1
|
Hesampour F, Bernstein CN, Ghia JE. Brain-Gut Axis: Invasive and Noninvasive Vagus Nerve Stimulation, Limitations, and Potential Therapeutic Approaches. Inflamm Bowel Dis 2024; 30:482-495. [PMID: 37738641 DOI: 10.1093/ibd/izad211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Indexed: 09/24/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing condition with no known etiology and is characterized by disrupted gut homeostasis, chronic inflammation, and ulcerative lesions. Although current treatments can reduce disease activity, IBD frequently recurs once treatments are discontinued, indicating that treatments are ineffective in providing long-term remission. The lack of responsiveness and reluctance of some affected persons to take medications because of potential adverse effects has enhanced the need for novel therapeutic approaches. The vagus nerve (VN) is likely important in the pathogenesis of IBD, considering the decreased activity of the parasympathetic nervous system, especially the VN, and the impaired interaction between the enteric nervous system and central nervous system in patients with IBD. Vagus nerve stimulation (VNS) has demonstrated anti-inflammatory effects in various inflammatory disorders, including IBD, by inhibiting the production of inflammatory cytokines by immune cells. It has been suggested that stimulating the vagus nerve to induce its anti-inflammatory effects may be a potential therapeutic approach for IBD. Noninvasive techniques for VNS have been developed. Considering the importance of VN function in the brain-gut axis, VNS is a promising treatment option for IBD. This review discusses the potential therapeutic advantages and drawbacks of VNS, particularly the use of noninvasive transcutaneous auricular vagus nerve stimulation.
Collapse
Affiliation(s)
| | - Charles N Bernstein
- Internal Medicine, University of Manitoba, Winnipeg, Canada
- Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, Canada
| | - Jean-Eric Ghia
- Immunology, University of Manitoba, Winnipeg, Canada
- Internal Medicine, University of Manitoba, Winnipeg, Canada
- Inflammatory Bowel Disease Clinical and Research Centre, University of Manitoba, Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| |
Collapse
|
2
|
Gabalski AH, Tynan A, Tsaava T, Li JH, Lee D, Hepler TD, Hide D, George S, Iñiguez CEB, Thompson DA, Zhu C, Wang H, Brines M, Tracey KJ, Chavan SS. Circulating extracellular choline acetyltransferase regulates inflammation. J Intern Med 2024; 295:346-356. [PMID: 38011942 PMCID: PMC10922394 DOI: 10.1111/joim.13750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
BACKGROUND Choline acetyltransferase (ChAT) is required for the biosynthesis of acetylcholine, the molecular mediator that inhibits cytokine production in the cholinergic anti-inflammatory pathway of the vagus nerve inflammatory reflex. Abundant work has established the biology of cytoplasmic ChAT in neurons, but much less is known about the potential presence and function of ChAT in the extracellular milieu. OBJECTIVES We evaluated the hypothesis that extracellular ChAT activity responds to inflammation and serves to inhibit cytokine release and attenuate inflammation. METHODS After developing novel methods for quantification of ChAT activity in plasma, we determined whether ChAT activity changes in response to inflammatory challenges. RESULTS Active ChAT circulates within the plasma compartment of mice and responds to immunological perturbations. Following the administration of bacterial endotoxin, plasma ChAT activity increases for 12-48 h, a time period that coincides with declining tumor necrosis factor (TNF) levels. Further, a direct activation of the cholinergic anti-inflammatory pathway by vagus nerve stimulation significantly increases plasma ChAT activity, whereas the administration of bioactive recombinant ChAT (r-ChAT) inhibits endotoxin-stimulated TNF production and anti-ChAT antibodies exacerbate endotoxin-induced TNF levels, results of which suggest that ChAT activity regulates endogenous TNF production. Administration of r-ChAT significantly attenuates pro-inflammatory cytokine production and disease activity in the dextran sodium sulfate preclinical model of inflammatory bowel disease. Finally, plasma ChAT levels are also elevated in humans with sepsis, with the highest levels observed in a patient who succumbed to infection. CONCLUSION As a group, these results support further investigation of ChAT as a counter-regulator of inflammation and potential therapeutic agent.
Collapse
Affiliation(s)
- Arielle H. Gabalski
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA
| | - Aisling Tynan
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Tea Tsaava
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Jian Hua Li
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Diana Lee
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA
| | - Tyler D. Hepler
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Daniel Hide
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Sam George
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Carlos E. Bravo Iñiguez
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Dane A Thompson
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Cassie Zhu
- Institute for Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Haichao Wang
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA
- Institute for Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Michael Brines
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Kevin J. Tracey
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA
| | - Sangeeta S. Chavan
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, NY 11549, USA
| |
Collapse
|
3
|
Brooke AK, Murrow DP, Caldwell KCN, Witt CE, Ross AE. Measuring neuron-regulated immune cell physiology via the alpha-2 adrenergic receptor in an ex vivo murine spleen model. Cell Mol Life Sci 2023; 80:354. [PMID: 37945921 PMCID: PMC11071927 DOI: 10.1007/s00018-023-05012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
The communication between the nervous and immune systems plays a crucial role in regulating immune cell function and inflammatory responses. Sympathetic neurons, which innervate the spleen, have been implicated in modulating immune cell activity. The neurotransmitter norepinephrine (NE), released by sympathetic neurons, influences immune cell responses by binding to adrenergic receptors on their surface. The alpha-2 adrenergic receptor (α2AR), expressed predominantly on sympathetic neurons, has received attention due to its autoreceptor function and ability to modulate NE release. In this study, we used fast-scan cyclic voltammetry (FSCV) to provide the first subsecond measurements of NE released in the white pulp region of the spleen and validated it with yohimbine, a known antagonist of α2AR. For further application of FSCV in neuroimmunology, we investigated the extent to which subsecond NE from sympathetic neurons is important for immune cell physiology and cytokine production, focusing on tumor necrosis factor-alpha (TNF-α), interleukin-10 (IL-10), and interleukin-6 (IL-6). Our findings provide insights into the regulatory mechanisms underlying sympathetic-immune interactions and show the significance of using FSCV, a traditional neurochemistry technique, to study these neuroimmune mechanisms.
Collapse
Affiliation(s)
- Alexandra K Brooke
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Daniel P Murrow
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Kaejaren C N Caldwell
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Colby E Witt
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Ashley E Ross
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA.
| |
Collapse
|
4
|
Alexandre YO, Mueller SN. Splenic stromal niches in homeostasis and immunity. Nat Rev Immunol 2023; 23:705-719. [PMID: 36973361 DOI: 10.1038/s41577-023-00857-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
The spleen is a gatekeeper of systemic immunity where immune responses against blood-borne pathogens are initiated and sustained. Non-haematopoietic stromal cells construct microanatomical niches in the spleen that make diverse contributions to physiological spleen functions and regulate the homeostasis of immune cells. Additional signals from spleen autonomic nerves also modify immune responses. Recent insight into the diversity of the splenic fibroblastic stromal cells has revised our understanding of how these cells help to orchestrate splenic responses to infection and contribute to immune responses. In this Review, we examine our current understanding of how stromal niches and neuroimmune circuits direct the immunological functions of the spleen, with a focus on T cell immunity.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
He S, Liu J, Xue Y, Fu T, Li Z. Sympathetic Nerves Coordinate Corneal Epithelial Wound Healing by Controlling the Mobilization of Ly6Chi Monocytes From the Spleen to the Injured Cornea. Invest Ophthalmol Vis Sci 2023; 64:13. [PMID: 37682569 PMCID: PMC10500368 DOI: 10.1167/iovs.64.12.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/10/2023] [Indexed: 09/09/2023] Open
Abstract
Purpose This study aims to investigate the potential involvement of spleen-derived monocytes in the repair process following corneal epithelial abrasion. Methods A corneal epithelial abrasion model was established in male C57BL/6J mice, and the dynamic changes of monocyte subpopulations in the injured cornea were analyzed using flow cytometry. The effects of Ly6Chi monocyte depletion and local adoptive transfer of purified Ly6Chi monocytes on wound closure and neutrophil recruitment to the injured cornea were observed. The effect of sympathetic nerves on the recruitment of spleen-derived Ly6Chi monocytes to the injured cornea was also investigated using multiple methods. The emigration of fluorescence-labeled monocytes to the injured cornea was validated through intravital microscopy. Finally, differential genes between different groups were identified through high-throughput RNA sequencing and analyzed for functional enrichment, followed by verification by quantitative PCR. Results Ly6Chi monocytes were present in large numbers in the injured cornea prior to neutrophil recruitment. Predepletion of Ly6Chi monocytes significantly inhibited neutrophil recruitment to the injured cornea. Furthermore, surgical removal of the spleen significantly reduced the number of Ly6Chi monocytes in the injured cornea. Further observations revealed that sympathetic blockade significantly reduced the number of Ly6Chi monocytes recruited to the injured cornea. In contrast, administration of the β2-adrenergic receptor agonist significantly increased the number of Ly6Chi monocytes recruited to the injured cornea in animals treated with sympathectomy and catecholamine synthesis inhibition. Conclusions Our results suggest that spleen-derived Ly6Chi monocytes, under the control of the sympathetic nervous system, play a critical role in the inflammatory response following corneal injury.
Collapse
Affiliation(s)
- Siyu He
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, Guangzhou, China
| | - Jun Liu
- International Ocular Surface Research Center, Institute of Ophthalmology, and Jinan University Medical School, Guangzhou, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Institute of Ophthalmology, and Jinan University Medical School, Guangzhou, China
| | - Ting Fu
- International Ocular Surface Research Center, Institute of Ophthalmology, and Jinan University Medical School, Guangzhou, China
| | - Zhijie Li
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, Guangzhou, China
- International Ocular Surface Research Center, Institute of Ophthalmology, and Jinan University Medical School, Guangzhou, China
| |
Collapse
|
6
|
Bravo-Iñiguez CE, Fritz JR, Shukla S, Sarangi S, Thompson DA, Amin SG, Tsaava T, Chaudhry S, Valentino SP, Hoffman HB, Imossi CW, Addorisio ME, Valdes-Ferrer SI, Chavan SS, Blanc L, Czura CJ, Tracey KJ, Huston JM. Vagus nerve stimulation primes platelets and reduces bleeding in hemophilia A male mice. Nat Commun 2023; 14:3122. [PMID: 37264009 PMCID: PMC10235098 DOI: 10.1038/s41467-023-38505-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Deficiency of coagulation factor VIII in hemophilia A disrupts clotting and prolongs bleeding. While the current mainstay of therapy is infusion of factor VIII concentrates, inhibitor antibodies often render these ineffective. Because preclinical evidence shows electrical vagus nerve stimulation accelerates clotting to reduce hemorrhage without precipitating systemic thrombosis, we reasoned it might reduce bleeding in hemophilia A. Using two different male murine hemorrhage and thrombosis models, we show vagus nerve stimulation bypasses the factor VIII deficiency of hemophilia A to decrease bleeding and accelerate clotting. Vagus nerve stimulation targets acetylcholine-producing T lymphocytes in spleen and α7 nicotinic acetylcholine receptors (α7nAChR) on platelets to increase calcium uptake and enhance alpha granule release. Splenectomy or genetic deletion of T cells or α7nAChR abolishes vagal control of platelet activation, thrombus formation, and bleeding in male mice. Vagus nerve stimulation warrants clinical study as a therapy for coagulation disorders and surgical or traumatic bleeding.
Collapse
Affiliation(s)
- Carlos E Bravo-Iñiguez
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Jason R Fritz
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shilpa Shukla
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Susmita Sarangi
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Dane A Thompson
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA
| | - Seema G Amin
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Tea Tsaava
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Saher Chaudhry
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sara P Valentino
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Hannah B Hoffman
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA
| | - Catherine W Imossi
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Meghan E Addorisio
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sergio I Valdes-Ferrer
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sangeeta S Chavan
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Lionel Blanc
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Departments of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Boulevard, Hempstead, NY, 11549, USA
| | - Christopher J Czura
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Jared M Huston
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA.
- Department of Science Education, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Boulevard, Hempstead, NY, 11549, USA.
| |
Collapse
|
7
|
Bremner JD, Piccinelli M, Garcia EV, Moncayo VM, Elon L, Nye JA, Cooke CD, Washington BP, Ortega RA, Desai SR, Okoh AK, Cheung B, Soyebo BO, Shallenberger LH, Raggi P, Shah AJ, Daaboul O, Jajeh MN, Ziegler C, Driggers EG, Murrah N, De Cecco CN, van Assen M, Krafty RT, Quyyumi AA, Vaccarino V. A Pilot Study of Neurobiological Mechanisms of Stress and Cardiovascular Risk. MEDICAL RESEARCH ARCHIVES 2023; 11:3787. [PMID: 37484871 PMCID: PMC10361343 DOI: 10.18103/mra.v11i4.3787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Objective Coronary heart disease is a leading cause of death and disability. Although psychological stress has been identified as an important potential contributor, mechanisms by which stress increases risk of heart disease and mortality are not fully understood. The purpose of this study was to assess mechanisms by which stress acts through the brain and heart to confer increased CHD risk. Methods Coronary Heart Disease patients (N=10) underwent cardiac imaging with [Tc-99m] sestamibi single photon emission tomography at rest and during a public speaking mental stress task. Patients returned for a second day and underwent positron emission tomography imaging of the brain, heart, bone marrow, aorta (indicating inflammation) and subcutaneous adipose tissue, after injection of [18F]2-fluoro-2-deoxyglucose for assessment of glucose uptake followed mental stress. Patients with (N=4) and without (N=6) mental stress-induced myocardial ischemia were compared for glucose uptake in brain, heart, adipose tissue and aorta with mental stress. Results Patients with mental stress-induced ischemia showed a pattern of increased uptake in the heart, medial prefrontal cortex, and adipose tissue with stress. In the heart disease group as a whole, activity increase with stress in the medial prefrontal brain and amygdala correlated with stress-induced increases in spleen (r=0.69, p=0.038; and r=0.69, p=0.04 respectfully). Stress-induced frontal lobe increased uptake correlated with stress-induced aorta uptake (r=0.71, p=0.016). Activity in insula and medial prefrontal cortex was correlated with post-stress activity in bone marrow and adipose tissue. Activity in other brain areas not implicated in stress did not show similar correlations. Increases in medial prefrontal activity with stress correlated with increased cardiac glucose uptake with stress, suggestive of myocardial ischemia (r=0.85, p=0.004). Conclusions These findings suggest a link between brain response to stress in key areas mediating emotion and peripheral organs involved in inflammation and hematopoietic activity, as well as myocardial ischemia, in Coronary Heart Disease patients.
Collapse
Affiliation(s)
- J. Douglas Bremner
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Marina Piccinelli
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Ernest V. Garcia
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Valeria M. Moncayo
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Lisa Elon
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| | - Jonathon A. Nye
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - C. David Cooke
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Brianna P. Washington
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Rebeca Alvarado Ortega
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Shivang R. Desai
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
| | - Alexis K. Okoh
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
| | - Brian Cheung
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
| | - Britt O. Soyebo
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| | | | - Paolo Raggi
- Mazankowski Alberta Heart Institute and the Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Amit J. Shah
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Emory University, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Obada Daaboul
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| | | | - Carrie Ziegler
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| | | | - Nancy Murrah
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| | - Carlo N. De Cecco
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Department Biomedical Informatics, Emory University School of Medicine, Atlanta, GA
| | - Marly van Assen
- Department Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Robert T. Krafty
- Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Arshed A. Quyyumi
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
| | - Viola Vaccarino
- Department Medicine (Cardiology), Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Carmon H, Haley EC, Parikh V, Tronson NC, Sarter M. Neuro-Immune Modulation of Cholinergic Signaling in an Addiction Vulnerability Trait. eNeuro 2023; 10:ENEURO.0023-23.2023. [PMID: 36810148 PMCID: PMC9997697 DOI: 10.1523/eneuro.0023-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Sign-tracking (ST) describes the propensity to approach and contact a Pavlovian reward cue. By contrast, goal-trackers (GTs) respond to such a cue by retrieving the reward. These behaviors index the presence of opponent cognitive-motivational traits, with STs exhibiting attentional control deficits, behavior dominated by incentive motivational processes, and vulnerability for addictive drug taking. Attentional control deficits in STs were previously attributed to attenuated cholinergic signaling, resulting from deficient translocation of intracellular choline transporters (CHTs) into synaptosomal plasma membrane. Here, we investigated a posttranslational modification of CHTs, poly-ubiquitination, and tested the hypothesis that elevated cytokine signaling in STs contributes to CHT modification. We demonstrated that intracellular CHTs, but not plasma membrane CHTs, are highly ubiquitinated in male and female sign-tracking rats when compared with GTs. Moreover, levels of cytokines measured in cortex and striatum, but not spleen, were higher in STs than in GTs. Activation of the innate immune system by systemic administration of the bacterial endotoxin lipopolysaccharide (LPS) elevated ubiquitinated CHT levels in cortex and striatum of GTs only, suggesting ceiling effects in STs. In spleen, LPS increased levels of most cytokines in both phenotypes. In cortex, LPS particularly robustly increased levels of the chemokines CCL2 and CXCL10. Phenotype-specific increases were restricted to GTs, again suggesting ceiling effects in STs. These results indicate that interactions between elevated brain immune modulator signaling and CHT regulation are essential components of the neuronal underpinnings of the addiction vulnerability trait indexed by sign-tracking.
Collapse
Affiliation(s)
- Hanna Carmon
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109
| | - Evan C Haley
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| | - Vinay Parikh
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109
- Department of Psychology and Neuroscience Program, University of Michigan, Ann Arbor, MI 48109
| | - Martin Sarter
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109
- Department of Psychology and Neuroscience Program, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
9
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
10
|
Alvarez MR, Alarcon JM, Roman CA, Lazaro D, Bobrowski-Khoury N, Baena-Caldas GP, Esber GR. Can a basic solution activate the inflammatory reflex? A review of potential mechanisms, opportunities, and challenges. Pharmacol Res 2023; 187:106525. [PMID: 36441036 DOI: 10.1016/j.phrs.2022.106525] [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: 08/31/2022] [Revised: 10/09/2022] [Accepted: 10/25/2022] [Indexed: 11/24/2022]
Abstract
Stimulation of the inflammatory reflex (IR) is a promising strategy to treat systemic inflammatory disorders. However, this strategy is hindered by the cost and side effects of traditional IR activators. Recently, oral intake of sodium bicarbonate (NaHCO3) has been suggested to activate the IR, providing a safe and inexpensive alternative. Critically, the mechanisms whereby NaHCO3 might achieve this effect and more broadly the pathways underlying the IR remain poorly understood. Here, we argue that the recognition of NaHCO3 as a potential IR activator presents exciting clinical and research opportunities. To aid this quest, we provide an integrative review of our current knowledge of the neural and cellular pathways mediating the IR and discuss the status of physiological models of IR activation. From this vantage point, we derive testable hypotheses on potential mechanisms whereby NaHCO3 might stimulate the IR and compare NaHCO3 with classic IR activators. Elucidation of these mechanisms will help determine the therapeutic value of NaHCO3 as an IR activator and provide new insights into the IR circuitry.
Collapse
Affiliation(s)
- Milena Rodriguez Alvarez
- Department of Internal Medicine, Division of Rheumatology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA.
| | - Juan Marcos Alarcon
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Christopher A Roman
- Department of Cell Biology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Deana Lazaro
- Division of Rheumatology, Department of Internal Medicine, Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY, USA
| | | | | | | |
Collapse
|
11
|
Cremin M, Schreiber S, Murray K, Tay EXY, Reardon C. The diversity of neuroimmune circuits controlling lung inflammation. Am J Physiol Lung Cell Mol Physiol 2023; 324:L53-L63. [PMID: 36410021 PMCID: PMC9829467 DOI: 10.1152/ajplung.00179.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
It is becoming increasingly appreciated that the nervous and immune systems communicate bidirectionally to regulate immunological outcomes in a variety of organs including the lung. Activation of neuronal signaling can be induced by inflammation, tissue damage, or pathogens to evoke or reduce immune cell activation in what has been termed a neuroimmune reflex. In the periphery, these reflexes include the cholinergic anti-inflammatory pathway, sympathetic reflex, and sensory nociceptor-immune cell pathways. Continual advances in neuroimmunology in peripheral organ systems have fueled small-scale clinical trials that have yielded encouraging results for a range of immunopathologies such as rheumatoid arthritis. Despite these successes, several limitations should give clinical investigators pause in the application of neural stimulation as a therapeutic for lung inflammation, especially if inflammation arises from a novel pathogen. In this review, the general mechanisms of each reflex, the evidence for these circuits in the control of lung inflammation, and the key knowledge gaps in our understanding of these neuroimmune circuits will be discussed. These limitations can be overcome not only through a better understanding of neuroanatomy but also through a systematic evaluation of stimulation parameters using immune activation in lung tissues as primary readouts. Our rapidly evolving understanding of the nervous and immune systems highlights the importance of communication between these cells in health and disease. This integrative approach has tremendous potential in the development of targeted therapeutics if specific challenges can be overcome.
Collapse
Affiliation(s)
- Michael Cremin
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Sierra Schreiber
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Kaitlin Murray
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Emmy Xue Yun Tay
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| |
Collapse
|
12
|
Kirkland LG, Garbe CG, Hadaya J, Benson PV, Wagener BM, Tankovic S, Hoover DB. Sympathetic innervation of human and porcine spleens: implications for between species variation in function. Bioelectron Med 2022; 8:20. [PMID: 36536461 PMCID: PMC9762010 DOI: 10.1186/s42234-022-00102-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The vagus nerve affects innate immune responses by activating spleen-projecting sympathetic neurons, which modulate leukocyte function. Recent basic and clinical research investigating vagus nerve stimulation to engage the cholinergic anti-inflammatory pathway (CAP) has shown promising therapeutic results for a variety of inflammatory diseases. Abundant sympathetic innervation occurs in rodent spleens, and use of these species has dominated mechanistic research investigating the CAP. However, previous neuroanatomical studies of human spleen found a more restricted pattern of innervation compared to rodents. Therefore, our primary goal was to establish the full extent of sympathetic innervation of human spleens using donor tissue with the shortest procurement to fixation time. Parallel studies of porcine spleen, a large animal model, were performed as a positive control and for comparison. METHODS Human and porcine spleen tissue were fixed immediately after harvest and prepared for immunohistochemistry. Human heart and porcine spleen were stained in conjunction as positive controls. Several immunohistochemical protocols were compared for best results. Tissue was stained for tyrosine hydroxylase (TH), a noradrenergic marker, using VIP purple chromogen. Consecutive tissue slices were stained for neuropeptide Y (NPY), which often co-localizes with TH, or double-labelled for TH and CD3, a T cell marker. High-magnification images and full scans of the tissue were obtained and analyzed for qualitative differences between species. RESULTS TH had dominant perivascular localization in human spleen, with negligible innervation of parenchyma, but such nerves were abundant throughout ventricular myocardium. In marked contrast, noradrenergic innervation was abundant in all regions of porcine spleen, with red pulp having more nerves than white pulp. NPY stain results were consistent with this pattern. In human spleen, noradrenergic nerves only ran close to T cells at the boundary of the periarterial lymphatic sheath and arteries. In porcine spleen, noradrenergic nerves were closely associated with T cells in both white and red pulp as well as other leukocytes in red pulp. CONCLUSION Sympathetic innervation of the spleen varies between species in both distribution and abundance, with humans and pigs being at opposite extremes. This has important implications for sympathetic regulation of neuroimmune interactions in the spleen of different species and focused targeting of the CAP in humans.
Collapse
Affiliation(s)
- Logan G. Kirkland
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Chloe G. Garbe
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Joseph Hadaya
- grid.19006.3e0000 0000 9632 6718UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA ,grid.19006.3e0000 0000 9632 6718Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA USA
| | - Paul V. Benson
- grid.265892.20000000106344187Department of Pathology, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Brant M. Wagener
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Sanjin Tankovic
- grid.265892.20000000106344187Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL 35249 USA
| | - Donald B. Hoover
- grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA ,grid.255381.80000 0001 2180 1673Department of Biomedical Sciences, Quillen College of Medicine and Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
13
|
Stout DM, Simmons AN, Nievergelt CM, Minassian A, Biswas N, Maihofer AX, Risbrough VB, Baker DG. Deriving psychiatric symptom-based biomarkers from multivariate relationships between psychophysiological and biochemical measures. Neuropsychopharmacology 2022; 47:2252-2260. [PMID: 35347268 PMCID: PMC9630445 DOI: 10.1038/s41386-022-01303-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/18/2022] [Accepted: 02/28/2022] [Indexed: 11/08/2022]
Abstract
Identification of biomarkers for psychiatric disorders remains very challenging due to substantial symptom heterogeneity and diagnostic comorbidity, limiting the ability to map symptoms to underlying neurobiology. Dimensional symptom clusters, such as anhedonia, hyperarousal, etc., are complex and arise due to interactions of a multitude of complex biological relationships. The primary aim of the current investigation was to use multi-set canonical correlation analysis (mCCA) to derive biomarkers (biochemical, physiological) linked to dimensional symptoms across the anxiety and depressive spectrum. Active-duty service members (N = 2,592) completed standardized depression, anxiety and posttraumatic stress questionnaires and several psychophysiological and biochemical assays. Using this approach, we identified two phenotype associations between distinct physiological and biological phenotypes. One was characterized by symptoms of dysphoric arousal (anhedonia, anxiety, hypervigilance) which was associated with low blood pressure and startle reactivity. This finding is in line with previous studies suggesting blunted physiological reactivity is associated with subpopulations endorsing anxiety with comorbid depressive features. A second phenotype of anxious fatigue (high anxiety and reexperiencing/avoidance symptoms coupled with fatigue) was associated with elevated blood levels of norepinephrine and the inflammatory marker C-reactive protein in conjunction with high blood pressure. This second phenotype may describe populations in which inflammation and high sympathetic outflow might contribute to anxious fatigue. Overall, these findings support the growing consensus that distinct neuropsychiatric symptom patterns are associated with differential physiological and blood-based biological profiles and highlight the potential of mCCA to reveal important psychiatric symptom biomarkers from several psychophysiological and biochemical measures.
Collapse
Affiliation(s)
- Daniel M Stout
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Alan N Simmons
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Caroline M Nievergelt
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Arpi Minassian
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nilima Biswas
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dewleen G Baker
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
14
|
Stavely R, Rahman AA, Sahakian L, Prakash MD, Robinson AM, Hassanzadeganroudsari M, Filippone RT, Fraser S, Eri R, Bornstein JC, Apostolopoulos V, Nurgali K. Divergent Adaptations in Autonomic Nerve Activity and Neuroimmune Signaling Associated With the Severity of Inflammation in Chronic Colitis. Inflamm Bowel Dis 2022; 28:1229-1243. [PMID: 35380670 DOI: 10.1093/ibd/izac060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The autonomic nervous system (ANS) is thought to play a critical role in the anti-inflammatory reflex pathway in acute colitis via its interaction with the spleen and colon. Inflammation in the intestine is associated with a blunting of vagal signaling and increased sympathetic activity. As a corollary, methods to restore sympatho-vagal balance are being investigated as therapeutic strategies for the treatment of intestinal inflammation. Nevertheless, it is indefinite whether these autonomic signaling adaptations in colitis are detrimental or beneficial to controlling intestinal inflammation. In this study, models of moderate and severe chronic colitis are utilized to resolve the correlations between sympatho-vagal signaling and the severity of intestinal inflammation. METHODS Spleens and colons were collected from Winnie (moderate colitis), Winnie-Prolapse (severe colitis), and control C57BL/6 mice. Changes to the size and histomorphology of spleens were evaluated. Flow cytometry was used to determine the expression of adrenergic and cholinergic signaling proteins in splenic B and T lymphocytes. The inflammatory profile of the spleen and colon was determined using a RT-PCR gene array. Blood pressure, heart rate, splanchnic sympathetic nerve and vagus nerve activity were recorded. RESULTS Spleens and colons from Winnie and Winnie-Prolapse mice exhibited gross abnormalities by histopathology. Genes associated with a pro-inflammatory response were upregulated in the colons from Winnie and further augmented in colons from Winnie-Prolapse mice. Conversely, many pro-inflammatory markers were downregulated in the spleens from Winnie-Prolapse mice. Heightened activity of the splanchnic nerve was observed in Winnie but not Winnie-Prolapse mice. Conversely, vagal nerve activity was greater in Winnie-Prolapse mice compared with Winnie mice. Splenic lymphocytes expressing α1 and β2 adrenoreceptors were reduced, but those expressing α7 nAChR and producing acetylcholine were increased in Winnie and Winnie-Prolapse mice. CONCLUSIONS Sympathetic activity may correlate with an adaptive mechanism to reduce the severity of chronic colitis. The Winnie and Winnie-Prolapse mouse models of moderate and severe chronic colitis are well suited to examine the pathophysiology of progressive chronic intestinal inflammation.
Collapse
Affiliation(s)
- Rhian Stavely
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmed A Rahman
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lauren Sahakian
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Monica D Prakash
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ainsley M Robinson
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Majid Hassanzadeganroudsari
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rhiannon T Filippone
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rajaraman Eri
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
| | - Joel C Bornstein
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Western Centre for Health Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Kelly MJ, Breathnach C, Tracey KJ, Donnelly SC. Manipulation of the inflammatory reflex as a therapeutic strategy. Cell Rep Med 2022; 3:100696. [PMID: 35858588 PMCID: PMC9381415 DOI: 10.1016/j.xcrm.2022.100696] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 06/20/2021] [Accepted: 06/23/2022] [Indexed: 02/07/2023]
Abstract
The cholinergic anti-inflammatory pathway is the efferent arm of the inflammatory reflex, a neural circuit through which the CNS can modulate peripheral immune responses. Signals communicated via the vagus and splenic nerves use acetylcholine, produced by Choline acetyltransferase (ChAT)+ T cells, to downregulate the inflammatory actions of macrophages expressing α7 nicotinic receptors. Pre-clinical studies using transgenic animals, cholinergic agonists, vagotomy, and vagus nerve stimulation have demonstrated this pathway's role and therapeutic potential in numerous inflammatory diseases. In this review, we summarize what is understood about the inflammatory reflex. We also demonstrate how pre-clinical findings are being translated into promising clinical trials, and we draw particular attention to innovative bioelectronic methods of harnessing the cholinergic anti-inflammatory pathway for clinical use.
Collapse
Affiliation(s)
- Mark J Kelly
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland; Tallaght University Hospital, Dublin, Ireland
| | | | - Kevin J Tracey
- Center for Biomedical Science and Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Seamas C Donnelly
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland; Tallaght University Hospital, Dublin, Ireland.
| |
Collapse
|
16
|
Zhu Y, Duan S, Wang M, Deng Z, Li J. Neuroimmune Interaction: A Widespread Mutual Regulation and the Weapons for Barrier Organs. Front Cell Dev Biol 2022; 10:906755. [PMID: 35646918 PMCID: PMC9130600 DOI: 10.3389/fcell.2022.906755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Since the embryo, the nervous system and immune system have been interacting to regulate each other’s development and working together to resist harmful stimuli. However, oversensitive neural response and uncontrolled immune attack are major causes of various diseases, especially in barrier organs, while neural-immune interaction makes it worse. As the first defense line, the barrier organs give a guarantee to maintain homeostasis in external environment. And the dense nerve innervation and abundant immune cell population in barrier organs facilitate the neuroimmune interaction, which is the physiological basis of multiple neuroimmune-related diseases. Neuroimmune-related diseases often have complex mechanisms and require a combination of drugs, posing challenges in finding etiology and treatment. Therefore, it is of great significance to illustrate the specific mechanism and exact way of neuro-immune interaction. In this review, we first described the mutual regulation of the two principal systems and then focused on neuro-immune interaction in the barrier organs, including intestinal tract, lungs and skin, to clarify the mechanisms and provide ideas for clinical etiology exploration and treatment.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Shixin Duan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Mei Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhili Deng, ; Ji Li,
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhili Deng, ; Ji Li,
| |
Collapse
|
17
|
Nava-Castro KE, Pavón L, Becerril-Villanueva LE, Ponce-Regalado MD, Aguilar-Díaz H, Segovia-Mendoza M, Morales-Montor J. Sexual Dimorphism of the Neuroimmunoendocrine Response in the Spleen during a Helminth Infection: A New Role for an Old Player? Pathogens 2022; 11:pathogens11030308. [PMID: 35335632 PMCID: PMC8955289 DOI: 10.3390/pathogens11030308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 02/05/2023] Open
Abstract
The interaction of the nervous, immune, and endocrine systems is crucial in maintaining homeostasis in vertebrates, and vital in mammals. The spleen is a key organ that regulates the neuroimmunoendocrine system. The Taenia crassiceps mouse system is an excellent experimental model to study the complex host–parasite relationship, particularly sex-associated susceptibility to infection. The present study aimed to determine the changes in neurotransmitters, cytokines, sex steroids, and sex-steroid receptors in the spleen of cysticercus-infected male and female mice and whole parasite counts. We found that parasite load was higher in females in comparison to male mice. The levels of the neurotransmitter epinephrine were significantly decreased in infected male animals. The expression of IL-2 and IL-4 in the spleen was markedly increased in infected mice; however, the expression of Interleukin (IL)-10 and interferon (IFN)-γ decreased. We also observed sex-associated differences between non-infected and infected mice. Interestingly, the data show that estradiol levels increased in infected males but decreased in females. Our studies provide evidence that infection leads to changes in neuroimmunoendocrine molecules in the spleen, and these changes are dimorphic and impact the establishment, growth, and reproduction of T. crassiceps. Our findings support the critical role of the neuroimmunoendocrine network in determining sex-associated susceptibility to the helminth parasite.
Collapse
Affiliation(s)
- Karen Elizabeth Nava-Castro
- Laboratorio de Biología y Química Atmosférica, Departamento de Ciencias Ambientales, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Lenin Pavón
- Laboratory of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Mexico City 14370, Mexico; (L.P.); (L.E.B.-V.)
| | - Luis Enrique Becerril-Villanueva
- Laboratory of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Mexico City 14370, Mexico; (L.P.); (L.E.B.-V.)
| | - María Dolores Ponce-Regalado
- Centro Universitario de los Altos, Departamento de Ciencias de la Salud, Universidad de Guadalajara, Jalisco 47610, Mexico;
| | - Hugo Aguilar-Díaz
- Centro Nacional de Investigaciones Disciplinarias en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales Agrícolas y Pecuarias (INIFAP), Morelos 50550, Mexico;
| | - Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico 00810, Mexico;
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico
- Correspondence: ; Tel.: +52-55-5622-3854 or +52-55-5622-3732; Fax: +52-55-5622-3369
| |
Collapse
|
18
|
Britt NM, Poston MD, Garbe CG, Miller MK, Peeters LD, Wills LJ, Schweitzer JB, Brown RW, Hoover DB. Localization of NGF expression in mouse spleen and salivary gland: Relevance to pleotropic functions. J Neuroimmunol 2022; 366:577846. [DOI: 10.1016/j.jneuroim.2022.577846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
|
19
|
Cleypool CGJ, Brinkman DJ, Mackaaij C, Nikkels PGJ, Nolte MA, Luyer MD, de Jonge WJ, Bleys RLAW. Age-Related Variation in Sympathetic Nerve Distribution in the Human Spleen. Front Neurosci 2021; 15:726825. [PMID: 34720859 PMCID: PMC8552063 DOI: 10.3389/fnins.2021.726825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction: The cholinergic anti-inflammatory pathway (CAIP) has been proposed as an efferent neural pathway dampening the systemic inflammatory response via the spleen. The CAIP activates the splenic neural plexus and a subsequent series of intrasplenic events, which at least require a close association between sympathetic nerves and T cells. Knowledge on this pathway has mostly been derived from rodent studies and only scarce information is available on the innervation of the human spleen. This study aimed to investigate the sympathetic innervation of different structures of the human spleen, the topographical association of nerves with T cells and age-related variations in nerve distribution. Materials and Methods: Spleen samples were retrieved from a diagnostic archive and were allocated to three age groups; neonates, 10–25 and 25–70 years of age. Sympathetic nerves and T cells were identified by immunohistochemistry for tyrosine hydroxylase (TH) and the membrane marker CD3, respectively. The overall presence of sympathetic nerves and T cells was semi-automatically quantified and expressed as total area percentage. A predefined scoring system was used to analyze the distribution of nerves within different splenic structures. Results: Sympathetic nerves were observed in all spleens and their number appeared to slightly increase from birth to adulthood and to decrease afterward. Irrespective to age, more than halve of the periarteriolar lymphatic sheaths (PALSs) contained sympathetic nerves in close association with T cells. Furthermore, discrete sympathetic nerves were observed in the capsule, trabeculae and red pulp and comparable to the total amount of sympathetic nerves, showed a tendency to decrease with age. No correlation was found between the number of T cells and sympathetic nerves. Conclusion: The presence of discrete sympathetic nerves in the splenic parenchyma, capsule and trabecular of human spleens could suggest a role in functions other than vasoregulation. In the PALS, sympathetic nerves were observed to be in proximity to T cells and is suggestive for the existence of the CAIP in humans. Since sympathetic nerve distribution shows interspecies and age-related variation, and our general understanding of the relative and spatial contribution of splenic innervation in immune regulation is incomplete, it remains difficult to estimate the anti-inflammatory potential of targeting splenic nerves in patients.
Collapse
Affiliation(s)
- Cindy G J Cleypool
- Division of Surgical Specialties, Department of Anatomy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - David J Brinkman
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Claire Mackaaij
- Division of Surgical Specialties, Department of Anatomy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Peter G J Nikkels
- Division of Laboratories, Pharmacy, Biomedical Genetics and Pathology, Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Martijn A Nolte
- Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Misha D Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Ronald L A W Bleys
- Division of Surgical Specialties, Department of Anatomy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
20
|
A differential DNA methylome signature of pulmonary immune cells from individuals converting to latent tuberculosis infection. Sci Rep 2021; 11:19418. [PMID: 34593857 PMCID: PMC8484443 DOI: 10.1038/s41598-021-98542-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, spreads via aerosols and the first encounter with the immune system is with the pulmonary-resident immune cells. The role of epigenetic regulations in the immune cells is emerging and we have previously shown that macrophages capacity to kill M. tuberculosis is reflected in the DNA methylome. The aim of this study was to investigate epigenetic modifications in alveolar macrophages and T cells in a cohort of medical students with an increased risk of TB exposure, longitudinally. DNA methylome analysis revealed that a unique DNA methylation profile was present in healthy subjects who later developed latent TB during the study. The profile was reflected in a different overall DNA methylation distribution as well as a distinct set of differentially methylated genes (DMGs). The DMGs were over-represented in pathways related to metabolic reprogramming of macrophages and T cell migration and IFN-γ production, pathways previously reported important in TB control. In conclusion, we identified a unique DNA methylation signature in individuals, with no peripheral immune response to M. tuberculosis antigen who later developed latent TB. Together the study suggests that the DNA methylation status of pulmonary immune cells can reveal who will develop latent TB infection.
Collapse
|
21
|
Cleypool CGJ, Mackaaij C, Verlinde-Schellekens SAMW, Bleys RLAW. A comparative anatomical and histological study on the presence of an apical splenic nerve in mice and humans. J Anat 2021; 240:296-304. [PMID: 34486109 PMCID: PMC8742969 DOI: 10.1111/joa.13541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
The cranial pole of the mouse spleen is considered to be parasympathetically innervated by a macroscopic observable nerve referred to as the apical splenic nerve (ASN). Electrical stimulation of the ASN resulted in increased levels of splenic acetylcholine, decreased lipopolysaccharide-induced levels of systemic tumor necrosis factor alpha and mitigated clinical symptoms in a mouse model of rheumatoid arthritis. If such a discrete ASN would be present in humans, this structure is of interest as it might represent a relatively easily accessible electrical stimulation target to treat immune-mediated inflammatory diseases. So far, it is unknown if a human ASN equivalent exists. This study aimed to provide a detailed description of the location and course of the ASN in mice. Subsequently, this information was used for a guided exploration of an equivalent structure in humans. Microscopic techniques were applied to confirm nerve identity and compare ASN composition. Six mice and six human cadavers were used to study and compare the ASN, both macro- and microscopically. Macroscopic morphological characteristics of the ASN in both mice and humans were described and photographs were taken. ASN samples were resected, embedded in paraffin, cut in 5 μm thin sections where after adjacent sections were stained with a general, sympathetic and parasympathetic nerve marker, respectively. Neural identity and nerve fiber composition was then evaluated microscopically. Macroscopically, the ASN could be clearly identified in all mice and was running in the phrenicosplenic ligament connecting the diaphragm and apical pole of the spleen. If a phrenicosplenic ligament was present in humans, a similar configuration of potential neural structures was observed. Since the gastrosplenic ligament was a continuation of the phrenicosplenic ligament, this ligament was explored as well and contained white, potential discrete nerve-like structures as well which could represent an ANS equivalent. Microscopic evaluation of the ASN in mice and human showed that this structure did not represent a nerve, but most likely connective tissue strains. White nerve-like structures, which could represent the ASN, were macroscopically observed in the phrenicosplenic ligament in both mice and human and in the gastrosplenic ligament in humans. The microscopic investigation did not confirm their neural identity and therefore, this study disclaims the existence of a parasympathetic ASN in both mice and human.
Collapse
Affiliation(s)
- Cindy G J Cleypool
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Claire Mackaaij
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Suzanne A M W Verlinde-Schellekens
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ronald L A W Bleys
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
22
|
Fujiu K, Manabe I. Nerve-macrophage interactions in cardiovascular disease. Int Immunol 2021; 34:81-95. [PMID: 34173833 DOI: 10.1093/intimm/dxab036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/25/2021] [Indexed: 01/09/2023] Open
Abstract
The heart is highly innervated by autonomic neurons, and dynamic autonomic regulation of the heart and blood vessels is essential for animals to carry out the normal activities of life. Cardiovascular diseases, including heart failure and myocardial infarction, are often characterized in part by an imbalance in autonomic nervous system activation, with excess sympathetic and diminished parasympathetic activation. Notably, however, this is often accompanied by chronic inflammation within the cardiovascular tissues, which suggests there are interactions between autonomic dysregulation and inflammation. Recent studies have been unraveling the mechanistic links between autonomic nerves and immune cells within cardiovascular disease. The autonomic nervous system and immune system also act in concert to coordinate the actions of multiple organs that not only maintain homeostasis but also likely play key roles in disease-disease interactions, such as cardiorenal syndrome and multimorbidity. In this review, we summarize the physiological and pathological interactions between autonomic nerves and macrophages in the context of cardiovascular disease.
Collapse
Affiliation(s)
- Katsuhito Fujiu
- Department of Cardiovascular Medicine, the University of Tokyo, Hongo, Bunkyo, Tokyo, Japan.,Department of Advanced Cardiology, the University of Tokyo, Hongo, Bunkyo, Tokyo, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Inohana, Chuo, Chiba, Chiba, Japan
| |
Collapse
|
23
|
Malekpour A, Rahmanifar F. Conventional histomorphometry and fast free of acrylamide clearing tissue (FACT) visualization of sciatic nerve in chicken ( Gallus domesticus). VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2021; 12:167-173. [PMID: 34345382 PMCID: PMC8328249 DOI: 10.30466/vrf.2019.98930.2361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/17/2019] [Indexed: 11/06/2022]
Abstract
Histomorphometry and use of the fast free of acrylamide clearing tissue (FACT) protocol were studied on the sciatic nerve in chicken (Gallus domesticus). In the first part of the study, the sciatic nerves of 20 chickens of four age groups (7, 14, 26 and 40 days) were studied (n=5 birds per age class). Their sciatic nerve samples were stained with Hematoxylin and Eosin and Masson's trichrome and were histomorphometrically evaluated. In the second part of the study, FACT protocol was applied on the sciatic nerve of a 26 days old chicken. After clearing of 1.00 mm-thick sciatic nerve sections, they were immunolabelled using Hoechst for nuclei staining and recorded by a Z-stack motorized fluorescent microscope. In the conventional histo-morphometry, the epineurium, perineurium and endoneurium were thicker and the nerve bundle diameter was bigger in the left sciatic nerve of chicken of all age groups compared to the right sciatic nerve. On the contrary, the axon diameter and the myelinated nerve fiber diameter were bigger, the myelin sheath was thicker, the nodes of Ranvier intervals were higher and the density of myelinated nerve fibers was also higher in the right sciatic nerve compared to the left one. In conclusion, histomorphometric parameters in the left and right sciatic nerve during chicken growth were significantly different. Furthermore, the FACT protocol could be used for the 3D imaging of the chicken sciatic nerve and its immunostained evaluation.
Collapse
Affiliation(s)
- Abdolrasoul Malekpour
- DVM Graduate, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.,Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
24
|
Ueno M. Restoring neuro-immune circuitry after brain and spinal cord injuries. Int Immunol 2021; 33:311-325. [PMID: 33851981 DOI: 10.1093/intimm/dxab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Neuro-immune interactions are essential for our body's defense and homeostasis. Anatomical and physiological analyses have shown that the nervous system comprises multiple pathways that regulate the dynamics and functions of immune cells, which are mainly mediated by the autonomic nervous system and adrenal signals. These are disturbed when the neurons and circuits are damaged by diseases of the central nervous system (CNS). Injuries caused by stroke or trauma often cause immune dysfunction by abrogation of the immune-regulating neural pathways, which leads to an increased risk of infections. Here, I review the structures and functions of the neural pathways connecting the brain and the immune system, and the neurogenic mechanisms of immune dysfunction that emerge after CNS injuries. Recent technological advances in manipulating specific neural circuits have added mechanistic aspects of neuro-immune interactions and their dysfunctions. Understanding the neural bases of immune control and their pathological processes will deepen our knowledge of homeostasis and lead to the development of strategies to cure immune deficiencies observed in various CNS disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan
| |
Collapse
|
25
|
Cleypool CGJ, Mackaaij C, Lotgerink Bruinenberg D, Schurink B, Bleys RLAW. Sympathetic nerve distribution in human lymph nodes. J Anat 2021; 239:282-289. [PMID: 33677834 PMCID: PMC8273593 DOI: 10.1111/joa.13422] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/01/2022] Open
Abstract
Various lymph node functions are regulated by the sympathetic nervous system as shown in rodent studies. If human lymph nodes show a comparable neural regulation, their afferent nerves could represent a potential therapeutic target to treat, for example, infectious or autoimmune disease. Little information is available on human lymph node innervation and the aim of this study is to establish a comprehensive and accurate representation of the presence and location of sympathetic nerves in human lymph nodes. Since previous studies mention sympathetic paravascular nerves to occasionally extent into T cell‐rich regions, the relation of these nerves with T cells was studied as well. A total number of 15 inguinal lymph nodes were resected from six donated human cadavers. Lymph node sections were stained with HE and a double T/B cell staining for evaluation of their morphology and to screen for general pathologies. A triple stain was used to identify blood vessels, sympathetic nerves and T cells, and, to study the presence and location of sympathetic nerves and their relation to T cells. To evaluate whether the observed nerves were en route to other structures or were involved in local processes, adjacent slides were stained with a marker for varicosities (synaptophysin), which presence is suggestive for synaptic activity. All lymph nodes contained sympathetic nerves, both as paravascular and discrete structures. In 15/15 lymph nodes, nerves were observed in their capsule, medulla and hilum, whereas only 13/15 lymph nodes contained nerves in their cortex. The amount of sympathetic nerves varied between compartments and between and within individuals. In general, if a lymph node contained more paravascular nerves in a specific compartment, more discrete nerves were observed as well. Occasionally, discrete nerves were observed in relation to T cells in lymphoid tissues of the cortex and medulla. Furthermore, discrete nerves were frequently present in the capsule and hilum. The presence of varicosities in a portion of these nerves, independently to their compartment, suggested a local regulatory function for these nerves. Human lymph nodes contain sympathetic nerves in their capsule, trabeculae, cortex, medulla and hilum, both as paravascular or as discrete structures. Discrete nerves were observed in relation to T cells and non‐T cell‐rich areas such as the hilar and capsular connective tissue. The presence of discrete structures suggests neural regulation of structures other than blood vessels, which was further supported by the presence of varicosities in a portion of these nerves. These observations are of relevance in further understanding neural regulation of lymph node immune responses and in the development of neuromodulatory immune therapies.
Collapse
Affiliation(s)
- Cindy G J Cleypool
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Claire Mackaaij
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Dyonne Lotgerink Bruinenberg
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bernadette Schurink
- Department of Pathology, Amsterdam University Medical Centre, Free University of Amsterdam, Amsterdam, the Netherlands
| | - Ronald L A W Bleys
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
26
|
Shouman K, Benarroch EE. Peripheral neuroimmune interactions: selected review and some clinical implications. Clin Auton Res 2021; 31:477-489. [PMID: 33641054 PMCID: PMC7914391 DOI: 10.1007/s10286-021-00787-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Purpose To provide a brief and focused review on peripheral neuroimmune interactions and their implications for some clinical disorders. Methods Narrative review of the literature including of English-language articles published between 1985 and 2021 using PubMed and MEDLINE. Results Many studies on experimental models and in vitro indicate that there are close interactions between the neural and immune systems. Processes from sensory afferents and autonomic efferents co-localize with immune cells and interact at discrete anatomical sites forming neuroimmune units. These neuroimmune interactions are bidirectional and mediated by a wide range of soluble factors including neuropeptides, classical neurotransmitters, cytokines, and other molecules that mediate complex cross-talk among nerves and immune cells. Small-diameter sensory afferents express a wide range of receptors that respond directly to tissue damage or pathogen signals and to chemokines, cytokines, or other molecules released from immune cells. Reciprocally, immune cells respond to neurotransmitters released from nociceptive and autonomic fibers. Neuroimmune interactions operate both at peripheral tissues and at the level of the central nervous system. Both centrally and peripherally, glial cells have a major active role in this bidirectional communication. Conclusions Peripheral neuroimmune interactions are complex and importantly contribute to the pathophysiology of several disorders, including skin, respiratory, and intestinal inflammatory disorders typically associated with pain and altered barrier function. These interactions may be relevant for persistence of symptoms in disorders associated with intense immune activation.
Collapse
Affiliation(s)
- Kamal Shouman
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Eduardo E Benarroch
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
27
|
Diaz-Salazar C, Bou-Puerto R, Mujal AM, Lau CM, von Hoesslin M, Zehn D, Sun JC. Cell-intrinsic adrenergic signaling controls the adaptive NK cell response to viral infection. J Exp Med 2020; 217:133715. [PMID: 32045471 PMCID: PMC7144534 DOI: 10.1084/jem.20190549] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/23/2019] [Accepted: 01/13/2020] [Indexed: 12/26/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that exhibit adaptive features, such as clonal expansion and memory, during viral infection. Although activating receptor engagement and proinflammatory cytokines are required to drive NK cell clonal expansion, additional stimulatory signals controlling their proliferation remain to be discovered. Here, we describe one such signal that is provided by the adrenergic nervous system, and demonstrate that cell-intrinsic adrenergic signaling is required for optimal adaptive NK cell responses. Early during mouse cytomegalovirus (MCMV) infection, NK cells up-regulated Adrb2 (which encodes the β2-adrenergic receptor), a process dependent on IL-12 and STAT4 signaling. NK cell-specific deletion of Adrb2 resulted in impaired NK cell expansion and memory during MCMV challenge, in part due to a diminished proliferative capacity. As a result, NK cell-intrinsic adrenergic signaling was required for protection against MCMV. Taken together, we propose a novel role for the adrenergic nervous system in regulating circulating lymphocyte responses to viral infection.
Collapse
Affiliation(s)
- Carlos Diaz-Salazar
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY
| | - Regina Bou-Puerto
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY
| | - Adriana M Mujal
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Colleen M Lau
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Madlaina von Hoesslin
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY.,Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
28
|
Salvo E, Stokes P, Keogh CE, Brust-Mascher I, Hennessey C, Knotts TA, Sladek JA, Rude KM, Swedek M, Rabasa G, Gareau MG. A murine model of pediatric inflammatory bowel disease causes microbiota-gut-brain axis deficits in adulthood. Am J Physiol Gastrointest Liver Physiol 2020; 319:G361-G374. [PMID: 32726162 PMCID: PMC7509259 DOI: 10.1152/ajpgi.00177.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel diseases (IBDs) are chronic intestinal diseases, frequently associated with comorbid psychological and cognitive deficits. These neuropsychiatric effects include anxiety, depression, and memory impairments that can be seen both during active disease and following remission and are more frequently seen in pediatric patients. The mechanism(s) through which these extraintestinal deficits develop remain unknown, and the study of these phenomenon is hampered by a lack of murine pediatric IBD models. Herein we describe microbiota-gut-brain (MGB) axis deficits following induction of colitis in a pediatric setting. Acute colitis was induced by administration of 2% dextran sodium sulfate (DSS) for 5 days starting at weaning [postnatal day (P)21] causing reduced weight gain, colonic shortening, and colonic inflammation by 8 days post-DSS (P29), which were mostly resolved in adult (P56) mice. Despite resolution of acute disease, cognitive deficits (novel object recognition task) and anxiety-like behavior (light/dark box) were identified in the absence of changes in exploratory behavior (open field test) in P56 mice previously treated with DSS at weaning. Behavioral deficits were found in conjunction with neuroinflammation, decreased neurogenesis, and altered expression of pattern recognition receptor genes in the hippocampus. Additionally, persistent alterations in the gut microbiota composition were observed at P56, including reduced butyrate-producing species. Taken together, these results describe for the first time the presence of MGB axis deficits following induction of colitis at weaning, which persist in adulthood.NEW & NOTEWORTHY Here we describe long-lasting impacts on the microbiota-gut-brain (MGB) axis following administration of low-dose dextran sodium sulfate (DSS) to weaning mice (P21), including gut dysbiosis, colonic inflammation, and brain/behavioral deficits in adulthood (P56). Early-life DSS leads to acute colonic inflammation, similar to adult mice; however, it results in long-lasting deficits in the MGB axis in adulthood (P56), in contrast to the transient deficits seen in adult DSS. This model highlights the unique features of pediatric inflammatory bowel disease.
Collapse
Affiliation(s)
- Eloisa Salvo
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Patricia Stokes
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Ciara E. Keogh
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Ingrid Brust-Mascher
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Carly Hennessey
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Trina A. Knotts
- 2Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California
| | - Jessica A. Sladek
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Kavi M. Rude
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Michelle Swedek
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Gonzalo Rabasa
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| | - Mélanie G. Gareau
- 1Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California
| |
Collapse
|
29
|
Cai J, Nash WT, Okusa MD. Ultrasound for the treatment of acute kidney injury and other inflammatory conditions: a promising path toward noninvasive neuroimmune regulation. Am J Physiol Renal Physiol 2020; 319:F125-F138. [PMID: 32508112 PMCID: PMC7468827 DOI: 10.1152/ajprenal.00145.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury (AKI) is an important clinical disorder with high prevalence, serious consequences, and limited therapeutic options. Modulation of neuroimmune interaction by nonpharmacological methods is emerging as a novel strategy for treating inflammatory diseases, including AKI. Recently, pulsed ultrasound (US) treatment was shown to protect from AKI by stimulating the cholinergic anti-inflammatory pathway. Because of the relatively simple, portable, and noninvasive nature of US procedures, US stimulation may be a valuable therapeutic option for treating inflammatory conditions. This review discusses potential impacts of US bioeffects on the nervous system and how this may generate feedback onto the immune system. We also discuss recent evidence supporting the use of US as a means to treat AKI and other inflammatory diseases.
Collapse
Affiliation(s)
- Jieru Cai
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virgnia
| | - William T Nash
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virgnia
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virgnia
| |
Collapse
|
30
|
Distribution of nerve fibers and nerve-immune cell association in mouse spleen revealed by immunofluorescent staining. Sci Rep 2020; 10:9850. [PMID: 32555231 PMCID: PMC7300136 DOI: 10.1038/s41598-020-66619-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/21/2020] [Indexed: 02/05/2023] Open
Abstract
The central nervous system regulates the immune system through the secretion of hormones from the pituitary gland and other endocrine organs, while the peripheral nervous system (PNS) communicates with the immune system through local nerve-immune cell interactions, including sympathetic/parasympathetic (efferent) and sensory (afferent) innervation to lymphoid tissue/organs. However, the precise mechanisms of this bi-directional crosstalk of the PNS and immune system remain mysterious. To study this kind of bi-directional crosstalk, we performed immunofluorescent staining of neurofilament and confocal microscopy to reveal the distribution of nerve fibers and nerve-immune cell associations inside mouse spleen. Our study demonstrates (i) extensive nerve fibers in all splenic compartments including the splenic nodules, periarteriolar lymphoid sheath, marginal zones, trabeculae, and red pulp; (ii) close associations of nerve fibers with blood vessels (including central arteries, marginal sinuses, penicillar arterioles, and splenic sinuses); (iii) close associations of nerve fibers with various subsets of dendritic cells, macrophages (Mac1+ and F4/80+), and lymphocytes (B cells, T helper cells, and cytotoxic T cells). Our data concerning the extensive splenic innervation and nerve-immune cell communication will enrich our knowledge of the mechanisms through which the PNS affects the cellular- and humoral-mediated immune responses in healthy and infectious/non-infectious states.
Collapse
|
31
|
Wei Y, Liang Y, Lin H, Dai Y, Yao S. Autonomic nervous system and inflammation interaction in endometriosis-associated pain. J Neuroinflammation 2020; 17:80. [PMID: 32145751 PMCID: PMC7060607 DOI: 10.1186/s12974-020-01752-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Endometriosis is a chronic inflammatory disease. Pain is the most common symptom in endometriosis. Endometriosis-associated pain is caused by inflammation, and is related to aberrant innervation. Although the specific mechanism between endometriosis-associated pain and the interaction of aberrant innervation and inflammation remains unclear, many studies have confirmed certain correlations between them. In addition, we found that some chronic inflammatory autoimmune diseases (AIDs) such as inflammatory bowel disease (IBD) and rheumatoid arthritis (RA) share similar characteristics: the changes in dysregulation of inflammatory factors as well as the function and innervation of the autonomic nervous system (ANS). The mechanisms underlying the interaction between the ANS and inflammation have provided new advances among these disorders. Therefore, the purpose of this review is to compare the changes in inflammation and ANS in endometriosis, IBD, and RA; and to explore the role and possible mechanism of sympathetic and parasympathetic nerves in endometriosis-associated inflammation by referring to IBD and RA studies to provide some reference for further endometriosis research and treatment.
Collapse
Affiliation(s)
- Yajing Wei
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Haishan Lin
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510089, China
| | - Yujing Dai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510089, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
32
|
Malin SG, Shavva VS, Tarnawski L, Olofsson PS. Functions of acetylcholine-producing lymphocytes in immunobiology. Curr Opin Neurobiol 2020; 62:115-121. [PMID: 32126362 DOI: 10.1016/j.conb.2020.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/26/2022]
Abstract
Recent advances in neuroscience and immunology have shown that cholinergic signals are vital in the regulation of inflammation and immunity. Choline acetyltransferase+ (ChAT+) lymphocytes have the capacity to biosynthesize and release acetylcholine, the cognate ligand for cholinergic receptors. Acetylcholine-producing T cells relay neural signals in the 'inflammatory reflex' that regulate cytokine release in spleen. Mice deficient in acetylcholine-producing T cells have increased blood pressure, show reduced local vasodilatation and viral control in lymphocytic choriomeningitis virus infection, and display changes in gut microbiota compared with littermates. These observations indicate that ChAT+ lymphocytes play physiologically important roles in regulation of inflammation and anti-microbial defense. However, the full scope and importance of ChAT+ lymphocytes in immunity and vascular biology remains to be elucidated. Here, we review key findings in this emerging area.
Collapse
Affiliation(s)
- Stephen G Malin
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vladmir S Shavva
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laura Tarnawski
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Regulatory B cells in infection, inflammation, and autoimmunity. Cell Immunol 2020; 352:104076. [PMID: 32143836 DOI: 10.1016/j.cellimm.2020.104076] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Regulatory B (Breg) cells are characterized by differential expression of CD5 and CD1d in mouse and CD24 and CD38 in human immune systems. The Breg family also includes LAG-3+CD138hi plasma cells, CD1d CD5 CD21 CD23 cells, Tim1, PD-L1, PD-L2, CD200- expressing B cells, and CD39hiKi67+ cells originating from the transitional, marginal zone or germinal centre of the spleen. Breg cells produce IL10 and IL35 and to cause immunosuppression. These cells respond to TLR2, TLR4, and TLR9 agonists, CD40 ligands, IL12p35 and heat shock proteins. Emerging evidence suggests that TLR signalling component Myd88 impacts the modulation of Breg cell responses and the host's susceptibility to infection. Breg cells are found to reduce relapsing-remitting experimental autoimmune encephalomyelitis. However, the Breg-mediated mechanism used to control T cell-mediated immune responses is still unclear. Here, we review the existing literature to find gaps in the current knowledge and to build a pathway to further research.
Collapse
|
34
|
Bonaz B. The vagus nerve and the sympathetic nervous system act in concert to modulate immunity. Brain Behav Immun 2020; 84:6-7. [PMID: 31785397 DOI: 10.1016/j.bbi.2019.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 01/03/2023] Open
Affiliation(s)
- Bruno Bonaz
- Service d'Hépato-Gastroentérologie, CHU Grenoble Alpes, 38000 Grenoble, France; University Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000 Grenoble, France.
| |
Collapse
|
35
|
Ren C, Yao RQ, Zhang H, Feng YW, Yao YM. Sepsis-associated encephalopathy: a vicious cycle of immunosuppression. J Neuroinflammation 2020; 17:14. [PMID: 31924221 PMCID: PMC6953314 DOI: 10.1186/s12974-020-1701-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is commonly complicated by septic conditions, and is responsible for increased mortality and poor outcomes in septic patients. Uncontrolled neuroinflammation and ischemic injury are major contributors to brain dysfunction, which arises from intractable immune malfunction and the collapse of neuroendocrine immune networks, such as the cholinergic anti-inflammatory pathway, hypothalamic-pituitary-adrenal axis, and sympathetic nervous system. Dysfunction in these neuromodulatory mechanisms compromised by SAE jeopardizes systemic immune responses, including those of neutrophils, macrophages/monocytes, dendritic cells, and T lymphocytes, which ultimately results in a vicious cycle between brain injury and a progressively aberrant immune response. Deep insight into the crosstalk between SAE and peripheral immunity is of great importance in extending the knowledge of the pathogenesis and development of sepsis-induced immunosuppression, as well as in exploring its effective remedies.
Collapse
Affiliation(s)
- Chao Ren
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, People's Republic of China
| | - Ren-Qi Yao
- Department of Burn Surgery, Changhai Hospital, The Navy Medical University, Shanghai, 200433, People's Republic of China
| | - Hui Zhang
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, People's Republic of China
| | - Yong-Wen Feng
- Department of Critical Care Medicine, The Second People's Hospital of Shenzhen, Shenzhen, 518035, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, People's Republic of China.
| |
Collapse
|
36
|
Pusceddu MM, Barboza M, Keogh CE, Schneider M, Stokes P, Sladek JA, Kim HJD, Torres-Fuentes C, Goldfild LR, Gillis SE, Brust-Mascher I, Rabasa G, Wong KA, Lebrilla C, Byndloss MX, Maisonneuve C, Bäumler AJ, Philpott DJ, Ferrero RL, Barrett KE, Reardon C, Gareau MG. Nod-like receptors are critical for gut-brain axis signalling in mice. J Physiol 2019; 597:5777-5797. [PMID: 31652348 DOI: 10.1113/jp278640] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS •Nucleotide binding oligomerization domain (Nod)-like receptors regulate cognition, anxiety and hypothalamic-pituitary-adrenal axis activation. •Nod-like receptors regulate central and peripheral serotonergic biology. •Nod-like receptors are important for maintenance of gastrointestinal physiology. •Intestinal epithelial cell expression of Nod1 receptors regulate behaviour. ABSTRACT Gut-brain axis signalling is critical for maintaining health and homeostasis. Stressful life events can impact gut-brain signalling, leading to altered mood, cognition and intestinal dysfunction. In the present study, we identified nucleotide binding oligomerization domain (Nod)-like receptors (NLR), Nod1 and Nod2, as novel regulators for gut-brain signalling. NLR are innate immune pattern recognition receptors expressed in the gut and brain, and are important in the regulation of gastrointestinal physiology. We found that mice deficient in both Nod1 and Nod2 (NodDKO) demonstrate signs of stress-induced anxiety, cognitive impairment and depression in the context of a hyperactive hypothalamic-pituitary-adrenal axis. These deficits were coupled with impairments in the serotonergic pathway in the brain, decreased hippocampal cell proliferation and immature neurons, as well as reduced neural activation. In addition, NodDKO mice had increased gastrointestinal permeability and altered serotonin signalling in the gut following exposure to acute stress. Administration of the selective serotonin reuptake inhibitor, fluoxetine, abrogated behavioural impairments and restored serotonin signalling. We also identified that intestinal epithelial cell-specific deletion of Nod1 (VilCre+ Nod1f/f ), but not Nod2, increased susceptibility to stress-induced anxiety-like behaviour and cognitive impairment following exposure to stress. Together, these data suggest that intestinal epithelial NLR are novel modulators of gut-brain communication and may serve as potential novel therapeutic targets for the treatment of gut-brain disorders.
Collapse
Affiliation(s)
- Matteo M Pusceddu
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Mariana Barboza
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Ciara E Keogh
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Melinda Schneider
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Patricia Stokes
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Jessica A Sladek
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Hyun Jung D Kim
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Cristina Torres-Fuentes
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA.,Department of Food Science & Technology, University of California Davis, Davis, CA, USA
| | - Lily R Goldfild
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Shane E Gillis
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Gonzalo Rabasa
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Kyle A Wong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Carlito Lebrilla
- Department of Chemistry, University of California Davis, Davis, CA, USA
| | - Mariana X Byndloss
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, USA
| | | | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, USA
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Richard L Ferrero
- Hudson Institute of Medical Research, Department of Molecular and Translational Science and Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Kim E Barrett
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Colin Reardon
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Mélanie G Gareau
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
37
|
Splenic artery denervation: target micro-anatomy, feasibility, and early preclinical experience. Transl Res 2019; 213:100-111. [PMID: 31415732 DOI: 10.1016/j.trsl.2019.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 01/28/2023]
Abstract
This study sought to evaluate perisplenic artery nerve distribution and the feasibility of splenic artery denervation (SDN). The NEXION radiofrequency catheter was used to perform SDN in healthy and inflammatory arthritis pigs. Splenic artery anatomy, nerve distribution, and splenic norepinephrine (NEPI) levels were evaluated before and after SDN. Perisplenic artery nerves were primarily distributed within 2.5 mm of the arterial lumen and were largely sympathetic on the basis of tyrosine hydroxylase expression. The pancreas, tended to be circumferentially positioned around the proximal splenic artery, typically >2.5 mm from the lumen, ensuring that most of the nerves could be targeted without affecting this sensitive organ. The mid segment of the splenic artery was relatively free of contact with the adjacent pancreas. Splenic NEPI levels and nerve abundance followed a decreasing gradient from the proximal to distal splenic artery. SDN resulted in significant reductions in splenic NEPI levels at day 14 (60.7%, P = 0.024) in naïve pigs and day 45 (100%, P = 0.001) in inflammatory arthritis pigs. There was no significant effect of SDN on joint soft tissue injury or circulating inflammatory markers in the inflammatory arthritis model. The majority of perisplenic arterial nerves are within close proximity of the lumen and are primarily sympathetic efferent fibers. Nerves in the mid-segment may be the preferred SDN target given their proximity to the artery and paucity of periarterial off-target organs. SDN appears safe and effective at reducing splenic NEPI levels.
Collapse
|
38
|
Murray K, Barboza M, Rude KM, Brust-Mascher I, Reardon C. Functional circuitry of neuro-immune communication in the mesenteric lymph node and spleen. Brain Behav Immun 2019; 82:214-223. [PMID: 31445965 PMCID: PMC6800652 DOI: 10.1016/j.bbi.2019.08.188] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022] Open
Abstract
The peripheral nervous system is an active participant in immune responses capable of blocking aberrant activation of a variety of immune cells. As one of these neuro-immune circuits, the cholinergic anti-inflammatory pathway has been well established to reduce the severity of several immunopathologies. While the activation of this pathway by vagal nerve stimulation requires sympathetic innervation of the spleen, the neuro-immune circuitry remains highly controversial. Neuro-immune pathways in other lymphoid tissues such as mesenteric lymph nodes (MLN) that are critical to the surveillance of the small intestine and proximal colon have not been assessed. Using conditionally expressed Channelrhodopsin, selective stimulation of sympathetic post-ganglionic neurons in the superior mesenteric ganglion (SMG) prevented macrophage activation and LPS-induced TNFα production in the spleen and MLN, but not in the inguinal LN. Site selective stimulation of the SMG induced the release of norepinephrine, resulting in β2AR dependent acetylcholine release in the MLN and spleen. VNS-evoked release of norepinephrine and acetylcholine in the MLN and spleen was significantly reduced using selective optogenetic blockade applied at the SMG. Additionally, this optogenetic blockade restored LPS-induced TNFα production, despite VNS. These studies identify the superior mesenteric ganglion as a critical node in a neuro-immune circuit that can inhibit immune function in the MLN and the spleen.
Collapse
Affiliation(s)
- Kaitlin Murray
- Department. of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Mariana Barboza
- Department. of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Kavi M. Rude
- Department. of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Ingrid Brust-Mascher
- Department. of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
| |
Collapse
|
39
|
Neuroimmune Interactions in the Gut and Their Significance for Intestinal Immunity. Cells 2019; 8:cells8070670. [PMID: 31269754 PMCID: PMC6679154 DOI: 10.3390/cells8070670] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBD) have a complex, multifactorial pathophysiology with an unmet need for effective treatment. This calls for novel strategies to improve disease outcome and quality of life for patients. Increasing evidence suggests that autonomic nerves and neurotransmitters, as well as neuropeptides, modulate the intestinal immune system, and thereby regulate the intestinal inflammatory processes. Although the autonomic nervous system is classically divided in a sympathetic and parasympathetic branch, both play a pivotal role in the crosstalk with the immune system, with the enteric nervous system acting as a potential interface. Pilot clinical trials that employ vagus nerve stimulation to reduce inflammation are met with promising results. In this paper, we review current knowledge on the innervation of the gut, the potential of cholinergic and adrenergic systems to modulate intestinal immunity, and comment on ongoing developments in clinical trials.
Collapse
|
40
|
Bleck D, Ma L, Erdene-Bymbadoo L, Brinks R, Schneider M, Tian L, Pongratz G. Introduction and validation of a new semi-automated method to determine sympathetic fiber density in target tissues. PLoS One 2019; 14:e0217475. [PMID: 31141555 PMCID: PMC6541301 DOI: 10.1371/journal.pone.0217475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/12/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years, the role of sympathetic nervous fibers in chronic inflammation has become increasingly evident. At the onset of inflammation, sympathetic activity is increased in the affected tissue. However, sympathetic fibers are largely absent from chronically inflamed tissue. Apparently, there is a very dynamic relationship between sympathetic innervation and the immune system in areas of inflammation, and hence a rapid and easy method for quantification of nerve fiber density of target organs is of great value to answer potential research questions. Currently, nervous fiber densities are either determined by tedious manual counting, which is not suitable for high throughput approaches, or by expensive automated processes relying on specialized software and high-end microscopy equipment. Usually, tyrosine hydroxylase (TH) is used as the marker for sympathetic fibers. In order to overcome the current quantification bottleneck with a cost-efficient alternative, an automated process was established and compared to the classic manual approach of counting TH-positive sympathetic fibers. Since TH is not exclusively expressed on sympathetic fibers, but also in a number of catecholamine-producing cells, a prerequisite for automated determination of fiber densities is to reliably distinct between cells and fibers. Therefore, an additional staining using peripherin exclusively expressed in nervous fibers as a secondary marker was established. Using this novel approach, we studied the spleens from a syndecan-3 knockout (SDC3KO) mouse line, and demonstrated equal results on SNS fiber density for both manual and automated counts (Manual counts: wildtype: 22.57 +/- 11.72 fibers per mm2; ko: 31.95 +/- 18.85 fibers per mm2; p = 0.05; Automated counts: wildtype: 31.6 +/- 18.98 fibers per mm2; ko: 45.49 +/- 19.65 fibers per mm2; p = 0.02). In conclusion, this new and simple method can be used as a high-throughput approach to reliably and quickly estimate SNS nerve fiber density in target tissues.
Collapse
Affiliation(s)
- Dennis Bleck
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Li Ma
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lkham Erdene-Bymbadoo
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ralph Brinks
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Schneider
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Li Tian
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine and Translational Medicine, Department of Physiology, Faculty of Medicine, University of Tartu, Tartu, Estonia
- * E-mail: (GP); (LT)
| | - Georg Pongratz
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail: (GP); (LT)
| |
Collapse
|
41
|
Splenic Blood Flow Increases after Hypothermic Stimulus (Cold Pressor Test): A Perfusion Magnetic Resonance Study. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8437927. [PMID: 31240227 PMCID: PMC6556242 DOI: 10.1155/2019/8437927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/03/2019] [Accepted: 05/06/2019] [Indexed: 02/01/2023]
Abstract
The Cold Pressor Test (CPT) is a novel diagnostic strategy to noninvasively assess the myocardial microvascular endothelial-dependent function using perfusion magnetic resonance imaging (MRI). Spleen perfusion is modulated by a complex combination of several mechanisms involving the autonomic nervous system and vasoactive mediators release. In this context, the effects of cold temperature on splenic blood flow (SBF) still need to be clarified. Ten healthy subjects were studied by MRI. MRI protocol included the acquisition of GRE T1-weighted sequence (“first pass perfusion”) during gadolinium administration (0.1mmol/kg of Gd-DOTA at flow of 3.0 ml/s), at rest and after CPT. Myocardial blood flow (MBF) and SBF were measured by applying Fermi function deconvolution, using the blood pool input function sampled from the left ventricle cavity. MBF and SBF values after performing CPT were significantly higher than rest values (SBF at rest: 0.65 ± 0.15 ml/min/g Vs. SBF after CPT: 0.90 ± 0.14 ml/min/g, p: <0.001; MBF at rest: 0.90 ± 0.068 ml/min/g Vs. MBF after CPT: 1.22 ± 0.098 ml/min/g, p<0.005). Both SBF and MBF increased in all patients during the CPT. In particular, the CPT-induced increase was 43% ± 29% for SBF and 36.5% ± 17% for MBF. CPT increases SBF in normal subjects. The characterization of a standard perfusion response to cold might allow the use of the spleen as reference marker for the adequacy of cold stimulation during myocardial perfusion MRI.
Collapse
|
42
|
Ramirez VT, Godinez DR, Brust-Mascher I, Nonnecke EB, Castillo PA, Gardner MB, Tu D, Sladek JA, Miller EN, Lebrilla CB, Bevins CL, Gareau MG, Reardon C. T-cell derived acetylcholine aids host defenses during enteric bacterial infection with Citrobacter rodentium. PLoS Pathog 2019; 15:e1007719. [PMID: 30973939 PMCID: PMC6478367 DOI: 10.1371/journal.ppat.1007719] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/23/2019] [Accepted: 03/20/2019] [Indexed: 12/14/2022] Open
Abstract
The regulation of mucosal immune function is critical to host protection from enteric pathogens but is incompletely understood. The nervous system and the neurotransmitter acetylcholine play an integral part in host defense against enteric bacterial pathogens. Here we report that acetylcholine producing-T-cells, as a non-neuronal source of ACh, were recruited to the colon during infection with the mouse pathogen Citrobacter rodentium. These ChAT+ T-cells did not exclusively belong to one Th subset and were able to produce IFNγ, IL-17A and IL-22. To interrogate the possible protective effect of acetylcholine released from these cells during enteric infection, T-cells were rendered deficient in their ability to produce acetylcholine through a conditional gene knockout approach. Significantly increased C. rodentium burden was observed in the colon from conditional KO (cKO) compared to WT mice at 10 days post-infection. This increased bacterial burden in cKO mice was associated with increased expression of the cytokines IL-1β, IL-6, and TNFα, but without significant changes in T-cell and ILC associated IL-17A, IL-22, and IFNγ, or epithelial expression of antimicrobial peptides, compared to WT mice. Despite the increased expression of pro-inflammatory cytokines during C. rodentium infection, inducible nitric oxide synthase (Nos2) expression was significantly reduced in intestinal epithelial cells of ChAT T-cell cKO mice 10 days post-infection. Additionally, a cholinergic agonist enhanced IFNγ-induced Nos2 expression in intestinal epithelial cell in vitro. These findings demonstrated that acetylcholine, produced by specialized T-cells that are recruited during C. rodentium infection, are a key mediator in host-microbe interactions and mucosal defenses.
Collapse
Affiliation(s)
- Valerie T. Ramirez
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Dayn R. Godinez
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Ingrid Brust-Mascher
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Eric B. Nonnecke
- Department of Microbiology & Immunology, UC Davis School of Medicine, UC Davis, Davis, California, United States of America
| | - Patricia A. Castillo
- Department of Microbiology & Immunology, UC Davis School of Medicine, UC Davis, Davis, California, United States of America
| | - Mariana Barboza Gardner
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
- Department of Chemistry, UC Davis, Davis, California, United States of America
| | - Diane Tu
- Department of Chemistry, UC Davis, Davis, California, United States of America
| | - Jessica A. Sladek
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Elaine N. Miller
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Carlito B. Lebrilla
- Department of Chemistry, UC Davis, Davis, California, United States of America
| | - Charles L. Bevins
- Department of Microbiology & Immunology, UC Davis School of Medicine, UC Davis, Davis, California, United States of America
| | - Melanie G. Gareau
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| | - Colin Reardon
- Department, of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, United States of America
| |
Collapse
|
43
|
Verlinden TJM, van Dijk P, Hikspoors J, Herrler A, Lamers WH, Köhler SE. Innervation of the human spleen: A complete hilum-embedding approach. Brain Behav Immun 2019; 77:92-100. [PMID: 30593840 DOI: 10.1016/j.bbi.2018.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION The spleen is hypothesized to play a role in the autonomic nervous system (ANS)-mediated control of host defence, but the neuroanatomical evidence for this assumption rests on a sparse number of studies, which mutually disagree with respect to the existence of cholinergic or vagal innervation. METHODS We conducted an immuno- and enzyme-histochemical study of the innervation of the human spleen using a complete hilum-embedding approach to ensure that only nerves that entered or left the spleen were studied, and that all splenic nerves were included in the sampled area. Furthermore, a complete embedded spleen was serially sectioned to prepare a 3D reconstruction of the hilar nerve plexus. RESULTS All detected nerves entering the spleen arise from the nerve plexus that surrounds branches of the splenic artery and are catecholaminergic. Inside the spleen these nerves continue within the adventitia of the white pulpal central arteries and red pulpal arterioles. Staining for either choline acetyltransferase or acetylcholinesterase did not reveal any evidence for cholinergic innervation of the human spleen, irrespective of the type of fixation (regularly fixed, fresh-frozen post-fixed or fresh-frozen cryoslides). Furthermore, no positive VIP staining was observed (VIP is often co-expressed in postganglionic parasympathetic nerves). CONCLUSION Our comprehensive approach did not produce any evidence for a direct cholinergic (or VIP-ergic) innervation of the spleen. This finding does not rule out (indirect) vagal innervation via postganglionic non-cholinergic periarterial fibres.
Collapse
Affiliation(s)
- Thomas J M Verlinden
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands.
| | - Paul van Dijk
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands
| | - Jill Hikspoors
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands
| | - Andreas Herrler
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands
| | - Wouter H Lamers
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands
| | - S Eleonore Köhler
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands
| |
Collapse
|
44
|
Mannon EC, Sun J, Wilson K, Brands M, Martinez-Quinones P, Baban B, O'Connor PM. A basic solution to activate the cholinergic anti-inflammatory pathway via the mesothelium? Pharmacol Res 2019; 141:236-248. [PMID: 30616018 DOI: 10.1016/j.phrs.2019.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/20/2018] [Accepted: 01/03/2019] [Indexed: 12/24/2022]
Abstract
Much research now indicates that vagal nerve stimulation results in a systemic reduction in inflammatory cytokine production and an increase in anti-inflammatory cell populations that originates from the spleen. Termed the 'cholinergic anti-inflammatory pathway', therapeutic activation of this innate physiological response holds enormous promise for the treatment of inflammatory disease. Much controversy remains however, regarding the underlying physiological pathways mediating this response. This controversy is anchored in the fact that the vagal nerve itself does not innervate the spleen. Recent research from our own laboratory indicating that oral intake of sodium bicarbonate stimulates splenic anti-inflammatory pathways, and that this effect may require transmission of signals to the spleen through the mesothelium, provide new insight into the physiological pathways mediating the cholinergic anti-inflammatory pathway. In this review, we examine proposed models of the cholinergic anti-inflammatory pathway and attempt to frame our recent results in relation to these hypotheses. Following this discussion, we then provide an alternative model of the cholinergic anti-inflammatory pathway which is consistent both with our recent findings and the published literature. We then discuss experimental approaches that may be useful to delineate these hypotheses. We believe the outcome of these experiments will be critical in identifying the most appropriate methods to harness the therapeutic potential of the cholinergic anti-inflammatory pathway for the treatment of disease and may also shed light on the etiology of other pathologies, such as idiopathic fibrosis.
Collapse
Affiliation(s)
- Elinor C Mannon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jingping Sun
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Katie Wilson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Michael Brands
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Patricia Martinez-Quinones
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Department of Surgery, Augusta University Medical Center, Augusta University, Augusta, GA, United States
| | - Babak Baban
- Department of Oral Biology, Augusta University, Augusta, GA, United States
| | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
45
|
Bottasso E. Toward the Existence of a Sympathetic Neuroplasticity Adaptive Mechanism Influencing the Immune Response. A Hypothetical View-Part I. Front Endocrinol (Lausanne) 2019; 10:632. [PMID: 31616373 PMCID: PMC6763740 DOI: 10.3389/fendo.2019.00632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/30/2019] [Indexed: 12/21/2022] Open
Abstract
The nervous system exerts a profound influence on the function of the immune system (IS), mainly through the sympathetic arm of the autonomic nervous system. In fact, the sympathetic nervous system richly innervates secondary lymphoid organs (SLOs) such as the spleen and lymph nodes. For decades, different research groups working in the field have consistently reported changes in the sympathetic innervation of the SLOs during the activation of the IS, which are characterized by a decreased noradrenergic activity and retraction of these fibers. Most of these groups interpreted these changes as a pathological phenomenon, referred to as "damage" or "injury" of the noradrenergic fibers. Some of them postulated that this "injury" was probably due to toxic effects of released endogenous mediators. Others, working on animal models of chronic stimulation of the IS, linked it to the very chronic nature of processes. Unlike these views, this first part of the present work reviews evidence which supports the hypothesis of a specific adaptive mechanism of neural plasticity from sympathetic fibers innervating SLOs, encompassing structural and functional changes of noradrenergic nerves. This plasticity mechanism would involve segmental retraction and degeneration of these fibers during the activation of the IS with subsequent regeneration once the steady state is recovered. The candidate molecules likely to mediate this phenomenon are also here introduced. The second part will extend this view as to the potential changes in sympathetic innervation likely to occur in inflamed non-lymphoid peripheral tissues and its possible immunological implications.
Collapse
|
46
|
Reardon C, Murray K, Lomax AE. Neuroimmune Communication in Health and Disease. Physiol Rev 2018; 98:2287-2316. [PMID: 30109819 PMCID: PMC6170975 DOI: 10.1152/physrev.00035.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/14/2022] Open
Abstract
The immune and nervous systems are tightly integrated, with each system capable of influencing the other to respond to infectious or inflammatory perturbations of homeostasis. Recent studies demonstrating the ability of neural stimulation to significantly reduce the severity of immunopathology and consequently reduce mortality have led to a resurgence in the field of neuroimmunology. Highlighting the tight integration of the nervous and immune systems, afferent neurons can be activated by a diverse range of substances from bacterial-derived products to cytokines released by host cells. While activation of vagal afferents by these substances dominates the literature, additional sensory neurons are responsive as well. It is becoming increasingly clear that although the cholinergic anti-inflammatory pathway has become the predominant model, a multitude of functional circuits exist through which neuronal messengers can influence immunological outcomes. These include pathways whereby efferent signaling occurs independent of the vagus nerve through sympathetic neurons. To receive input from the nervous system, immune cells including B and T cells, macrophages, and professional antigen presenting cells express specific neurotransmitter receptors that affect immune cell function. Specialized immune cell populations not only express neurotransmitter receptors, but express the enzymatic machinery required to produce neurotransmitters, such as acetylcholine, allowing them to act as signaling intermediaries. Although elegant experiments have begun to decipher some of these interactions, integration of these molecules, cells, and anatomy into defined neuroimmune circuits in health and disease is in its infancy. This review describes these circuits and highlights continued challenges and opportunities for the field.
Collapse
Affiliation(s)
- Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Kaitlin Murray
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Alan E Lomax
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California ; and Department of Biomedical and Molecular Sciences and Department of Medicine, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
47
|
Niche-induced extramedullary hematopoiesis in the spleen is regulated by the transcription factor Tlx1. Sci Rep 2018; 8:8308. [PMID: 29844356 PMCID: PMC5974313 DOI: 10.1038/s41598-018-26693-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 05/18/2018] [Indexed: 12/12/2022] Open
Abstract
Extramedullary hematopoiesis (EMH) in postnatal life is a pathological process in which the differentiation of hematopoietic stem/progenitor cells (HSPCs) occurs outside the bone marrow (BM) to respond to hematopoietic emergencies. The spleen is a major site for EMH; however, the cellular and molecular nature of the stromal cell components supporting HSPC maintenance, the niche for EMH in the spleen remain poorly understood compared to the growing understanding of the BM niche at the steady-state as well as in emergency hematopoiesis. In the present study, we demonstrate that mesenchymal progenitor-like cells expressing Tlx1, an essential transcription factor for spleen organogenesis, and selectively localized in the perifollicular region of the red pulp of the spleen, are a major source of HSPC niche factors. Consistently, overexpression of Tlx1 in situ induces EMH, which is associated with mobilization of HSPC into the circulation and their recruitment into the spleen where they proliferate and differentiate. The alterations in the splenic microenvironment induced by Tlx1 overexpression in situ phenocopy lipopolysaccharide (LPS)-induced EMH, and the conditional loss of Tlx1 abolished LPS-induced splenic EMH. These findings indicate that activation of Tlx1 expression in the postnatal splenic mesenchymal cells is critical for the development of splenic EMH.
Collapse
|
48
|
Hu D, Nicholls PK, Yin C, Kelman K, Yuan Q, Greene WK, Shi Z, Ma B. Immunofluorescent Localization of Non-myelinating Schwann Cells and Their Interactions With Immune Cells in Mouse Thymus. J Histochem Cytochem 2018; 66:775-785. [PMID: 29775115 DOI: 10.1369/0022155418778543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The thymus is innervated by sympathetic/parasympathetic nerve fibers from the peripheral nervous system (PNS), suggesting a neural regulation of thymic function including T-cell development. Despite some published studies, data on the innervation and nerve-immune interaction inside the thymus remain limited. In the present study, we used immunofluorescent staining of glial fibrillary acidic protein (GFAP) coupled with confocal microscopy/three-dimensional (3D) reconstruction to reveal the distribution of non-myelinating Schwann cells (NMSC) and their interactions with immune cells inside mouse thymus. Our results demonstrate (1) the presence of an extensive network of NMSC processes in all compartments of the thymus including the capsule, subcapsular region, cortex, cortico-medullary junction, and medulla; (2) close associations/interactions of NMSC processes with blood vessels, indicating the neural control of blood flow inside the thymus; (3) the close "synapse-like" association of NMSC processes with various subsets of dendritic cells (DC; e.g., B220+ DCs, CD4+ DCs, and CD8+ DCs), and lymphocytes (B cells, CD4+/CD8+ thymocytes). Our novel findings concerning the distribution of NMSCs and the associations of NMSCs and immune cells inside mouse thymus should help us understand the anatomical basis and the mechanisms through which the PNS affects T-cell development and thymic endocrine function in health and disease.
Collapse
Affiliation(s)
- Dailun Hu
- Clinical College, Hebei Medical University, Shijiazhuang, China
| | - Philip K Nicholls
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Australia
| | - Changfu Yin
- Clinical College, Hebei Medical University, Shijiazhuang, China
| | - Khama Kelman
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Australia
| | - Qionglan Yuan
- School of Medicine, Tongji University, Shanghai, China
| | - Wayne K Greene
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Australia
| | - Zhongli Shi
- Clinical College, Hebei Medical University, Shijiazhuang, China
| | - Bin Ma
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Australia
| |
Collapse
|
49
|
Ma B, Yin C, Hu D, Newman M, Nicholls PK, Wu Z, Greene WK, Shi Z. Distribution of non-myelinating Schwann cells and their associations with leukocytes in mouse spleen revealed by immunofluorescence staining. Eur J Histochem 2018; 62:2890. [PMID: 29943953 PMCID: PMC6038114 DOI: 10.4081/ejh.2018.2890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/06/2018] [Accepted: 03/09/2018] [Indexed: 12/31/2022] Open
Abstract
The nervous system and the immune system communicate extensively with each other in order to maintain homeostasis and to regulate the immune response. The peripheral nervous system (PNS) communicates specifically with the immune system according to local interactions, including the “hardwiring” of sympathetic/parasympathetic (efferent) and sensory nerves (afferent) to lymphoid tissue and organs. To reveal this type of bidirectional neuroimmune interaction at the microscopic level, we used immunofluorescent staining of glial fibrillary acidic protein (GFAP) coupled with confocal microscopy/3D reconstruction to reveal the distribution of nonmyelinating Schwann cells (NMSCs) and their interactions with immune cells inside mouse spleen. Our results demonstrate i) the presence of an extensive network of NMSC processes in all splenic compartments including the splenic nodules, periarteriolar lymphoid sheath (PALS), marginal zone, trabecula, and red pulp; ii) the close association of NMSC processes with blood vessels (including central arteries and their branches, marginal sinuses, penicillar arterioles and splenic sinuses); iii) the close “synapse-like” interaction/association of NMSC processes with various subsets of dendritic cells (DCs; e.g., CD4+CD11c+ DCs, B220+CD11c+ DCs, and F4/80+ CD11c+ DCs), macrophages (F4/80+), and lymphocytes (B cells, CD4+ T helper cells). Our novel findings concerning the distribution of NMSCs and NMSC-leukocytes interactions inside mouse spleen should improve our understanding of the mechanisms through which the PNS affects cellular- and humoral-mediated immune responses in a variety of health conditions and infectious/non-infectious diseases.
Collapse
Affiliation(s)
- Bin Ma
- Murdoch University, School of Veterinary and Life Sciences.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hoover DB, Brown TC, Miller MK, Schweitzer JB, Williams DL. Loss of Sympathetic Nerves in Spleens from Patients with End Stage Sepsis. Front Immunol 2017; 8:1712. [PMID: 29270174 PMCID: PMC5723638 DOI: 10.3389/fimmu.2017.01712] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/20/2017] [Indexed: 01/22/2023] Open
Abstract
The spleen is an important site for central regulation of immune function by noradrenergic sympathetic nerves, but little is known about this major region of neuroimmune communication in humans. Experimental studies using animal models have established that sympathetic innervation of the spleen is essential for cholinergic anti-inflammatory responses evoked by vagal nerve stimulation, and clinical studies are evaluating this approach for treating inflammatory diseases. Most data on sympathetic nerves in spleen derive from rodent studies, and this work has established that remodeling of sympathetic innervation can occur during inflammation. However, little is known about the effects of sepsis on spleen innervation. Our primary goals were to (i) localize noradrenergic nerves in human spleen by immunohistochemistry for tyrosine hydroxylase (TH), a specific noradrenergic marker, (ii) determine if nerves occur in close apposition to leukocytes, and (iii) determine if splenic sympathetic innervation is altered in patients who died from end stage sepsis. Staining for vesicular acetylcholine transporter (VAChT) was done to screen for cholinergic nerves. Archived paraffin tissue blocks were used. Control samples were obtained from trauma patients or patients who died after hemorrhagic stroke. TH + nerves were associated with arteries and arterioles in all control spleens, occurring in bundles or as nerve fibers. Individual TH + nerve fibers entered the perivascular region where some appeared in close apposition to leukocytes. In marked contrast, spleens from half of the septic patients lacked TH + nerves fibers and the average abundance of TH + nerves for the septic group was only 16% of that for the control group (control: 0.272 ± 0.060% area, n = 6; sepsis: 0.043 ± 0.026% area, n = 8; P < 0.005). All spleens lacked cholinergic innervation. Our results provide definitive evidence for the distribution of noradrenergic nerves in normal human spleen and the first evidence for direct sympathetic innervation of leukocytes in human spleen. We also provide the first evidence for marked loss of noradrenergic nerves in patients who died from sepsis. Such nerve loss could impair neuroimmunomodulation and may not be limited to the spleen.
Collapse
Affiliation(s)
- Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Thomas Christopher Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Madeleine K Miller
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - John B Schweitzer
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L Williams
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|