1
|
Wang Y, Anesi JC, Panicker IS, Cook D, Bista P, Fang Y, Oqueli E. Neuroimmune Interactions and Their Role in Immune Cell Trafficking in Cardiovascular Diseases and Cancer. Int J Mol Sci 2025; 26:2553. [PMID: 40141195 PMCID: PMC11941982 DOI: 10.3390/ijms26062553] [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: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Sympathetic nerves innervate bone marrow and various immune organs, where norepinephrine-the primary sympathetic neurotransmitter-directly interacts with immune cells that express adrenergic receptors. This article reviewed the key molecular pathways triggered by sympathetic activation and explored how sympathetic activity influences immune cell migration. Norepinephrine serves as a chemoattractant for monocytes, macrophages, and stem cells, promoting the migration of myeloid cells while inhibiting the migration of lymphocytes at physiological concentrations. We also examined the role of immune cell infiltration in cardiovascular diseases and cancer. Evidence suggests that sympathetic activation increases myeloid cell infiltration into target tissues across various cardiovascular diseases, including atherosclerosis, hypertension, cardiac fibrosis, cardiac hypertrophy, arrhythmia, myocardial infarction, heart failure, and stroke. Conversely, inhibiting sympathetic activity may serve as a potential therapeutic strategy to treat these conditions by reducing macrophage infiltration. Furthermore, sympathetic activation promotes macrophage accumulation in cancer tissues, mirroring its effects in cardiovascular diseases, while suppressing T lymphocyte infiltration into cancerous sites. These changes contribute to increased cancer growth and metastasis. Thus, inhibiting sympathetic activation could help to protect against cancer by enhancing T cell infiltration and reducing macrophage presence in tumors.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Jack C. Anesi
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Indu S. Panicker
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Darcy Cook
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Prapti Bista
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Yan Fang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Ernesto Oqueli
- Cardiology Department, Grampians Health Ballarat, Ballarat, VIC 3353, Australia
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3217, Australia
| |
Collapse
|
2
|
Lauten TH, Elkhatib SK, Natour T, Reed EC, Jojo CN, Case AJ. T H17/Treg lymphocyte balance is regulated by beta adrenergic and cAMP signaling. Brain Behav Immun 2025; 123:1061-1070. [PMID: 39542072 PMCID: PMC11967417 DOI: 10.1016/j.bbi.2024.11.013] [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: 06/06/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is a debilitating psychological disorder that also presents with neuroimmune irregularities. Patients display elevated sympathetic tone and are at an increased risk of developing secondary autoimmune diseases. Previously, using a mouse model of repeated social defeat stress (RSDS) that recapitulates certain features of PTSD, we demonstrated that elimination of sympathetic signaling to T-lymphocytes specifically limited their ability to produce pro-inflammatory interleukin 17A (IL-17A); a cytokine implicated in the development of many autoimmune disorders. However, the mechanism linking sympathetic signaling to T-lymphocyte IL-17A production remained unclear. METHODS Using a modified version of RSDS that allows for both males and females, as well as ex vivo models of T-lymphocyte polarization, we assessed the impact and mechanism of adrenergic receptor blockade (genetically and pharmacologically) and catecholamine depletion on T-lymphocyte differentiation to IL-17A-producing subtypes (i.e., TH17). RESULTS Only pharmacological inhibition of the beta 1 and 2 adrenergic receptors (β1/2) significantly decreased circulating IL-17A levels after RSDS, but did not impact other pro-inflammatory cytokines (e.g.,IL-6, TNF-α, and IL-10). This finding was confirmed using RSDS with both global β1/2 receptor knock-out mice, as well as by adoptively transferring β1/2 knock-out T-lymphocytes into immunodeficient hosts. Ex vivo polarized T-lymphocytes produced significantly less IL-17A with the blockade of β1/2 signaling, even in the absence of exogenous sympathetic neurotransmitter supplementation, which suggested T-lymphocyte-produced catecholamines may be involved in IL-17A production. Furthermore, cyclic AMP (cAMP) was demonstrated to be mechanistically involved in driving IL-17A production in T-lymphocytes, and amplifying cAMP signaling could restore IL-17A deficits caused by the absence of β1/2 signaling. Last, removal of β1/2 and cAMP signaling, even in IL-17A polarizing conditions, promoted regulatory T-lymphocyte (Treg) polarization, suggesting adrenergic signaling plays a role in the switching between pro- and anti-inflammatory T-lymphocyte subtypes. CONCLUSIONS Our data depict a novel role for β1/2 adrenergic and cAMP signaling in the balance of TH17/Treg lymphocytes. These findings provide a new target for pharmacological therapy in both psychiatric and autoimmune diseases associated with IL-17A-related pathology.
Collapse
MESH Headings
- Animals
- Mice
- Th17 Cells/metabolism
- Th17 Cells/immunology
- Signal Transduction
- Cyclic AMP/metabolism
- Male
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/immunology
- Female
- Receptors, Adrenergic, beta-2/metabolism
- Mice, Inbred C57BL
- Interleukin-17/metabolism
- Mice, Knockout
- Receptors, Adrenergic, beta-1/metabolism
- Stress, Psychological/metabolism
- Stress, Psychological/immunology
- Social Defeat
- Disease Models, Animal
- Cell Differentiation
- Adrenergic beta-Antagonists/pharmacology
- Receptors, Adrenergic, beta/metabolism
Collapse
Affiliation(s)
- Tatlock H Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Safwan K Elkhatib
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Emily C Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Caroline N Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States.
| |
Collapse
|
3
|
Wu X, Ying H, Yang Q, Yang Q, Liu H, Ding Y, Zhao H, Chen Z, Zheng R, Lin H, Wang S, Li M, Wang T, Zhao Z, Xu M, Chen Y, Xu Y, Vincent EE, Borges MC, Gaunt TR, Ning G, Wang W, Bi Y, Zheng J, Lu J. Transcriptome-wide Mendelian randomization during CD4 + T cell activation reveals immune-related drug targets for cardiometabolic diseases. Nat Commun 2024; 15:9302. [PMID: 39468075 PMCID: PMC11519452 DOI: 10.1038/s41467-024-53621-7] [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: 10/29/2023] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
Immunity has shown potentials in informing drug development for cardiometabolic diseases, such as type 2 diabetes (T2D) and coronary artery disease (CAD). Here, we performed a transcriptome-wide Mendelian randomization (MR) study to estimate the putative causal effects of 11,021 gene expression profiles during CD4+ T cells activation on the development of T2D and CAD. Robust MR and colocalization evidence was observed for 162 genes altering T2D risk and 80 genes altering CAD risk, with 12% and 16% respectively demonstrating CD4+ T cell specificity. We observed temporal causal patterns during T cell activation in 69 gene-T2D pairs and 34 gene-CAD pairs. These genes were eight times more likely to show robust genetic evidence. We further identified 25 genes that were targets for drugs under clinical investigation, including LIPA and GCK. This study provides evidence to support immune-to-metabolic disease connections, and prioritises immune-mediated drug targets for cardiometabolic diseases.
Collapse
Affiliation(s)
- Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Ying
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianqian Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Yang
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Haoyu Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilan Ding
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiling Zhao
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Zhihe Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Emma E Vincent
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, Bristol, UK
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Straub RH, Cutolo M. A History of Psycho-Neuro-Endocrine Immune Interactions in Rheumatic Diseases. Neuroimmunomodulation 2024; 31:183-210. [PMID: 39168106 DOI: 10.1159/000540959] [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] [Received: 05/24/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND All active scientists stand on the shoulders of giants and many other more anonymous scientists, and this is not different in our field of psycho-neuro-endocrine immunology in rheumatic diseases. Too often, the modern world of publishing forgets about the collective enterprise of scientists. Some journals advise the authors to present only literature from the last decade, and it has become a natural attitude of many scientists to present only the latest publications. In order to work against this general unempirical behavior, neuroimmunomodulation devotes the 30th anniversary issue to the history of medical science in psycho-neuro-endocrine immunology. SUMMARY Keywords were derived from the psycho-neuro-endocrine immunology research field very well known to the authors (R.H.S. has collected a list of keywords since 1994). We screened PubMed, the Cochran Library of Medicine, Embase, Scopus database, and the ORCID database to find relevant historical literature. The Snowballing procedure helped find related work. According to the historical appearance of discoveries in the field, the order of presentation follows the subsequent scheme: (1) the sensory nervous system, (2) the sympathetic nervous system, (3) the vagus nerve, (4) steroid hormones (glucocorticoids, androgens, progesterone, estrogens, and the vitamin D hormone), (5) afferent pathways involved in fatigue, anxiety, insomnia, and depression (includes pathophysiology), and (6) evolutionary medicine and energy regulation - an umbrella theory. KEY MESSAGES A brief history on psycho-neuro-endocrine immunology cannot address all relevant aspects of the field. The authors are aware of this shortcoming. The reader must see this review as a viewpoint through the biased eyes of the authors. Nevertheless, the text gives an overview of the history in psycho-neuro-endocrine immunology of rheumatic diseases.
Collapse
Affiliation(s)
- Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Maurizio Cutolo
- Research Laboratories and Academic Division of Clinical Rheumatology, Department of Internal Medicine DIMI, Postgraduate School of Rheumatology, University of Genova, Genoa, Italy
| |
Collapse
|
5
|
Lauten TH, Elkhatib SK, Natour T, Reed EC, Jojo CN, Case AJ. Beta-adrenergic signaling and T-lymphocyte-produced catecholamines are necessary for interleukin 17A synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597633. [PMID: 38895227 PMCID: PMC11185643 DOI: 10.1101/2024.06.05.597633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Post-traumatic stress disorder (PTSD) is a debilitating psychological disorder that also presents with neuroimmune irregularities. Patients display elevated sympathetic tone and are at an increased risk of developing secondary autoimmune diseases. Previously, using a preclinical model of PTSD, we demonstrated that elimination of sympathetic signaling to T-lymphocytes specifically limited their ability to produce pro-inflammatory interleukin 17A (IL-17A); a cytokine implicated in the development of many autoimmune disorders. However, the mechanism linking sympathetic signaling to T-lymphocyte IL-17A production remained unclear. Methods Using a modified version of repeated social defeat stress (RSDS) that allows for both males and females, we assessed the impact of adrenergic receptor blockade (genetically and pharmacologically) and catecholamine depletion on T-lymphocyte IL-17A generation. Additionally, we explored the impact of adrenergic signaling and T-lymphocyte-produced catecholamines on both CD4+ and CD8+ T-lymphocytes polarized to IL-17A-producing phenotypes ex vivo. Results Only pharmacological inhibition of the beta 1 and 2 adrenergic receptors (β1/2) significantly decreased circulating IL-17A levels after RSDS, but did not impact other pro-inflammatory cytokines (e.g., IL-6, TNF-α, and IL-10). This finding was confirmed using RSDS with both global β1/2 receptor knock-out mice, as well as by adoptively transferring β1/2 knock-out T-lymphocytes into immunodeficient hosts. Furthermore, ex vivo polarized T-lymphocytes produced significantly less IL-17A with the blockade of β1/2 signaling, even in the absence of exogenous sympathetic neurotransmitter supplementation, which suggested T-lymphocyte-produced catecholamines may be involved in IL-17A production. Indeed, pharmacological depletion of catecholamines both in vivo and ex vivo abrogated T-lymphocyte IL-17A production demonstrating the importance of immune-generated neurotransmission in pro-inflammatory cytokine generation. Conclusions Our data depict a novel role for β1/2 adrenergic receptors and autologous catecholamine signaling during T-lymphocyte IL-17A production. These findings provide a new target for pharmacological therapy in both psychiatric and autoimmune diseases associated with IL-17A-related pathology.
Collapse
Affiliation(s)
- Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Safwan K. Elkhatib
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Caroline N. Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
6
|
Perrotta S, Carnevale D. Brain-Splenic Immune System Interactions in Hypertension: Cellular and Molecular Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:65-75. [PMID: 37942610 DOI: 10.1161/atvbaha.123.318230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
Hypertension represents a major worldwide cause of death and disability, and it is becoming increasingly clear that available therapies are not sufficient to reduce the risk of major cardiovascular events. Various mechanisms contribute to blood pressure increase: neurohormonal activation, autonomic nervous system imbalance, and immune activation. Of note, the brain is an important regulator of blood pressure levels; it recognizes the peripheral perturbation and organizes a reflex response by modulating immune system and hormonal release to attempt at restoring the homeostasis. The connection between the brain and peripheral organs is mediated by the autonomic nervous system, which also modulates immune and inflammatory responses. Interestingly, an increased autonomic nervous system activity has been correlated with an altered immune response in cardiovascular diseases. The spleen is the largest immune organ exerting a potent influence on the cardiovascular system during disease and is characterized by a dense noradrenergic innervation. Taken together, these aspects led to hypothesize a key role of neuroimmune mechanisms in the onset and progression of hypertension. This review discusses how the nervous and splenic immune systems interact and how the mechanisms underlying the neuroimmune cross talk influence the disease progression.
Collapse
Affiliation(s)
- Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy (D.C.)
| |
Collapse
|
7
|
Qiao H, Xu Q, Xu Y, Zhao Y, He N, Tang J, Zhao J, Liu Y. Molecular chaperones in stroke-induced immunosuppression. Neural Regen Res 2023; 18:2638-2644. [PMID: 37449602 DOI: 10.4103/1673-5374.373678] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Stroke-induced immunosuppression is a process that leads to peripheral suppression of the immune system after a stroke and belongs to the central nervous system injury-induced immunosuppressive syndrome. Stroke-induced immunosuppression leads to increased susceptibility to post-stroke infections, such as urinary tract infections and stroke-associated pneumonia, worsening prognosis. Molecular chaperones are a large class of proteins that are able to maintain proteostasis by directing the folding of nascent polypeptide chains, refolding misfolded proteins, and targeting misfolded proteins for degradation. Various molecular chaperones have been shown to play roles in stroke-induced immunosuppression by modulating the activity of other molecular chaperones, cochaperones, and their associated pathways. This review summarizes the role of molecular chaperones in stroke-induced immunosuppression and discusses new approaches to restore host immune defense after stroke.
Collapse
Affiliation(s)
- Haoduo Qiao
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Yunfei Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Yao Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University; Department of Pathophysiology, Xiangya School of Medicine, Central South University; Sepsis Translational Medicine Key Laboratory of Hunan Province; National Medicine Functional Experimental Teaching Center, Changsha, Hunan Province, China
| |
Collapse
|
8
|
Control of lymph node activity by direct local innervation. Trends Neurosci 2022; 45:704-712. [PMID: 35820971 DOI: 10.1016/j.tins.2022.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022]
Abstract
The nervous system detects environmental and internal stimuli and relays this information to immune cells via neurotransmitters and neuropeptides. This is essential to respond appropriately to immunogenic threats and to support system homeostasis. Lymph nodes (LNs) act as sentinels where adaptive immune responses are generated. They are richly innervated by peripheral sympathetic and sensory nerves, which are responsible for the local secretion of neurotransmitters by sympathetic fibers, such as norepinephrine, and neuropeptides by sensory fibers, including calcitonin gene-related peptide (CGRP) and substance P. Additionally, time-of-day-dependent oscillations in nerve activity are associated with differential immune responses, suggesting a potential role for neuroimmune interactions in coordinating immunity in a circadian fashion. Here, we discuss how LN activity is controlled by local innervation.
Collapse
|
9
|
Carnevale D. Neuroimmune axis of cardiovascular control: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:379-394. [PMID: 35301456 DOI: 10.1038/s41569-022-00678-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2022] [Indexed: 12/21/2022]
Abstract
Cardiovascular diseases (CVDs) make a substantial contribution to the global burden of disease. Prevention strategies have succeeded in reducing the effect of acute CVD events and deaths, but the long-term consequences of cardiovascular risk factors still represent the major cause of disability and chronic illness, suggesting that some pathophysiological mechanisms might not be adequately targeted by current therapies. Many of the underlying causes of CVD have now been recognized to have immune and inflammatory components. However, inflammation and immune activation were mostly regarded as a consequence of target-organ damage. Only more recent findings have indicated that immune dysregulation can be pathogenic for CVD, identifying a need for novel immunomodulatory therapeutic strategies. The nervous system, through an array of afferent and efferent arms of the autonomic nervous system, profoundly affects cardiovascular function. Interestingly, the autonomic nervous system also innervates immune organs, and neuroimmune interactions that are biologically relevant to CVD have been discovered, providing the foundation to target neural reflexes as an immunomodulatory therapeutic strategy. This Review summarizes how the neural regulation of immunity and inflammation participates in the onset and progression of CVD and explores promising opportunities for future therapeutic strategies.
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Molecular Medicine, Sapienza University, Rome, Italy. .,Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy.
| |
Collapse
|
10
|
Priyanka HP, Thiyagaraj A, Krithika G, Nair RS, Hopper W, ThyagaRajan S. 17β-Estradiol Concentration and Direct β 2-Adrenoceptor Inhibition Determine Estrogen-Mediated Reversal of Adrenergic Immunosuppression. Ann Neurosci 2022; 29:32-52. [PMID: 35875427 PMCID: PMC9305908 DOI: 10.1177/09727531211070541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Sympathetic innervation of lymphoid organs, and the presence of 17β-estradiol (estrogen or E2) and adrenergic receptors (ARs) on lymphocytes, suggests that sympathetic stimulation and hormonal activation may influence immune functions. Purpose: Modeling and simulating these pathways may help to understand the dynamics of neuroendocrine-immune modulation at the cellular and molecular levels. Methods: Dose- and receptor-dependent effects of E2 and AR subtype-specific agonists were established in vitro on lymphocytes from young male Sprague-Dawley rats and were modeled in silico using the MATLAB Simbiology toolbox. Kinetic principles were assigned to define receptor–ligand dynamics, and concentration/time plots were obtained using Ode15s solvers at different time intervals for key regulatory molecules. Comparisons were drawn between in silico and in vitro data for validating the constructed model with sensitivity analysis of key regulatory molecules to assess their individual impacts on the dynamics of the system. Finally, docking studies were conducted with key ligands E2 and norepinephrine (NE) to understand the mechanistic principles underlying their interactions. Results: Adrenergic activation triggered proapoptotic signals, while E2 enhanced survival signals, showing opposing effects as observed in vitro. Treatment of lymphocytes with E2 shows a 10-fold increase in survival signals in a dose-dependent manner. Cyclic adenosine monophosphate (cAMP) activation is crucial for the activation of survival signals through extracellular signal-regulated kinase (p-ERK) and cAMP responsive element binding (p-CREB) protein. Docking studies showed the direct inhibition of ERK by NE and β2-AR by E2 explaining how estrogen signaling overrides NE-mediated immunosuppression in vitro. Conclusion: The cross-talk between E2 and adrenergic signaling pathways determines lymphocyte functions in a receptor subtype and coactivation-dependent manner in health and disease.
Collapse
Affiliation(s)
- Hannah P. Priyanka
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Inspire Lab, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Specialty Hospital, Chennai, Tamil Nadu, India
| | - A. Thiyagaraj
- Department of Bioinformatics, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - G. Krithika
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras Guindy, Campus, Chennai, Tamil Nadu, India
| | - R. S. Nair
- Inspire Lab, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Specialty Hospital, Chennai, Tamil Nadu, India
| | - W. Hopper
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Department of Bioinformatics, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - S. ThyagaRajan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
11
|
Sahbaie P, Li WW, Guo TZ, Shi XY, Kingery WS, Clark JD. Autonomic Regulation of Nociceptive and Immunologic Changes in a Mouse Model of Complex Regional Pain Syndrome. THE JOURNAL OF PAIN 2022; 23:472-486. [PMID: 34699985 PMCID: PMC8920776 DOI: 10.1016/j.jpain.2021.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 09/27/2021] [Indexed: 01/16/2023]
Abstract
Chronic pain frequently develops after limb injuries, and its pathogenesis is poorly understood. We explored the hypothesis that the autonomic nervous system regulates adaptive immune system activation and nociceptive sensitization in a mouse model of chronic post-traumatic pain with features of complex regional pain syndrome (CRPS). In studies sympathetic signaling was reduced using 6-hydroxydopamine (6-OHDA) or lofexidine, while parasympathetic signaling was augmented by nicotine administration. Hindpaw allodynia, unweighting, skin temperature, and edema were measured at 3 and 7 weeks after fracture. Hypertrophy of regional lymph nodes and IgM deposition in the skin of injured limbs were followed as indices of adaptive immune system activation. Passive transfer of serum from fracture mice to recipient B cell deficient (muMT) mice was used to assess the formation of pain-related autoantibodies. We observed that 6-OHDA or lofexidine reduced fracture-induced hindpaw nociceptive sensitization and unweighting. Nicotine had similar effects. These treatments also prevented IgM deposition, hypertrophy of popliteal lymph nodes, and the development of pronociceptive serum transfer effects. We conclude that inhibiting sympathetic or augmenting parasympathetic signaling inhibits pro-nociceptive immunological changes accompanying limb fracture. These translational results support the use of similar approaches in trials potentially alleviating persistent post-traumatic pain and, possibly, CRPS. PERSPECTIVE: Selective treatments aimed at autonomic nervous system modulation reduce fracture-related nociceptive and functional sequelae. The same treatment strategies limit pain-supporting immune system activation and the production of pro-nociceptive antibodies. Thus, the therapeutic regulation of autonomic activity after limb injury may reduce the incidence of chronic pain.
Collapse
Affiliation(s)
- Peyman Sahbaie
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Department of Anesthesia, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California.
| | - Wen-Wu Li
- Anesthesiology Service; Veterans Affairs Palo Alto Health Care System; 3801 Miranda Ave, Palo Alto, CA 94304, U.S.A,Department of Anesthesia, Perioperative and Pain Medicine; Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tian-Zhi Guo
- Anesthesiology Service; Veterans Affairs Palo Alto Health Care System; 3801 Miranda Ave, Palo Alto, CA 94304, U.S.A,Palo Alto Veterans Institute for Research, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - Xiao-you Shi
- Anesthesiology Service; Veterans Affairs Palo Alto Health Care System; 3801 Miranda Ave, Palo Alto, CA 94304, U.S.A,Department of Anesthesia, Perioperative and Pain Medicine; Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wade S. Kingery
- Palo Alto Veterans Institute for Research, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - J David Clark
- Anesthesiology Service; Veterans Affairs Palo Alto Health Care System; 3801 Miranda Ave, Palo Alto, CA 94304, U.S.A,Department of Anesthesia, Perioperative and Pain Medicine; Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Allegra A, Sant'Antonio E, Musolino C, Ettari R. New insights into neuropeptides regulation of immune system and hemopoiesis: effects on hematologic malignancies. Curr Med Chem 2021; 29:2412-2437. [PMID: 34521320 DOI: 10.2174/0929867328666210914120228] [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: 03/29/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 11/22/2022]
Abstract
Several neurotransmitters and neuropeptides were reported to join to or to cooperate with different cells of the immune system, bone marrow, and peripheral cells and numerous data support that neuroactive molecules might control immune system activity and hemopoiesis operating on lymphoid organs, and the primary hematopoietic unit, the hematopoietic niche. Furthermore, many compounds seem to be able to take part to the leukemogenesis and lymphomagenesis process, and in the onset of multiple myeloma. In this review, we will assess the possibility that neurotransmitters and neuropeptides may have a role in the onset of haematological neoplasms, may affect the response to treatment or may represent a useful starting point for a new therapeutic approach. More in vivo investigations are needed to evaluate neuropeptide's role in haematological malignancies and the possible utilization as an antitumor therapeutic target. Comprehending the effect of the pharmacological administration of neuropeptide modulators on hematologic malignancies opens up new possibilities in curing clonal hematologic diseases to achieve more satisfactory outcomes.
Collapse
Affiliation(s)
- Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood, University of Messina. Italy
| | | | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood, University of Messina. Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina. Italy
| |
Collapse
|
13
|
Elkhatib SK, Moshfegh CM, Watson GF, Schwab AD, Katsurada K, Patel KP, Case AJ. Splenic denervation attenuates repeated social defeat stress-induced T-lymphocyte inflammation. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 1:190-200. [PMID: 35330608 PMCID: PMC8941638 DOI: 10.1016/j.bpsgos.2021.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/12/2021] [Accepted: 05/06/2021] [Indexed: 11/28/2022] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is a devastating psychological disorder. Patients with PTSD canonically demonstrate an increased risk for inflammatory diseases, as well as increased sympathetic tone and norepinephrine (NE) outflow. Yet, the exact etiology and causal nature of these physiologic changes remain unclear. Previously, we demonstrated that exogenous NE alters mitochondrial superoxide in T-lymphocytes to produce a pro-inflammatory T-helper 17 (TH17) phenotype, and observed similar TH17 polarization in a preclinical model of PTSD. Therefore, we hypothesized sympathetic-driven neuroimmune interactions could mediate psychological trauma-induced T-lymphocyte inflammation. Methods Repeated social defeat stress (RSDS) is a preclinical murine model that recapitulates the behavioral, autonomic, and inflammatory aspects of PTSD. Targeted splenic denervation (Dnx) was performed to deduce the contribution of splenic sympathetic nerves to RSDS-induced inflammation. Eighty-five C57BL/6J mice underwent Dnx or sham-operation, followed by RSDS or control paradigms. Animals were assessed for behavioral, autonomic, inflammatory, and redox profiles. Results Dnx did not alter the antisocial or anxiety-like behavior induced by RSDS. In circulation, RSDS Dnx animals exhibited diminished levels of T-lymphocyte-specific cytokines (IL-2, IL-17A, and IL-22) compared to intact animals, whereas other non-specific inflammatory cytokines (e.g., IL-6, TNF-α, and IL-10) were unaffected by Dnx. Importantly, Dnx specifically ameliorated the increases in RSDS-induced T-lymphocyte mitochondrial superoxide, TH17 polarization, and pro-inflammatory gene expression with minimal impact to non-T-lymphocyte immune populations. Conclusions Overall, our data suggest that sympathetic nerves regulate RSDS-induced splenic T-lymphocyte inflammation, but play less of a role in the behavioral and non-T-lymphocyte inflammatory phenotypes induced by this psychological trauma paradigm.
Collapse
Affiliation(s)
- Safwan K. Elkhatib
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Cassandra M. Moshfegh
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gabrielle F. Watson
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Aaron D. Schwab
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Adam J. Case
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| |
Collapse
|
14
|
Chen CS, Weber J, Holtkamp SJ, Ince LM, de Juan A, Wang C, Lutes L, Barnoud C, Kizil B, Hergenhan SM, Salvermoser J, Lasch M, Deindl E, Schraml B, Baumjohann D, Scheiermann C. Loss of direct adrenergic innervation after peripheral nerve injury causes lymph node expansion through IFN-γ. J Exp Med 2021; 218:e20202377. [PMID: 34086056 PMCID: PMC8185988 DOI: 10.1084/jem.20202377] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/19/2021] [Accepted: 05/03/2021] [Indexed: 11/04/2022] Open
Abstract
Peripheral nerve injury can cause debilitating disease and immune cell-mediated destruction of the affected nerve. While the focus has been on the nerve-regenerative response, the effect of loss of innervation on lymph node function is unclear. Here, we show that the popliteal lymph node (popLN) receives direct neural input from the sciatic nerve and that sciatic denervation causes lymph node expansion. Loss of sympathetic, adrenergic tone induces the expression of IFN-γ in LN CD8 T cells, which is responsible for LN expansion. Surgery-induced IFN-γ expression and expansion can be rescued by β2 adrenergic receptor agonists but not sensory nerve agonists. These data demonstrate the mechanisms governing the pro-inflammatory effect of loss of direct adrenergic input on lymph node function.
Collapse
Affiliation(s)
- Chien-Sin Chen
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Jasmin Weber
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Stephan Jonas Holtkamp
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Louise Madeleine Ince
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alba de Juan
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Chen Wang
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lydia Lutes
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Coline Barnoud
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Burak Kizil
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sophia Martina Hergenhan
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Johanna Salvermoser
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Manuel Lasch
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximillians-Universität München, Munich, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig-Maximillians-Universität München, Munich, Germany
| | - Elisabeth Deindl
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximillians-Universität München, Munich, Germany
| | - Barbara Schraml
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
| | - Dirk Baumjohann
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Christoph Scheiermann
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximillians-Universität München, Planegg-Martinsried, Germany
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
O'Neill E, Griffin ÉW, O'Sullivan R, Murray C, Ryan L, Yssel J, Harkin A, Cunningham C. Acute neuroinflammation, sickness behavior and working memory responses to acute systemic LPS challenge following noradrenergic lesion in mice. Brain Behav Immun 2021; 94:357-368. [PMID: 33307172 DOI: 10.1016/j.bbi.2020.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Locus coeruleus (LC)-derived noradrenaline is important in cognition and decreases with age, but the impact of prior noradrenaline deficiency on vulnerability to inflammation-induced acute cognitive dysfunction is unclear. Here we assessed whether noradrenergic depletion, in female mice, impacted upon inflammation, locomotor activity and working memory directly after acute systemic immune challenge with bacterial lipopolysaccharide (LPS), a paradigm we have previously used to capture delirium-like acute cognitive deficits. Mice received 2 doses of the LC-selective noradrenergic toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4; 50 mg/kg i.p.) and were challenged, 2 weeks later, with LPS (100 μg/kg i.p.). DSP-4 dramatically reduced noradrenaline concentrations and tyrosine hydroxylase-positive afferents in the frontal cortex and hippocampus. This did not significantly alter numbers of Pu.1-positive microglia, Iba1-positive microglial morphology or mRNA expression of microglia-associated gene transcripts (Tyrobp, Sall1, Cd68, Sra2, Clec7a) in the hippocampus or frontal cortex and produced modest reductions in Cx3cr1 and P2ry12. LPS induced blood and brain cytokine levels, cFOS activation and locomotor responses that were highly similar in DSP-4- and vehicle-treated mice, although LPS-induced plasma TNF-α was significantly reduced in those treated with DSP-4. Importantly, prior noradrenergic depletion did not predispose to LPS-induced T-maze working memory deficits. The data demonstrate that significant depletion of noradrenaline in the hippocampus and frontal cortex does not prompt acutely exaggerated neuroinflammation or leave the brain vulnerable to acute, transient working memory deficits upon low dose LPS challenge. These findings have implications for our understanding of the impact of systemic inflammation on the aging and vulnerable brain during septic encephalopathy and delirium.
Collapse
Affiliation(s)
- Eoin O'Neill
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Éadaoin W Griffin
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Ruairi O'Sullivan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Carol Murray
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Lucy Ryan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Justin Yssel
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew Harkin
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
16
|
Devi S, Alexandre YO, Loi JK, Gillis R, Ghazanfari N, Creed SJ, Holz LE, Shackleford D, Mackay LK, Heath WR, Sloan EK, Mueller SN. Adrenergic regulation of the vasculature impairs leukocyte interstitial migration and suppresses immune responses. Immunity 2021; 54:1219-1230.e7. [PMID: 33915109 DOI: 10.1016/j.immuni.2021.03.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
The sympathetic nervous system (SNS) controls various physiological functions via the neurotransmitter noradrenaline. Activation of the SNS in response to psychological or physical stress is frequently associated with weakened immunity. Here, we investigated how adrenoceptor signaling influences leukocyte behavior. Intravital two-photon imaging after injection of noradrenaline revealed transient inhibition of CD8+ and CD4+ T cell locomotion in tissues. Expression of β-adrenergic receptor in hematopoietic cells was not required for NA-mediated inhibition of motility. Rather, chemogenetic activation of the SNS or treatment with adrenergic receptor agonists induced vasoconstriction and decreased local blood flow, resulting in abrupt hypoxia that triggered rapid calcium signaling in leukocytes and halted cell motility. Oxygen supplementation reversed these effects. Treatment with adrenergic receptor agonists impaired T cell responses induced in response to viral and parasitic infections, as well as anti-tumor responses. Thus, stimulation of the SNS impairs leukocyte mobility, providing a mechanistic understanding of the link between adrenergic receptors and compromised immunity.
Collapse
Affiliation(s)
- Sapna Devi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Joon Keit Loi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Ryan Gillis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Nazanin Ghazanfari
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Sarah J Creed
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - David Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052 Australia; Division of Surgery, Peter MacCallum Cancer Center, Victoria, 3000, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia; The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, Victoria, 3000, Australia.
| |
Collapse
|
17
|
Lucas TA, Zhu L, Buckwalter MS. Spleen glia are a transcriptionally unique glial subtype interposed between immune cells and sympathetic axons. Glia 2021; 69:1799-1815. [DOI: 10.1002/glia.23993] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Tawaun A. Lucas
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
| | - Li Zhu
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
- Department of Neurosurgery Stanford School of Medicine Stanford California USA
| |
Collapse
|
18
|
Hima L, Patel MN, Kannan T, Gour S, Pratap UP, Priyanka HP, Vasantharekha R, ThyagaRajan S. Age-associated decline in neural, endocrine, and immune responses in men and women: Involvement of intracellular signaling pathways. J Neuroimmunol 2020; 345:577290. [DOI: 10.1016/j.jneuroim.2020.577290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
|
19
|
Matt SM, Gaskill PJ. Where Is Dopamine and how do Immune Cells See it?: Dopamine-Mediated Immune Cell Function in Health and Disease. J Neuroimmune Pharmacol 2020; 15:114-164. [PMID: 31077015 PMCID: PMC6842680 DOI: 10.1007/s11481-019-09851-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023]
Abstract
Dopamine is well recognized as a neurotransmitter in the brain, and regulates critical functions in a variety of peripheral systems. Growing research has also shown that dopamine acts as an important regulator of immune function. Many immune cells express dopamine receptors and other dopamine related proteins, enabling them to actively respond to dopamine and suggesting that dopaminergic immunoregulation is an important part of proper immune function. A detailed understanding of the physiological concentrations of dopamine in specific regions of the human body, particularly in peripheral systems, is critical to the development of hypotheses and experiments examining the effects of physiologically relevant dopamine concentrations on immune cells. Unfortunately, the dopamine concentrations to which these immune cells would be exposed in different anatomical regions are not clear. To address this issue, this comprehensive review details the current information regarding concentrations of dopamine found in both the central nervous system and in many regions of the periphery. In addition, we discuss the immune cells present in each region, and how these could interact with dopamine in each compartment described. Finally, the review briefly addresses how changes in these dopamine concentrations could influence immune cell dysfunction in several disease states including Parkinson's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, as well as the collection of pathologies, cognitive and motor symptoms associated with HIV infection in the central nervous system, known as NeuroHIV. These data will improve our understanding of the interactions between the dopaminergic and immune systems during both homeostatic function and in disease, clarify the effects of existing dopaminergic drugs and promote the creation of new therapeutic strategies based on manipulating immune function through dopaminergic signaling. Graphical Abstract.
Collapse
Affiliation(s)
- S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
20
|
Huang YY, Li X, Li X, Sheng YY, Zhuang PW, Zhang YJ. Neuroimmune crosstalk in central nervous system injury-induced infection and pharmacological intervention. Brain Res Bull 2019; 153:232-238. [PMID: 31536756 DOI: 10.1016/j.brainresbull.2019.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 12/30/2022]
Abstract
Infection (such as pneumonia and urinary tract infection) is one of the leading causes of death in patients with acute central nervous system (CNS) injury, which also greatly affects the patients' prognosis and quality of life. Antibiotics are commonly used for the treatment of various infections, however, available evidence demonstrate that prophylactic antibiotic treatments for CNS injury-induced infection have been unsuccessful. Effective approaches for prevention of CNS injury induced-infection remain scarce, therefore, better understanding the molecular and cellular mechanisms of infection post-CNS injury may aid in the development of efficacious therapeutic options. CNS injury-induced infection is confirmed affected by the sympathetic/parasympathetic nervous system, hypothalamic-pituitary-adrenal axis, and even brain-gut axis. In this review, we summarized the mechanisms of CNS injury- induced infection, crosstalk between the CNS and the immune system and current pharmacological intervention to provide ideas for the development of new anti- infective therapeutic strategies.
Collapse
Affiliation(s)
- Ying-Ying Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Xueli Li
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Xiaojin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Yuan-Yuan Sheng
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Peng-Wei Zhuang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Yan-Jun Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| |
Collapse
|
21
|
Glucocorticoids and Catecholamines Affect in Vitro Functionality of Porcine Blood Immune Cells. Animals (Basel) 2019; 9:ani9080545. [PMID: 31408932 PMCID: PMC6720833 DOI: 10.3390/ani9080545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/28/2022] Open
Abstract
Simple Summary In modern livestock husbandry, animals may face stressful events like weaning, regrouping, or transportation, all of which can impair animal welfare and health. Research in model organisms has revealed that stress hormones, such as glucocorticoids and catecholamines, strongly modulate the immune system and thus the animals’ ability to fight infections. In the pig, knowledge about this relationship is rare, and results from rodents cannot readily be transferred due to some physiological differences. Therefore, the effects of glucocorticoids and catecholamines on porcine immune cell proliferation and the production of the pro-inflammatory cytokine TNFα were investigated in an in vitro study. Blood was obtained from catheterized pigs to exclude pre-exposure to stress hormones. Glucocorticoids exerted inhibitory effects on both investigated immune functions. Catecholamines, on the other hand, showed diverse effects on lymphocyte proliferation and TNFα production of particular immune cell types. This suggests that studies from model species are not entirely transferrable to pigs. Future research should extend the preliminary findings on cytokine production and focus on the molecular mechanisms and health impacts of stress hormones in pigs. Abstract Stress hormones exert important modulating influences on the functionality of immune cells. Despite its major role as a livestock animal and its increasing use as an animal model, knowledge about this relationship in the domestic pig is rare. This study therefore aimed to characterize the effect of glucocorticoids and catecholamines on the proliferation and cytokine production of porcine peripheral blood mononuclear cells (PBMC). Blood was obtained from donor pigs equipped with indwelling catheters to exclude stress hormone exposition before in vitro testing. PBMC were stimulated in the presence of cortisol, adrenaline or noradrenaline at concentrations resembling low to high stress conditions. Proliferation was determined via 3H-thymidine incorporation, and TNFα producers were quantified by intracellular cytokine staining. Cortisol led to a decrease in mitogen-induced lymphocyte proliferation and the number of TNFα producing cells. In contrast, catecholamines increased proliferation while exerting repressive or no effects on the number of cytokine producers. Remarkably, in concentrations presumably found in lymphatic tissue in stress situations, noradrenaline suppressed lymphocyte proliferation completely. The shown repressive effects might especially have implications on health and welfare in pigs. The obtained results provide a preliminary database for extended studies on the molecular mechanisms of glucocorticoid and catecholamine actions on porcine immune cells.
Collapse
|
22
|
Seicol BJ, Bejarano S, Behnke N, Guo L. Neuromodulation of metabolic functions: from pharmaceuticals to bioelectronics to biocircuits. J Biol Eng 2019; 13:67. [PMID: 31388355 PMCID: PMC6676523 DOI: 10.1186/s13036-019-0194-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022] Open
Abstract
Neuromodulation of central and peripheral neural circuitry brings together neurobiologists and neural engineers to develop advanced neural interfaces to decode and recapitulate the information encoded in the nervous system. Dysfunctional neuronal networks contribute not only to the pathophysiology of neurological diseases, but also to numerous metabolic disorders. Many regions of the central nervous system (CNS), especially within the hypothalamus, regulate metabolism. Recent evidence has linked obesity and diabetes to hyperactive or dysregulated autonomic nervous system (ANS) activity. Neural regulation of metabolic functions provides access to control pathology through neuromodulation. Metabolism is defined as cellular events that involve catabolic and/or anabolic processes, including control of systemic metabolic functions, as well as cellular signaling pathways, such as cytokine release by immune cells. Therefore, neuromodulation to control metabolic functions can be used to target metabolic diseases, such as diabetes and chronic inflammatory diseases. Better understanding of neurometabolic circuitry will allow for targeted stimulation to modulate metabolic functions. Within the broad category of metabolic functions, cellular signaling, including the production and release of cytokines and other immunological processes, is regulated by both the CNS and ANS. Neural innervations of metabolic (e.g. pancreas) and immunologic (e.g. spleen) organs have been understood for over a century, however, it is only now becoming possible to decode the neuronal information to enable exogenous controls of these systems. Future interventions taking advantage of this progress will enable scientists, engineering and medical doctors to more effectively treat metabolic diseases.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH USA
- Department of Neuroscience, The Ohio State University, Columbus, OH USA
| | | | - Nicholas Behnke
- Department of Food, Agricultural, and Biological Engineering, The Ohio State University, Columbus, OH USA
| | - Liang Guo
- Department of Neuroscience, The Ohio State University, Columbus, OH USA
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH USA
| |
Collapse
|
23
|
Pathological cardiac hypertrophy: the synergy of adenylyl cyclases inhibition in cardiac and immune cells during chronic catecholamine stress. J Mol Med (Berl) 2019; 97:897-907. [PMID: 31062036 DOI: 10.1007/s00109-019-01790-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022]
Abstract
Response to stressors in our environment and daily lives is an adaptation conserved through evolution as it is beneficial in enhancing the survival and continuity of humans. Although stressors have evolved, the drastic physiological response they elicit still remains unchanged. The chronic secretion and circulation of catecholamines to produce physical responses when they are not required may result in pathological consequences which affect cardiac function drastically. This review seeks to point out the probable implication of chronic stress in inducing an inflammation disorder in the heart. We discussed the likely synergy of a G protein-independent stimuli signaling via β2-adrenergic receptors in both cardiomyocytes and immune cells during chronic catecholamine stress. To explain this synergy, we hypothesized the possibility of adenylyl cyclases having a regulatory effect on G protein-coupled receptor kinases. This was based on the negative correlations they exhibit during normal cardiac function and heart failures. As such, the downregulation of adenylyl cyclases in cardiomyocytes and immune cells during chronic catecholamine stress enhances the expressions of G protein-coupled receptor kinases. In addition, we explain the maladaptive roles played by G protein-coupled receptor kinase and extracellular signal-regulated kinase in the synergistic cascade that pathologically remodels the heart. Finally, we highlighted the therapeutic potentials of an adenylyl cyclases stimulator to attenuate pathological cardiac hypertrophy (PCH) and improve cardiac function in patients developing cardiac disorders due to chronic catecholamine stress.
Collapse
|
24
|
Epigenetic Signature of Chronic Maternal Stress Load During Pregnancy Might be a Potential Biomarker for Spontaneous Preterm Birth. Balkan J Med Genet 2018; 21:27-33. [PMID: 30984521 PMCID: PMC6454236 DOI: 10.2478/bjmg-2018-0023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Preterm birth is the leading cause of mortality in newborn infants and can lead to significant neonatal morbidities. Spontaneous preterm birth accounts for at least 50.0% of all preterm births. We argue that chronic maternal stress load, which is an important risk factor for spontaneous preterm birth, could be represented by epigenetic signature of several specific genetic loci in the mother’s blood. A literature search was done in PubMed with the following keywords: “DNA methylation,” “epigenetics,” “maternal stress” and “preterm birth” from year 2000 to 2017. We suggest that these genetic loci might be related to vulnerability and hypersensibility of stress response during pregnancy in women with preterm births. The mother’s epi-genetic stress bioprofile was supposed to be a result of chronic maternal stress load since her birth. This epigenetic bioprofile might also be a potential biomarker for spontaneous preterm birth. DNA methylation changes are tissue-specific and human stress response manifests mostly through the central nervous system (CNS). Nevertheless, we found evidence that methylation changes of DNA isolated from blood leucocytes might be a reliable measure of stress-related epigenetic changes that occur in the CNS. Evaluating biological mechanisms through the development of simple assays based on epigenetic changes to measure chronic stress loads in expectant mothers can lead to our ability to prepare more effective measures for the prevention of preterm births, as well as leading to more effective treatment strategies for both expectant mothers and their newborns.
Collapse
|
25
|
Reardon C, Murray K, Lomax AE. Neuroimmune Communication in Health and Disease. Physiol Rev 2018; 98:2287-2316. [PMID: 30109819 PMCID: PMC6170975 DOI: 10.1152/physrev.00035.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022] Open
Abstract
The immune and nervous systems are tightly integrated, with each system capable of influencing the other to respond to infectious or inflammatory perturbations of homeostasis. Recent studies demonstrating the ability of neural stimulation to significantly reduce the severity of immunopathology and consequently reduce mortality have led to a resurgence in the field of neuroimmunology. Highlighting the tight integration of the nervous and immune systems, afferent neurons can be activated by a diverse range of substances from bacterial-derived products to cytokines released by host cells. While activation of vagal afferents by these substances dominates the literature, additional sensory neurons are responsive as well. It is becoming increasingly clear that although the cholinergic anti-inflammatory pathway has become the predominant model, a multitude of functional circuits exist through which neuronal messengers can influence immunological outcomes. These include pathways whereby efferent signaling occurs independent of the vagus nerve through sympathetic neurons. To receive input from the nervous system, immune cells including B and T cells, macrophages, and professional antigen presenting cells express specific neurotransmitter receptors that affect immune cell function. Specialized immune cell populations not only express neurotransmitter receptors, but express the enzymatic machinery required to produce neurotransmitters, such as acetylcholine, allowing them to act as signaling intermediaries. Although elegant experiments have begun to decipher some of these interactions, integration of these molecules, cells, and anatomy into defined neuroimmune circuits in health and disease is in its infancy. This review describes these circuits and highlights continued challenges and opportunities for the field.
Collapse
Affiliation(s)
- Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Kaitlin Murray
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Alan E Lomax
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
26
|
Szpotowicz-Czech B, Wiecek M, Szymura J, Maciejczyk M, Szygula Z. Changes in chosen immune system indicators and the level of HSP-70 after single whole-body cryostimulation in healthy men. Cent Eur J Immunol 2018; 43:186-193. [PMID: 30135632 PMCID: PMC6102624 DOI: 10.5114/ceji.2018.77389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 09/25/2017] [Indexed: 01/20/2023] Open
Abstract
AIM OF THE STUDY The aim of our research was to examine the influence of single whole-body cryostimulation (WBC) on chosen immune system indicators including the heat shock protein HSP-70. MATERIAL AND METHODS The study was carried out among ten young and healthy men (mean age 22.4 ±1.65, with a body mass index of 22.91 ±2.39 kg/m2). The participants were subjected to single whole-body cryostimulation (at -130°C temperatures) in a special cryogenic chamber for 3 minutes. Blood samples were collected three times: before cryostimulation, 30 minutes and 24 hours after WBC. Immunoglobulins (IgA, IgG, IgM), interleukins (IL-6, IL-10, IL-1β) and the heat shock protein (HSP-70) were determined in the blood serum. RESULTS As a result of a single exposure to cryogenic temperatures, a significant increase in the level of IL-6 was observed 30 minutes after the WBC (p < 0.05) and a decrease in the level of HSP-70 24 hours after the treatment (p < 0.05). There were no significant changes in the level of interleukins (IL-10, IL-1β) or immunoglobulins 30 minutes after a single WBC treatment or 24 hours later. CONCLUSIONS Detailed analysis of the issue shows that a single application of whole-body cryostimulation causes a small, modulating effect on the IL-6 level. Single whole-body cryostimulation treatment has also a slight silencing effect on the HSP-70 level in healthy, young men. Reduction in the concentration of HSP-70 24 hours after WBC may indicate lack of the damaging impact on the spatial structure of the protein due to cryogenic temperatures.
Collapse
Affiliation(s)
| | - Magdalena Wiecek
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education, Krakow, Poland
| | - Jadwiga Szymura
- Department of Clinical Rehabilitation, Faculty of Motor Rehabilitation, University of Physical Education, Krakow, Poland
| | - Marcin Maciejczyk
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education, Krakow, Poland
| | - Zbigniew Szygula
- Department of Sports Medicine and Human Nutrition, Faculty of Physical Education and Sport, University of Physical Education in Krakow, Krakow, Poland
| |
Collapse
|
27
|
Shi K, Wood K, Shi FD, Wang X, Liu Q. Stroke-induced immunosuppression and poststroke infection. Stroke Vasc Neurol 2018; 3:34-41. [PMID: 29600006 PMCID: PMC5870641 DOI: 10.1136/svn-2017-000123] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 12/03/2017] [Accepted: 12/11/2017] [Indexed: 02/04/2023] Open
Abstract
Infections occur commonly after stroke and are strongly associated with an unfavourable functional outcome of these patients. Approaches for effective management of poststroke infection remain scarce, presenting an urgent need for preventive anti-infection strategies for patients who have suffered a stroke. Emerging evidence indicates that stroke impairs systemic immune responses and increases the susceptibility to infections, suggesting that the modification of impaired immune defence could be beneficial. In this review, we summarised previous attempts to prevent poststroke infections using prophylactic antibiotics and the current understanding of stroke-induced immunosuppression. Further elucidation of the immune mechanisms of stroke will pave the way to tailored design of new treatment to combat poststroke infection via modifying the immune system.
Collapse
Affiliation(s)
- Kaibin Shi
- Departments of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Departments of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Kristofer Wood
- Departments of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Fu-Dong Shi
- Departments of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Departments of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Xiaoying Wang
- Departments of Neurology and Radiology, Neuroprotection Research Laboratory, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiang Liu
- Departments of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Departments of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
28
|
Qiao G, Chen M, Bucsek MJ, Repasky EA, Hylander BL. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front Immunol 2018; 9:164. [PMID: 29479349 PMCID: PMC5812031 DOI: 10.3389/fimmu.2018.00164] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
An immune response must be tightly controlled so that it will be commensurate with the level of response needed to protect the organism without damaging normal tissue. The roles of cytokines and chemokines in orchestrating these processes are well known, but although stress has long been thought to also affect immune responses, the underlying mechanisms were not as well understood. Recently, the role of nerves and, specifically, the sympathetic nervous system, in regulating immune responses is being revealed. Generally, an acute stress response is beneficial but chronic stress is detrimental because it suppresses the activities of effector immune cells while increasing the activities of immunosuppressive cells. In this review, we first discuss the underlying biology of adrenergic signaling in cells of both the innate and adaptive immune system. We then focus on the effects of chronic adrenergic stress in promoting tumor growth, giving examples of effects on tumor cells and immune cells, explaining the methods commonly used to induce stress in preclinical mouse models. We highlight how this relates to our observations that mandated housing conditions impose baseline chronic stress on mouse models, which is sufficient to cause chronic immunosuppression. This problem is not commonly recognized, but it has been shown to impact conclusions of several studies of mouse physiology and mouse models of disease. Moreover, the fact that preclinical mouse models are chronically immunosuppressed has critical ramifications for analysis of any experiments with an immune component. Our group has found that reducing adrenergic stress by housing mice at thermoneutrality or treating mice housed at cooler temperatures with β-blockers reverses immunosuppression and significantly improves responses to checkpoint inhibitor immunotherapy. These observations are clinically relevant because there are numerous retrospective epidemiological studies concluding that cancer patients who were taking β-blockers have better outcomes. Clinical trials testing whether β-blockers can be repurposed to improve the efficacy of traditional and immunotherapies in patients are on the horizon.
Collapse
Affiliation(s)
- Guanxi Qiao
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Minhui Chen
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark J. Bucsek
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Elizabeth A. Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bonnie L. Hylander
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
29
|
Ben-Shaanan T, Schiller M, Rolls A. Studying brain-regulation of immunity with optogenetics and chemogenetics; A new experimental platform. Brain Behav Immun 2017; 65:1-8. [PMID: 27890661 DOI: 10.1016/j.bbi.2016.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
The interactions between the brain and the immune system are bidirectional. Nevertheless, we have far greater understanding of how the immune system affects the brain than how the brain affects immunity. New technological developments such as optogenetics and chemogenetics (using DREADDs; Designer Receptors Exclusively Activated by Designer Drugs) can bridge this gap in our understanding, as they enable an unprecedented mechanistic and systemic analysis of the communication between the brain and the immune system. In this review, we discuss new experimental approaches for revealing neuronal circuits that can participate in regulation of immunity. In addition, we discuss methods, specifically optogenetics and chemogenetics, that enable targeted neuronal manipulation to reveal how different brain regions affect immunity. We describe how these techniques can be used as an experimental platform to address fundamental questions in psychoneuroimmunology and to understand how neuronal circuits associate with different psychological states can affect physiology.
Collapse
Affiliation(s)
- Tamar Ben-Shaanan
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Maya Schiller
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Asya Rolls
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
30
|
Sleep influences the immune response and the rejection process alters sleep pattern: Evidence from a skin allograft model in mice. Brain Behav Immun 2017; 61:274-288. [PMID: 28069386 DOI: 10.1016/j.bbi.2016.12.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/24/2016] [Accepted: 12/31/2016] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Sleep generally regulates immune functions in a supportive manner and can affect parameters that are directly involved in the rejection process. STUDY OBJECTIVES The first objective was to assess whether sleep deprivation (SD) or sleep restriction (SR) affects the allograft rejection process in mice. The second objective was to investigate whether the rejection process itself modulates the sleep pattern of allografted mice. DESIGN Adult BALB/c and C57BL/6J male mice were used as the donors and recipients, respectively, except for the syngeneic group (ISOTX), which received skin from mice of the same strain (C57BL/6J). The recipients were randomly assigned to either one of two control groups - TX (allogenic) or ISOTX (syngeneic) - which underwent stereotaxic surgery to enable sleep recording prior to the allograft but were not sleep deprived; one of two paradoxical sleep deprived groups - SDTX and TXSD - which underwent 72h of continuous SD either before or after the allograft respectively, and one of two sleep restricted groups - SRTX and TXSR - which underwent 21h of SD and 3h of sleep for 15days either before or after the allograft respectively. INTERVENTIONS The skin allograft was inspected daily to determine the survival time, expected as 8.0±0.4days in this transplant model under no treatment. The sleep pattern was controlled throughout the rejection process in the SD and SR groups. Draining lymph nodes, spleen, blood and skin grafts were harvested on the 5th day after transplantation for evaluation of the immune parameters related to allograft rejection. MEASUREMENTS AND RESULTS In the control groups, we observed a reduction in paradoxical sleep throughout the entire allograft rejection process. Acute and chronic experimental sleep loss in the SD and SR groups produced marked alterations in the immune response. Both SD and SR prolonged allograft survival compared to the non-sleep-deprived group. There were reductions in the following parameters involved in the allograft rejection under sleep loss: CD4+ and CD8+ T cell subpopulations in the peripheral lymph organs and spleen, circulating sIL-2R levels, graft-infiltrating CD4+ T cells and skin allograft global gene expression. CONCLUSIONS We provide, as far as we are aware, the first evidence in vivo that the immune response can alter the normal sleep pattern, and that sleep loss can conversely affect the immune response related to graft rejection.
Collapse
|
31
|
Rollins-Smith LA. Amphibian immunity-stress, disease, and climate change. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:111-119. [PMID: 27387153 DOI: 10.1016/j.dci.2016.07.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/25/2016] [Accepted: 07/01/2016] [Indexed: 05/22/2023]
Abstract
Like all other vertebrate groups, amphibian responses to the environment are mediated through the brain (hypothalamic)-pituitary-adrenal/interrenal (HPA/I) axis and the sympathetic nervous system. Amphibians are facing historically unprecedented environmental stress due to climate change that will involve unpredictable temperature and rainfall regimes and possible nutritional deficits due to extremes of temperature and drought. At the same time, amphibians in all parts of the world are experiencing unprecedented declines due to the emerging diseases, chytridiomycosis (caused by Batrachochytrium dendrobatidis and Batrachochytrium salamandrivorans) and ranavirus diseases due to viruses of the genus Ranavirus in the family Iridoviridae. Other pathogens and parasites also afflict amphibians, but here I will limit myself to a review of recent literature linking stress and these emerging diseases (chytridiomycosis and ranavirus disease) in order to better predict how environmental stressors and disease will affect global amphibian populations.
Collapse
Affiliation(s)
- Louise A Rollins-Smith
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
32
|
Verburg-van Kemenade BML, Cohen N, Chadzinska M. Neuroendocrine-immune interaction: Evolutionarily conserved mechanisms that maintain allostasis in an ever-changing environment. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:2-23. [PMID: 27296493 DOI: 10.1016/j.dci.2016.05.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 05/02/2023]
Abstract
It has now become accepted that the immune system and neuroendocrine system form an integrated part of our physiology. Immunological defense mechanisms act in concert with physiological processes like growth and reproduction, energy intake and metabolism, as well as neuronal development. Not only are psychological and environmental stressors communicated to the immune system, but also, vice versa, the immune response and adaptation to a current pathogen challenge are communicated to the entire body, including the brain, to evoke adaptive responses (e.g., fever, sickness behavior) that ensure allocation of energy to fight the pathogen. This phenomenon is evolutionarily conserved. Hence it is both interesting and important to consider the evolutionary history of this bi-directional neuroendocrine-immune communication to reveal phylogenetically ancient or relatively recently acquired mechanisms. Indeed, such considerations have already disclosed an extensive "common vocabulary" of information pathways as well as molecules and their receptors used by both the neuroendocrine and immune systems. This review focuses on the principal mechanisms of bi-directional communication and the evidence for evolutionary conservation of the important physiological pathways involved.
Collapse
Affiliation(s)
- B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept. of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
| | - Nicholas Cohen
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| |
Collapse
|
33
|
Dhawan S, De Palma G, Willemze RA, Hilbers FW, Verseijden C, Luyer MD, Nuding S, Wehkamp J, Souwer Y, de Jong EC, Seppen J, van den Wijngaard RM, Wehner S, Verdu E, Bercik P, de Jonge WJ. Acetylcholine-producing T cells in the intestine regulate antimicrobial peptide expression and microbial diversity. Am J Physiol Gastrointest Liver Physiol 2016; 311:G920-G933. [PMID: 27514477 DOI: 10.1152/ajpgi.00114.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/10/2016] [Indexed: 01/31/2023]
Abstract
The cholinergic anti-inflammatory pathway reduces systemic tumor necrosis factor (TNF) via acetylcholine-producing memory T cells in the spleen. These choline acetyltransferase (ChAT)-expressing T cells are also found in the intestine, where their function is unclear. We aimed to characterize these cells in mouse and human intestine and delineate their function. We made use of the ChAT-enhanced green fluorescent protein (eGFP) reporter mice. CD4Cre mice were crossed to ChATfl/fl mice to achieve specific deletion of ChAT in CD4+ T cells. We observed that the majority of ChAT-expressing T cells in the human and mouse intestine have characteristics of Th17 cells and coexpress IL17A, IL22, and RORC The generation of ChAT-expressing T cells was skewed by dendritic cells after activation of their adrenergic receptor β2 To evaluate ChAT T cell function, we generated CD4-specific ChAT-deficient mice. CD4ChAT-/- mice showed a reduced level of epithelial antimicrobial peptides lysozyme, defensin A, and ang4, which was associated with an enhanced bacterial diversity and richness in the small intestinal lumen in CD4ChAT-/- mice. We conclude that ChAT-expressing T cells in the gut are stimulated by adrenergic receptor activation on dendritic cells. ChAT-expressing T cells may function to mediate the host AMP secretion, microbial growth and expansion.
Collapse
Affiliation(s)
- Shobhit Dhawan
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Giada De Palma
- Farncombe Institute, McMaster University, Hamilton, Ontario, Canada
| | - Rose A Willemze
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Francisca W Hilbers
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Caroline Verseijden
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Misha D Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Sabine Nuding
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology Stuttgart, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Jan Wehkamp
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology Stuttgart, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Yuri Souwer
- Department of Cell Biology and Histology, Academic Medical Centrum, Amsterdam, The Netherlands; and
| | - Esther C de Jong
- Department of Cell Biology and Histology, Academic Medical Centrum, Amsterdam, The Netherlands; and
| | - J Seppen
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - René M van den Wijngaard
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Sven Wehner
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Elena Verdu
- Farncombe Institute, McMaster University, Hamilton, Ontario, Canada
| | - Premek Bercik
- Farncombe Institute, McMaster University, Hamilton, Ontario, Canada
| | - Wouter J de Jonge
- Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands;
| |
Collapse
|
34
|
Barrios-Payán J, Revuelta A, Mata-Espinosa D, Marquina-Castillo B, Villanueva EB, Gutiérrez MEH, Pérez-Sánchez G, Pavón L, Hernandez-Pando R. The contribution of the sympathetic nervous system to the immunopathology of experimental pulmonary tuberculosis. J Neuroimmunol 2016; 298:98-105. [PMID: 27609282 DOI: 10.1016/j.jneuroim.2016.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 11/24/2022]
Abstract
The role of norepinephrine (NE) in the immunopathology of experimental tuberculosis (TB) was studied by measuring pulmonary NE and determining its cellular sources and targets. Functional studies were performed administrating adrenergic and anti-adrenergic drugs at different TB phases. Results showed high production of NE during early infection by adrenergic nerve terminals and lymphocytes located in the lungs and mediastinal lymph nodes, these cells highly expressed β2 adreno-receptors (β2AR) which by an autocrine mechanism promote Th-1 cell differentiation favoring protection. During advanced infection, the production of NE and β2AR sharply decreased, suggesting that adrenergic activity is less important during late TB.
Collapse
Affiliation(s)
- Jorge Barrios-Payán
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico.
| | - Alberto Revuelta
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico.
| | - Dulce Mata-Espinosa
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico.
| | - Brenda Marquina-Castillo
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico.
| | - Enrique Becerril Villanueva
- Department of Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Deleg. Tlalpan, 14370 México City, Mexico.
| | - María Eugenia Hernández Gutiérrez
- Department of Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Deleg. Tlalpan, 14370 México City, Mexico.
| | - Gilberto Pérez-Sánchez
- Department of Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Deleg. Tlalpan, 14370 México City, Mexico.
| | - Lenin Pavón
- Department of Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Deleg. Tlalpan, 14370 México City, Mexico.
| | - Rogelio Hernandez-Pando
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico.
| |
Collapse
|
35
|
Strahler J, Skoluda N, Rohleder N, Nater UM. Dysregulated stress signal sensitivity and inflammatory disinhibition as a pathophysiological mechanism of stress-related chronic fatigue. Neurosci Biobehav Rev 2016; 68:298-318. [DOI: 10.1016/j.neubiorev.2016.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/03/2016] [Accepted: 05/10/2016] [Indexed: 01/20/2023]
|
36
|
Balivada S, Pawar HN, Montgomery S, Kenney MJ. Effect of ghrelin on regulation of splenic sympathetic nerve discharge. Auton Neurosci 2016; 201:68-71. [PMID: 27554768 DOI: 10.1016/j.autneu.2016.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/22/2016] [Accepted: 08/18/2016] [Indexed: 10/21/2022]
Abstract
Ghrelin influences immune system function and modulates the sympathetic nervous system; however, the contribution of ghrelin to neural-immune interactions is not well-established because the effect of ghrelin on splenic sympathetic nerve discharge (SND) is not known. This study tested the hypothesis that central ghrelin administration would inhibit splenic SND in anesthetized rats. Rats received intracerebroventricular (ICV) injections of ghrelin (1nmol/kg) or aCSF. Lumbar SND recordings provided a non-visceral nerve control. The ICV ghrelin administration significantly increased splenic and lumbar SND, whereas mean arterial pressure (MAP) was not altered. These findings provide fundamental information regarding the nature of sympathetic-immune interactions.
Collapse
Affiliation(s)
- Sivasai Balivada
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States; Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, United States
| | - Hitesh N Pawar
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States; Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, United States.
| | - Shawnee Montgomery
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States
| | - Michael J Kenney
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States; Department of Biological Sciences, College of Science, University of Texas at El Paso, El Paso, TX 79968, United States
| |
Collapse
|
37
|
Reardon C. Neuro-immune interactions in the cholinergic anti-inflammatory reflex. Immunol Lett 2016; 178:92-6. [PMID: 27542331 DOI: 10.1016/j.imlet.2016.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/12/2016] [Accepted: 08/15/2016] [Indexed: 10/21/2022]
Abstract
Communication between the nervous and immune systems can significantly alter immune cell function in a number of inflammatory diseases. Elegant studies have defined a basic functional circuit in a "cholinergic anti-inflammatory pathway" that highlights a unique role for the vagus nerve, and has brought about a resurgence in the field of neuro-immunology. This research has further identified that in addition to tonic signals that can restrain immune cell activation; the anti-inflammatory reflex arc is amiable to targeted stimulation as a therapeutic modality. The success of vagal electrical neural stimulation in a plethora of pre-clinical inflammation models has spurred the development of "electroceuticals" or neurostimulatory devices in the treatment of chronic inflammation. This development has begun despite addressing of fundamental questions such as the functional neural circuitry being crudely mapped and unresolved mechanisms of action of acetylcholine on target immune cells. Perhaps fortuitously, rapid advances in neuroscience techniques may allow us to begin to answer some of these longstanding questions and clarify recent controversies.
Collapse
Affiliation(s)
- Colin Reardon
- University of California Davis, School of Veterinary Medicine, Department of Anatomy, Physiology, and Cell Biology, 1089 Veterinary Medicine Drive, VM3B, Room 2007, Davis, CA 95616, United States.
| |
Collapse
|
38
|
Hua S. Neuroimmune Interaction in the Regulation of Peripheral Opioid-Mediated Analgesia in Inflammation. Front Immunol 2016; 7:293. [PMID: 27532001 PMCID: PMC4970451 DOI: 10.3389/fimmu.2016.00293] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/20/2016] [Indexed: 12/12/2022] Open
Abstract
Peripheral immune cell-mediated analgesia in inflammation is an important endogenous mechanism of pain control. Opioid receptors localized on peripheral sensory nerve terminals are activated by endogenous opioid peptides released from immune cells to produce significant analgesia. Following transendothelial migration of opioid-containing leukocytes into peripheral sites of inflammation, opioid peptides are released into a harsh milieu associated with an increase in temperature, low pH, and high proteolytic activity. Together, this microenvironment has been suggested to increase the activity of opioid peptide metabolism. Therefore, the proximity of immune cells and nerve fibers may be essential to produce adequate analgesic effects. Close associations between opioid-containing immune cells and peripheral nerve terminals have been observed. However, it is not yet determined whether these immune cells actually form synaptic-like contacts with peripheral sensory terminals and/or whether they secrete opioids in a paracrine manner. This review will provide novel insight into the peripheral mechanisms of immune-derived analgesia in inflammation, in particular, the importance of direct interactions between immune cells and the peripheral nervous system.
Collapse
Affiliation(s)
- Susan Hua
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
39
|
A mechanistic approach for modulation of chlorpyrifos-induced toxicity in human lymphocytes by melatonin, coenzyme Q10, and vinpocetine. Hum Exp Toxicol 2016; 35:839-50. [DOI: 10.1177/0960327115607945] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Chlorpyrifos (CP) is an organophosphorus pesticide that induces oxidative stress through the production of free radicals and depletes intracellular antioxidant reserves. In this study, the efficacy of three antioxidants (melatonin, coenzyme Q10 (CoQ10), and vinpocetine) on alleviation of toxic effects of CP was evaluated. Materials and Methods: Cytotoxicity of CP, in the presence or absence of effective doses of melatonin, CoQ10, and vinpocetine, was determined in human peripheral blood lymphocytes after 72-h exposure. The levels of acetylcholinesterase (AChE) activity along with tumor necrosis factor α (TNF-α), as inflammatory index, were measured. Further, the viability and oxidative stress markers including cellular mitochondrial activity, cell death modes (apoptosis vs. necrosis), total antioxidant power (TAP), total thiol molecules (TTM), lipid peroxidation (LPO), and myeloperoxidase (MPO) activity were measured. Results: CoQ10 and also the combination of the three antioxidants were the most notable in opposing toxicity of CP and led to increasing TAP and TTM; improvement of AChE activity; and lowering LPO, MPO, TNF-α, and apoptosis compared to CP alone. Conclusion: CP toxicity overwhelms the intracellular antioxidant defense mechanisms. Exogenous supplementation with antioxidants, such as the ones we have investigated, seems to be effective in the prevention of cytotoxicity of CP.
Collapse
|
40
|
Superior Cervical Ganglia Neurons Induce Foxp3+ Regulatory T Cells via Calcitonin Gene-Related Peptide. PLoS One 2016; 11:e0152443. [PMID: 27022966 PMCID: PMC4811438 DOI: 10.1371/journal.pone.0152443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 03/14/2016] [Indexed: 01/21/2023] Open
Abstract
The nervous and immune systems communicate bidirectionally, utilizing diverse molecular signals including cytokines and neurotransmitters to provide an integrated response to changes in the body’s internal and external environment. Although, neuro-immune interactions are becoming better understood under inflammatory circumstances and it has been evidenced that interaction between neurons and T cells results in the conversion of encephalitogenic T cells to T regulatory cells, relatively little is known about the communication between neurons and naïve T cells. Here, we demonstrate that following co-culture of naïve CD4+ T cells with superior cervical ganglion neurons, the percentage of Foxp3 expressing CD4+CD25+ cells significantly increased. This was mediated in part by immune-regulatory cytokines TGF-β and IL-10, as well as the neuropeptide calcitonin gene-related peptide while vasoactive intestinal peptide was shown to play no role in generation of T regulatory cells. Additionally, T cells co-cultured with neurons showed a decrease in the levels of pro-inflammatory cytokine IFN-γ released upon in vitro stimulation. These findings suggest that the generation of Tregs may be promoted by naïve CD4+ T cell: neuron interaction through the release of neuropeptide CGRP.
Collapse
|
41
|
Kim H, Kwon YA, Ahn IS, Kim S, Kim S, Jo SA, Kim DK. Overexpression of Cell Cycle Proteins of Peripheral Lymphocytes in Patients with Alzheimer's Disease. Psychiatry Investig 2016; 13:127-34. [PMID: 26766955 PMCID: PMC4701676 DOI: 10.4306/pi.2016.13.1.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 05/19/2015] [Accepted: 06/13/2015] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Biological markers for Alzheimer's disease (AD) will help clinicians make objective diagnoses early during the course of dementia. Previous studies have suggested that cell cycle dysregulation begins earlier than the onset of clinical manifestations in AD. METHODS We examined the lymphocyte expression of cell cycle proteins in AD patients, dementia controls (DC), and normal controls (NC). One-hundred seventeen subjects (36 AD, 31 DC, and 50 NC) were recruited. The cell cycle proteins CDK2, CDK4, CDK6, cyclin B, and cyclin D were measured in peripheral lymphocytes. Cell cycle protein expression in the three groups was compared after adjusting for age and sex. RESULTS The levels of cell cycle proteins CDK2, CDK4, CDK6, cyclin B, and cyclin D were significantly higher in AD patients than in the NC subjects. The DC group manifested intermediate levels of cell cycle proteins compared with the AD patients and the NC subjects. The present study indicates that cell cycle proteins are upregulated in the peripheral lymphocytes of AD patients. CONCLUSION Cell cycle dysregulation in peripheral lymphocytes may present a promising starting point for identifying peripheral biomarkers of AD.
Collapse
Affiliation(s)
- Hyeran Kim
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Ah Kwon
- Center for Clinical Research, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Inn Sook Ahn
- Center for Clinical Research, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Sangha Kim
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seonwoo Kim
- Biostatistics Unit, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Sangmee Ahn Jo
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Doh Kwan Kim
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Clinical Research, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| |
Collapse
|
42
|
Case AJ, Zimmerman MC. Sympathetic-mediated activation versus suppression of the immune system: consequences for hypertension. J Physiol 2015; 594:527-36. [PMID: 26830047 DOI: 10.1113/jp271516] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/17/2015] [Indexed: 01/08/2023] Open
Abstract
It is generally well-accepted that the immune system is a significant contributor in the pathogenesis of hypertension. Specifically, activated and pro-inflammatory T-lymphocytes located primarily in the vasculature and kidneys appear to have a causal role in exacerbating elevated blood pressure. It has been proposed that increased sympathetic nerve activity and noradrenaline outflow associated with hypertension may be primary contributors to the initial activation of the immune system early in the disease progression. However, it has been repeatedly demonstrated in many different human and experimental diseases that sympathoexcitation is immunosuppressive in nature. Moreover, human hypertensive patients have demonstrated increased susceptibility to secondary immune insults like infections. Thus, it is plausible, and perhaps even likely, that in diseases like hypertension, specific immune cells are activated by increased noradrenaline, while others are in fact suppressed. We propose a model in which this differential regulation is based upon activation status of the immune cell as well as the resident organ. With this, the concept of global immunosuppression is obfuscated as a viable target for hypertension treatment, and we put forth the concept of focused organ-specific immunotherapy as an alternative option.
Collapse
Affiliation(s)
- Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Centre, Omaha, NE, USA
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Centre, Omaha, NE, USA
| |
Collapse
|
43
|
Abstract
Over the past decades evidence has accumulated clearly demonstrating a pivotal role for the sympathetic nervous system (SNS) and its neurotransmitters in regulating inflammation. The first part of this review provides the reader with an overview showing that the interaction of the SNS with the immune system to control inflammation is strongly context-dependent (for example, depending on the activation state of the immune cell or neuro-transmitter concentration). In the second part we focus on autoimmune arthritis as a well investigated example for sympathetically controlled inflammation to show that the SNS and catecholamines play a differential role depending on the time point of ongoing disease. A model will be developed to explain the proinflammatory effects of the SNS in the early phase and the anti-inflammatory effects of catecholamines in the later phase of autoimmune arthritis. In the final part, a conceptual framework is discussed that shows that a major purpose of increased SNS activity is nourishment of a continuously activated immune system at a systemic level using energy-rich fuels (glucose, amino acids, lipids), while uncoupling from central nervous regulation occurs at sites of inflammation by repulsion of sympathetic fibers and local adrenoceptor regulation. This creates zones of ‘permitted local inflammation’. However, if this ‘inflammatory configuration’ persists and is strong, as in autoimmunity, the effects are detrimental because of the resultant chronic catabolic state, leading to cachexia, high blood pressure, insulin resistance, and increased cardiovascular mortality, and so on. Today, the challenge is to translate this conceptual knowledge into clinical benefit.
Collapse
|
44
|
Bowyer JF, Tranter KM, Hanig JP, Crabtree NM, Schleimer RP, George NI. Evaluating the Stability of RNA-Seq Transcriptome Profiles and Drug-Induced Immune-Related Expression Changes in Whole Blood. PLoS One 2015; 10:e0133315. [PMID: 26177368 PMCID: PMC4503719 DOI: 10.1371/journal.pone.0133315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/24/2015] [Indexed: 12/01/2022] Open
Abstract
Methods were developed to evaluate the stability of rat whole blood expression obtained from RNA sequencing (RNA-seq) and assess changes in whole blood transcriptome profiles in experiments replicated over time. Expression was measured in globin-depleted RNA extracted from the whole blood of Sprague-Dawley rats, given either saline (control) or neurotoxic doses of amphetamine (AMPH). The experiment was repeated four times (paired control and AMPH groups) over a 2-year span. The transcriptome of the control and AMPH-treated groups was evaluated on: 1) transcript levels for ribosomal protein subunits; 2) relative expression of immune-related genes; 3) stability of the control transcriptome over 2 years; and 4) stability of the effects of AMPH on immune-related genes over 2 years. All, except one, of the 70 genes that encode the 80s ribosome had levels that ranked in the top 5% of all mean expression levels. Deviations in sequencing performance led to significant changes in the ribosomal transcripts. The overall expression profile of immune-related genes and genes specific to monocytes, T-cells or B-cells were well represented and consistent within treatment groups. There were no differences between the levels of ribosomal transcripts in time-matched control and AMPH groups but significant differences in the expression of immune-related genes between control and AMPH groups. AMPH significantly increased expression of some genes related to monocytes but down-regulated those specific to T-cells. These changes were partially due to changes in the two types of leukocytes present in blood, which indicate an activation of the innate immune system by AMPH. Thus, the stability of RNA-seq whole blood transcriptome can be verified by assessing ribosomal protein subunits and immune-related gene expression. Such stability enables the pooling of samples from replicate experiments to carry out differential expression analysis with acceptable power.
Collapse
Affiliation(s)
- John F. Bowyer
- Division of Neurotoxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| | - Karen M. Tranter
- Division of Neurotoxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Joseph P. Hanig
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Nathaniel M. Crabtree
- Division of Neurotoxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Robert P. Schleimer
- Division of Allergy and Immunology, Northwestern Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Nysia I. George
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, Arkansas, United States of America
| |
Collapse
|
45
|
Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front Neurosci 2015; 9:172. [PMID: 26089771 PMCID: PMC4453474 DOI: 10.3389/fnins.2015.00172] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
As we age, the immune system undergoes a process of senescence accompanied by the increased production of proinflammatory cytokines, a chronic subclinical condition named as “inflammaging”. Emerging evidence from human and experimental models suggest that immune senescence also affects the central nervous system and promotes neuronal dysfunction, especially within susceptible neuronal populations. In this review we discuss the potential role of immune aging, inflammation and metabolic derangement in neurological diseases. The discovery of novel therapeutic strategies targeting age-linked inflammation may promote healthy brain aging and the treatment of neurodegenerative as well as neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michela Deleidi
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Madeline Jäggle
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Graziella Rubino
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen Tübingen, Germany
| |
Collapse
|
46
|
de Jonge WJ. Neuronal Regulation of Mucosal Immune Responses. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00046-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Estrogen modulates β2-adrenoceptor-induced cell-mediated and inflammatory immune responses through ER-α involving distinct intracellular signaling pathways, antioxidant enzymes, and nitric oxide. Cell Immunol 2014; 292:1-8. [DOI: 10.1016/j.cellimm.2014.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/06/2014] [Indexed: 11/16/2022]
|
48
|
Abstract
The interaction between the sympathetic nervous system and the immune system has been documented over the last several decades. In this review, the neuroanatomical, cellular, and molecular evidence for neuroimmune regulation in the maintenance of immune homeostasis will be discussed, as well as the potential impact of neuroimmune dysregulation in health and disease.
Collapse
Affiliation(s)
- Caroline J Padro
- The Biomedical Sciences Graduate Program, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States.
| | - Virginia M Sanders
- The Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States; The Institute of Behavioral Medicine Research, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States.
| |
Collapse
|
49
|
CNS inflammation and bone marrow neuropathy in type 1 diabetes. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 183:1608-20. [PMID: 24160325 DOI: 10.1016/j.ajpath.2013.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 06/28/2013] [Accepted: 07/12/2013] [Indexed: 02/06/2023]
Abstract
By using pseudorabies virus expressing green fluorescence protein, we found that efferent bone marrow-neural connections trace to sympathetic centers of the central nervous system in normal mice. However, this was markedly reduced in type 1 diabetes, suggesting a significant loss of bone marrow innervation. This loss of innervation was associated with a change in hematopoiesis toward generation of more monocytes and an altered diurnal release of monocytes in rodents and patients with type 1 diabetes. In the hypothalamus and granular insular cortex of mice with type 1 diabetes, bone marrow-derived microglia/macrophages were activated and found at a greater density than in controls. Infiltration of CD45(+)/CCR2(+)/GR-1(+)/Iba-1(+) bone marrow-derived monocytes into the hypothalamus could be mitigated by treatment with minocycline, an anti-inflammatory agent capable of crossing the blood-brain barrier. Our studies suggest that targeting central inflammation may facilitate management of microvascular complications.
Collapse
|
50
|
O'Connor TG, Moynihan JA, Caserta MT. Annual research review: The neuroinflammation hypothesis for stress and psychopathology in children--developmental psychoneuroimmunology. J Child Psychol Psychiatry 2014; 55:615-31. [PMID: 24372371 PMCID: PMC4029900 DOI: 10.1111/jcpp.12187] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2013] [Indexed: 12/14/2022]
Abstract
Experimental animal and adult human data suggest that stress exposure is associated with alterations in immune system function that may underlie increased susceptibility to disease and behavioral disorders. The implications of these data for child psychology and psychiatry are not yet clear. The current review seeks to distil and translate the relevant animal and adult human work to children to advance a developmental model of psychoneuroimmunology. In addition to reviewing key specific findings, we consider biological/conceptual models and technical aspects of psychoneuroimmunology work in pediatric populations, and outline the rationales and advantages of integrating hypotheses concerning neuroinflammation in developmental studies of psychopathology.
Collapse
|