1
|
Nayak TK, Parasania D, Tilley DG. Adrenergic orchestration of immune cell dynamics in response to cardiac stress. J Mol Cell Cardiol 2024; 196:115-124. [PMID: 39303854 DOI: 10.1016/j.yjmcc.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Immune cells contribute approximately 5-10 % of the heart's total cell population, including several myeloid cell and lymphocyte cell subsets, which, despite their relatively small percentages, play important roles in cardiac homeostasis and remodeling responses to various forms of injury and long-term stress. Pathological cardiac stress activates the sympathetic nervous system (SNS), resulting in the release of the catecholamines epinephrine and norepinephrine either systemically or from sympathetic nerve terminals within various lymphoid organs. Acting at α- or β-adrenergic receptors (αAR, βAR), catecholamines regulate immune cell hematopoiesis, egress and migration in response to stress. Classically, αAR stimulation tends to promote inflammatory responses while βAR stimulation has typically been shown to be immunosuppressive, though the effects can be nuanced depending on the immune cells subtype, the site of regulation and pathophysiological context. Herein, we will discuss several facets of SNS-mediated regulation of immune cells and their response to cardiac stress, including: catecholamine response to cardiovascular stress and action at their receptors, adrenergic regulation of hematopoiesis, immune cell retention and release from the bone marrow, adrenergic regulation of splenic immune cells and their retention, as well as adrenergic regulation of immune cell recruitment to the injured heart, including neutrophils, monocytes and macrophages. A particular focus will be given to βAR-mediated effects on myeloid cells in response to acute or chronic cardiac stress.
Collapse
Affiliation(s)
- Tapas K Nayak
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Dev Parasania
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
2
|
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
|
3
|
Daria C, Lancaster G, Murphy AJ, Henderson LA, Dawood T, Macefield VG. Relationship between muscle sympathetic nerve activity and rapid increases in circulating leukocytes during experimental muscle pain. Clin Auton Res 2024; 34:227-231. [PMID: 38227276 DOI: 10.1007/s10286-023-01012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Affiliation(s)
- Camille Daria
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Graeme Lancaster
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Luke A Henderson
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tye Dawood
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Vaughan G Macefield
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Neuroscience, Monash University Central Clinical School, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
4
|
Yuan X, Wan L, Chen Z, Long Z, Chen D, Liu P, Fu Y, Zhu S, Peng L, Qiu R, Tang B, Jiang H. Peripheral Inflammatory and Immune Landscape in Multiple System Atrophy: A Cross-Sectional Study. Mov Disord 2024; 39:391-399. [PMID: 38155513 DOI: 10.1002/mds.29674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/16/2023] [Accepted: 11/09/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Neuroinflammation might contribute to the pathogenesis of multiple systemic atrophy (MSA). However, specific alterations in the peripheral inflammatory and immune profiles of patients with MSA remain unclear. OBJECTIVES To determine the peripheral inflammatory and immune profiles of patients with MSA and their potential value as biomarkers for facilitating clinical diagnosis and monitoring disease severity. METHODS This cross-sectional study included 235, 240, and 235 patients with MSA, patients with Parkinson's disease (PD), and healthy controls (HCs), respectively. Inflammatory and immune parameters were measured in peripheral blood, differences between groups were assessed, and clusters were analyzed. Associations between the parameters and clinical characteristics of MSA were assessed using Spearman and partial correlation analyses. RESULTS Significant differences were observed especially in monocytes, neutrophils-to-lymphocyte ratio (NLR) and neutrophils-to-lymphocyte ratio (MPV) between MSA patients and HCs (P < 0.01). Monocytes and uric acid (UA) levels were also significantly different between the MSA and PD patients (P < 0.05). The combination of NLR and MPV distinguished MSA-P patients from HCs (areas under the curve = 0.824). In addition, complement components C4 and C3 were significantly correlated with the Scale Outcomes in PD for Autonomic Symptoms and Wexner scale, whereas immunoglobulin G (IgG) was significantly correlated with scores of Unified Multiple System Atrophy Rating Scale (P < 0.05). CONCLUSIONS In MSA patients, monocytes, NLR and MPV might serve as potential diagnostic biomarkers, whereas MLR, C3, C4, and IgG significantly correlate with disease severity. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xinrong Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Panyan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - You Fu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Khanmammadova N, Islam S, Sharma P, Amit M. Neuro-immune interactions and immuno-oncology. Trends Cancer 2023; 9:636-649. [PMID: 37258398 PMCID: PMC10524972 DOI: 10.1016/j.trecan.2023.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/24/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023]
Abstract
The nervous system is an important component of the tumor microenvironment (TME), driving tumorigenesis and tumor progression. Neuronal cues (e.g., neurotransmitters and neuropeptides) in the TME cause phenotypic changes in immune cells, such as increased exhaustion and inhibition of effector cells, which promote immune evasion and cancer progression. Two types of immune regulation by tumor-associated nerves are discussed in this review: regulation via neuronal stimuli (i.e., by neural transmission) and checkpoint-mediated neuronal immune regulation. The latter occurs via the expression of immune checkpoints on the membranes of intratumoral nerves and glial cells. Here, we summarize novel findings regarding the neuroimmune circuits in the tumor milieu, while emphasizing the potential targets of new and affordable anticancer therapeutic approaches.
Collapse
Affiliation(s)
- Narmina Khanmammadova
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shajedul Islam
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunobiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Houston Health Science Center Graduate School of Biomedical Sciences, Department of Neuroscience, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
6
|
Dervishi E, Hailemariam D, Goldansaz SA, Ametaj BN. Early-Life Exposure to Lipopolysaccharide Induces Persistent Changes in Gene Expression Profiles in the Liver and Spleen of Female FVB/N Mice. Vet Sci 2023; 10:445. [PMID: 37505851 PMCID: PMC10384579 DOI: 10.3390/vetsci10070445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
The objective of this study was to investigate how subcutaneous (sc) lipopolysaccharide (LPS) administration affects the gene expression profiles of insulin signaling as well as innate and adaptive immunity genes in mouse livers and spleens. FVB/N female mice were randomly assigned to one of two treatment groups at 5 weeks of age: (1) a six-week subcutaneous injection of saline at 11 μL/h (control-CON), or (2) a six-week subcutaneous injection of LPS from Escherichia coli 0111:B4 at 0.1 μg/g body weight at 11 μL/h. At 106 weeks (i.e., 742 days) after the last treatment, mice were euthanized. Following euthanasia, liver and spleen samples were collected, snap frozen, and stored at -80 °C until gene expression profiling. LPS upregulated nine genes in the liver, according to the findings (Pparg, Frs3, Kras, Raf1, Gsk3b, Rras2, Hk2, Pik3r2, and Myd88). With a 4.18-fold increase over the CON group, Pparg was the most up-regulated gene in the liver. Based on the annotation cluster analysis, LPS treatment upregulated liver genes which are involved in pathways associated with hepatic steatosis, B- and T-cell receptor signaling, chemokine signaling, as well as other types of cancers such as endometrial cancer, prostate cancer, and colorectal cancer. LPS increased the spleen expression of Ccl11, Ccl25, Il6, Cxcl5, Pparg, Tlr4, Nos2, Cxcl11, Il1a, Ccl17, and Fcgr3, all of which are involved in innate and adaptive immune responses and the regulation of cytokine production. Furthermore, functional analysis revealed that cytokine-cytokine receptor interaction and chemokine signaling pathways were the most enriched in LPS-treated mice spleen tissue. Our findings support the notion that early-life LPS exposure can result in long-term changes in gene expression profiling in the liver and spleen tissues of FVB/N female mice.
Collapse
Affiliation(s)
- Elda Dervishi
- Department of Agricultural Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Dagnachew Hailemariam
- Department of Agricultural Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Seyed Ali Goldansaz
- Department of Agricultural Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Burim N Ametaj
- Department of Agricultural Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
7
|
Mposhi A, Turner JD. How can early life adversity still exert an effect decades later? A question of timing, tissues and mechanisms. Front Immunol 2023; 14:1215544. [PMID: 37457711 PMCID: PMC10348484 DOI: 10.3389/fimmu.2023.1215544] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to any number of stressors during the first 1000 days from conception to age 2 years is important in shaping an individual's life trajectory of health and disease. Despite the expanding range of stressors as well as later-life phenotypes and outcomes, the underlying molecular mechanisms remain unclear. Our previous data strongly suggests that early-life exposure to a stressor reduces the capacity of the immune system to generate subsequent generations of naïve cells, while others have shown that, early life stress impairs the capacity of neuronal stem cells to proliferate as they age. This leads us to the "stem cell hypothesis" whereby exposure to adversity during a sensitive period acts through a common mechanism in all the cell types by programming the tissue resident progenitor cells. Furthermore, we review the mechanistic differences observed in fully differentiated cells and suggest that early life adversity (ELA) may alter mitochondria in stem cells. This may consequently alter the destiny of these cells, producing the lifelong "supply" of functionally altered fully differentiated cells.
Collapse
|
8
|
Pongratz G, Straub RH. Chronic Effects of the Sympathetic Nervous System in Inflammatory Models. Neuroimmunomodulation 2023; 30:113-134. [PMID: 37231902 DOI: 10.1159/000530969] [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: 01/16/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The immune system is embedded in a network of regulatory systems to keep homeostasis in case of an immunologic challenge. Neuroendocrine immunologic research has revealed several aspects of these interactions over the past decades, e.g., between the autonomic nervous system and the immune system. This review will focus on evidence revealing the role of the sympathetic nervous system (SNS) in chronic inflammation, like colitis, multiple sclerosis, systemic sclerosis, lupus erythematodes, and arthritis with a focus on animal models supported by human data. A theory of the contribution of the SNS in chronic inflammation will be presented that spans these disease entities. One major finding is the biphasic nature of the sympathetic contribution to inflammation, with proinflammatory effects until the point of disease outbreak and mainly anti-inflammatory influence thereafter. Since sympathetic nerve fibers are lost from sites of inflammation during inflammation, local cells and immune cells achieve the capability to endogenously produce catecholamines to fine-tune the inflammatory response independent of brain control. On a systemic level, it has been shown across models that the SNS is activated in inflammation as opposed to the parasympathetic nervous system. Permanent overactivity of the SNS contributes to many of the known disease sequelae. One goal of neuroendocrine immune research is defining new therapeutic targets. In this respect, it will be discussed that at least in arthritis, it might be beneficial to support β-adrenergic and inhibit α-adrenergic activity besides restoring autonomic balance. Overall, in the clinical setting, we now need controlled interventional studies to successfully translate the theoretical knowledge into benefits for patients.
Collapse
Affiliation(s)
- Georg Pongratz
- Department of Gastroenterology, Division of Rheumatology and Clinical Immunology, St. John of God Hospital, Regensburg, Germany
- Medical Faculty of the University of Regensburg, Regensburg, Germany
| | - Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrino-Immunology, Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
9
|
Wulf MJ, Tom VJ. Consequences of spinal cord injury on the sympathetic nervous system. Front Cell Neurosci 2023; 17:999253. [PMID: 36925966 PMCID: PMC10011113 DOI: 10.3389/fncel.2023.999253] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Spinal cord injury (SCI) damages multiple structures at the lesion site, including ascending, descending, and propriospinal axons; interrupting the conduction of information up and down the spinal cord. Additionally, axons associated with the autonomic nervous system that control involuntary physiological functions course through the spinal cord. Moreover, sympathetic, and parasympathetic preganglionic neurons reside in the spinal cord. Thus, depending on the level of an SCI, autonomic function can be greatly impacted by the trauma resulting in dysfunction of various organs. For example, SCI can lead to dysregulation of a variety of organs, such as the pineal gland, the heart and vasculature, lungs, spleen, kidneys, and bladder. Indeed, it is becoming more apparent that many disorders that negatively affect quality-of-life for SCI individuals have a basis in dysregulation of the sympathetic nervous system. Here, we will review how SCI impacts the sympathetic nervous system and how that negatively impacts target organs that receive sympathetic innervation. A deeper understanding of this may offer potential therapeutic insight into how to improve health and quality-of-life for those living with SCI.
Collapse
Affiliation(s)
| | - Veronica J. Tom
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Jin X, Wang X, Sun J, Tan W, Zhang G, Han J, Xie M, Zhou L, Yu Z, Xu T, Wang C, Wang Y, Zhou X, Jiang H. Subthreshold splenic nerve stimulation prevents myocardial Ischemia-Reperfusion injury via neuroimmunomodulation of proinflammatory factor levels. Int Immunopharmacol 2023; 114:109522. [PMID: 36502595 DOI: 10.1016/j.intimp.2022.109522] [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: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Clinical outcomes following myocardial ischemia-reperfusion (I/R) injury are strongly related to the intensity and duration of inflammation. The splenic nerve (SpN) is indispensable for the anti-inflammatory reflex. This study aimed to investigate whether splenic nerve stimulation (SpNS) plays a cardioprotective role in myocardial I/R injury and the potential underlying mechanism. METHODS Sprague-Dawley rats were randomly divided into four groups: sham group, I/R group, SpNS group, and I/R plus SpNS group. The highest SpNS intensity that did not influence heart rate was identified, and SpNS at this intensity was used as the subthreshold stimulus. Continuous subthreshold SpNS was applied for 1 h before ligation of the left coronary artery for 45 min. After 72 h of reperfusion, samples were collected for analysis. RESULTS SpN activity and splenic concentrations of cholinergic anti-inflammatory pathway (CAP)-related neurotransmitters were significantly increased by SpNS. The infarct size, oxidative stress, sympathetic tone, and the levels of proinflammatory cytokines, including TNF-α, IL-1β, and IL-6, were significantly reduced in rats subjected to subthreshold SpNS after myocardial I/R injury compared with those subjected to I/R injury alone. CONCLUSIONS Subthreshold SpNS ameliorates myocardial damage, the inflammatory response, and cardiac remodelling induced by myocardial I/R injury via neuroimmunomodulation of proinflammatory factor levels. SpNS is a potential therapeutic strategy for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Xiaoxing Jin
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Guocheng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Jiapeng Han
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Zhiyao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Tianyou Xu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Changyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| |
Collapse
|
11
|
Floreste FR, Titon B, Titon SCM, Muxel SM, Figueiredo ACD, Gomes FR, Assis VR. Liver vs. spleen: Time course of organ-dependent immune gene expression in an LPS-stimulated toad (Rhinella diptycha). Comp Biochem Physiol B Biochem Mol Biol 2023; 263:110784. [PMID: 35931313 DOI: 10.1016/j.cbpb.2022.110784] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 10/16/2022]
Abstract
The inflammatory response comprises highly orchestrated events that are conserved amongst vertebrate groups. Hepatic and splenic cytokines are major mediators of the systemic inflammatory processes. However, the liver is still neglected as an immune organ in amphibians. This study reports organ-dependent gene expression using an anuran model. We tracked mRNA levels of immune proteins [C1s (subcomponent S of the complement protein 1), IFN-γ, IL-1β, IL-6, and IL-10] at four time-points (1 h, 3 h, 6 h, and 18 h post-injection) in spleens and livers of intraperitoneal LPS-challenged (2 mg/kg) adult male toads (Rhinella diptycha) using independent samples. We found acute C1s up-regulation in the liver 1 h post-injection, with no treatment effect in the spleen. The LPS injection did not show any effect in splenic IFN-γ gene expression while eliciting only a marginal effect in the hepatic tissue. IL-1β was up-regulated in both organs, with the liver initially displaying early expression (1 h and 3 h) and the spleen taking over late expression (18 h). Both organs exhibited similar patterns for IL-6, with early up-regulation (1 h and 3 h) and late peak (18 h). Although IL-10 was early detected and up-regulated only in the liver, both organs showed up-regulation in 6 h and 18 h post-injection. Our results show an exclusive hepatic prominence in complement protein expression during the acute-phase response. Furthermore, hepatic pro-inflammatory cytokine expression was more pronounced in earliest time-points, while the spleen offers a slower and more consistent response overall. Our data provide an organ-integrative outlook into the initial hours of the inflammation in amphibians, confirming the liver's pivotal role as a regulator in the acute-phase of the inflammatory response in amphibians.
Collapse
Affiliation(s)
- Felipe R Floreste
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil.
| | - Braz Titon
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil
| | - Stefanny C M Titon
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil. https://twitter.com/StefannyTiton
| | - Sandra M Muxel
- Laboratory of Neuroimmunology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil. https://twitter.com/SandraMuxel
| | - Aymam C de Figueiredo
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil
| | - Fernando R Gomes
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil
| | - Vania R Assis
- Laboratory of Behavior and Evolutionary Physiology, Institute of Biosciences, Department of Physiology, University of São Paulo, São Paulo, Brazil. https://twitter.com/VaniaRAssis1
| |
Collapse
|
12
|
Yan JT, Zhu YZ, Liang L, Feng XY. NE-activated β 2-AR/β-arrestin 2/Src pathway mediates duodenal hyperpermeability induced by water-immersion restraint stress. Am J Physiol Cell Physiol 2023; 324:C133-C141. [PMID: 36440855 DOI: 10.1152/ajpcell.00412.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Stress causes a rapid spike in norepinephrine (NE) levels, leading to gastrointestinal dysfunction. NE reduces the expression of tight junctions (TJs) and aggravates intestinal mucosal damage, but the regulatory mechanism is still unclear. The present study aimed to investigate the molecular mechanisms underlying the regulation of stress-associated duodenal hyperpermeability by NE. Fluorescein isothiocyanate-dextran permeability, transepithelial resistance, immunofluorescence, Western blot, and high-performance liquid chromatography analysis were used in water-immersion restraint stress (WIRS) rats in this study. The results indicate that the duodenal permeability, degradation of TJs, mucosal NE, and β2-adrenergic receptor (β2-AR) increased in WIRS rats. The duodenal intracellular cyclic adenosine monophosphate levels were decreased, whereas the expression of β-arrestin 2 negatively regulates G protein-coupled receptors signaling, was significantly increased. Src recruitment was mediated by β-arrestin; thus, the levels of Src kinase activation were enhanced in WIRS rats. NE depletion, β2-AR, or β-arrestin 2 blockade significantly decreased mucosal permeability and increased TJs expression, suggesting improved mucosal barrier function. Moreover, NE induced an increased duodenal permeability of normal rats with activated β-arrestin 2/Src signaling, which was significantly inhibited by β2-AR blockade. The present findings demonstrate that the enhanced NE induced an increased duodenal permeability in WIRS rats through the activated β2-AR/β-arrestin 2/Src pathway. This study provides novel insight into the molecular mechanism underlying the regulation of NE on the duodenal mucosal barrier and a new target for treating duodenal ulcers induced by stress.
Collapse
Affiliation(s)
- Jing-Ting Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| | - Yin-Zhe Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Liang Liang
- Grade 2020 Pediatrics, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Xiao-Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Floreste FR, Titon B, Titon SCM, Muxel SM, Gomes FR, Assis VR. Time Course of Splenic Cytokine mRNA and Hormones during a Lipopolysaccharide-Induced Inflammation in Toads. Integr Comp Biol 2022; 62:1618-1628. [PMID: 35362514 DOI: 10.1093/icb/icac013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 01/05/2023] Open
Abstract
Inflammation comprises alterations in glucocorticoids (in amphibians, corticosterone-CORT) and melatonin (MEL) levels, two hormones with immunomodulatory effects on cytokine production in several vertebrates. Cytokines mediate inflammation progress differently depending on their function. While some are secreted during the acute phase of the immune response, others prevail during the resolution phase. Major efforts have been made to understand the interaction of endocrine mediators and cytokine production in endotherms, but little is known for ectotherms so far. Characterizing the stages of inflammation and their interplay with endocrine mediators is crucial for an assertive and integrative approach to amphibian physiology and ecoimmunology. Herein, we investigated CORT and MEL plasma levels as well as splenic cytokine (IL-1β, IL-6, and IL-10) mRNA levels during the progression of the inflammatory response in toads (Rhinella diptycha) in four time-points (1, 3, 6, and 18 h) after an immune challenge with lipopolysaccharide (LPS) using independent samples. Toads were responsive to LPS, with all hormones and cytokines affected by LPS. IL-1β and IL-6 were up-regulated after 1 h, but IL-1β decreased right after 3 h, while IL-6 sustained up-regulation throughout all time-points. IL-10 had not been detected until 6 h post-LPS-stimulation, when it showed up-regulation, along with a CORT increase at the same time-point. After 18 h, CORT levels were still high, and IL-1β was up-regulated again, along with up-regulated IL-6 and an IL-10 decrease. We also found positive correlations between IL-1β with IL-6 for LPS and saline groups. LPS-treated individuals showed an overall decrease in MEL plasma levels compared to saline counterparts. Our results showcase the early endocrine and molecular events of the amphibian immune response. We also report activation of the hypothalamus-pituitary-interrenal (HPI) axis during inflammation and increasing evidence for an immune-pineal axis to be described in amphibians.
Collapse
Affiliation(s)
- Felipe R Floreste
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| | - Braz Titon
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| | - Stefanny C M Titon
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| | - Sandra M Muxel
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| | - Fernando R Gomes
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| | - Vania R Assis
- Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, SP 05508090, Brazil
| |
Collapse
|
14
|
Li YW, Li W, Wang ST, Gong YN, Dou BM, Lyu ZX, Ulloa L, Wang SJ, Xu ZF, Guo Y. The autonomic nervous system: A potential link to the efficacy of acupuncture. Front Neurosci 2022; 16:1038945. [PMID: 36570846 PMCID: PMC9772996 DOI: 10.3389/fnins.2022.1038945] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
The autonomic nervous system (ANS) is a diffuse network that regulates physiological systems to maintain body homeostasis by integrating inputs from the internal and external environment, including the sympathetic, parasympathetic, and enteric nervous systems (ENS). Recent evidence suggests that ANS is one of the key neural pathways for acupuncture signal transduction, which has attracted worldwide attention in the acupuncture field. Here, we reviewed the basic and clinical research published in PubMed over the past 20 years on the effects of acupuncture on ANS regulation and homeostasis maintenance. It was found that acupuncture effectively alleviates ANS dysfunction-associated symptoms in its indications, such as migraine, depression, insomnia, functional dyspepsia, functional constipation. Acupuncture stimulation on some specific acupoints activates sensory nerve fibers, the spinal cord, and the brain. Using information integration and efferents from a complex network of autonomic nuclei of the brain, such as the insular cortex (IC), prefrontal cortex, anterior cingulate cortex (ACC), amygdala (AMG), hypothalamus, periaqueductal gray (PAG), nucleus tractus solitarius (NTS), ventrolateral medulla (VLM), nucleus ambiguus (AMB), acupuncture alleviates visceral dysfunction, inflammation via efferent autonomic nerves, and relieves pain and pain affect. The modulating pattern of sympathetic and parasympathetic nerves is associated with acupuncture stimulation on specific acupoints, intervention parameters, and disease models, and the relationships among them require further exploration. In conclusion, ANS is one of the therapeutic targets for acupuncture and mediates acupuncture's actions, which restores homeostasis. A systemic study is needed to determine the rules and mechanisms underlying the effects of acupoint stimulation on corresponding organs mediated by specific central nervous networks and the efferent ANS.
Collapse
Affiliation(s)
- Yan-Wei Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Song-Tao Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi-Nan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Bao-Min Dou
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhong-Xi Lyu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Luis Ulloa
- Department of Anesthesiology, Center for Perioperative Organ Protection, Duke University, Durham, NC, United States
| | - Shen-Jun Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Shen-Jun Wang,
| | - Zhi-Fang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Zhi-Fang Xu,
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,*Correspondence: Yi Guo,
| |
Collapse
|
15
|
Bouras M, Asehnoune K, Roquilly A. Immune modulation after traumatic brain injury. Front Med (Lausanne) 2022; 9:995044. [PMID: 36530909 PMCID: PMC9751027 DOI: 10.3389/fmed.2022.995044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/14/2022] [Indexed: 07/20/2023] Open
Abstract
Traumatic brain injury (TBI) induces instant activation of innate immunity in brain tissue, followed by a systematization of the inflammatory response. The subsequent response, evolved to limit an overwhelming systemic inflammatory response and to induce healing, involves the autonomic nervous system, hormonal systems, and the regulation of immune cells. This physiological response induces an immunosuppression and tolerance state that promotes to the occurrence of secondary infections. This review describes the immunological consequences of TBI and highlights potential novel therapeutic approaches using immune modulation to restore homeostasis between the nervous system and innate immunity.
Collapse
Affiliation(s)
- Marwan Bouras
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Karim Asehnoune
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| |
Collapse
|
16
|
Honke N, Wiest CJ, Pongratz G. β2-Adrenergic Receptor Expression and Intracellular Signaling in B Cells Are Highly Dynamic during Collagen-Induced Arthritis. Biomedicines 2022; 10:biomedicines10081950. [PMID: 36009497 PMCID: PMC9406045 DOI: 10.3390/biomedicines10081950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The sympathetic nervous system (SNS) has either a pro-inflammatory or anti-inflammatory effect, depending on the stage of arthritis. In the past, treatment of arthritic B cells with a β2-adrenergic receptor (β2-ADR) agonist has been shown to attenuate arthritis. In this study, the expression and signaling of β2-ADR in B cells during collagen-induced arthritis (CIA) were investigated to provide an explanation of why only B cells from arthritic mice are able to improve CIA. Splenic B cells were isolated via magnetic-activated cell sorting (MACS). Adrenergic receptors on B cells and intracellular β2-ADR downstream molecules (G protein-coupled receptor kinase 2 (GRK-2), β-Arrestin 2, p38 MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2) and cAMP response element-binding protein (CREB)) were analyzed at different time points in naïve and arthritic B cells with and without stimulation of β2-ADR agonist terbutaline by flow cytometry. β2-ADR-expressing B cells increase during CIA without a change in receptor density. Moreover, we observed a profound downregulation of GRK-2 shortly after induction of arthritis and an increase in β-Arrestin 2 only at late stage of arthritis. The second messengers studied (p38, ERK1/2 and CREB) followed a biphasic course, characterized by a reduction at onset and an increase in established arthritis. Stimulation of CIA B cells with the β-ADR agonist terbutaline increased pp38 MAPK independent of the timepoint, while pERK1/2 and pCREB were enhanced only in the late phase of arthritis. The phosphorylation of p38 MAPK, ERK1/2 and CREB in the late phase of arthritis was associated with increased IL-10 produced by B10 cells. The change of β2-ADR expression and signaling during sustained inflammation might be an integral part of the switch from pro- to anti-inflammatory action of sympathetic mechanisms in late arthritis.
Collapse
Affiliation(s)
- Nadine Honke
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: (N.H.); (G.P.); Tel.: +49-(0)-2118106149 (N.H.); +49-(0)-9405-18-1078 (G.P.)
| | - Clemens J. Wiest
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Georg Pongratz
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
- Center for Rheumatologic Rehabilitation, Asklepios Clinic, 93077 Bad Abbach, Germany
- Medical Faculty of the University of Regensburg, 93053 Regensburg, Germany
- Correspondence: (N.H.); (G.P.); Tel.: +49-(0)-2118106149 (N.H.); +49-(0)-9405-18-1078 (G.P.)
| |
Collapse
|
17
|
Kwak SB, Kim SJ, Kim J, Kang YL, Ko CW, Kim I, Park JW. Tumor regionalization after surgery: Roles of the tumor microenvironment and neutrophil extracellular traps. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:720-729. [PMID: 35764882 PMCID: PMC9256747 DOI: 10.1038/s12276-022-00784-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 11/09/2022]
Abstract
Surgery is unanimously regarded as the primary strategy to cure solid tumors in the early stages but is not always used in advanced cases. However, tumor surgery must be carefully considered because the risk of metastasis could be increased by the surgical procedure. Tumor surgery may result in a deep wound, which induces many biological responses favoring tumor metastasis. In particular, NETosis, which is the process of forming neutrophil extracellular traps (NETs), has received attention as a risk factor for surgery-induced metastasis. To reduce cancer mortality, researchers have made efforts to prevent secondary metastasis after resection of the primary tumor. From this point of view, a better understanding of surgery-induced metastasis might provide new strategies for more effective and safer surgical approaches. In this paper, recent insights into the surgical effects on metastasis will be reviewed. Moreover, in-depth opinions about the effects of NETs on metastasis will be discussed. Therapies that limit the formation of web-like structures formed by white cells known as neutrophils may lower the risk of cancer spread (metastasis) following surgical tumor removal. Removing solid tumors remains a key cancer treatment, but in some cases surgery itself increases the risk of metastasis. Jong-Wan Park at Seoul National University, South Korea, and co-workers reviewed current understanding of metastasis following surgery. Surgical removal destroys the architecture supporting cancer cells but this can release tumor cells into blood vessels. The stress of deep wounds also affects immune responses, most notably neutrophil extracellular traps (NETs), web-like structures formed by neutrophils to trap and kill pathogens. NETs have previously been implicated in metastasis. In a post-surgical environment enriched in neutrophils and pro-inflammatory cytokines, NET formation may help cancer cells thrive, promoting metastasis.
Collapse
Affiliation(s)
- Su-Bin Kwak
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Sang Jin Kim
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Ye-Lim Kang
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Chang Woo Ko
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Iljin Kim
- Department of Pharmacology, Inha University College of Medicine, Inha-ro, Michuhol-gu, Incheon, 22212, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
18
|
Grant CE, Flis A, Ryan BM. Understanding the Role of Dopamine in Cancer: Past, Present, and Future. Carcinogenesis 2022; 43:517-527. [PMID: 35616105 DOI: 10.1093/carcin/bgac045] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 05/12/2022] [Accepted: 05/23/2022] [Indexed: 11/14/2022] Open
Abstract
Dopamine (DA, 3-hydroxytyramine) is member of the catecholamine family and is classically characterized according to its role in the central nervous system as a neurotransmitter. In recent decades, many novel and intriguing discoveries have been made about the peripheral expression of DA receptors (DRs) and the role of DA signaling in both normal and pathological processes. Drawing from decades of evidence suggesting a link between DA and cancer, the DA pathway (DAP) has recently emerged as a potential target in antitumor therapies. Due to the onerous, expensive, and frequently unsuccessful nature of drug development, the repurposing of dopaminergic drugs for cancer therapy has the potential to greatly benefit patients and drug developers alike. However, the lack of clear mechanistic data supporting the direct involvement of DRs and their downstream signaling components in cancer represents an ongoing challenge that has limited the translation of these drugs to the clinic. Despite this, the breadth of evidence linking DA to cancer and non-tumor cells in the tumor microenvironment (TME) justifies further inquiry into the potential applications of this treatment modality in cancer. Herein, we review the literature characterizing the interplay between the DA signaling axis and cancer, highlighting key findings, and then propose rational lines of investigation to follow.
Collapse
Affiliation(s)
- Christopher E Grant
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Amy Flis
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
19
|
Rudzki S. Is PTSD an Evolutionary Survival Adaptation Initiated by Unrestrained Cytokine Signaling and Maintained by Epigenetic Change? Mil Med 2022; 188:usac095. [PMID: 35446412 DOI: 10.1093/milmed/usac095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Treatment outcomes for PTSD with current psychological therapies are poor, with very few patients achieving sustained symptom remission. A number of authors have identified physiological and immune disturbances in Post Traumatic Stress Disorder (PTSD) patients, but there is no unifying hypothesis that explains the myriad features of the disorder. MATERIALS AND METHODS The medical literature was reviewed over a 6-year period primarily using the medical database PUBMED. RESULTS The literature contains numerous papers that have identified a range of physiological and immune dysfunction in association with PTSD. This paper proposes that unrestrained cytokine signaling induces epigenetic changes that promote an evolutionary survival adaptation, which maintains a defensive PTSD phenotype. The brain can associate immune signaling with past threat and initiate a defensive behavioral response. The sympathetic nervous system is pro-inflammatory, while the parasympathetic nervous system is anti-inflammatory. Prolonged cholinergic withdrawal will promote a chronic inflammatory state. The innate immune cytokine IL-1β has pleiotropic properties and can regulate autonomic, glucocorticoid, and glutamate receptor functions, sleep, memory, and epigenetic enzymes. Changes in epigenetic enzyme activity can potentially alter phenotype and induce an adaptation. Levels of IL-1β correlate with severity and duration of PTSD and PTSD can be prevented by bolus administration of hydrocortisone in acute sepsis, consistent with unrestrained inflammation being a risk factor for PTSD. The nervous and immune systems engage in crosstalk, governed by common receptors. The benefits of currently used psychiatric medication may arise from immune, as well as synaptic, modulation. The psychedelic drugs (3,4-Methylenedioxymethamphetamine (MDMA), psilocybin, and ketamine) have potent immunosuppressive and anti-inflammatory effects on the adaptive immune system, which may contribute to their reported benefit in PTSD. There may be distinct PTSD phenotypes induced by innate and adaptive cytokine signaling. CONCLUSION In order for an organism to survive, it must adapt to its environment. Cytokines signal danger to the brain and can induce epigenetic changes that result in a persistent defensive phenotype. PTSD may be the price individuals pay for the genomic flexibility that promotes adaptation and survival.
Collapse
Affiliation(s)
- Stephan Rudzki
- Canberra Sports Medicine, Deakin, Australian Capital Territory 2600, Australia
| |
Collapse
|
20
|
Bourhy L, Mazeraud A, Bozza FA, Turc G, Lledo PM, Sharshar T. Neuro-Inflammatory Response and Brain-Peripheral Crosstalk in Sepsis and Stroke. Front Immunol 2022; 13:834649. [PMID: 35464410 PMCID: PMC9022190 DOI: 10.3389/fimmu.2022.834649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/07/2022] [Indexed: 12/18/2022] Open
Abstract
Despite recent therapeutic advances, ischemic stroke is still a leading cause of death and disability. There is renewed attention on peripheral inflammatory signaling as a way of modulating the post-ischemic neuro-inflammatory process. The immune-brain crosstalk has long been the focus for understanding the mechanisms of sickness behavior, which is an adaptive autonomic, neuroendocrine, and behavioral response to a peripheral inflammation. It is mediated by humoral and neural pathways that mainly involve the circumventricular organs and vagal nerve, respectively. In this review we address the question of how sepsis and stroke can dysregulate this adaptive response, notably by impairing the central integration of peripheral signaling, but also by efferent control of the immune response. We highlight the potential role of gut-brain and brain-spleen signaling in stroke.
Collapse
Affiliation(s)
- Lena Bourhy
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
| | - Aurélien Mazeraud
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
- Neuro-Anesthesiology and Intensive Care Medicine, Groupe Hospitalier Universitaire (GHU) Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas (INI), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Guillaume Turc
- Department of Neurology, GHU Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université de Paris, Centre National de Recherche Scientifique, Unité Mixte de Recherche (CNRS UMR) 3571, Perception and Memory Unit, Paris, France
| | - Tarek Sharshar
- Neuro-Anesthesiology and Intensive Care Medicine, Groupe Hospitalier Universitaire (GHU) Paris Psychiatrie et Neurosciences, Université de Paris, Paris, France
| |
Collapse
|
21
|
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: 23] [Impact Index Per Article: 11.5] [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
|
22
|
Effects of Exercise Training on the Autonomic Nervous System with a Focus on Anti-Inflammatory and Antioxidants Effects. Antioxidants (Basel) 2022; 11:antiox11020350. [PMID: 35204231 PMCID: PMC8868289 DOI: 10.3390/antiox11020350] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
Studies show that the autonomic nervous system (ANS) has an important impact on health in general. In response to environmental demands, homeostatic processes are often compromised, therefore determining an increase in the sympathetic nervous system (SNS)’s functions and a decrease in the parasympathetic nervous system (PNS)’s functions. In modern societies, chronic stress associated with an unhealthy lifestyle contributes to ANS dysfunction. In this review, we provide a brief introduction to the ANS network, its connections to the HPA axis and its stress responses and give an overview of the critical implications of ANS in health and disease—focused specifically on the immune system, cardiovascular, oxidative stress and metabolic dysregulation. The hypothalamic–pituitary–adrenal axis (HPA), the SNS and more recently the PNS have been identified as regulating the immune system. The HPA axis and PNS have anti-inflammatory effects and the SNS has been shown to have both pro- and anti-inflammatory effects. The positive impact of physical exercise (PE) is well known and has been studied by many researchers, but its negative impact has been less studied. Depending on the type, duration and individual characteristics of the person doing the exercise (age, gender, disease status, etc.), PE can be considered a physiological stressor. The negative impact of PE seems to be connected with the oxidative stress induced by effort.
Collapse
|
23
|
Honke N, Lowin T, Opgenoorth B, Shaabani N, Lautwein A, Teijaro JR, Schneider M, Pongratz G. Endogenously produced catecholamines improve the regulatory function of TLR9-activated B cells. PLoS Biol 2022; 20:e3001513. [PMID: 35073310 PMCID: PMC8786184 DOI: 10.1371/journal.pbio.3001513] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
The sympathetic nervous system (SNS) contributes to immune balance by promoting anti-inflammatory B cells. However, whether B cells possess a self-regulating mechanism by which they modulate regulatory B cell (Breg) function is not well understood. In this study, we investigated the ability of B cells to synthesize their own catecholamines upon stimulation with different B cell activators and found that expression of the enzyme tyrosine hydroxylase (TH), required to generate catecholamines, is up-regulated by Toll-like receptor (TLR)9. This TLR9-dependent expression of TH correlated with up-regulation of adrenergic receptors (ADRs), enhanced interleukin (IL)-10 production, and overexpression of the co-inhibitory ligands programmed death ligand 1 (PD-L1) and Fas ligand (FasL). Moreover, concomitant stimulation of ß1-3-ADRs together with a B cell receptor (BCR)/TLR9 stimulus clearly enhances the anti-inflammatory potential of Bregs to suppress CD4 T cells, a crucial population in the pathogenesis of autoimmune diseases, like rheumatoid arthritis (RA). Furthermore, TH up-regulation was also demonstrated in B cells during the course of collagen-induced arthritis (CIA), a mouse model for the investigation of RA. In conclusion, our data show that B cells possess an autonomous mechanism to modulate their regulatory function in an autocrine and/or paracrine manner. These findings help to better understand the function of B cells in the regulation of autoimmune diseases and the interplay of SNS. The sympathetic nervous system produces neurotransmitters such as catecholamines which contribute to immune balance by promoting anti-inflammatory B cells. This study shows that mouse B cells can themselves synthesize, sense, and transport catecholamines, which in turn modulate regulatory B cell function in an autocrine and/or paracrine manner to suppress T cell proliferation.
Collapse
Affiliation(s)
- Nadine Honke
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
- * E-mail: (NH); (GP)
| | - Torsten Lowin
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Birgit Opgenoorth
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Namir Shaabani
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Alexander Lautwein
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - John R. Teijaro
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthias Schneider
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Georg Pongratz
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
- * E-mail: (NH); (GP)
| |
Collapse
|
24
|
Khan N, Kaur S, Knuth CM, Jeschke MG. CNS-Spleen Axis - a Close Interplay in Mediating Inflammatory Responses in Burn Patients and a Key to Novel Burn Therapeutics. Front Immunol 2021; 12:720221. [PMID: 34539655 PMCID: PMC8448279 DOI: 10.3389/fimmu.2021.720221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
Severe burn-induced inflammation and subsequent hypermetabolic response can lead to profound infection and sepsis, resulting in multiple organ failure and high mortality risk in patients. This represents an extremely challenging issue for clinicians as sepsis is the leading cause of mortality in burn patients. Since hyperinflammation and immune dysfunction are a result of an immune imbalance, restoring these conditions seem to have promising benefits for burn patients. A key network that modulates the immune balance is the central nervous system (CNS)-spleen axis, which coordinates multiple signaling pathways, including sympathetic and parasympathetic pathways. Modulating inflammation is a key strategy that researchers use to understand neuroimmunomodulation in other hyperinflammatory disease models and modulating the CNS-spleen axis has led to improved clinical outcomes in patients. As the immune balance is paramount for recovery in burn-induced sepsis and patients with hyperinflammatory conditions, it appears that severe burn injuries substantially alter this CNS-spleen axis. Therefore, it is essential to address and discuss the potential therapeutic techniques that target the CNS-spleen axis that aim to restore homeostasis in burn patients. To understand this in detail, we have conducted a systematic review to explore the role of the CNS-spleen axis and its impact on immunomodulation concerning the burn-induced hypermetabolic response and associated sepsis complications. Furthermore, this thorough review explores the role of the spleen, CNS-spleen axis in the ebb and flow phases following a severe burn, how this axis induces metabolic factors and immune dysfunction, and therapeutic techniques and chemical interventions that restore the immune balance via neuroimmunomodulation.
Collapse
Affiliation(s)
- Noorisah Khan
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Supreet Kaur
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Carly M Knuth
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Stakenborg N, Boeckxstaens GE. Bioelectronics in the brain-gut axis: focus on inflammatory bowel disease (IBD). Int Immunol 2021; 33:337-348. [PMID: 33788920 PMCID: PMC8183669 DOI: 10.1093/intimm/dxab014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence shows that intestinal homeostasis is mediated by cross-talk between the nervous system, enteric neurons and immune cells, together forming specialized neuroimmune units at distinct anatomical locations within the gut. In this review, we will particularly discuss how the intrinsic and extrinsic neuronal circuitry regulates macrophage function and phenotype in the gut during homeostasis and aberrant inflammation, such as observed in inflammatory bowel disease (IBD). Furthermore, we will provide an overview of basic and translational IBD research using these neuronal circuits as a novel therapeutic tool. Finally, we will highlight the different challenges ahead to make bioelectronic neuromodulation a standard treatment for intestinal immune-mediated diseases.
Collapse
Affiliation(s)
- Nathalie Stakenborg
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Herestraat 49, O&N1 bus 701, Leuven 3000, Belgium
| | - Guy E Boeckxstaens
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Herestraat 49, O&N1 bus 701, Leuven 3000, Belgium
| |
Collapse
|
26
|
Priyanka HP, Nair RS, Kumaraguru S, Saravanaraj K, Ramasamy V. Insights on neuroendocrine regulation of immune mediators in female reproductive aging and cancer. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
27
|
Lim GN, Regan SL, Ross AE. Subsecond spontaneous catecholamine release in mesenteric lymph node ex vivo. J Neurochem 2020; 155:417-429. [DOI: 10.1111/jnc.15115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Gary N. Lim
- Department of Chemistry University of Cincinnati Cincinnati OH USA
| | - Samantha L. Regan
- Department of Pediatrics University of CincinnatiCollege of Medicine and Division of NeurologyCincinnati Children’s Research Foundation Cincinnati OH USA
- Neuroscience Graduate Program University of Cincinnati Cincinnati OH USA
| | - Ashley E. Ross
- Department of Chemistry University of Cincinnati Cincinnati OH USA
- Neuroscience Graduate Program University of Cincinnati Cincinnati OH USA
| |
Collapse
|
28
|
Zera KA, Buckwalter MS. The Local and Peripheral Immune Responses to Stroke: Implications for Therapeutic Development. Neurotherapeutics 2020; 17:414-435. [PMID: 32193840 PMCID: PMC7283378 DOI: 10.1007/s13311-020-00844-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The immune response to stroke is an exciting target for future stroke therapies. Stroke is a leading cause of morbidity and mortality worldwide, and clot removal (mechanical or pharmacological) to achieve tissue reperfusion is the only therapy currently approved for patient use. Due to a short therapeutic window and incomplete effectiveness, however, many patients are left with infarcted tissue that stimulates inflammation. Although this is critical to promote repair, it can also damage surrounding healthy brain tissue. In addition, acute immunodepression and subsequent infections are common and are associated with worse patient outcomes. Thus, the acute immune response is a major focus of researchers attempting to identify ways to amplify its benefits and suppress its negative effects to improve short-term recovery of patients. Here we review what is known about this powerful process. This includes the role of brain resident cells such as microglia, peripherally activated cells such as macrophages and neutrophils, and activated endothelium. The role of systemic immune activation and subsequent immunodepression in the days after stroke is also discussed, as is the chronic immune responses and its effects on cognitive function. The biphasic role of inflammation, as well as complex timelines of cell production, differentiation, and trafficking, suggests that the relationship between the acute and chronic phases of stroke recovery is complex. Gaining a more complete understanding of this intricate process by which inflammation is initiated, propagated, and terminated may potentially lead to therapeutics that can treat a larger population of stroke patients than what is currently available. The immune response plays a critical role in patient recovery in both the acute and chronic phases after stroke. In patients, the immune response can be beneficial by promoting repair and recovery, and also detrimental by propagating a pro-inflammatory microenvironment. Thus, it is critical to understand the mechanisms of immune activation following stroke in order to successfully design therapeutics.
Collapse
Affiliation(s)
- Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford Univeristy School of Medicine, Stanford, CA, USA.
| |
Collapse
|
29
|
Conaghan PG, Cook AD, Hamilton JA, Tak PP. Therapeutic options for targeting inflammatory osteoarthritis pain. Nat Rev Rheumatol 2020; 15:355-363. [PMID: 31068673 DOI: 10.1038/s41584-019-0221-y] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pain is the major symptom of osteoarthritis (OA) and is an important factor in strategies to manage this disease. However, the current standard of care does not provide satisfactory pain relief for many patients. The pathophysiology of OA is complex, and its presentation as a clinical syndrome is associated with pathologies of multiple joint tissues. Inflammation is associated with both OA pain and disease outcome and is therefore a major treatment target for OA and OA pain. Unlike TNF inhibitors and IL-1 inhibitors, established drugs such as glucocorticoids and methotrexate can reduce OA pain. Although central nociceptive pathways contribute to OA pain, crosstalk between the immune system and nociceptive neurons is central to inflammatory pain; therefore, new therapies might target this crosstalk. Newly identified drug targets, including neurotrophins and the granulocyte-macrophage colony-stimulating factor (GM-CSF)-CC-chemokine ligand 17 (CCL17) chemokine axis, offer the hope of better results but require clinical validation.
Collapse
Affiliation(s)
- Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute of Health Research Leeds Biomedical Research Centre, Leeds, UK
| | - Andrew D Cook
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St Albans, Victoria, Australia
| | - Paul P Tak
- Department of Clinical Immunology & Rheumatology, Academic Medical Centre, Amsterdam University Medical Centre, Amsterdam, Netherlands. .,Department of Rheumatology, Ghent University, Ghent, Belgium. .,Department of Medicine, Cambridge University, Cambridge, UK. .,Flagship Pioneering, Cambridge, MA, USA.
| |
Collapse
|
30
|
Li X, Gao J, Yu Z, Jiang W, Sun W, Yu C, Sun J, Wang C, Chen J, Jing S, Li H. Regulatory Effect of Anwulignan on the Immune Function Through Its Antioxidation and Anti-Apoptosis in D-Galactose-Induced Aging Mice. Clin Interv Aging 2020; 15:97-110. [PMID: 32099340 PMCID: PMC6996228 DOI: 10.2147/cia.s237601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/01/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Aging is a spontaneous and inevitable phenomenon of biology, which can lead to the gradual deterioration of tissues and organs. One of the age-related deterioration processes is immunosenescence, which leads to changes in the function of immune systems, including immune cells and associated cytokines. A proper modulation of immune responses can improve the age-related immunosenescence process and then reach healthy aging. Schisandra sphenanthera, a traditional Chinese medicine, has been used as both a medicine and a nutritional supplement for thousands of years. Anwulignan, a monomer compound of Schisandra sphenanthera lignans, has been reported to possess an immunomodulatory effect. Therefore, this study was designed to further explore whether Anwulignan could also modulate the immune functions in aging model mice and the underlying mechanism. METHODS D-galactose (D-gal) is often used as an inducer of immunosenescence in animals. In this study, a mice model was created by subcutaneous D-gal (220 mg kg-1) for successive 42 days. Then, the blood and spleen tissue samples were taken for the analysis and observation of cytokine levels, immunoglobulin levels, leukocyte numbers, and the phagocytic activity of macrophages, as well as the histological changes, the proliferation ability of lymphocytes, and the biochemical parameters in the spleen tissue. RESULTS Anwulignan significantly increased the serum levels of IL-2, IL-4, IFN-γ, lgG, lgM, and lgA, decreased the content of TNF-α and IL-6 in the aging mice, and increased the blood leukocyte number, the phagocytic activity, the lymphocyte proliferation, and the spleen index in vitro. Anwulignan also significantly increased the activities of SOD and GSH-Px, decreased the contents of MDA and 8-OHdG in the spleen tissue, up-regulated the expressions of Nrf2, HO-1, and Bcl2, down-regulated the expressions of Keap1, Caspase-3, and Bax in the spleen cells, and reduced the apoptosis of spleen lymphocytes. CONCLUSION Anwulignan can restore the immune function that is declined in D-gal-induced aging mice partly related to its antioxidant capacity by activating the Nrf2/ARE pathway and downstream enzymes, as well as its anti-apoptotic effect by regulating Caspase-3 and the ratio of Bcl2 to Bax in the spleen.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Jiaqi Gao
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Zepeng Yu
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Weihai Jiang
- Affiliated Hospital of Beihua University, Jilin, Jilin132011, People’s Republic of China
| | - Wei Sun
- Affiliated Hospital of Beihua University, Jilin, Jilin132011, People’s Republic of China
| | - Chunyan Yu
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Jinghui Sun
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Jianguang Chen
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| | - Shu Jing
- Affiliated Hospital of Beihua University, Jilin, Jilin132011, People’s Republic of China
| | - He Li
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, People’s Republic of China
| |
Collapse
|
31
|
Krishna G, Beitchman JA, Bromberg CE, Currier Thomas T. Approaches to Monitor Circuit Disruption after Traumatic Brain Injury: Frontiers in Preclinical Research. Int J Mol Sci 2020; 21:ijms21020588. [PMID: 31963314 PMCID: PMC7014469 DOI: 10.3390/ijms21020588] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/03/2020] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (TBI) often results in pathophysiological damage that can manifest as both acute and chronic neurological deficits. In an attempt to repair and reconnect disrupted circuits to compensate for loss of afferent and efferent connections, maladaptive circuitry is created and contributes to neurological deficits, including post-concussive symptoms. The TBI-induced pathology physically and metabolically changes the structure and function of neurons associated with behaviorally relevant circuit function. Complex neurological processing is governed, in part, by circuitry mediated by primary and modulatory neurotransmitter systems, where signaling is disrupted acutely and chronically after injury, and therefore serves as a primary target for treatment. Monitoring of neurotransmitter signaling in experimental models with technology empowered with improved temporal and spatial resolution is capable of recording in vivo extracellular neurotransmitter signaling in behaviorally relevant circuits. Here, we review preclinical evidence in TBI literature that implicates the role of neurotransmitter changes mediating circuit function that contributes to neurological deficits in the post-acute and chronic phases and methods developed for in vivo neurochemical monitoring. Coupling TBI models demonstrating chronic behavioral deficits with in vivo technologies capable of real-time monitoring of neurotransmitters provides an innovative approach to directly quantify and characterize neurotransmitter signaling as a universal consequence of TBI and the direct influence of pharmacological approaches on both behavior and signaling.
Collapse
Affiliation(s)
- Gokul Krishna
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Joshua A. Beitchman
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Caitlin E. Bromberg
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix VA Healthcare System, Phoenix, AZ 85012, USA
- Correspondence: ; Tel.: +1-602-827-2348
| |
Collapse
|
32
|
Al-Shalan HAM, Hu D, Nicholls PK, Greene WK, Ma B. Immunofluorescent characterization of innervation and nerve-immune cell neighborhood in mouse thymus. Cell Tissue Res 2019; 378:239-254. [PMID: 31230166 DOI: 10.1007/s00441-019-03052-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/23/2019] [Indexed: 12/29/2022]
Abstract
The central nervous system impacts the immune system mainly by regulating the systemic concentration of humoral substances, whereas the peripheral nervous system (PNS) communicates with the immune system specifically according to local "hardwiring" of sympathetic/parasympathetic (efferent) and sensory (afferent) nerves to the primary and secondary lymphoid tissue/organs (e.g., thymus spleen and lymph nodes). In the present study, we use immunofluorescent staining of neurofilament-heavy to reveal the distribution of nerve fibers and the nerve-immune cell neighborhood inside the mouse thymus. Our results demonstrate (a) the presence of an extensive meshwork of nerve fibers in all thymic compartments, including the capsule, subcapsular region, cortex, cortico-medullary junction and medulla; (b) close associations of nerve fibers with blood vessels (including the postcapillary venules), indicating the neural control of blood circulation and immune cell dynamics inside the thymus; (c) the close proximity of nerve fibers to various subsets of thymocytes (e.g., CD4+, CD8+ and CD4+CD8+), dendritic cells (e.g., B220+, CD4+, CD8+ and F4/80+), macrophages (Mac1+ and F4/80+) and B cells. Our novel findings concerning thymic innervation and the nerve-immune cell neighborhood in situ should facilitate the understanding of bi-directional communications between the PNS and primary lymphoid organs. Since the innervation of lymphoid organs, including the thymus, may play essential roles in the pathogenesis and progression of some neuroimmune, infectious and autoimmune diseases, better knowledge of PNS-immune system crosstalk should benefit the development of potential therapies for these diseases.
Collapse
Affiliation(s)
- Huda A M Al-Shalan
- Discipline of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, 6150, Australia.,Department of Microbiology/Virology, College of Veterinary Medicine, Baghdad University, Baghdad, 10070, Iraq
| | - Dailun Hu
- Clinical College, Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Philip K Nicholls
- Discipline of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, 6150, Australia
| | - Wayne K Greene
- Discipline of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, 6150, Australia
| | - Bin Ma
- Discipline of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, 6150, Australia.
| |
Collapse
|
33
|
Kerage D, Sloan EK, Mattarollo SR, McCombe PA. Interaction of neurotransmitters and neurochemicals with lymphocytes. J Neuroimmunol 2019; 332:99-111. [PMID: 30999218 DOI: 10.1016/j.jneuroim.2019.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Neurotransmitters and neurochemicals can act on lymphocytes by binding to receptors expressed by lymphocytes. This review describes lymphocyte expression of receptors for a selection of neurotransmitters and neurochemicals, the anatomical locations where lymphocytes can interact with neurotransmitters, and the effects of the neurotransmitters on lymphocyte function. Implications for health and disease are also discussed.
Collapse
Affiliation(s)
- Daniel Kerage
- The University of Queensland Diamantina Institute, Brisbane, Australia; Transplant Research Program, Boston Children's Hospital, Boston, MA, United States of America
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Division of Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Cousins Center for Neuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, USA
| | | | - Pamela A McCombe
- The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Brisbane, Australia; Royal Brisbane and Women's Hospital, Herston, Brisbane, Australia.
| |
Collapse
|
34
|
Isoproterenol-induced beta-2 adrenergic receptor activation negatively regulates interleukin-2 signaling. Biochem J 2018; 475:2907-2923. [PMID: 30120106 DOI: 10.1042/bcj20180503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
Regulation of intracellular signaling pathways in lymphocytes is critical for cell homeostasis and immune response. Interleukin-2 (IL-2), a key regulator of lymphocytes, signals following receptor-ligand engagement and subsequent recruitment and activation of effector proteins including JAKs and STATs. Lymphocytes can also be regulated by the central nervous system through the β2 adrenergic receptor (β2AR) pathway which can affect cell trafficking, proliferation, differentiation, and cytokine production. The cross-talk between these two signaling pathways represents an important mechanism that has yet to be fully elucidated. The present study provides evidence for communication between the IL-2 receptor (IL-2R) and β2AR. Treatment of human lymphoid cell lines with the β2AR agonist isoproterenol (ISO) alone increased cAMP levels and mediated a stimulatory response by activating AKT and ERK to promote cell viability. Interestingly, ISO activation of β2AR also induced threonine phosphorylation of the IL-2Rβ. In contrast, ISO treatment prior to IL-2 stimulation produced an inhibitory signal that disrupted IL-2 induced activation of the JAK/STAT, MEK/ERK, and PI3K pathways by inhibiting the formation of the IL-2R beta-gamma chain complex, and subsequently cell proliferation. Moreover, γc-family cytokines-mediated STAT5 activation was also inhibited by ISO. These results suggest a molecular mechanism by which β2AR signaling can both stimulate and suppress lymphocyte responses and thus explain how certain therapeutic agents, such as vasodilators, may impact immune responsiveness.
Collapse
|
35
|
Figueroa F, Mendoza G, Cardozo D, Mohamed F, Oliveros L, Forneris M. Sympathetic innervation regulates macrophage activity in rats with polycystic ovary. J Endocrinol 2018; 238:33-45. [PMID: 29720538 DOI: 10.1530/joe-17-0736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/02/2018] [Indexed: 01/14/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is a low-grade inflammatory disease characterized by hyperandrogenism and ovarian hyperinnervation. The aim of this work is to investigate whether in vivo bilateral superior ovarian nerve (SON) section in adult rats with estradiol valerate-induced PCOS (PCO rats) affects macrophage spleen cells (MФ) and modifies the steroidogenic ability of their secretions. Culture media of MФ from PCO rats and PCO rats with SON section (PCO-SON rats) were used to stimulate in vitro intact ovaries. Compared with macrophages PCO, macrophages from PCO-SON rats released less tumor necrosis factor-α and nitric oxide, expressed lower Bax and Nfkb mRNA and showed reduced TUNEL staining. Also, in PCO rats, the SON section decreased kisspeptin and nerve growth factor mRNA expressions, without changes in Trka receptor mRNA levels. Macrophage secretions from PCO-SON rats decreased androstenedione and stimulated progesterone release in PCO ovaries, compared to macrophage secretions from PCO rats. No changes were observed in ovarian estradiol response. These findings emphasize the importance of the SON in spleen MΦ, since its manipulation leads to secondary modifications of immunological and neural mediators, which might influence ovarian steroidogenesis. In PCO ovaries, the reduction of androstenedione and the improvement of progesterone release induced by PCO-SON MΦ secretion, might be beneficial considering the hormonal anomalies characteristic of PCOS. We present functional evidence that modulation of the immune-endocrine function by peripheral sympathetic nervous system might have implications for understanding the pathophysiology of PCOS.
Collapse
Affiliation(s)
- Florencia Figueroa
- Laboratorio de Biología de la ReproducciónFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Gisela Mendoza
- Laboratorio de Biología de la ReproducciónFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Darío Cardozo
- Laboratorio de Biología de la ReproducciónFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Fabián Mohamed
- Area MorfologíaFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Liliana Oliveros
- Laboratorio de Biología de la ReproducciónFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Myriam Forneris
- Laboratorio de Biología de la ReproducciónFacultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
36
|
Noble BT, Brennan FH, Popovich PG. The spleen as a neuroimmune interface after spinal cord injury. J Neuroimmunol 2018; 321:1-11. [PMID: 29957379 DOI: 10.1016/j.jneuroim.2018.05.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 01/17/2023]
Abstract
Traumatic spinal cord injury (SCI) causes widespread damage to neurons, glia and endothelia located throughout the spinal parenchyma. In response to the injury, resident and blood-derived leukocytes orchestrate an intraspinal inflammatory response that propagates secondary neuropathology and also promotes tissue repair. SCI also negatively affects autonomic control over peripheral immune organs, notably the spleen. The spleen is the largest secondary lymphoid organ in mammals, with major roles in blood filtration and host defense. Splenic function is carefully regulated by neuroendocrine mechanisms that ensure that the immune responses to infection or injury are proportionate to the initiating stimulus, and can be terminated when the stimulus is cleared. After SCI, control over the viscera, including endocrine and lymphoid tissues is lost due to damage to spinal autonomic (sympathetic) circuitry. This review begins by examining the normal structure and function of the spleen including patterns of innervation and the role played by the nervous system in regulating spleen function. We then describe how after SCI, loss of proper neural control over splenic function leads to systems-wide neuropathology, immune suppression and autoimmunity. We conclude by discussing opportunities for targeting the spleen to restore immune homeostasis, reduce morbidity and mortality, and improve functional recovery after SCI.
Collapse
Affiliation(s)
- Benjamin T Noble
- Neuroscience Graduate Studies Program, Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, Columbus 43210, OH, USA
| | - Faith H Brennan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus 43210, OH, USA
| | - Phillip G Popovich
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus 43210, OH, USA.
| |
Collapse
|
37
|
Abstract
The nervous system regulates immunity and inflammation. The molecular detection of pathogen fragments, cytokines, and other immune molecules by sensory neurons generates immunoregulatory responses through efferent autonomic neuron signaling. The functional organization of this neural control is based on principles of reflex regulation. Reflexes involving the vagus nerve and other nerves have been therapeutically explored in models of inflammatory and autoimmune conditions, and recently in clinical settings. The brain integrates neuro-immune communication, and brain function is altered in diseases characterized by peripheral immune dysregulation and inflammation. Here we review the anatomical and molecular basis of the neural interface with immunity, focusing on peripheral neural control of immune functions and the role of the brain in the model of the immunological homunculus. Clinical advances stemming from this knowledge within the framework of bioelectronic medicine are also briefly outlined.
Collapse
Affiliation(s)
- Valentin A Pavlov
- Center for Biomedical Science and Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York 11030, USA; , ,
| | - Sangeeta S Chavan
- Center for Biomedical Science and Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York 11030, USA; , ,
| | - Kevin J Tracey
- Center for Biomedical Science and Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York 11030, USA; , ,
| |
Collapse
|
38
|
Cook AD, Christensen AD, Tewari D, McMahon SB, Hamilton JA. Immune Cytokines and Their Receptors in Inflammatory Pain. Trends Immunol 2018; 39:240-255. [DOI: 10.1016/j.it.2017.12.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/28/2017] [Accepted: 12/13/2017] [Indexed: 01/23/2023]
|
39
|
Nicholls AJ, Wen SW, Hall P, Hickey MJ, Wong CHY. Activation of the sympathetic nervous system modulates neutrophil function. J Leukoc Biol 2017; 103:295-309. [PMID: 29345350 PMCID: PMC6635748 DOI: 10.1002/jlb.3ma0517-194rr] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/04/2017] [Accepted: 12/06/2017] [Indexed: 01/19/2023] Open
Abstract
Emerging evidence has revealed that noradrenaline (NA), the main neurotransmitter of the sympathetic nervous system (SNS), regulates a variety of immune functions via binding to adrenergic receptors present on immune cells. In this study, we examined the role of NA in the regulation of neutrophil functions. Neutrophils were isolated from the bone marrow of naïve mice and treated with NA at various concentrations to assess the effect on various neutrophil functions. Additionally, we performed cremaster intravital microscopy to examine neutrophil‐endothelial cell interactions following NA superfusion in vivo. In a separate group of animals, mice were subjected to an experimental model of stroke and at 4 and 24 h neutrophils were isolated for assessment on their ability to migrate toward various chemokines. Treatment of neutrophils with NA for 4 h significantly impaired neutrophil chemotaxis and induced an N2 neutrophil phenotype with reduced expression of the genes critical for cytoskeleton remodeling and inflammation. Prolonged NA administration promoted neutrophils to release myeloperoxidase and IL‐6, but suppressed the production of interferon‐γ and IL‐10, reduced neutrophil activation and phagocytosis. Superfusion of NA over the cremaster muscle almost completely inhibited fMLP‐induced neutrophil adhesion/arrest and transmigration. Furthermore, using a mouse model of stroke, a pathological condition in which SNS activation is evident, neutrophils isolated from poststroke mice showed markedly reduced chemotaxis toward all of the chemokines tested. The findings from our study indicate that neutrophil chemotaxis, activation, and phagocytosis can all be negatively regulated in an NA‐dependent manner. A better understanding of the relationship between sympathetic activation and neutrophil function will be important for the development of effective antibacterial interventions.
Collapse
Affiliation(s)
- Alyce J Nicholls
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Australia
| | - Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Australia
| | - Pam Hall
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Australia
| |
Collapse
|
40
|
The Spleen: A Hub Connecting Nervous and Immune Systems in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2017; 18:ijms18061216. [PMID: 28590409 PMCID: PMC5486039 DOI: 10.3390/ijms18061216] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic disorders have been identified as major health problems affecting a large portion of the world population. In addition, obesity and insulin resistance are principal risk factors for the development of cardiovascular diseases. Altered immune responses are common features of both hypertension and obesity and, moreover, the involvement of the nervous system in the modulation of immune system is gaining even more attention in both pathophysiological contexts. For these reasons, during the last decades, researches focused their efforts on the comprehension of the molecular mechanisms connecting immune system to cardiovascular and metabolic diseases. On the other hand, it has been reported that in these pathological conditions, central neural pathways modulate the activity of the peripheral nervous system, which is strongly involved in onset and progression of the disease. It is interesting to notice that neural reflex can also participate in the modulation of immune functions. In this scenario, the spleen becomes the crucial hub allowing the interaction of different systems differently involved in metabolic and cardiovascular diseases. Here, we summarize the major findings that dissect the role of the immune system in disorders related to metabolic and cardiovascular dysfunctions, and how this could also be influenced by neural reflexes.
Collapse
|
41
|
NGF and Its Receptors in the Regulation of Inflammatory Response. Int J Mol Sci 2017; 18:ijms18051028. [PMID: 28492466 PMCID: PMC5454940 DOI: 10.3390/ijms18051028] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/31/2017] [Accepted: 05/03/2017] [Indexed: 12/28/2022] Open
Abstract
There is growing interest in the complex relationship between the nervous and immune systems and how its alteration can affect homeostasis and result in the development of inflammatory diseases. A key mediator in cross-talk between the two systems is nerve growth factor (NGF), which can influence both neuronal cell function and immune cell activity. The up-regulation of NGF described in inflamed tissues of many diseases can regulate innervation and neuronal activity of peripheral neurons, inducing the release of immune-active neuropeptides and neurotransmitters, but can also directly influence innate and adaptive immune responses. Expression of the NGF receptors tropomyosin receptor kinase A (TrkA) and p75 neurotrophin receptor (p75NTR) is dynamically regulated in immune cells, suggesting a varying requirement for NGF depending on their state of differentiation and functional activity. NGF has a variety of effects that can be either pro-inflammatory or anti-inflammatory. This apparent contradiction can be explained by considering NGF as part of an endogenous mechanism that, while activating immune responses, also activates pathways necessary to dampen the inflammatory response and limit tissue damage. Decreases in TrkA expression, such as that recently demonstrated in immune cells of arthritis patients, might prevent the activation by NGF of regulatory feed-back mechanisms, thus contributing to the development and maintenance of chronic inflammation.
Collapse
|
42
|
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: 3.1] [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
|
43
|
Martelli D, Farmer DGS, Yao ST. The splanchnic anti-inflammatory pathway: could it be the efferent arm of the inflammatory reflex? Exp Physiol 2016; 101:1245-1252. [PMID: 27377300 DOI: 10.1113/ep085559] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 07/01/2016] [Indexed: 12/13/2022]
Abstract
What is the topic of this review? We review the current literature on the neural reflex termed the 'inflammatory reflex' that inhibits an excessive release of inflammatory mediators in response to an immune challenge. What advances does it highlight? The original model proposed that the inflammatory reflex is a vago-vagal reflex that controls immune function. We posit that, in the endotoxaemic animal model, the vagus nerves do not appear to play a role. The evidence suggests that the efferent motor pathway, termed here the 'splanchnic anti-inflammatory pathway', is purely sympathetic, travelling via the greater splanchnic nerves to regulate the ensuing inflammatory response to immune challenges. Exposure to immune challenges results in the development of inflammation. An insufficient inflammatory response can be life-threatening, whereas an exaggerated response is also detrimental because it causes tissue damage and, in extreme cases, septic shock that can lead to death. Hence, inflammation must be finely regulated. It is generally accepted that the brain inhibits inflammation induced by an immune challenge in two main ways: humorally, by activating the hypothalamic-pituitary-adrenal axis to release glucocorticoids; and neurally, via a mechanism that has been termed the 'inflammatory reflex'. The efferent arm of this reflex (the neural-to-immune link) was thought to be the 'cholinergic anti-inflammatory pathway'. Here, we discuss data that support the hypothesis that the vagus nerves play no role in the control of inflammation in the endotoxaemic animal model. We have shown and posit that it is the greater splanchnic nerves that are activated in response to the immune challenge and that, in turn, drive postganglionic sympathetic neurons to inhibit inflammation.
Collapse
Affiliation(s)
- D Martelli
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, 3010, Australia. .,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy.
| | - D G S Farmer
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, 3010, Australia
| | - S T Yao
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, 3010, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, VIC, 3010, Australia
| |
Collapse
|
44
|
Swaminathan S, Rosner MH, Okusa MD. Emerging therapeutic targets of sepsis-associated acute kidney injury. Semin Nephrol 2015; 35:38-54. [PMID: 25795498 DOI: 10.1016/j.semnephrol.2015.01.005] [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] [Indexed: 12/12/2022]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is linked to high morbidity and mortality. To date, singular approaches to target specific pathways known to contribute to the pathogenesis of SA-AKI have failed. Because of the complexity of the pathogenesis of SA-AKI, a reassessment necessitates integrative approaches to therapeutics of SA-AKI that include general supportive therapies such as the use of vasopressors, fluids, antimicrobials, and target-specific and time-dependent therapeutics. There has been recent progress in our understanding of the pathogenesis and treatment of SA-AKI including the temporal nature of proinflammatory and anti-inflammatory processes. In this review, we discuss the clinical and experimental basis of emerging therapeutic approaches that focus on targeting early proinflammatory and late anti-inflammatory processes, as well as therapeutics that may enhance cellular survival and recovery. Finally, we include ongoing clinical trials in sepsis.
Collapse
Affiliation(s)
- Sundararaman Swaminathan
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA
| | - Mitchell H Rosner
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA.
| |
Collapse
|
45
|
Lei R, Zhao T, Li Q, Wang X, Ma H, Deng Y. Carbon Ion Irradiated Neural Injury Induced the Peripheral Immune Effects in Vitro or in Vivo. Int J Mol Sci 2015; 16:28334-46. [PMID: 26633364 PMCID: PMC4691056 DOI: 10.3390/ijms161226109] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/11/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022] Open
Abstract
Carbon ion radiation is a promising treatment for brain cancer; however, the immune system involved long-term systemic effects evoke a concern of complementary and alternative therapies in clinical treatment. To clarify radiotherapy caused fundamental changes in peripheral immune system, examinations were performed based on established models in vitro and in vivo. We found that brain-localized carbon ion radiation of neural cells induced complex changes in the peripheral blood, thymus, and spleen at one, two, and three months after its application. Atrophy, apoptosis, and abnormal T-cell distributions were observed in rats receiving a single high dose of radiation. Radiation downregulated the expression of proteins involved in T-cell development at the transcriptional level and increased the proportion of CD3⁺CD4(-)CD8⁺ T-cells in the thymus and the proportion of CD3⁺CD4⁺CD8(-) T-cells in the spleen. These data show that brain irradiation severely affects the peripheral immune system, even at relatively long times after irradiation. In addition, they provide valuable information that will implement the design of biological-based strategies that will aid brain cancer patients suffering from the long-term side effects of radiation.
Collapse
Affiliation(s)
- Runhong Lei
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Tuo Zhao
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Qiang Li
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.
| | - Xiao Wang
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China.
| | - Hong Ma
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
46
|
Di Giovangiulio M, Verheijden S, Bosmans G, Stakenborg N, Boeckxstaens GE, Matteoli G. The Neuromodulation of the Intestinal Immune System and Its Relevance in Inflammatory Bowel Disease. Front Immunol 2015; 6:590. [PMID: 26635804 PMCID: PMC4653294 DOI: 10.3389/fimmu.2015.00590] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/03/2015] [Indexed: 12/18/2022] Open
Abstract
One of the main tasks of the immune system is to discriminate and appropriately react to “danger” or “non-danger” signals. This is crucial in the gastrointestinal tract, where the immune system is confronted with a myriad of food antigens and symbiotic microflora that are in constant contact with the mucosa, in addition to any potential pathogens. This large number of antigens and commensal microflora, which are essential for providing vital nutrients, must be tolerated by the intestinal immune system to prevent aberrant inflammation. Hence, the balance between immune activation versus tolerance should be tightly regulated to maintain intestinal homeostasis and to prevent immune activation indiscriminately against all luminal antigens. Loss of this delicate equilibrium can lead to chronic activation of the intestinal immune response resulting in intestinal disorders, such as inflammatory bowel diseases (IBD). In order to maintain homeostasis, the immune system has evolved diverse regulatory strategies including additional non-immunological actors able to control the immune response. Accumulating evidence strongly indicates a bidirectional link between the two systems in which the brain modulates the immune response via the detection of circulating cytokines and via direct afferent input from sensory fibers and from enteric neurons. In the current review, we will highlight the most recent findings regarding the cross-talk between the nervous system and the mucosal immune system and will discuss the potential use of these neuronal circuits and neuromediators as novel therapeutic tools to reestablish immune tolerance and treat intestinal chronic inflammation.
Collapse
Affiliation(s)
- Martina Di Giovangiulio
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Simon Verheijden
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Goele Bosmans
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Nathalie Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Guy E Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Gianluca Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| |
Collapse
|
47
|
Scanzano A, Cosentino M. Adrenergic regulation of innate immunity: a review. Front Pharmacol 2015; 6:171. [PMID: 26321956 PMCID: PMC4534859 DOI: 10.3389/fphar.2015.00171] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
The sympathetic nervous system has a major role in the brain-immune cross-talk, but few information exist on the sympathoadrenergic regulation of innate immune system. The aim of this review is to summarize available knowledge regarding the sympathetic modulation of the innate immune response, providing a rational background for the possible repurposing of adrenergic drugs as immunomodulating agents. The cells of immune system express adrenoceptors (AR), which represent the target for noradrenaline and adrenaline. In human neutrophils, adrenaline and noradrenaline inhibit migration, CD11b/CD18 expression, and oxidative metabolism, possibly through β-AR, although the role of α1- and α2-AR requires further investigation. Natural Killer express β-AR, which are usually inhibitory. Monocytes express β-AR and their activation is usually antiinflammatory. On murine Dentritic cells (DC), β-AR mediate sympathetic influence on DC-T cells interactions. In human DC β2-AR may affect Th1/2 differentiation of CD4+ T cells. In microglia and in astrocytes, β2-AR dysregulation may contribute to neuroinflammation in autoimmune and neurodegenerative disease. In conclusion, extensive evidence supports a critical role for adrenergic mechanisms in the regulation of innate immunity, in peripheral tissues as well as in the CNS. Sympathoadrenergic pathways in the innate immune system may represent novel antiinflammatory and immunomodulating targets with significant therapeutic potential.
Collapse
Affiliation(s)
- Angela Scanzano
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| |
Collapse
|
48
|
Cosentino M, Marino F, Maestroni GJM. Sympathoadrenergic modulation of hematopoiesis: a review of available evidence and of therapeutic perspectives. Front Cell Neurosci 2015; 9:302. [PMID: 26300737 PMCID: PMC4525045 DOI: 10.3389/fncel.2015.00302] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/23/2015] [Indexed: 12/18/2022] Open
Abstract
Innervation of the bone marrow (BM) has been described more than one century ago, however the first in vivo evidence that sympathoadrenergic fibers have a role in hematopoiesis dates back to less than 25 years ago. Evidence has since increased showing that adrenergic nerves in the BM release noradrenaline and possibly also dopamine, which act on adrenoceptors and dopaminergic receptors (DR) expressed on hematopoietic cells and affect cell survival, proliferation, migration and engraftment ability. Remarkably, dysregulation of adrenergic fibers to the BM is associated with hematopoietic disturbances and myeloproliferative disease. Several adrenergic and dopaminergic agents are already in clinical use for non-hematological indications and with a usually favorable risk-benefit profile, and are therefore potential candidates for non-conventional modulation of hematopoiesis.
Collapse
Affiliation(s)
- Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| | - Franca Marino
- Center for Research in Medical Pharmacology, University of Insubria Varese, Italy
| | | |
Collapse
|
49
|
Abstract
The present review assesses the current state of literature defining integrative autonomic-immune physiological processing, focusing on studies that have employed electrophysiological, pharmacological, molecular biological, and central nervous system experimental approaches. Central autonomic neural networks are informed of peripheral immune status via numerous communicating pathways, including neural and non-neural. Cytokines and other immune factors affect the level of activity and responsivity of discharges in sympathetic and parasympathetic nerves innervating diverse targets. Multiple levels of the neuraxis contribute to cytokine-induced changes in efferent parasympathetic and sympathetic nerve outflows, leading to modulation of peripheral immune responses. The functionality of local sympathoimmune interactions depends on the microenvironment created by diverse signaling mechanisms involving integration between sympathetic nervous system neurotransmitters and neuromodulators; specific adrenergic receptors; and the presence or absence of immune cells, cytokines, and bacteria. Functional mechanisms contributing to the cholinergic anti-inflammatory pathway likely involve novel cholinergic-adrenergic interactions at peripheral sites, including autonomic ganglion and lymphoid targets. Immune cells express adrenergic and nicotinic receptors. Neurotransmitters released by sympathetic and parasympathetic nerve endings bind to their respective receptors located on the surface of immune cells and initiate immune-modulatory responses. Both sympathetic and parasympathetic arms of the autonomic nervous system are instrumental in orchestrating neuroimmune processes, although additional studies are required to understand dynamic and complex adrenergic-cholinergic interactions. Further understanding of regulatory mechanisms linking the sympathetic nervous, parasympathetic nervous, and immune systems is critical for understanding relationships between chronic disease development and immune-associated changes in autonomic nervous system function.
Collapse
Affiliation(s)
- M J Kenney
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | | |
Collapse
|
50
|
Gigliotti JC, Huang L, Bajwa A, Ye H, Mace EH, Hossack JA, Kalantari K, Inoue T, Rosin DL, Okusa MD. Ultrasound Modulates the Splenic Neuroimmune Axis in Attenuating AKI. J Am Soc Nephrol 2015; 26:2470-81. [PMID: 25644106 DOI: 10.1681/asn.2014080769] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/11/2014] [Indexed: 11/03/2022] Open
Abstract
We showed previously that prior exposure to a modified ultrasound regimen prevents kidney ischemia-reperfusion injury (IRI) likely via the splenic cholinergic anti-inflammatory pathway (CAP) and α7 nicotinic acetylcholine receptors (α7nAChR). However, it is unclear how ultrasound stimulates the splenic CAP. Further investigating the role of the spleen in ischemic injury, we found that prior splenectomy (-7d) or chemical sympathectomy of the spleen with 6-hydroxydopamine (6OHDA; -14d) exacerbated injury after subthreshold (24-minute ischemia) IRI. 6-OHDA-induced splenic denervation also prevented ultrasound-induced protection of kidneys from moderate (26-minute ischemia) IRI. Ultrasound-induced protection required hematopoietic but not parenchymal α7nAChRs, as shown by experiments in bone marrow chimeras generated with wild-type and α7nAChR(-/-) mice. Ultrasound protection was associated with reduced expression of circulating and kidney-derived cytokines. However, splenocytes isolated from mice 24 hours after ultrasound treatment released more IL-6 ex vivo in response to LPS than splenocytes from sham mice. Adoptive transfer of splenocytes from ultrasound-treated (but not sham) mice to naïve mice was sufficient to protect kidneys of recipient mice from IRI. Ultrasound treatment 24 hours before cecal ligation puncture-induced sepsis was effective in reducing plasma creatinine in this model of AKI. Thus, splenocytes of ultrasound-treated mice are capable of modulating IRI in vivo, supporting our ongoing hypothesis that a modified ultrasound regimen has therapeutic potential for AKI and other inflammatory conditions.
Collapse
Affiliation(s)
- Joseph C Gigliotti
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Liping Huang
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Amandeep Bajwa
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Hong Ye
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Eric H Mace
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | | | - Kambiz Kalantari
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Tsuyoshi Inoue
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine
| | - Diane L Rosin
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine; Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia
| | - Mark D Okusa
- Department of Medicine, Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine;
| |
Collapse
|