasing the chances to reach the extreme limit of human lifespan in healthy status.
Collapse
Affiliation(s)
- Sergio Giunta
- Casa Di Cura Prof. Nobili (Gruppo Garofalo (GHC)), Castiglione Dei Pepoli, Bologna, Italy
| | - Shijin Xia
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, China
| | | | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica Delle Marche, Via Tronto 10/A, 60126, Ancona, Italy.
- Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
35
|
Bauer KC, Trehan R, Ruf B, Myojin Y, Benmebarek MR, Ma C, Seifert M, Nur A, Qi J, Huang P, Soliman M, Green BL, Wabitsch S, Springer DA, Rodriguez-Matos FJ, Ghabra S, Gregory SN, Matta J, Dawson B, Golino J, Xie C, Dzutsev A, Trinchieri G, Korangy F, Greten TF. The Gut Microbiome Controls Liver Tumors via the Vagus Nerve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576951. [PMID: 38328040 PMCID: PMC10849697 DOI: 10.1101/2024.01.23.576951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Liver cancer ranks amongst the deadliest cancers. Nerves have emerged as an understudied regulator of tumor progression. The parasympathetic vagus nerve influences systemic immunity via acetylcholine (ACh). Whether cholinergic neuroimmune interactions influence hepatocellular carcinoma (HCC) remains uncertain. Liver denervation via hepatic vagotomy (HV) significantly reduced liver tumor burden, while pharmacological enhancement of parasympathetic tone promoted tumor growth. Cholinergic disruption in Rag1KO mice revealed that cholinergic regulation requires adaptive immunity. Further scRNA-seq and in vitro studies indicated that vagal ACh dampens CD8+ T cell activity via muscarinic ACh receptor (AChR) CHRM3. Depletion of CD8+ T cells abrogated HV outcomes and selective deletion of Chrm3 on CD8 + T cells inhibited liver tumor growth. Beyond tumor-specific outcomes, vagotomy improved cancer-associated fatigue and anxiety-like behavior. As microbiota transplantation from HCC donors was sufficient to impair behavior, we investigated putative microbiota-neuroimmune crosstalk. Tumor, rather than vagotomy, robustly altered fecal bacterial composition, increasing Desulfovibrionales and Clostridial taxa. Strikingly, in tumor-free mice, vagotomy permitted HCC-associated microbiota to activate hepatic CD8+ T cells. These findings reveal that gut bacteria influence behavior and liver anti-tumor immunity via a dynamic and pharmaceutically targetable, vagus-liver axis.
Collapse
|
36
|
Nicolini P, Malfatto G, Lucchi T. Heart Rate Variability and Cognition: A Narrative Systematic Review of Longitudinal Studies. J Clin Med 2024; 13:280. [PMID: 38202287 PMCID: PMC10780278 DOI: 10.3390/jcm13010280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Heart rate variability (HRV) is a reliable and convenient method to assess autonomic function. Cross-sectional studies have established a link between HRV and cognition. Longitudinal studies are an emerging area of research with important clinical implications in terms of the predictive value of HRV for future cognition and in terms of the potential causal relationship between HRV and cognition. However, they have not yet been the objective of a systematic review. Therefore, the aim of this systematic review was to investigate the association between HRV and cognition in longitudinal studies. METHODS The review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The Embase, PsycINFO and PubMed databases were searched from the earliest available date to 26 June 2023. Studies were included if they involved adult human subjects and evaluated the longitudinal association between HRV and cognition. The risk of bias was assessed with the Newcastle-Ottawa Scale for Cohort Studies. The results were presented narratively. RESULTS Of 14,359 records screened, 12 studies were included in this systematic review, with a total of 24,390 participants. Two thirds of the studies were published from 2020 onwards. All studies found a longitudinal relationship between HRV and cognition. There was a consistent association between higher parasympathetic nervous system (PNS) activity and better cognition, and some association between higher sympathetic nervous system activity and worse cognition. Also, higher PNS activity persistently predicted better executive functioning, while data on episodic memory and language were more scant and/or controversial. CONCLUSIONS Our results support the role of HRV as a biomarker of future cognition and, potentially, as a therapeutic target to improve cognition. They will need confirmation by further, more comprehensive studies also including unequivocal non-HRV sympathetic measures and meta-analyses.
Collapse
Affiliation(s)
- Paola Nicolini
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Geriatric Unit, Internal Medicine Department, 20122 Milan, Italy;
| | - Gabriella Malfatto
- Istituto Auxologico Italiano IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, Ospedale San Luca, 20149 Milan, Italy;
| | - Tiziano Lucchi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Geriatric Unit, Internal Medicine Department, 20122 Milan, Italy;
| |
Collapse
|
37
|
Andersson U, Tracey KJ. Vagus nerve SARS-CoV-2 infection and inflammatory reflex dysfunction: Is there a causal relationship? J Intern Med 2024; 295:91-102. [PMID: 38018736 DOI: 10.1111/joim.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Autonomic dysfunction is a clinical hallmark of infection caused by SARS-CoV-2, but the underlying mechanisms are unknown. The vagus nerve inflammatory reflex is an important, well-characterized mechanism for the reflexive suppression of cytokine storm, and its experimental or clinical impairment facilitates the onset and progression of hyperinflammation. Recent pathological evidence from COVID-19 victims reveals viral infection and inflammation in the vagus nerve and associated nuclei in the medulla oblongata. Although it has been suggested that vagus nerve inflammation in these patients mediates dysregulated respiration, whether it also contributes to dysfunction of the vagus nerve inflammatory reflex has not been addressed. Because lethality and tissue injury in acute COVID-19 are characterized by cytokine storm, it is plausible to consider evidence that impairment of the inflammatory reflex may contribute to overproduction of cytokines and resultant hyperinflammatory pathogenesis. Accordingly, here the authors discuss the inflammatory reflex, the consequences of its dysfunction in COVID-19, and whether there are opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| |
Collapse
|
38
|
Huang Y, Dong S, Li X, Shi J, Zhang Y, Liu S, Zhang Y, Yu J. VNS-mediated α7nAChR signaling promotes SPM synthesis via regulation of netrin-1 expression during LPS-induced ALI. FASEB J 2024; 38:e9664. [PMID: 38038805 DOI: 10.1096/fj.202301623r] [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: 08/09/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023]
Abstract
The α7 nicotinic acetylcholine receptor (α7nAChR) plays a crucial role in the cholinergic anti-inflammatory pathway (CAP) during sepsis-associated acute lung injury (ALI). Increasing evidence suggests that specialized pro-resolving mediators (SPMs) are important in resolving α7nAChR-mediated ALI resolution. Our study aims to elucidate the pivotal role of α7nAChR in the CAP during LPS-associated acute lung injury (ALI). By employing vagus nerve stimulation (VNS), we identified α7nAChR as the key CAP subunit in ALI mice, effectively reducing lung permeability and the release of inflammatory cytokines. We further investigated the alterations in SPMs regulated by α7nAChR, revealing a predominant synthesis of lipoxin A4 (LXA4). The significance of α7nAChR-netrin-1 pathway in governing SPM synthesis was confirmed through the use of netrin-1 knockout mice and siRNA-transfected macrophages. Additionally, our evaluation identified a synchronous alteration of LXA4 synthesis in the α7nAChR-netrin-1 pathway accompanied by 5-lipoxygenase (5-LOX), thereby confirming an ameliorative effect of LXA4 on lung injury and macrophage inflammatory response. Concurrently, inhibiting the function of LXA4 annulled the lung-protective effect of VNS. As a result, our findings reveal a novel anti-inflammatory pathway wherein VNS modulates netrin-1 expression via α7nAChR, ultimately leading to LXA4 synthesis and subsequent lung protection.
Collapse
Affiliation(s)
- Yan Huang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shasha Liu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Ye Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
39
|
Brooke AK, Murrow DP, Caldwell KCN, Witt CE, Ross AE. Measuring neuron-regulated immune cell physiology via the alpha-2 adrenergic receptor in an ex vivo murine spleen model. Cell Mol Life Sci 2023; 80:354. [PMID: 37945921 PMCID: PMC11071927 DOI: 10.1007/s00018-023-05012-2] [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: 07/10/2023] [Revised: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
The communication between the nervous and immune systems plays a crucial role in regulating immune cell function and inflammatory responses. Sympathetic neurons, which innervate the spleen, have been implicated in modulating immune cell activity. The neurotransmitter norepinephrine (NE), released by sympathetic neurons, influences immune cell responses by binding to adrenergic receptors on their surface. The alpha-2 adrenergic receptor (α2AR), expressed predominantly on sympathetic neurons, has received attention due to its autoreceptor function and ability to modulate NE release. In this study, we used fast-scan cyclic voltammetry (FSCV) to provide the first subsecond measurements of NE released in the white pulp region of the spleen and validated it with yohimbine, a known antagonist of α2AR. For further application of FSCV in neuroimmunology, we investigated the extent to which subsecond NE from sympathetic neurons is important for immune cell physiology and cytokine production, focusing on tumor necrosis factor-alpha (TNF-α), interleukin-10 (IL-10), and interleukin-6 (IL-6). Our findings provide insights into the regulatory mechanisms underlying sympathetic-immune interactions and show the significance of using FSCV, a traditional neurochemistry technique, to study these neuroimmune mechanisms.
Collapse
Affiliation(s)
- Alexandra K Brooke
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Daniel P Murrow
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Kaejaren C N Caldwell
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Colby E Witt
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Ashley E Ross
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA.
| |
Collapse
|
40
|
Yang D, Almanzar N, Chiu IM. The role of cellular and molecular neuroimmune crosstalk in gut immunity. Cell Mol Immunol 2023; 20:1259-1269. [PMID: 37336989 PMCID: PMC10616093 DOI: 10.1038/s41423-023-01054-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/01/2023] [Indexed: 06/21/2023] Open
Abstract
The gastrointestinal tract is densely innervated by the peripheral nervous system and populated by the immune system. These two systems critically coordinate the sensations of and adaptations to dietary, microbial, and damaging stimuli from the external and internal microenvironment during tissue homeostasis and inflammation. The brain receives and integrates ascending sensory signals from the gut and transduces descending signals back to the gut via autonomic neurons. Neurons regulate intestinal immune responses through the action of local axon reflexes or through neuronal circuits via the gut-brain axis. This neuroimmune crosstalk is critical for gut homeostatic maintenance and disease resolution. In this review, we discuss the roles of distinct types of gut-innervating neurons in the modulation of intestinal mucosal immunity. We will focus on the molecular mechanisms governing how different immune cells respond to neural signals in host defense and inflammation. We also discuss the therapeutic potential of strategies targeting neuroimmune crosstalk for intestinal diseases.
Collapse
Affiliation(s)
- Daping Yang
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Nicole Almanzar
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Abraham MN, Nedeljkovic-Kurepa A, Fernandes T, Yaipen O, Brewer MR, Taylor MD, Deutschman C. M1 Cholinergic Signaling Modulates Cytokine Levels and Splenocyte Sub-Phenotypes Following Cecal Ligation and Puncture. RESEARCH SQUARE 2023:rs.3.rs-3353062. [PMID: 37886474 PMCID: PMC10602092 DOI: 10.21203/rs.3.rs-3353062/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Background The contribution of the central nervous system to sepsis pathobiology is incompletely understood. In previous studies, administration of endotoxin to mice decreased activity of the vagus anti-inflammatory reflex. Treatment with the centrally-acting M1/M4 muscarinic acetylcholine (ACh) receptor (M1/M4AChR) attenuated this endotoxin-mediated change. We hypothesize that decreased M1/M4AChR-mediated activity contributes to inflammation following cecal ligation and puncture (CLP), a mouse model of sepsis. Methods Basal forebrain cholinergic activity (immunostaining), serum cytokine/chemokine levels (ELISA) and splenocyte subtypes (flow cytometry) were examined at baseline and following CLP in male C57BL/6 male mice. Rersults At 48hrs. post-CLP, activity in basal forebrain cells expressing choline acetyltransferase (ChAT) was half of that observed at baseline. Lower activity was also noted in the hippocampus, which contains projections from ChAT-expressing basal forebrain neurons. Serum levels of TNFα, IL-1β, MIP-1α, IL-6, KC and G-CSF were higher post-CLP than at baseline. Post-CLP numbers of splenic macrophages and inflammatory monocytes, TNFa+ and ILb+ neutrophils and ILb+ monocytes were higher than baseline while numbers of central Dendritic Cells (cDCs), CD4+ and CD8+ T cells were lower. When, following CLP, mice were treated with xanomeline, a central-acting M1AChR agonist, activity in basal forebrain ChAT-expressing neurons and in the hippocampus was significantly higher than in untreated animals. Post-CLP serum concentrations of TNFα, IL-1β, and MIP-1α, but not of IL-6, KC and G-CSF, were significantly lower in xanomline-treated mice than in untreated mice. Post-CLP numbers of splenic neutrophils, macrophages, inflammatory monocytes and TNFα+ neutrophils also were lower in xanomeline-treated mice than in untreated animals. The effects of CLP on percentages of IL-1β+ neutrophils, IL-1β+ monocytes, cDCs, CD4+ T cells and CD8+ T cells were similar in xanomeline - treated and untreated post-CLP mice. Conclusion Our findings indicate that M1/M4AChR-mediated responses modulate CLP-induced alterations in the distribution of some, but not all, leukocyte phenotypes and certain cytokines and chemokines.
Collapse
Affiliation(s)
| | | | | | - Omar Yaipen
- Northwell Health Feinstein Institutes for Medical Research
| | | | | | - Clifford Deutschman
- Hofstra Northwell School of Medicine at Hofstra University: Donald and Barbara Zucker School of Medicine at Hofstra/Northwell
| |
Collapse
|
42
|
Boahen A, Hu D, Adams MJ, Nicholls PK, Greene WK, Ma B. Bidirectional crosstalk between the peripheral nervous system and lymphoid tissues/organs. Front Immunol 2023; 14:1254054. [PMID: 37767094 PMCID: PMC10520967 DOI: 10.3389/fimmu.2023.1254054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The central nervous system (CNS) influences the immune system generally by regulating the systemic concentration of humoral substances (e.g., cortisol and epinephrine), whereas the peripheral nervous system (PNS) communicates specifically with the immune system according to local interactions/connections. An imbalance between the components of the PNS might contribute to pathogenesis and the further development of certain diseases. In this review, we have explored the "thread" (hardwiring) of the connections between the immune system (e.g., primary/secondary/tertiary lymphoid tissues/organs) and PNS (e.g., sensory, sympathetic, parasympathetic, and enteric nervous systems (ENS)) in health and disease in vitro and in vivo. Neuroimmune cell units provide an anatomical and physiological basis for bidirectional crosstalk between the PNS and the immune system in peripheral tissues, including lymphoid tissues and organs. These neuroimmune interactions/modulation studies might greatly contribute to a better understanding of the mechanisms through which the PNS possibly affects cellular and humoral-mediated immune responses or vice versa in health and diseases. Physical, chemical, pharmacological, and other manipulations of these neuroimmune interactions should bring about the development of practical therapeutic applications for certain neurological, neuroimmunological, infectious, inflammatory, and immunological disorders/diseases.
Collapse
Affiliation(s)
- Angela Boahen
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri-Kembangan, Selangor, Malaysia
| | - Dailun Hu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Murray J. Adams
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Philip K. Nicholls
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Wayne K. Greene
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
43
|
Mazzone GL, Coronel MF, Mladinic M, Sámano C. An update to pain management after spinal cord injury: from pharmacology to circRNAs. Rev Neurosci 2023; 34:599-611. [PMID: 36351309 DOI: 10.1515/revneuro-2022-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/19/2022] [Indexed: 08/04/2023]
Abstract
Neuropathic pain (NP) following a spinal cord injury (SCI) is often hard to control and therapies should be focused on the physical, psychological, behavioral, social, and environmental factors that may contribute to chronic sensory symptoms. Novel therapeutic treatments for NP management should be based on the combination of pharmacological and nonpharmacological options. Some of them are addressed in this review with a focus on mechanisms and novel treatments. Several reports demonstrated an aberrant expression of non-coding RNAs (ncRNAs) that may represent key regulatory factors with a crucial role in the pathophysiology of NP and as potential diagnostic biomarkers. This review analyses the latest evidence for cellular and molecular mechanisms associated with the role of circular RNAs (circRNAs) in the management of pain after SCI. Advantages in the use of circRNA are their stability (up to 48 h), and specificity as sponges of different miRNAs related to SCI and nerve injury. The present review discusses novel data about deregulated circRNAs (up or downregulated) that sponge miRNAs, and promote cellular and molecular interactions with mRNAs and proteins. This data support the concept that circRNAs could be considered as novel potential therapeutic targets for NP management especially after spinal cord injuries.
Collapse
Affiliation(s)
- Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500, B1629AHJ, Pilar, Buenos Aires, Argentina
| | - María F Coronel
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500, B1629AHJ, Pilar, Buenos Aires, Argentina
| | - Miranda Mladinic
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Cynthia Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa. Avenida Vasco de Quiroga 4871, Col. Santa Fe Cuajimalpa. Alcaldía Cuajimalpa de Morelos, C.P. 05348, Ciudad de México, México
| |
Collapse
|
44
|
Zouali M. Pharmacological and Electroceutical Targeting of the Cholinergic Anti-Inflammatory Pathway in Autoimmune Diseases. Pharmaceuticals (Basel) 2023; 16:1089. [PMID: 37631004 PMCID: PMC10459025 DOI: 10.3390/ph16081089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Continuous dialogue between the immune system and the brain plays a key homeostatic role in various immune responses to environmental cues. Several functions are under the control of the vagus nerve-based inflammatory reflex, a physiological mechanism through which nerve signals regulate immune functions. In the cholinergic anti-inflammatory pathway, the vagus nerve, its pivotal neurotransmitter acetylcholine, together with the corresponding receptors play a key role in modulating the immune response of mammals. Through communications of peripheral nerves with immune cells, it modulates proliferation and differentiation activities of various immune cell subsets. As a result, this pathway represents a potential target for treating autoimmune diseases characterized by overt inflammation and a decrease in vagal tone. Consistently, converging observations made in both animal models and clinical trials revealed that targeting the cholinergic anti-inflammatory pathway using pharmacologic approaches can provide beneficial effects. In parallel, bioelectronic medicine has recently emerged as an alternative approach to managing systemic inflammation. In several studies, nerve electrostimulation was reported to be clinically relevant in reducing chronic inflammation in autoimmune diseases, including rheumatoid arthritis and diabetes. In the future, these new approaches could represent a major therapeutic strategy for autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
45
|
Fang YT, Lin YT, Tseng WL, Tseng P, Hua GL, Chao YJ, Wu YJ. Neuroimmunomodulation of vagus nerve stimulation and the therapeutic implications. Front Aging Neurosci 2023; 15:1173987. [PMID: 37484689 PMCID: PMC10358778 DOI: 10.3389/fnagi.2023.1173987] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Vagus nerve stimulation (VNS) is a technology that provides electrical stimulation to the cervical vagus nerve and can be applied in the treatment of a wide variety of neuropsychiatric and systemic diseases. VNS exerts its effect by stimulating vagal afferent and efferent fibers, which project upward to the brainstem nuclei and the relayed circuits and downward to the internal organs to influence the autonomic, neuroendocrine, and neuroimmunology systems. The neuroimmunomodulation effect of VNS is mediated through the cholinergic anti-inflammatory pathway that regulates immune cells and decreases pro-inflammatory cytokines. Traditional and non-invasive VNS have Food and Drug Administration (FDA)-approved indications for patients with drug-refractory epilepsy, treatment-refractory major depressive disorders, and headaches. The number of clinical trials and translational studies that explore the therapeutic potentials and mechanisms of VNS is increasing. In this review, we first introduced the anatomical and physiological bases of the vagus nerve and the immunomodulating functions of VNS. We covered studies that investigated the mechanisms of VNS and its therapeutic implications for a spectrum of brain disorders and systemic diseases in the context of neuroimmunomodulation.
Collapse
Affiliation(s)
- Yi-Ting Fang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ye-Ting Lin
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Lung Tseng
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Philip Tseng
- Cross College Elite Program, National Cheng Kung University, Tainan, Taiwan
- Research Center for Mind, Brain and Learning, National Chengchi University, Taipei, Taiwan
| | - Gia-Linh Hua
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jui Chao
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Jen Wu
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
46
|
Cai H, Du Z, Lin X, Lawrence WR, Hopke PK, Rich DQ, Lin S, Xiao J, Deng X, Qu Y, Lin Z, Wang X, Ju X, Chen S, Zhang Y, Wu W, Wang Y, Gu J, Hao Y, Zhang W. Interactions between long-term ambient particle exposures and lifestyle on the prevalence of hypertension and diabetes: insight from a large community-based survey. J Epidemiol Community Health 2023; 77:440-446. [PMID: 37094940 PMCID: PMC10330163 DOI: 10.1136/jech-2023-220480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/01/2023] [Indexed: 04/26/2023]
Abstract
INTRODUCTION Evidence on the interaction of lifestyle and long-term ambient particle (PM) exposure on the prevalence of hypertension, diabetes, particularly their combined condition is limited. We investigate the associations between PM and these outcomes and whether the associations were modified by various lifestyles. METHODS This was a large population-based survey during 2019-2021 in Southern China. The concentrations of PM were interpolated and assigned to participants by the residential address. Hypertension and diabetes status were from questionnaires and confirmed with the community health centres. Logistic regression was applied to examine the associations, followed by a comprehensive set of stratified analyses by the lifestyles including diet, smoking, drinking, sleeping and exercise. RESULTS A total of 82 345 residents were included in the final analyses. For each 1 μg/m3 increase in PM2.5, the adjusted OR for the prevalence of hypertension, diabetes and their combined condition were 1.05 (95% CI 1.05 to 1.06), 1.07 (95% CI 1.06 to 1.08) and 1.05 (95% CI 1.04 to 1.06), respectively. We observed that the association between PM2.5 and the combined condition was greatest in the group with 4-8 unhealthy lifestyles (OR=1.09, 95% CI 1.06 to 1.13) followed by the group with 2-3 and those with 0-1 unhealthy lifestyle (P interaction=0.026). Similar results and trends were observed in PM10 and/or in those with hypertension or diabetes. Individuals who consumed alcohol, had inadequate sleep duration or had poor quality sleep were more vulnerable. CONCLUSION Long-term PM exposure was associated with increased prevalence of hypertension, diabetes and their combined condition, and those with unhealthy lifestyles suffered greater risks of these conditions.
Collapse
Affiliation(s)
- Huanle Cai
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhicheng Du
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao Lin
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wayne R Lawrence
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip K Hopke
- Institute for a Sustainable Environment, Clarkson University, Potsdam, New York, USA
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - David Q Rich
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Shao Lin
- Department of Environmental Health Sciences, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Jianpeng Xiao
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Xinlei Deng
- Department of Environmental Health Sciences, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Yanji Qu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, Guangdong, China
| | - Ziqiang Lin
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xinran Wang
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xu Ju
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shirui Chen
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuqin Zhang
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenjing Wu
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ying Wang
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Gu
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuantao Hao
- Peking University Center for Public Health and Epidemic Preparedness & Response, Peking University, Beijing, China
| | - Wangjian Zhang
- School of Public Health/Sun Yat-sen Global Health Institute/Center for Health Information Research, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
47
|
Husain Syed F, Okusa MD. Neuroimmune Control of Inflammation in Acute Kidney Injury: From Mouse Models to Human Disease. Nephron Clin Pract 2023; 147:754-758. [PMID: 37257419 DOI: 10.1159/000531293] [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: 04/17/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Inflammation is common in patients with acute kidney injury (AKI) and contributes to increased risk of morbidity and mortality. The central nervous system plays an important role in the immune and inflammatory pathways of AKI. In this review, we discuss the preclinical evidence for the neural pathways associated with neuromodulation in AKI, as well as clinical trials that translate these observations into the clinical context. The ultimate goal of these trials is to design strategies using noninvasive approaches, such as splenic pulsed ultrasonography, to prevent or attenuate inflammatory conditions at the bedside, including AKI.
Collapse
Affiliation(s)
- Faeq Husain Syed
- Division of Nephrology, University of Virginia, Charlottesville, Virginia, USA
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Mark D Okusa
- Division of Nephrology, University of Virginia, Charlottesville, Virginia, USA
- The Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
48
|
Montanari M, Imbriani P, Bonsi P, Martella G, Peppe A. Beyond the Microbiota: Understanding the Role of the Enteric Nervous System in Parkinson's Disease from Mice to Human. Biomedicines 2023; 11:1560. [PMID: 37371655 DOI: 10.3390/biomedicines11061560] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The enteric nervous system (ENS) is a nerve network composed of neurons and glial cells that regulates the motor and secretory functions of the gastrointestinal (GI) tract. There is abundant evidence of mutual communication between the brain and the GI tract. Dysfunction of these connections appears to be involved in the pathophysiology of Parkinson's disease (PD). Alterations in the ENS have been shown to occur very early in PD, even before central nervous system (CNS) involvement. Post-mortem studies of PD patients have shown aggregation of α-synuclein (αS) in specific subtypes of neurons in the ENS. Subsequently, αS spreads retrogradely in the CNS through preganglionic vagal fibers to this nerve's dorsal motor nucleus (DMV) and other central nervous structures. Here, we highlight the role of the ENS in PD pathogenesis based on evidence observed in animal models and using a translational perspective. While acknowledging the putative role of the microbiome in the gut-brain axis (GBA), this review provides a comprehensive view of the ENS not only as a "second brain", but also as a window into the "first brain", a potentially crucial element in the search for new therapeutic approaches that can delay and even cure the disease.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Neuroscience, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Clinical Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Antonella Peppe
- Clinical Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| |
Collapse
|
49
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
50
|
Scherlinger M, Tsokos GC. Neurotransmitters arrive to control systemic autoimmunity. Cell Metab 2023; 35:728-729. [PMID: 37137285 DOI: 10.1016/j.cmet.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/05/2023]
Abstract
Immune cell microenvironment plays a major role in the aberrant function of immune cells in systemic lupus erythematosus. Zeng and co-authors show that in human and murine lupus, splenic stromal cell-derived acetylcholine switches B cell metabolism to fatty acid oxidation and promotes B cell autoreactivity and disease development.
Collapse
Affiliation(s)
- Marc Scherlinger
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France; Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Strasbourg, France
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|