1
|
Zhao C, Pan M, Chen J, Li L, Zhang Y, Liu W, Matthay MA, Wang H, Jin X, Xu JF, Su X. Vagal-α7 nicotinic acetylcholine receptor signaling exacerbates influenza severity by promoting lung epithelial cell infection. J Med Virol 2024; 96:e29768. [PMID: 38978388 DOI: 10.1002/jmv.29768] [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: 04/01/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
The vagus nerve circuit, operating through the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), regulates the inflammatory response by influencing immune cells. However, the role of vagal-α7 nAChR signaling in influenza virus infection is unclear. In particular, does vagal-α7 nAChR signaling impact the infection of alveolar epithelial cells (AECs), the primary target cells of influenza virus? Here, we demonstrated a distinct role of α7 nAChR in type II AECs compared to its role in immune cells during influenza infection. We found that deletion of Chrna7 (encoding gene of α7 nAChR) in type II AECs or disruption of vagal circuits reduced lung influenza infection and protected mice from influenza-induced lung injury. We further unveiled that activation of α7 nAChR enhanced influenza infection through PTP1B-NEDD4L-ASK1-p38MAPK pathway. Mechanistically, activation of α7 nAChR signaling decreased p38MAPK phosphorylation during infection, facilitating the nuclear export of influenza viral ribonucleoproteins and thereby promoting infection. Taken together, our findings reveal a mechanism mediated by vagal-α7 nAChR signaling that promotes influenza viral infection and exacerbates disease severity. Targeting vagal-α7 nAChR signaling may offer novel strategies for combating influenza virus infections.
Collapse
Affiliation(s)
- Caiqi Zhao
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengyao Pan
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ling Li
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjun Liu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Michael A Matthay
- Department of Medicine, Department of Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Haichao Wang
- Department of Emergency Medicine, North Shore University Hospital, Manhasset, New York, USA
| | - Xia Jin
- Shanghai Serum Bio-Technology Co., Ltd., Shanghai, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
2
|
Peng Q, Luo X, Mo L, Xu X, Liu Y, Liu D, Yang P. TRIM41 contributes to the pathogenesis of airway allergy by compromising dendritic cells' tolerogenic properties. iScience 2024; 27:110067. [PMID: 38883815 PMCID: PMC11176661 DOI: 10.1016/j.isci.2024.110067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Dendritic cells (DC) play a crucial role in the initiation of immune responses. TRIM41, an E3 ubiquitin ligase, can facilitate targeting protein degradation. The purpose of this study is to analyze the role of TRIM41 in the pathogenesis of airway allergy (AA) and the impact of regulating TRIM41 on suppressing AA. We observed that the airway DCs of AA mice had a higher expression of Trim41. The expression of Trim41 in airway DCs was associated with the DCs' tolerogenic functions of AA mice. The AA responses, including increased amounts of eosinophil peroxidase, mast cell protease-1, Th2 cytokines, and specific IgE in bronchoalveolar lavage fluids, were positively correlated with the Trim41 expression in mouse airway DCs. TRIM41 induced c-Maf degradation and interfered with the Il10 expression in airway DCs, which could be counteracted by inhibiting TRIM41. Regulation of TRIM41 mitigated experimental AA responses.
Collapse
Affiliation(s)
- Qiuying Peng
- Department of Pediatric Otolaryngology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
- Department of Pediatrics, Guangzhou Panyu Maternal and Children Health Hospital, Guangzhou, China
| | - Xiangqian Luo
- Department of Pediatric Otolaryngology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Lihua Mo
- Department of Pediatric Otolaryngology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University and State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xuejie Xu
- Institute of Allergy & Immunology of Shenzhen University and State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Yu Liu
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dabo Liu
- Department of Pediatric Otolaryngology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University and State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Ni S, Yuan X, Cao Q, Chen Y, Peng X, Lin J, Li Y, Ma W, Gao S, Chen D. Gut microbiota regulate migration of lymphocytes from gut to lung. Microb Pathog 2023; 183:106311. [PMID: 37625662 DOI: 10.1016/j.micpath.2023.106311] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/10/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
The community of microorganisms known as gut microbiota that lives in the intestine confers significant health benefits on its host, primarily in the form of immunological homeostasis regulation. Gut microbiota not only can shape immune responses in the gut but also in other organs. This review focus on the gut-lung axis. Aberrant gut microbiota development is associated with greater lung disease susceptibility and respiratory disease induced by a variety of pathogenic bacteria. They are known to cause changes in gut microbiota. Recent research has found that immune cells in the intestine migrate to distant lung to exert anti-infective effects. Moreover, evidence indicates that the gut microbiota and their metabolites influence intestinal immune cells. Therefore, we suspect that intestine-derived immune cells may play a significant role against pulmonary pathogenic infections by receiving instructions from gut microbiota.
Collapse
Affiliation(s)
- Silu Ni
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Xiulei Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Qihang Cao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Yiming Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Xingyu Peng
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Jingyi Lin
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Yanyan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Wentao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Shikong Gao
- Shenmu Animal Husbandry Development Center, Shenmu, 719399, Shaanxi, China.
| | - Dekun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
4
|
Li L, Xu X, Wang X, Zhang S, Yao W, Liu J, Liu Z, Yang P. Galectin-9 in synergy with NF-κB inhibition restores immune regulatory capability in dendritic cells of subjects with food allergy. Clin Exp Immunol 2023; 213:155-163. [PMID: 37279535 PMCID: PMC10361740 DOI: 10.1093/cei/uxad062] [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: 11/03/2022] [Revised: 03/08/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023] Open
Abstract
The pathogenesis of immune tolerance disruption is not fully understood. Galectin-9 (Gal9) has immune regulatory functions. The objective of the present study is to assess the role of Gal9 in maintaining immune tolerance. Blood and intestinal biopsies were taken from patients with food allergy (FA). The status of tolerogenic dendritic cells (tDC) and type 1 regulatory T cells (Tr1 cells) in the samples was evaluated and used as representative parameters of immune tolerance. An FA mouse model was established to assess the role of Gal9 in maintaining immune tolerance. We found that peripheral CD11c+ CD5+ CD1d+ tDC frequency was significantly lower in FA patients as compared to health control (HC) subjects. There was no significant change in CD11c+ DC frequency between the FA group and the HC group. The expression of IL-10 in peripheral tDCs was lower in the FA group than that in the HC group. A positive correlation was detected between the serum levels of IL-10 and Gal9. The expression of Gal9 was observed in intestinal biopsies, which was positively correlated with the serum levels of Gal9 as well as serum IL-10 levels. Peripheral Tr1 cells had lower frequencies in the FA group than in the non-FA (Con) group. tDCs demonstrated the ability to generate Tr1 cells, which was weaker in the FA group as compared with the Con group. Exposure of FA tDCs to Gal9 in culture restored the ability to generate Tr1 cells. In summary, the lower frequency of tDC and Tr1 cell of FA patients was associated with the levels of Gal9. The presence of Gal9 restored the capacity of tDC to generate Tr1 cells.
Collapse
Affiliation(s)
- Linjing Li
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuejie Xu
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Xinxin Wang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Shuang Zhang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Wenkai Yao
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Jiangqi Liu
- Department of Allergy, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Zhiqiang Liu
- Department of Allergy, Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Pingchang Yang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| |
Collapse
|
5
|
Jiang M, Li Z, Zhang F, Li Z, Xu D, Jing J, Li F, Wang J, Ding J. Butyrate inhibits iILC2-mediated lung inflammation via lung-gut axis in chronic obstructive pulmonary disease (COPD). BMC Pulm Med 2023; 23:163. [PMID: 37173731 PMCID: PMC10182695 DOI: 10.1186/s12890-023-02438-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND The study investigated the effects and underlying mechanisms of intestinal flora metabolite butyrate on inflammatory ILC2 cells (iILC2s)-mediated lung inflammation in chronic obstructive pulmonary disease (COPD). METHODS Mouse models of COPD and acute exacerbation of COPD (AECOPD) were established. Flow cytometry was used to detect natural ILC2 cells (nILC2s) and iILC2s in lung and colon tissues. The 16s rRNA and GC-MS were used to detect microbial flora and short chain fatty acids (SCFAs) in feces. ELISA was used to detect IL-13 and IL-4. Western blot and qRT-PCR were used to detect the relative protein and mRNA levels, respectively. In vitro experiments were performed with sorted ILC2s from colon tissues of control mice. Mice with AECOPD were treated with butyrate. RESULTS The nILC2s and iILC2s in lung and colon tissues of AECOPD mice were significantly higher than control groups. The abundance of the flora Clostridiaceae was significantly reduced, and the content of SCFAs, including acetate and butyrate, was significantly reduced. The in vitro experiments showed that butyrate inhibited iILC2 cell phenotype and cytokine secretion. Butyrate treatment reduced the proportion of iILC2 cells in the colon and lung tissues of mice with AECOPD. CONCLUSIONS The nILC2s and iILC2s in the colon tissues are involved in the course of COPD. Decreased Clostridiaceae and butyrate in AECOPD mice caused the accumulation of iILC2 cells in the intestines and lungs. Supplementation of butyrate can reduce iILC2 in the intestine and lung tissues. Our data may provide new ideas for prevention and treatment of COPD.
Collapse
Affiliation(s)
- Min Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China
| | - Zhiwei Li
- Clinical Laboratory Center, People's Hospital of Xinjiang Uygur Autonomous, Urumqi, 830001, Xinjiang, China
| | - Fengbo Zhang
- Department of Clinical Laboratory, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Zheng Li
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China
| | - Dan Xu
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China
| | - Jing Jing
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China
| | - Fengsen Li
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China
| | - Jing Wang
- Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region, No. 116, Huanghe Road, Urumqi, 830011, Xinjiang, China.
| | - Jianbing Ding
- Department of Immunology, College of Basic Medicine, Xinjiang Medical University, No. 4, Xinyi Road, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
6
|
Xiong L, Nutt SL, Seillet C. Innate lymphoid cells: More than just immune cells. Front Immunol 2022; 13:1033904. [PMID: 36389661 PMCID: PMC9643152 DOI: 10.3389/fimmu.2022.1033904] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/10/2022] [Indexed: 11/21/2022] Open
Abstract
Since their discovery, innate lymphoid cells (ILCs) have been described as the innate counterpart of the T cells. Indeed, ILCs and T cells share many features including their common progenitors, transcriptional regulation, and effector cytokine secretion. Several studies have shown complementary and redundant roles for ILCs and T cells, leaving open questions regarding why these cells would have been evolutionarily conserved. It has become apparent in the last decade that ILCs, and rare immune cells more generally, that reside in non-lymphoid tissue have non-canonical functions for immune cells that contribute to tissue homeostasis and function. Viewed through this lens, ILCs would not be just the innate counterpart of T cells, but instead act as a link between sensory cells that monitor any changes in the environment that are not necessarily pathogenic and instruct effector cells that act to maintain body homeostasis. As these non-canonical functions of immune cells are operating in absence of pathogenic signals, it opens great avenues of research for immunologists that they now need to identify the physiological cues that regulate these cells and how the process confers a finer level of control and a greater flexibility that enables the organism to adapt to changing environmental conditions. In the review, we highlight how ILCs participate in the physiologic function of the tissue in which they reside and how physiological cues, in particular neural inputs control their homeostatic activity.
Collapse
Affiliation(s)
- Le Xiong
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen L. Nutt
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Cyril Seillet
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Cyril Seillet,
| |
Collapse
|
7
|
Tian X, Wei J, Yang M, Niu Y, Liu M, Du Y, Jin Y. An integrated strategy to reveal the potential anti-asthma mechanism of peimine by metabolite profiling, network pharmacology, and molecular docking. J Sep Sci 2022; 45:2819-2832. [PMID: 35638750 DOI: 10.1002/jssc.202200128] [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: 02/16/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 11/10/2022]
Abstract
Peimine, one of the major quality markers in Fritillaria Cirrhosae Bulbus, was expected to become a new anti-asthma drug. However, its metabolic profiles and anti-asthma mechanism have not been clarified previously. In this study, a method was developed for the detection of peimine metabolites in vitro by ultra-high-performance liquid chromatography coupled with hybrid triple quadrupole time-of-flight mass spectrometry. The potential anti-asthma mechanism was predicted by an integrated analysis of network pharmacology and molecular docking. A total of 19 metabolites were identified with the aid of software and molecular networking. The metabolic profiles of peimine elucidated that the metabolism was a multi-pathway process with characteristics of species difference. The network pharmacology results showed that peimine and its metabolites could regulate multiple asthma-related targets. The above targets were involved in various regulatory pathways linked to asthma. Moreover, the results of molecular docking showed that both peimine and its metabolites had a certain affinity with the β2 adrenergic receptor. The results provided not only important references to understand the metabolism and pharmacodynamic changes of peimine in vitro, but also supporting data for further pharmacological evaluation. It also provided a new perspective for clarifying the functional changes of traditional Chinese medicine in vitro.
Collapse
Affiliation(s)
- Xi Tian
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei, Medical University, Shijiazhuang, P. R. China
| | - Jinhuan Wei
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei, Medical University, Shijiazhuang, P. R. China
| | - Mengxin Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei, Medical University, Shijiazhuang, P. R. China
| | - Yukun Niu
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei, Medical University, Shijiazhuang, P. R. China
| | - Minyan Liu
- Chemical Engineering Institute, Shijiazhuang University, Shijiazhuang, P. R. China
| | - Yingfeng Du
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei, Medical University, Shijiazhuang, P. R. China
| | - Yiran Jin
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| |
Collapse
|
8
|
Irie M, Sasahara K, Artis D, Kabata H. Current overview of the role of neuropeptides in ILC2s and future directions. Allergol Int 2022; 71:294-300. [PMID: 35367135 DOI: 10.1016/j.alit.2022.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Indexed: 12/22/2022] Open
Abstract
The neural and immune systems are closely connected, and recently, their molecular mechanisms and relationships with diseases have attracted substantial attention. Particularly, it has been increasingly reported that ILC2s, which produce type 2 cytokines independent of acquired immunity, are regulated by neuropeptides such as catecholamines, acetylcholine, vasoactive intestinal peptide, neuromedins, and calcitonin gene-related peptide. However, the regulatory mechanisms in this regard are only partially understood, implying that further studies are still needed to clarify the complete mechanisms and processes. In this review, we summarize current reports on the regulatory effect of neuropeptides on ILC2s, some of which have conflicting results, possibly owing to the complexity of G-protein coupled receptors. By summarizing the current evidence, we hope to be able to identify what is currently unknown as well as what needs to be clarified in the future.
Collapse
|
9
|
Meng Z, Chen H, Deng C, Meng S. Potential cellular endocrinology mechanisms underlying the effects of Chinese herbal medicine therapy on asthma. Front Endocrinol (Lausanne) 2022; 13:916328. [PMID: 36051395 PMCID: PMC9424672 DOI: 10.3389/fendo.2022.916328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Asthma is a complex syndrome with polygenetic tendency and multiple phenotypes, which has variable expiratory airflow limitation and respiratory symptoms that vary over time and in intensity. In recent years, continuous industrial development has seriously impacted the climate and air quality at a global scale. It has been verified that climate change can induce asthma in predisposed individuals and that atmospheric pollution can exacerbate asthma severity. At present, a subset of patients is resistant to the drug therapy for asthma. Hence, it is urgent to find new ideas for asthma prevention and treatment. In this review, we discuss the prescription, composition, formulation, and mechanism of traditional Chinese medicine monomer, traditional Chinese medicine monomer complex, single herbs, and traditional Chinese patent medicine in the treatment of asthma. We also discuss the effects of Chinese herbal medicine on asthma from the perspective of cellular endocrinology in the past decade, emphasizing on the roles as intracellular and extracellular messengers of three substances-hormones, substances secreted by pulmonary neuroendocrine cells, and neuroendocrine-related signaling protein-which provide the theoretical basis for clinical application and new drug development.
Collapse
Affiliation(s)
- Zeyu Meng
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Chujun Deng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Shengxi Meng,
| |
Collapse
|
10
|
La Flamme AC. Immunology & Cell Biology's top 10 original research articles 2020-2021. Immunol Cell Biol 2021; 100:6-8. [PMID: 34939210 DOI: 10.1111/imcb.12514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Anne C La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
11
|
Hollenhorst MI, Krasteva-Christ G. Nicotinic Acetylcholine Receptors in the Respiratory Tract. Molecules 2021; 26:6097. [PMID: 34684676 PMCID: PMC8539672 DOI: 10.3390/molecules26206097] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023] Open
Abstract
Nicotinic acetylcholine receptors (nAChR) are widely distributed in neuronal and non-neuronal tissues, where they play diverse physiological roles. In this review, we highlight the recent findings regarding the role of nAChR in the respiratory tract with a special focus on the involvement of nAChR in the regulation of multiple processes in health and disease. We discuss the role of nAChR in mucociliary clearance, inflammation, and infection and in airway diseases such as asthma, chronic obstructive pulmonary disease, and cancer. The subtype diversity of nAChR enables differential regulation, making them a suitable pharmaceutical target in many diseases. The stimulation of the α3β4 nAChR could be beneficial in diseases accompanied by impaired mucociliary clearance, and the anti-inflammatory effect due to an α7 nAChR stimulation could alleviate symptoms in diseases with chronic inflammation such as chronic obstructive pulmonary disease and asthma, while the inhibition of the α5 nAChR could potentially be applied in non-small cell lung cancer treatment. However, while clinical studies targeting nAChR in the airways are still lacking, we suggest that more detailed research into this topic and possible pharmaceutical applications could represent a valuable tool to alleviate the symptoms of diverse airway diseases.
Collapse
|
12
|
Bai MT, Li Y, Hu ZL. Ragweed pollen induces allergic conjunctivitis immune tolerance in mice via regulation of the NF-κB signal pathway. Int J Ophthalmol 2021; 14:955-964. [PMID: 34282378 DOI: 10.18240/ijo.2021.07.01] [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: 08/24/2020] [Accepted: 03/25/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the feasibility and mechanism of immune tolerance in allergic conjunctivitis. METHODS The allergic conjunctivitis immune tolerance mice model was established by ragweed pollen (RW) and the related cytokines were detected. The mice were divided into 9 groups and the maslinic acid (MA) or PBS were given for different group after modeling. The expression levels of chemokine ligand 5 (CCL5) and P-65 in the conjunctival tissue were analyzed by immunohistochemistry, quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot. The percentage of interleukin-17 (IL-17) and CD4+CD25+ in the splenocyte supernatant was analyzed by flow cytometry. Furthermore, the serum and splenocyte supernatant concentration of total-IgE, interleukin-10 (IL-10), and IL-17 was analyzed by enzyme linked immune response (ELISA). RESULTS After the model was established, symptoms of conjunctivitis were alleviated, the level of P-65, CCL5, IL-17, and total-IgE was raised, while the expression of IL-10, CD4+CD25+ was decreased. This result fully demonstrated that a typical IL-17/regulatory-T-cells (Treg cells) imbalance and NF-κB activation. When the NF-κB signal pathway was suppressed, it showed that there was a further relief of conjunctivitis in mice. At the same time, the expression of total-IgE, IL-17, and CCL5 was decreased and the expression of anti-inflammatory factor (IL-10, CD4+CD25+) was increased. CONCLUSION In the state of immune tolerance, symptoms of conjunctivitis in mice are alleviated, the Th-17 cells of allergic conjunctivitis mice are inhibited, and Treg cells activity is enhanced.
Collapse
Affiliation(s)
- Meng-Tian Bai
- Department of Ophthalmology, Fourth Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China.,Yunnan Eye Institute, Kunming 650032, Yunnan Province, China.,Key Laboratory of Yunnan Province for the Prevention and Treatment of Ophthalmology, Kunming 650032, Yunnan Province, China.,Provincial Innovation Team for Cataract and Ocular Fundus Disease, Fourth Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China.,Expert Workstation of Yao Ke, Kunming 650032, Yunnan Province, China.,Department of Ophthalmology, Suining Central Hospital, Suining 629000, Sichuan Province, China
| | - Yun Li
- Department of Oncology, Suining Central Hospital, Suining 629000, Sichuan Province, China
| | - Zhu-Lin Hu
- Department of Ophthalmology, Fourth Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China.,Yunnan Eye Institute, Kunming 650032, Yunnan Province, China.,Key Laboratory of Yunnan Province for the Prevention and Treatment of Ophthalmology, Kunming 650032, Yunnan Province, China.,Provincial Innovation Team for Cataract and Ocular Fundus Disease, Fourth Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China.,Expert Workstation of Yao Ke, Kunming 650032, Yunnan Province, China
| |
Collapse
|
13
|
Sbierski-Kind J, Mroz N, Molofsky AB. Perivascular stromal cells: Directors of tissue immune niches. Immunol Rev 2021; 302:10-31. [PMID: 34075598 DOI: 10.1111/imr.12984] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/05/2021] [Accepted: 05/09/2021] [Indexed: 12/12/2022]
Abstract
Perivascular niches are specialized microenvironments where stromal and immune cells interact with vasculature to monitor tissue status. Adventitial perivascular niches surround larger blood vessels and other boundary sites, supporting collections of immune cells, stromal cells, lymphatics, and neurons. Adventitial fibroblasts (AFs), a subtype of mesenchymal stromal cell, are the dominant constituents in adventitial spaces, regulating vascular integrity while organizing the accumulation and activation of a variety of interacting immune cells. In contrast, pericytes are stromal mural cells that support microvascular capillaries and surround organ-specific parenchymal cells. Here, we outline the unique immune and non-immune composition of perivascular tissue immune niches, with an emphasis on the heterogeneity and immunoregulatory functions of AFs and pericytes across diverse organs. We will discuss how perivascular stromal cells contribute to the regulation of innate and adaptive immune responses and integrate immunological signals to impact tissue health and disease.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas Mroz
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.,Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.,Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Santana FPR, Ricardo-da-Silva FY, Fantozzi ET, Pinheiro NM, Tibério IFLC, Moreira LFP, Prado MAM, Prado VF, Tavares-de-Lima W, Prado CM, Breithaupt-Faloppa AC. Lung Edema and Mortality Induced by Intestinal Ischemia and Reperfusion Is Regulated by VAChT Levels in Female Mice. Inflammation 2021; 44:1553-1564. [PMID: 33715111 DOI: 10.1007/s10753-021-01440-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/11/2020] [Accepted: 02/19/2021] [Indexed: 12/22/2022]
Abstract
Acute lung injury induced by intestinal ischemia/reperfusion (I/R) is a relevant clinical condition. Acetylcholine (ACh) and the α7 nicotinic ACh receptor (nAChRα-7) are involved in the control of inflammation. Mice with reduced levels of the vesicular ACh transporter (VAChT), a protein responsible for controlling ACh release, were used to test the involvement of cholinergic signaling in lung inflammation due to intestinal I/R. Female mice with reduced levels of VAChT (VAChT-KDHOM) or wild-type littermate controls (WT) were submitted to intestinal I/R followed by 2 h of reperfusion. Mortality, vascular permeability, and recruitment of inflammatory cells into the lung were investigated. Parts of mice were submitted to ovariectomy (OVx) to study the effect of sex hormones or treated with PNU-282,987 (nAChRα-7 agonist). A total of 43.4% of VAChT-KDHOM-I/R mice died in the reperfusion period compared to 5.2% of WT I/R mice. The I/R increased lung inflammation in both genotypes. In VAChT-KDHOM mice, I/R increased vascular permeability and decreased the release of cytokines in the lung compared to WT I/R mice. Ovariectomy reduced lung inflammation and permeability compared to non-OVx, but it did not avoid mortality in VAChT-KDHOM-I/R mice. PNU treatment reduced lung permeability, increased the release of proinflammatory cytokines and the myeloperoxidase activity in the lungs, and prevented the increased mortality observed in VAChT-KDHOM mice. Cholinergic signaling is an important component of the lung protector response against intestinal I/R injury. Decreased cholinergic signaling seems to increase pulmonary edema and dysfunctional cytokine release that increased mortality, which can be prevented by increasing activation of nAChRα-7.
Collapse
Affiliation(s)
- Fernanda P R Santana
- Department of Biological Science, Federal University of São Paulo, Diadema, Brazil
| | - Fernanda Y Ricardo-da-Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Evelyn T Fantozzi
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Nathália M Pinheiro
- Department of Bioscience, Federal University of São Paulo, Campus Baixada Santista, Rua Silva Jardim, 136 - Vila Mathias, Santos, SP, Brazil
- Department of Internal Medicine, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tibério
- Department of Internal Medicine, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marco Antônio M Prado
- Robarts Research Institute, Department of Physiology & Pharmacology, The University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, The University of Western Ontario, London, Canada
| | - Vânia F Prado
- Robarts Research Institute, Department of Physiology & Pharmacology, The University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, The University of Western Ontario, London, Canada
| | - Wothan Tavares-de-Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Carla Máximo Prado
- Department of Biological Science, Federal University of São Paulo, Diadema, Brazil.
- Department of Bioscience, Federal University of São Paulo, Campus Baixada Santista, Rua Silva Jardim, 136 - Vila Mathias, Santos, SP, Brazil.
- Department of Internal Medicine, School of Medicine, University of São Paulo, São Paulo, Brazil.
| | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Shouman K, Benarroch EE. Peripheral neuroimmune interactions: selected review and some clinical implications. Clin Auton Res 2021; 31:477-489. [PMID: 33641054 PMCID: PMC7914391 DOI: 10.1007/s10286-021-00787-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Purpose To provide a brief and focused review on peripheral neuroimmune interactions and their implications for some clinical disorders. Methods Narrative review of the literature including of English-language articles published between 1985 and 2021 using PubMed and MEDLINE. Results Many studies on experimental models and in vitro indicate that there are close interactions between the neural and immune systems. Processes from sensory afferents and autonomic efferents co-localize with immune cells and interact at discrete anatomical sites forming neuroimmune units. These neuroimmune interactions are bidirectional and mediated by a wide range of soluble factors including neuropeptides, classical neurotransmitters, cytokines, and other molecules that mediate complex cross-talk among nerves and immune cells. Small-diameter sensory afferents express a wide range of receptors that respond directly to tissue damage or pathogen signals and to chemokines, cytokines, or other molecules released from immune cells. Reciprocally, immune cells respond to neurotransmitters released from nociceptive and autonomic fibers. Neuroimmune interactions operate both at peripheral tissues and at the level of the central nervous system. Both centrally and peripherally, glial cells have a major active role in this bidirectional communication. Conclusions Peripheral neuroimmune interactions are complex and importantly contribute to the pathophysiology of several disorders, including skin, respiratory, and intestinal inflammatory disorders typically associated with pain and altered barrier function. These interactions may be relevant for persistence of symptoms in disorders associated with intense immune activation.
Collapse
Affiliation(s)
- Kamal Shouman
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Eduardo E Benarroch
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Nicotinic Acetylcholine Receptor Involvement in Inflammatory Bowel Disease and Interactions with Gut Microbiota. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031189. [PMID: 33572734 PMCID: PMC7908252 DOI: 10.3390/ijerph18031189] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
The gut-brain axis describes a complex interplay between the central nervous system and organs of the gastrointestinal tract. Sensory neurons of dorsal root and nodose ganglia, neurons of the autonomic nervous system, and immune cells collect and relay information about the status of the gut to the brain. A critical component in this bi-directional communication system is the vagus nerve which is essential for coordinating the immune system’s response to the activities of commensal bacteria in the gut and to pathogenic strains and their toxins. Local control of gut function is provided by networks of neurons in the enteric nervous system also called the ‘gut-brain’. One element common to all of these gut-brain systems is the expression of nicotinic acetylcholine receptors. These ligand-gated ion channels serve myriad roles in the gut-brain axis including mediating fast synaptic transmission between autonomic pre- and postganglionic neurons, modulation of neurotransmitter release from peripheral sensory and enteric neurons, and modulation of cytokine release from immune cells. Here we review the role of nicotinic receptors in the gut-brain axis with a focus on the interplay of these receptors with the gut microbiome and their involvement in dysregulation of gut function and inflammatory bowel diseases.
Collapse
|
17
|
Chen W, Shu Q, Fan J. Neural Regulation of Interactions Between Group 2 Innate Lymphoid Cells and Pulmonary Immune Cells. Front Immunol 2020; 11:576929. [PMID: 33193374 PMCID: PMC7658006 DOI: 10.3389/fimmu.2020.576929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence supports the involvement of nervous system in the regulation of immune responses. Group 2 innate lymphoid cells (ILC2), which function as a crucial bridge between innate and adaptive immunity, are present in large numbers in barrier tissues. Neuropeptides and neurotransmitters have been found to participate in the regulation of ILC2, adding a new dimension to neuroimmunity. However, a comprehensive and detailed overview of the mechanisms of neural regulation of ILC2, associated with previous findings and prospects for future research, is still lacking. In this review, we compile existing information that supports neurons as yet poorly understood regulators of ILC2 in the field of lung innate and adaptive immunity, focusing on neural regulation of the interaction between ILC2 and pulmonary immune cells.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiang Shu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|