1
|
Falvey A, Palandira SP, Chavan SS, Brines M, Dantzer R, Tracey KJ, Pavlov VA. Electrical stimulation of the dorsal motor nucleus of the vagus in male mice can regulate inflammation without affecting the heart rate. Brain Behav Immun 2024; 120:630-639. [PMID: 38670240 DOI: 10.1016/j.bbi.2024.04.027] [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: 11/09/2023] [Revised: 04/01/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The vagus nerve plays an important role in neuroimmune interactions and in the regulation of inflammation. A major source of efferent vagus nerve fibers that contribute to the regulation of inflammation is the brainstem dorsal motor nucleus of the vagus (DMN), as recently shown using optogenetics. In contrast to optogenetics, electrical neuromodulation has broad therapeutic implications. However, the anti-inflammatory effectiveness of electrical stimulation of the DMN (eDMNS) and the possible heart rate (HR) alterations associated with this approach have not been investigated. Here, we examined the effects of eDMNS on HR and cytokine levels in mice administered with lipopolysaccharide (LPS, endotoxin) and in mice subjected to cecal ligation and puncture (CLP) sepsis. METHODS Anesthetized male 8-10-week-old C57BL/6 mice on a stereotaxic frame were subjected to eDMNS using a concentric bipolar electrode inserted into the left or right DMN or sham stimulation. eDMNS (500, 250 or 50 μA at 30 Hz, for 1 min) was performed and HR recorded. In endotoxemia experiments, sham or eDMNS utilizing 250 μA or 50 μA was performed for 5 mins and was followed by LPS (0.5 mg/kg) i.p. administration. eDMNS was also applied in mice with cervical unilateral vagotomy or sham operation. In CLP experiments sham or left eDMNS was performed immediately post CLP. Cytokines and corticosterone were analyzed 90 mins after LPS administration or 24 h after CLP. CLP survival was monitored for 14 days. RESULTS Either left or right eDMNS at 500 μA and 250 μA decreased HR, compared with baseline pre-stimulation. This effect was not observed at 50 μA. Left side eDMNS at 50 μA, compared with sham stimulation, significantly decreased serum and splenic levels of the pro-inflammatory cytokine TNF and increased serum levels of the anti-inflammatory cytokine IL-10 during endotoxemia. The anti-inflammatory effect of eDMNS was abrogated in mice with unilateral vagotomy and was not associated with serum corticosterone alterations. Right side eDMNS in endotoxemic mice suppressed serum TNF and increased serum IL-10 levels but had no effects on splenic cytokines. In mice with CLP, left side eDMNS suppressed serum IL-6, as well as splenic IL-6 and increased splenic IL-10 and significantly improved the survival rate of CLP mice. CONCLUSIONS For the first time we show that a regimen of eDMNS which does not cause bradycardia alleviates LPS-induced inflammation. These eDMNS anti-inflammatory effects require an intact vagus nerve and are not associated with corticosteroid alterations. eDMNS also decreases inflammation and improves survival in a model of polymicrobial sepsis. These findings are of interest for further studies exploring bioelectronic anti-inflammatory approaches targeting the brainstem DMN.
Collapse
Affiliation(s)
- Aidan Falvey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Santhoshi P Palandira
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Sangeeta S Chavan
- Institute of Bioelectronic Medicine, 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; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Michael Brines
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Robert Dantzer
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, 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; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Valentin A Pavlov
- Institute of Bioelectronic Medicine, 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; Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA.
| |
Collapse
|
2
|
Zou N, Zhou Q, Zhang Y, Xin C, Wang Y, Claire-Marie R, Rong P, Gao G, Li S. Transcutaneous auricular vagus nerve stimulation as a novel therapy connecting the central and peripheral systems: a review. Int J Surg 2024; 110:4993-5006. [PMID: 38729100 PMCID: PMC11326027 DOI: 10.1097/js9.0000000000001592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
Currently, clinical practice and scientific research mostly revolve around a single disease or system, but the single disease-oriented diagnostic and therapeutic paradigm needs to be revised. This review describes how transcutaneous auricular vagus nerve stimulation (taVNS), a novel non-invasive neuromodulation approach, connects the central and peripheral systems of the body. Through stimulation of the widely distributed vagus nerve from the head to the abdominal cavity, this therapy can improve and treat central system disorders, peripheral system disorders, and central-peripheral comorbidities caused by autonomic dysfunction. In the past, research on taVNS has focused on the treatment of central system disorders by modulating this brain nerve. As the vagus nerve innervates the heart, lungs, liver, pancreas, gastrointestinal tract, spleen and other peripheral organs, taVNS could have an overall modulatory effect on the region of the body where the vagus nerve is widespread. Based on this physiological basis, the authors summarize the existing evidence of the taVNS ability to regulate cardiac function, adiposity, glucose levels, gastrointestinal function, and immune function, among others, to treat peripheral system diseases, and complex diseases with central and peripheral comorbidities. This review shows the successful examples and research progress of taVNS using peripheral neuromodulation mechanisms from more perspectives, demonstrating the expanded scope and value of taVNS to provide new ideas and approaches for holistic therapy from both central and peripheral perspectives.
Collapse
Affiliation(s)
- Ningyi Zou
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
| | - Qing Zhou
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
| | - Yuzhengheng Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
| | - Chen Xin
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
| | - Yifei Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
| | | | - Peijing Rong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences
| | - Guojian Gao
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaoyuan Li
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences
| |
Collapse
|
3
|
Trevizan-Baú P, McAllen RM. What is the Vagal-Adrenal Axis? J Comp Neurol 2024; 532:e25656. [PMID: 38980012 DOI: 10.1002/cne.25656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Some recent publications have used the term "vagal-adrenal axis" to account for mechanisms involved in the regulation of inflammation by electroacupuncture. This concept proposes that efferent parasympathetic nerve fibers in the vagus directly innervate the adrenal glands to influence catecholamine secretion. Here, we discuss evidence for anatomical and functional links between the vagi and adrenal glands that may be relevant in the context of inflammation and its neural control by factors, including acupuncture. First, we find that evidence for any direct vagal parasympathetic efferent innervation of the adrenal glands is weak and likely artifactual. Second, we find good evidence that vagal afferent fibers directly innervate the adrenal gland, although their function is uncertain. Third, we highlight a wealth of evidence for indirect pathways, whereby vagal afferent signals act via the central nervous system to modify adrenal-dependent anti-inflammatory responses. Vagal afferents, not efferents, are thus the likely key to these phenomena.
Collapse
Affiliation(s)
- Pedro Trevizan-Baú
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Robin M McAllen
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Trevizan-Baú P, Ringuet MT, Stebbing MJ, McAllen RM, Furness JB, Mueller SN. Protocol for the isolation of the mouse sympathetic splanchnic-celiac-superior mesenteric ganglion complex. STAR Protoc 2024; 5:103036. [PMID: 38676927 PMCID: PMC11067456 DOI: 10.1016/j.xpro.2024.103036] [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: 02/20/2024] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
Neurons that originate from pre-vertebral sympathetic ganglia, the splanchnic-celiac-superior mesenteric ganglion complex (SCSMG) in mouse, have important roles in control of organs of the upper abdomen. Here, we present a protocol for the isolation of the mouse sympathetic SCSMG. We describe steps for surgical incision, ganglia isolation, ganglia fine dissection, and whole-mount SCSMG after clearing-enhanced 3D (Ce3D) clearing method and immunohistochemistry. Given the importance of mice in studies of that control, this protocol aims to assist biomedical researchers in the dissection of the mouse SCSMG.
Collapse
Affiliation(s)
- Pedro Trevizan-Baú
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Department of Physiological Sciences, University of Florida, Gainesville, FL, USA.
| | - Mitchell Ty Ringuet
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Martin J Stebbing
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Robin M McAllen
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - John B Furness
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
González-González MA, Conde SV, Latorre R, Thébault SC, Pratelli M, Spitzer NC, Verkhratsky A, Tremblay MÈ, Akcora CG, Hernández-Reynoso AG, Ecker M, Coates J, Vincent KL, Ma B. Bioelectronic Medicine: a multidisciplinary roadmap from biophysics to precision therapies. Front Integr Neurosci 2024; 18:1321872. [PMID: 38440417 PMCID: PMC10911101 DOI: 10.3389/fnint.2024.1321872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/10/2024] [Indexed: 03/06/2024] Open
Abstract
Bioelectronic Medicine stands as an emerging field that rapidly evolves and offers distinctive clinical benefits, alongside unique challenges. It consists of the modulation of the nervous system by precise delivery of electrical current for the treatment of clinical conditions, such as post-stroke movement recovery or drug-resistant disorders. The unquestionable clinical impact of Bioelectronic Medicine is underscored by the successful translation to humans in the last decades, and the long list of preclinical studies. Given the emergency of accelerating the progress in new neuromodulation treatments (i.e., drug-resistant hypertension, autoimmune and degenerative diseases), collaboration between multiple fields is imperative. This work intends to foster multidisciplinary work and bring together different fields to provide the fundamental basis underlying Bioelectronic Medicine. In this review we will go from the biophysics of the cell membrane, which we consider the inner core of neuromodulation, to patient care. We will discuss the recently discovered mechanism of neurotransmission switching and how it will impact neuromodulation design, and we will provide an update on neuronal and glial basis in health and disease. The advances in biomedical technology have facilitated the collection of large amounts of data, thereby introducing new challenges in data analysis. We will discuss the current approaches and challenges in high throughput data analysis, encompassing big data, networks, artificial intelligence, and internet of things. Emphasis will be placed on understanding the electrochemical properties of neural interfaces, along with the integration of biocompatible and reliable materials and compliance with biomedical regulations for translational applications. Preclinical validation is foundational to the translational process, and we will discuss the critical aspects of such animal studies. Finally, we will focus on the patient point-of-care and challenges in neuromodulation as the ultimate goal of bioelectronic medicine. This review is a call to scientists from different fields to work together with a common endeavor: accelerate the decoding and modulation of the nervous system in a new era of therapeutic possibilities.
Collapse
Affiliation(s)
- María Alejandra González-González
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatric Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Silvia V. Conde
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NOVA University, Lisbon, Portugal
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Stéphanie C. Thébault
- Laboratorio de Investigación Traslacional en salud visual (D-13), Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Marta Pratelli
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Nicholas C. Spitzer
- Neurobiology Department, Kavli Institute for Brain and Mind, UC San Diego, La Jolla, CA, United States
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Collaborative Center on Big Science Plan for Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Cuneyt G. Akcora
- Department of Computer Science, University of Central Florida, Orlando, FL, United States
| | | | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX, United States
| | | | - Kathleen L. Vincent
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, United States
| | - Brandy Ma
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
6
|
Lataro RM, Brognara F, Iturriaga R, Paton JFR. Inflammation of some visceral sensory systems and autonomic dysfunction in cardiovascular disease. Auton Neurosci 2024; 251:103137. [PMID: 38104365 DOI: 10.1016/j.autneu.2023.103137] [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: 07/27/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
The sensitization and hypertonicity of visceral afferents are highly relevant to the development and progression of cardiovascular and respiratory disease states. In this review, we described the evidence that the inflammatory process regulates visceral afferent sensitivity and tonicity, affecting the control of the cardiovascular and respiratory system. Some inflammatory mediators like nitric oxide, angiotensin II, endothelin-1, and arginine vasopressin may inhibit baroreceptor afferents and contribute to the baroreflex impairment observed in cardiovascular diseases. Cytokines may act directly on peripheral afferent terminals that transmit information to the central nervous system (CNS). TLR-4 receptors, which recognize lipopolysaccharide, were identified in the nodose and petrosal ganglion and have been implicated in disrupting the blood-brain barrier, which can potentiate the inflammatory process. For example, cytokines may cross the blood-brain barrier to access the CNS. Additionally, pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and some of their receptors have been identified in the nodose ganglion and carotid body. These pro-inflammatory cytokines also sensitize the dorsal root ganglion or are released in the nucleus of the solitary tract. In cardiovascular disease, pro-inflammatory mediators increase in the brain, heart, vessels, and plasma and may act locally or systemically to activate/sensitize afferent nervous terminals. Recent evidence demonstrated that the carotid body chemoreceptor cells might sense systemic pro-inflammatory molecules, supporting the novel proposal that the carotid body is part of the afferent pathway in the central anti-inflammatory reflexes. The exact mechanisms of how pro-inflammatory mediators affects visceral afferent signals and contribute to the pathophysiology of cardiovascular diseases awaits future research.
Collapse
Affiliation(s)
- R M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - F Brognara
- Department of Nursing, General and Specialized, Nursing School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Iturriaga
- Facultad de Ciencias Biológicas, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Investigación en Fisiología y Medicina en Altura - FIMEDALT, Universidad de Antofagasta, Antofagasta, Chile
| | - J F R Paton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
7
|
Andalib S, Divani AA, Ayata C, Baig S, Arsava EM, Topcuoglu MA, Cáceres EL, Parikh V, Desai MJ, Majid A, Girolami S, Di Napoli M. Vagus Nerve Stimulation in Ischemic Stroke. Curr Neurol Neurosci Rep 2023; 23:947-962. [PMID: 38008851 PMCID: PMC10841711 DOI: 10.1007/s11910-023-01323-w] [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] [Accepted: 10/30/2023] [Indexed: 11/28/2023]
Abstract
PURPOSE OF REVIEW Vagus nerve stimulation (VNS) has emerged as a potential therapeutic approach for neurological and psychiatric disorders. In recent years, there has been increasing interest in VNS for treating ischemic stroke. This review discusses the evidence supporting VNS as a treatment option for ischemic stroke and elucidates its underlying mechanisms. RECENT FINDINGS Preclinical studies investigating VNS in stroke models have shown reduced infarct volumes and improved neurological deficits. Additionally, VNS has been found to reduce reperfusion injury. VNS may promote neuroprotection by reducing inflammation, enhancing cerebral blood flow, and modulating the release of neurotransmitters. Additionally, VNS may stimulate neuroplasticity, thereby facilitating post-stroke recovery. The Food and Drug Administration has approved invasive VNS (iVNS) combined with rehabilitation for ischemic stroke patients with moderate to severe upper limb deficits. However, iVNS is not feasible in acute stroke due to its time-sensitive nature. Non-invasive VNS (nVNS) may be an alternative approach for treating ischemic stroke. While the evidence from preclinical studies and clinical trials of nVNS is promising, the mechanisms through which VNS exerts its beneficial effects on ischemic stroke are still being elucidated. Therefore, further research is needed to better understand the efficacy and underlying mechanisms of nVNS in ischemic stroke. Moreover, large-scale randomized clinical trials are necessary to determine the optimal nVNS protocols, assess its long-term effects on stroke recovery and outcomes, and identify the potential benefits of combining nVNS with other rehabilitation strategies.
Collapse
Affiliation(s)
- Sasan Andalib
- Research Unit of Neurology, Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Afshin A Divani
- Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA.
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology and Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sheharyar Baig
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Ethem Murat Arsava
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | | | - Vinay Parikh
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Masoom J Desai
- Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Sara Girolami
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| |
Collapse
|
8
|
Münzberg H, Berthoud HR, Neuhuber WL. Sensory spinal interoceptive pathways and energy balance regulation. Mol Metab 2023; 78:101817. [PMID: 37806487 PMCID: PMC10590858 DOI: 10.1016/j.molmet.2023.101817] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
Interoception plays an important role in homeostatic regulation of energy intake and metabolism. Major interoceptive pathways include gut-to-brain and adipose tissue-to brain signaling via vagal sensory nerves and hormones, such as leptin. However, signaling via spinal sensory neurons is rapidly emerging as an additional important signaling pathway. Here we provide an in-depth review of the known anatomy and functions of spinal sensory pathways and discuss potential mechanisms relevant for energy balance homeostasis in health and disease. Because sensory innervation by dorsal root ganglia (DRG) neurons goes far beyond vagally innervated viscera and includes adipose tissue, skeletal muscle, and skin, it is in a position to provide much more complete metabolic information to the brain. Molecular and anatomical identification of function specific DRG neurons will be important steps in designing pharmacological and neuromodulation approaches to affect energy balance regulation in disease states such as obesity, diabetes, and cancer.
Collapse
Affiliation(s)
- Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Winfried L Neuhuber
- Institute for Anatomy and Cell Biology, Friedrich-Alexander University, Erlangen, Germany.
| |
Collapse
|
9
|
Rivera-Castro ME, Pastelín CF, Bravo-Benítez J, Morán C. Organization of the Subdiaphragmatic Vagus Nerve and Its Connection with the Celiac Plexus and the Ovaries in the Female Rat. Brain Sci 2023; 13:1032. [PMID: 37508964 PMCID: PMC10377505 DOI: 10.3390/brainsci13071032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Communication between the ovaries and the central nervous system occurs by peripheral innervation through the celiac plexus, superior ovarian nerve, and ovarian plexus nerve. The vagus nerve is involved in regulating the ovaries, but the neuroanatomical pathway that links them is not clear. Adult female rats were used for gross anatomy, acetylcholinesterase histochemistry, and the immunofluorescence analysis of tyrosine hydroxylase (TH), choline acetyltransferase (ChAT), and tryptophan hydroxylase 2 (TPH). The results obtained indicate that the right vagus nerve (RVN) travels parallel and caudal to the esophagus, where three nerve branches were identified. Also, a right vagal plexus (RVP) formed by microganglia was described, establishing communication with the celiac plexus, and was mainly reactive to tyrosine hydroxylase (TH); some serotoninergic and cholinergic neurons were also found. The left vagus nerve (LVN) travels over the esophagus, bifurcates before its insertion into the stomach and enters the RCG. This neuroanatomical and biochemical description of the RVN and LVN in the rat suggests the RVP is formed by presynaptic catecholaminergic terminals and cholinergic neurons. This information could support detailed studies of communication between the vagus nerve and the ovaries and identify the type of neural signaling involved in abdominal control of the vagus nerve.
Collapse
Affiliation(s)
- María E Rivera-Castro
- Doctorado en Investigaciones Cerebrales, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz 91190, Mexico
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla 72960, Mexico
| | - César F Pastelín
- Facultad de Medicina Veterinaria y Zootecnia, Benemérita Universidad Autónoma de Puebla, Tecamachalco, Puebla 75460, Mexico
| | - Juan Bravo-Benítez
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla 72960, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla 72960, Mexico
| |
Collapse
|
10
|
Falvey A, Palandira SP, Chavan SS, Brines M, Tracey KJ, Pavlov VA. Electrical stimulation of the dorsal motor nucleus of the vagus regulates inflammation without affecting the heart rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541191. [PMID: 37292846 PMCID: PMC10245723 DOI: 10.1101/2023.05.17.541191] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background The vagus nerve plays an important role in neuroimmune interactions and in the regulation of inflammation. A major source of efferent vagus nerve fibers that contribute to the regulation of inflammation is the brainstem dorsal motor nucleus of the vagus (DMN) as recently shown using optogenetics. In contrast to optogenetics, electrical neuromodulation has broad therapeutic implications, but the anti-inflammatory efficacy of electrical DMN stimulation (eDMNS) was not previously investigated. Here, we examined the effects of eDMNS on heart rate (HR) and cytokine levels in murine endotoxemia as well as the cecal ligation and puncture (CLP) model of sepsis. Methods Anesthetized male 8-10-week-old C57BL/6 mice on a stereotaxic frame were subjected to eDMNS using a concentric bipolar electrode inserted into the left or right DMN or sham stimulation. eDMNS (50, 250 or 500 μA and 30 Hz, for 1 min) was performed and HR recorded. In endotoxemia experiments, sham or eDMNS utilizing 250 μA or 50 μA was performed for 5 mins and was followed by LPS (0.5 mg/kg) i.p. administration. eDMNS was also applied in mice with cervical unilateral vagotomy or sham operation. In CLP experiments sham or left eDMNS was performed immediately post CLP. Cytokines and corticosterone were analyzed 90 mins after LPS administration or 24h after CLP. CLP survival was monitored for 14 days. Results Either left or right eDMNS at 250 μA and 500 μA decreased HR, compared with pre- and post-stimulation. This effect was not observed at 50 μA. Left side eDMNS at 50 μA, compared with sham stimulation, significantly decreased serum and splenic levels of the pro-inflammatory cytokine TNF and increased serum levels of the anti-inflammatory cytokine IL-10 during endotoxemia. The anti-inflammatory effect of eDMNS was abrogated in mice with unilateral vagotomy and were not associated with serum corticosterone alterations. Right side eDMNS suppressed serum TNF levels but had no effects on serum IL-10 and on splenic cytokines. In mice with CLP, left side eDMNS suppressed serum TNF and IL-6, as well as splenic IL-6 and increased splenic IL-10 and significantly improved the survival rate of CLP mice. Conclusions For the first time we show that a regimen of eDMNS which does not cause bradycardia alleviates LPS-induced inflammation and these effects require an intact vagus nerve and are not associated with corticosteroid alterations. eDMNS also decreases inflammation and improves survival in a model of polymicrobial sepsis. These findings are of interest for further studies exploring bioelectronic anti-inflammatory approaches targeting the brainstem DMN.
Collapse
Affiliation(s)
- Aidan Falvey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Santhoshi P. Palandira
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Sangeeta S. Chavan
- Institute of Bioelectronic Medicine, 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, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Michael Brines
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Kevin J. Tracey
- Institute of Bioelectronic Medicine, 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, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| | - Valentin A. Pavlov
- Institute of Bioelectronic Medicine, 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, New York 11549, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, USA
| |
Collapse
|
11
|
Adori M, Bhat S, Gramignoli R, Valladolid-Acebes I, Bengtsson T, Uhlèn M, Adori C. Hepatic Innervations and Nonalcoholic Fatty Liver Disease. Semin Liver Dis 2023; 43:149-162. [PMID: 37156523 PMCID: PMC10348844 DOI: 10.1055/s-0043-57237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder. Increased sympathetic (noradrenergic) nerve tone has a complex role in the etiopathomechanism of NAFLD, affecting the development/progression of steatosis, inflammation, fibrosis, and liver hemodynamical alterations. Also, lipid sensing by vagal afferent fibers is an important player in the development of hepatic steatosis. Moreover, disorganization and progressive degeneration of liver sympathetic nerves were recently described in human and experimental NAFLD. These structural alterations likely come along with impaired liver sympathetic nerve functionality and lack of adequate hepatic noradrenergic signaling. Here, we first overview the anatomy and physiology of liver nerves. Then, we discuss the nerve impairments in NAFLD and their pathophysiological consequences in hepatic metabolism, inflammation, fibrosis, and hemodynamics. We conclude that further studies considering the spatial-temporal dynamics of structural and functional changes in the hepatic nervous system may lead to more targeted pharmacotherapeutic advances in NAFLD.
Collapse
Affiliation(s)
- Monika Adori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sadam Bhat
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ismael Valladolid-Acebes
- Department of Molecular Medicine and Surgery, The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Mathias Uhlèn
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Csaba Adori
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
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
|
13
|
Pavlov VA, Tracey KJ. Bioelectronic medicine: Preclinical insights and clinical advances. Neuron 2022; 110:3627-3644. [PMID: 36174571 PMCID: PMC10155266 DOI: 10.1016/j.neuron.2022.09.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/28/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022]
Abstract
The nervous system maintains homeostasis and health. Homeostatic disruptions underlying the pathobiology of many diseases can be controlled by bioelectronic devices targeting CNS and peripheral neural circuits. New insights into the regulatory functions of the nervous system and technological developments in bioelectronics drive progress in the emerging field of bioelectronic medicine. Here, we provide an overview of key aspects of preclinical research, translation, and clinical advances in bioelectronic medicine.
Collapse
Affiliation(s)
- Valentin A Pavlov
- Institute of Bioelectronic Medicine, the Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, the Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
14
|
Mota CMD, Madden CJ. Neural control of the spleen as an effector of immune responses to inflammation: mechanisms and treatments. Am J Physiol Regul Integr Comp Physiol 2022; 323:R375-R384. [PMID: 35993560 PMCID: PMC9485006 DOI: 10.1152/ajpregu.00151.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022]
Abstract
Immune system responses are a vital defense mechanism against pathogens. Inflammatory mediators finely regulate complex inflammatory responses from initiation to resolution. However, in certain conditions, the inflammation is initiated and amplified, but not resolved. Understanding the biological mechanisms underlying the regulation of the immune response is critical for developing therapeutic alternatives, including pharmaceuticals and bioelectronic tools. The spleen is an important immune effector organ since it orchestrates innate and adaptive immune responses such as pathogen clearance, cytokine production, and differentiation of cells, therefore playing a modulatory role that balances pro- and anti-inflammatory responses. However, modulation of splenic immune activity is a largely unexplored potential therapeutic tool that could be used for the treatment of inflammatory and life-threatening conditions. This review discusses some of the mechanisms controlling neuroimmune communication and the brain-spleen axis.
Collapse
Affiliation(s)
- Clarissa M D Mota
- Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
15
|
Newell-Rogers MK, Duong A, Nazarali R, Tobin RP, Rogers SK, Shapiro LA. Unilateral Cervical Vagotomy Modulates Immune Cell Profiles and the Response to a Traumatic Brain Injury. Int J Mol Sci 2022; 23:9851. [PMID: 36077246 PMCID: PMC9456009 DOI: 10.3390/ijms23179851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
TBI induces splenic B and T cell expansion that contributes to neuroinflammation and neurodegeneration. The vagus nerve, the longest of the cranial nerves, is the predominant parasympathetic pathway allowing the central nervous system (CNS) control over peripheral organs, including regulation of inflammatory responses. One way this is accomplished is by vagus innervation of the celiac ganglion, from which the splenic nerve innervates the spleen. This splenic innervation enables modulation of the splenic immune response, including splenocyte selection, activation, and downstream signaling. Considering that the left and right vagus nerves have distinct courses, it is possible that they differentially influence the splenic immune response following a CNS injury. To test this possibility, immune cell subsets were profiled and quantified following either a left or a right unilateral vagotomy. Both unilateral vagotomies caused similar effects with respect to the percentage of B cells and in the decreased percentage of macrophages and T cells following vagotomy. We next tested the hypothesis that a left unilateral vagotomy would modulate the splenic immune response to a traumatic brain injury (TBI). Mice received a left cervical vagotomy or a sham vagotomy 3 days prior to a fluid percussion injury (FPI), a well-characterized mouse model of TBI that consistently elicits an immune and neuroimmune response. Flow cytometric analysis showed that vagotomy prior to FPI resulted in fewer CLIP+ B cells, and CD4+, CD25+, and CD8+ T cells. Vagotomy followed by FPI also resulted in an altered distribution of CD11bhigh and CD11blow macrophages. Thus, transduction of immune signals from the CNS to the periphery via the vagus nerve can be targeted to modulate the immune response following TBI.
Collapse
Affiliation(s)
- M. Karen Newell-Rogers
- School of Medicine, Texas A&M University, 8447 Riverside Parkway, Bryan, TX 77807, USA
- BCell Solutions, Inc., Colorado Springs, CO 80907, USA
| | - Amanda Duong
- School of Medicine, Texas A&M University, 8447 Riverside Parkway, Bryan, TX 77807, USA
| | - Rizwan Nazarali
- Department of Anesthesiology, School of Medicine, University of Colorado, Denver, CO 80309, USA
| | - Richard P. Tobin
- Department of Surgery-Surgical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Susannah K. Rogers
- School of Medicine, Texas A&M University, 8447 Riverside Parkway, Bryan, TX 77807, USA
| | - Lee A. Shapiro
- School of Medicine, Texas A&M University, 8447 Riverside Parkway, Bryan, TX 77807, USA
| |
Collapse
|
16
|
Liu YX, Yu Y, Liu JP, Liu WJ, Cao Y, Yan RM, Yao YM. Neuroimmune Regulation in Sepsis-Associated Encephalopathy: The Interaction Between the Brain and Peripheral Immunity. Front Neurol 2022; 13:892480. [PMID: 35832175 PMCID: PMC9271799 DOI: 10.3389/fneur.2022.892480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE), the most popular cause of coma in the intensive care unit (ICU), is the diffuse cerebral damage caused by the septic challenge. SAE is closely related to high mortality and extended cognitive impairment in patients in septic shock. At present, many studies have demonstrated that SAE might be mainly associated with blood–brain barrier damage, abnormal neurotransmitter secretion, oxidative stress, and neuroimmune dysfunction. Nevertheless, the precise mechanism which initiates SAE and contributes to the long-term cognitive impairment remains largely unknown. Recently, a growing body of evidence has indicated that there is close crosstalk between SAE and peripheral immunity. The excessive migration of peripheral immune cells to the brain, the activation of glia, and resulting dysfunction of the central immune system are the main causes of septic nerve damage. This study reviews the update on the pathogenesis of septic encephalopathy, focusing on the over-activation of immune cells in the central nervous system (CNS) and the “neurocentral–endocrine–immune” networks in the development of SAE, aiming to further understand the potential mechanism of SAE and provide new targets for diagnosis and management of septic complications.
Collapse
Affiliation(s)
- Yu-xiao Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Yang Yu
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jing-peng Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Wen-jia Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Cao
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Run-min Yan
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
- *Correspondence: Yong-ming Yao
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Run-min Yan
| |
Collapse
|
17
|
Khodadadi F, Ketabchi F, Khodabandeh Z, Tavassoli A, Lewis GF, Bahaoddini A. The effect of subdiaphragmatic vagotomy on heart rate variability and lung inflammation in rats with severe hemorrhagic shock. BMC Cardiovasc Disord 2022; 22:181. [PMID: 35439928 PMCID: PMC9020113 DOI: 10.1186/s12872-022-02594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background The influence of cutting the sub-diaphragmatic branch of the vagus nerve on heart rate variability (HRV) and inflammatory reaction to severe hemorrhagic shock has not been determined prior to this study. Methods Male Sprague–Dawley rats were divided into four groups of Sham, sub-diaphragmatic vagotomized (Vag), subacute (135 ± 2 min) hemorrhagic shock (SHS), and sub-diaphragmatic vagotomized with SHS (Vag + SHS). Hemodynamic parameters were recorded and HRV calculated during multiple phases in a conscious model of hemorrhagic shock. The expressions of TNF-α and iNOS were measured in the spleen and lung tissues at the conclusion of the protocol. Results Decreases in blood pressure during blood withdrawal were identical in the SHS and Vag + SHS groups. However, heart rate only decreased in the Nadir-1 phase of the SHS group. HRV indicated increased power in the very-low, low, and high (VLF, LF, and HF) frequency bands during the Nadir-1 phase of the SHS and Vag + SHS groups, albeit the values were higher in the SHS group. In the recovery phase, the HF bands were only lower in the SHS group. After hemorrhagic shock followed by resuscitation, the expression of TNF-α and iNOS increased in the spleen and lung of the SHS group, and the expression of these genes was significantly lower in the Vag + SHS group than in the SHS group. Conclusion Parasympathetic activity increases during the hypotensive phase of hemorrhagic shock, whereas the cardiac vagal tone decreases in the recovery phase. Sub-diapragmatic vagotomy blunts the cardiac vagal tone during hemorrhagic shock, but its effect is reversed in the recovery phase. The vagus nerve plays a role in proinflammatory responses in the lungs and spleen in subacute hemorrhagic shock followed by resuscitation.
Collapse
Affiliation(s)
- Fateme Khodadadi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Farzaneh Ketabchi
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Alireza Tavassoli
- Department of Pathology, Fasa University of Medical Sciences, Fasa, Iran
| | - Gregory F Lewis
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.,The Traumatic Stress Research Consortium at the Kinsey Institute, Indiana University, Bloomington, IN, USA
| | | |
Collapse
|
18
|
Perelló M, Cornejo MP, De Francesco PN, Fernandez G, Gautron L, Valdivia LS. The controversial role of the vagus nerve in mediating ghrelin´s actions: gut feelings and beyond. IBRO Neurosci Rep 2022; 12:228-239. [PMID: 35746965 PMCID: PMC9210457 DOI: 10.1016/j.ibneur.2022.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a stomach-derived peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) and displays a plethora of neuroendocrine, metabolic, autonomic and behavioral actions. It has been proposed that some actions of ghrelin are exerted via the vagus nerve, which provides a bidirectional communication between the central nervous system and peripheral systems. The vagus nerve comprises sensory fibers, which originate from neurons of the nodose and jugular ganglia, and motor fibers, which originate from neurons of the medulla. Many anatomical studies have mapped GHSR expression in vagal sensory or motor neurons. Also, numerous functional studies investigated the role of the vagus nerve mediating specific actions of ghrelin. Here, we critically review the topic and discuss the available evidence supporting, or not, a role for the vagus nerve mediating some specific actions of ghrelin. We conclude that studies using rats have provided the most congruent evidence indicating that the vagus nerve mediates some actions of ghrelin on the digestive and cardiovascular systems, whereas studies in mice resulted in conflicting observations. Even considering exclusively studies performed in rats, the putative role of the vagus nerve in mediating the orexigenic and growth hormone (GH) secretagogue properties of ghrelin remains debated. In humans, studies are still insufficient to draw definitive conclusions regarding the role of the vagus nerve mediating most of the actions of ghrelin. Thus, the extent to which the vagus nerve mediates ghrelin actions, particularly in humans, is still uncertain and likely one of the most intriguing unsolved aspects of the field.
Collapse
|
19
|
Falvey A, Metz CN, Tracey KJ, Pavlov VA. Peripheral nerve stimulation and immunity: the expanding opportunities for providing mechanistic insight and therapeutic intervention. Int Immunol 2022; 34:107-118. [PMID: 34498051 PMCID: PMC8783605 DOI: 10.1093/intimm/dxab068] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/07/2021] [Indexed: 12/29/2022] Open
Abstract
Pre-clinical research advances our understanding of the vagus nerve-mediated regulation of immunity and clinical trials successfully utilize electrical vagus nerve stimulation in the treatment of patients with inflammatory disorders. This symbiotic relationship between pre-clinical and clinical research exploring the vagus nerve-based 'inflammatory reflex' has substantially contributed to establishing the field of bioelectronic medicine. Recent studies identify a crosstalk between the vagus nerve and other neural circuitries in controlling inflammation and delineate new neural immunoregulatory pathways. Here we outline current mechanistic insights into the role of vagal and non-vagal neural pathways in neuro-immune communication and inflammatory regulation. We also provide a timely overview of expanding opportunities for bioelectronic neuromodulation in the treatment of various inflammatory disorders.
Collapse
Affiliation(s)
- Aidan Falvey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Christine N Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kevin J Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Valentin A Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
20
|
Somatosensory and autonomic neuronal regulation of the immune response. Nat Rev Neurosci 2022; 23:157-171. [PMID: 34997214 DOI: 10.1038/s41583-021-00555-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
Bidirectional communication between the peripheral nervous system (PNS) and the immune system is a crucial part of an effective but balanced mammalian response to invading pathogens, tissue damage and inflammatory stimuli. Here, we review how somatosensory and autonomic neurons regulate immune cellular responses at barrier tissues and in peripheral organs. Immune cells express receptors for neuronal mediators, including neuropeptides and neurotransmitters, allowing neurons to influence their function in acute and chronic inflammatory diseases. Distinct subsets of peripheral sensory, sympathetic, parasympathetic and enteric neurons are able to signal to innate and adaptive immune cells to modulate their cellular functions. In this Review, we highlight recent studies defining the molecular mechanisms by which neuroimmune signalling mediates tissue homeostasis and pathology. Understanding the neural circuitry that regulates immune responses can offer novel targets for the treatment of a wide array of diseases.
Collapse
|
21
|
Neuhuber WL, Berthoud HR. Functional anatomy of the vagus system - Emphasis on the somato-visceral interface. Auton Neurosci 2021; 236:102887. [PMID: 34634680 PMCID: PMC8627476 DOI: 10.1016/j.autneu.2021.102887] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/02/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022]
Abstract
Due to its pivotal role in autonomic networks, the vagus attracts continuous interest from both basic scientists and clinicians. In particular, recent advances in vagus nerve stimulation strategies and their application to pathological conditions beyond epilepsy provide a good opportunity to recall basic features of vagal peripheral and central anatomy. In addition to the "classical" vagal brainstem nuclei, i.e., dorsal motor nucleus, nucleus ambiguus and nucleus tractus solitarii, the spinal trigeminal and paratrigeminal nuclei come into play as targets of vagal afferents. On the other hand, the nucleus of the solitary tract receives and integrates not only visceral but also somatic afferents. Thus, the vagus system participates significantly in what may be defined as "somato-visceral interface".
Collapse
Affiliation(s)
- Winfried L Neuhuber
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University, Krankenhausstrasse 9, Erlangen, Germany.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
22
|
Gonzalez-Gonzalez MA, Bendale GS, Wang K, Wallace GG, Romero-Ortega M. Platinized graphene fiber electrodes uncover direct spleen-vagus communication. Commun Biol 2021; 4:1097. [PMID: 34535751 PMCID: PMC8448843 DOI: 10.1038/s42003-021-02628-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/30/2021] [Indexed: 11/21/2022] Open
Abstract
Neural interfacing nerve fascicles along the splenic neurovascular plexus (SNVP) is needed to better understand the spleen physiology, and for selective neuromodulation of this major organ. However, their small size and anatomical location have proven to be a significant challenge. Here, we use a reduced liquid crystalline graphene oxide (rGO) fiber coated with platinum (Pt) as a super-flexible suture-like electrode to interface multiple SNVP. The Pt-rGO fibers work as a handover knot electrodes over the small SNVP, allowing sensitive recording from four splenic nerve terminal branches (SN 1–4), to uncover differential activity and axon composition among them. Here, the asymmetric defasciculation of the SN branches is revealed by electron microscopy, and the functional compartmentalization in spleen innervation is evidenced in response to hypoxia and pharmacological modulation of mean arterial pressure. We demonstrate that electrical stimulation of cervical and sub-diaphragmatic vagus nerve (VN), evokes activity in a subset of SN terminal branches, providing evidence for a direct VN control over the spleen. This notion is supported by adenoviral tract-tracing of SN branches, revealing an unconventional direct brain-spleen projection. High-performance Pt-rGO fiber electrodes, may be used for the fine neural modulation of other small neurovascular plexus at the point of entry of major organs as a bioelectronic medical alternative. Gonzalez-Gonzalez et al. use high-performance platinized graphene fiber electrodes to interface individual neurovascular plexus that innervate the spleen. Their approach provides evidence for distinct function of individual spleen terminal branches in organ function.
Collapse
Affiliation(s)
- Maria A Gonzalez-Gonzalez
- Biomedical Engineering and Biomedical Sciences, University of Houston, Health 2, 4849 Calhoun Rd., Room 6014, Houston, TX, 77204-6064, USA
| | - Geetanjali S Bendale
- Biomedical Engineering and Biomedical Sciences, University of Houston, Health 2, 4849 Calhoun Rd., Room 6014, Houston, TX, 77204-6064, USA
| | - Kezhong Wang
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Gordon G Wallace
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Mario Romero-Ortega
- Biomedical Engineering and Biomedical Sciences, University of Houston, Health 2, 4849 Calhoun Rd., Room 6014, Houston, TX, 77204-6064, USA.
| |
Collapse
|
23
|
Vagus nerve stimulation activates two distinct neuroimmune circuits converging in the spleen to protect mice from kidney injury. Proc Natl Acad Sci U S A 2021; 118:2021758118. [PMID: 33737395 DOI: 10.1073/pnas.2021758118] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury is highly prevalent and associated with high morbidity and mortality, and there are no approved drugs for its prevention and treatment. Vagus nerve stimulation (VNS) alleviates inflammatory diseases including kidney disease; however, neural circuits involved in VNS-induced tissue protection remain poorly understood. The vagus nerve, a heterogeneous group of neural fibers, innervates numerous organs. VNS broadly stimulates these fibers without specificity. We used optogenetics to selectively stimulate vagus efferent or afferent fibers. Anterograde efferent fiber stimulation or anterograde (centripetal) sensory afferent fiber stimulation both conferred kidney protection from ischemia-reperfusion injury. We identified the C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis as the downstream pathway of vagus afferent fiber stimulation. Our study provides a map of the neural circuits important for kidney protection induced by VNS, which is critical for the safe and effective clinical application of VNS for protection from acute kidney injury.
Collapse
|
24
|
Hypothalamic detection of macronutrients via multiple gut-brain pathways. Cell Metab 2021; 33:676-687.e5. [PMID: 33450178 PMCID: PMC7933100 DOI: 10.1016/j.cmet.2020.12.018] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022]
Abstract
Food intake is tightly regulated by complex and coordinated gut-brain interactions. Nutrients rapidly modulate activity in key populations of hypothalamic neurons that regulate food intake, including hunger-sensitive agouti-related protein (AgRP)-expressing neurons. Because individual macronutrients engage specific receptors in the gut to communicate with the brain, we reasoned that macronutrients may utilize different pathways to reduce activity in AgRP neurons. Here, we revealed that AgRP neuron activity in hungry mice is inhibited by site-specific intestinal detection of different macronutrients. We showed that vagal gut-brain signaling is required for AgRP neuron inhibition by fat. In contrast, spinal gut-brain signaling relays the presence of intestinal glucose. Further, we identified glucose sensors in the intestine and hepatic portal vein that mediate glucose-dependent AgRP neuron inhibition. Therefore, distinct pathways are activated by individual macronutrients to inhibit AgRP neuron activity.
Collapse
|
25
|
Medeiros KA, Siqueira BS, Urrutia MAD, Porto EM, Grassiolli S, Amorim JPDA. Vagotomy associated with splenectomy reduces lipid accumulation and causes kidneys histological changes in rats with hypothalamic obesity. Acta Cir Bras 2021; 36:e360205. [PMID: 33624722 PMCID: PMC7902077 DOI: 10.1590/acb360205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/13/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose To evaluate the influence of autonomic vagal and splenic activities on renal
histomorphometric aspects in obese rats. Methods Thirty male Wistar rats were used, of which, 24 received subcutaneous
injections of monosodium glutamate (MSG) during the first 5 days of life (4
g/kg body weight) and six control animals received injections of saline
solution (CON). Five experimental groups were organized (n = 6/group):
falsely-operated control (CON-FO); falsely-operated obese (MSG-FO);
vagotomized obese (MSG-VAG); splenectomized obese (MSG-SPL); vagotomized and
splenectomized obese (MSG-VAG-SPL). Results The MSG-FO group animals showed a significant reduction in body weight and
nasal-anal length when compared to CON-FO group animals (p < 0.05). The
MSG-VAG-SPL group showed significant reduced in most biometric parameters
associated with obesity. Falsely-operated obese animals showed a significant
reduction in renal weight, glomerular diameters, glomerular tuff and capsule
areas and Bowman’s space compared to CON-FO group animals (p < 0.05).
There was a significant reduction in diameter, glomerular tuft and capsule
areas, and Bowman’s space in MSG-VAG, MSG-SPL, MSG-VAG-SPL groups when
compared to the MSG-FO group. Conclusions Vagotomy associated with splenectomy induces a reduction in the adiposity and
causes histological changes in the kidney of obese rats.
Collapse
|
26
|
Courties A, Berenbaum F, Sellam J. Vagus nerve stimulation in musculoskeletal diseases. Joint Bone Spine 2021; 88:105149. [PMID: 33548494 DOI: 10.1016/j.jbspin.2021.105149] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/21/2021] [Indexed: 01/21/2023]
Abstract
The vagus nerve is the main nerve of the parasympathetic autonomic nervous system. Beyond its vegetative functions, the vagus nerve possesses anti-inflammatory and analgesic properties. Initially developed in the treatment of refractory epilepsy, vagus nerve stimulation (VNS) is currently being evaluated in several musculoskeletal diseases. VNS can be invasive by placing an electrode around the cervical vagus nerve and connected to a generator implanted subcutaneously or non-invasive stimulating the cervical vagus nerve branch percutaneously (auricular or cervical). In rheumatoid arthritis (RA) patients, VNS has been shown to dampen the inflammatory response of circulatory peripheral cells. Several open-labeled small pilot studies have demonstrated that VNS, either invasive or transcutaneous, is associated with a significant decrease of RA disease activity. As well, other studies have shown that VNS could limit fatigue in Sjogren's syndrome and systemic lupus, or decrease pain in fibromyalgia as well as in erosive hand osteoarthritis. However, some questions remain, such as the settings of stimulation, the duration of treatment, or the optimal stimulation route. Finally, randomized controlled trials versus sham stimulation with large samples of patients are mandatory to definitively conclude about the efficacy of VNS.
Collapse
Affiliation(s)
- Alice Courties
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France
| | - Francis Berenbaum
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jérémie Sellam
- INSERM UMRS 938, service de rhumatologie, Sorbonne Université, hôpital Saint-Antoine, AP-HP, Paris, France.
| |
Collapse
|
27
|
Geng QS, Shen ZB, Zheng YY, Xue WH, Li LF, Zhao J. Precise medication for tumor patients in the context of mental stress. Cell Transplant 2021; 30:9636897211049813. [PMID: 34719974 PMCID: PMC8564128 DOI: 10.1177/09636897211049813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 11/17/2022] Open
Abstract
Cancer is the leading cause of disease-related death worldwide due to its late diagnosis and poor outcomes. Precision medicine plays an important role in the treatment of tumors. As found for many types of tumors, mental stress plays a vital role in the promotion and progression of tumors. In this paper, we briefly introduce the manifestation and effects of mental symptoms in tumor patients. We next specifically discuss the multiple roles of precision medicine in the tumor therapy. Finally, we also highlight the precision medicine strategy for psychiatric symptoms in tumor patients, which promises to enhance the efficacy of tumor therapy.
Collapse
Affiliation(s)
- Qi-Shun Geng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou
University, Zhengzhou, China
- Qi-Shun Geng and Zhi-Bo Shen are co-first author and equally
contributed to this work
| | - Zhi-Bo Shen
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou
University, Zhengzhou, China
- Qi-Shun Geng and Zhi-Bo Shen are co-first author and equally
contributed to this work
| | - Yuan-Yuan Zheng
- Internet Medical and System Applications of National Engineering
Laboratory, Zhengzhou, Henan, China
| | - Wen-Hua Xue
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou
University, Zhengzhou, China
| | - Li-Feng Li
- Internet Medical and System Applications of National Engineering
Laboratory, Zhengzhou, Henan, China
| | - Jie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou
University, Zhengzhou, China
- Internet Medical and System Applications of National Engineering
Laboratory, Zhengzhou, Henan, China
| |
Collapse
|
28
|
Kressel AM, Tsaava T, Levine YA, Chang EH, Addorisio ME, Chang Q, Burbach BJ, Carnevale D, Lembo G, Zador AM, Andersson U, Pavlov VA, Chavan SS, Tracey KJ. Identification of a brainstem locus that inhibits tumor necrosis factor. Proc Natl Acad Sci U S A 2020; 117:29803-29810. [PMID: 33168718 PMCID: PMC7703602 DOI: 10.1073/pnas.2008213117] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the brain, compact clusters of neuron cell bodies, termed nuclei, are essential for maintaining parameters of host physiology within a narrow range optimal for health. Neurons residing in the brainstem dorsal motor nucleus (DMN) project in the vagus nerve to communicate with the lungs, liver, gastrointestinal tract, and other organs. Vagus nerve-mediated reflexes also control immune system responses to infection and injury by inhibiting the production of tumor necrosis factor (TNF) and other cytokines in the spleen, although the function of DMN neurons in regulating TNF release is not known. Here, optogenetics and functional mapping reveal cholinergic neurons in the DMN, which project to the celiac-superior mesenteric ganglia, significantly increase splenic nerve activity and inhibit TNF production. Efferent vagus nerve fibers terminating in the celiac-superior mesenteric ganglia form varicose-like structures surrounding individual nerve cell bodies innervating the spleen. Selective optogenetic activation of DMN cholinergic neurons or electrical activation of the cervical vagus nerve evokes action potentials in the splenic nerve. Pharmacological blockade and surgical transection of the vagus nerve inhibit vagus nerve-evoked splenic nerve responses. These results indicate that cholinergic neurons residing in the brainstem DMN control TNF production, revealing a role for brainstem coordination of immunity.
Collapse
Affiliation(s)
- Adam M Kressel
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY 11030
| | - Tea Tsaava
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | | | - Eric H Chang
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Meghan E Addorisio
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Qing Chang
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | | | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | | | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Valentin A Pavlov
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549
| | - Sangeeta S Chavan
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549
| |
Collapse
|
29
|
Wasinski F, Klein MO, Bittencourt JC, Metzger M, Donato J. Distribution of growth hormone-responsive cells in the brain of rats and mice. Brain Res 2020; 1751:147189. [PMID: 33152340 DOI: 10.1016/j.brainres.2020.147189] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
A growth hormone (GH) injection is able to induce the phosphorylated form of the signal transducer and activator of transcription 5 (pSTAT5) in a large number of cells throughout the mouse brain. The present study had the objective to map the distribution of GH-responsive cells in the brain of rats that received an intracerebroventricular injection of GH and compare it to the pattern found in mice. We observed that rats and mice exhibited a similar distribution of GH-induced pSTAT5 in the majority of areas of the telencephalon, hypothalamus and brainstem. However, rats exhibited a higher density of GH-responsive cells than mice in the horizontal limb of the diagonal band of Broca (HDB), supraoptic and suprachiasmatic nuclei, whereas mice displayed more GH-responsive cells than rats in the hippocampus, lateral hypothalamic area and dorsal motor nucleus of the vagus (DMX). Since both HDB and DMX contain acetylcholine-producing neurons, pSTAT5 was co-localized with choline acetyltransferase in GH-injected animals. We found that 50.0 ± 4.5% of cholinergic neurons in the rat HDB coexpressed GH-induced pSTAT5, whereas very few co-localizations were observed in the mouse HDB. In contrast, rats displayed fewer cholinergic neurons responsive to GH in the DMX at the level of the area postrema. In summary, pSTAT5 can be used as a marker of GH-responsive cells in the rat brain. Although rats and mice exhibit a relatively similar distribution of GH-responsive neurons, some species-specific differences exist, as exemplified for the responsiveness to GH in distinct populations of cholinergic neurons.
Collapse
Affiliation(s)
- Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Marianne O Klein
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Jackson C Bittencourt
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Martin Metzger
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo, Brazil.
| |
Collapse
|
30
|
Cawthon CR, Kirkland RA, Pandya S, Brinson NA, de La Serre CB. Non-neuronal crosstalk promotes an inflammatory response in nodose ganglia cultures after exposure to byproducts from gram positive, high-fat-diet-associated gut bacteria. Physiol Behav 2020; 226:113124. [PMID: 32763334 PMCID: PMC7530053 DOI: 10.1016/j.physbeh.2020.113124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
Vagal afferent neurons (VAN) projecting to the lamina propria of the digestive tract are the primary source of gut-originating signals to the central nervous system (CNS). VAN cell bodies are found in the nodose ganglia (NG). Responsiveness of VAN to gut-originating signals is altered by feeding status with sensitivity to satiety signals such as cholecystokinin (CCK) increasing in the fed state. Chronic high-fat (HF) feeding results in inflammation at the level of the NG associated with a loss of VAN ability to switch phenotype from the fasted to the fed state. HF feeding also leads to compositional changes in the gut microbiota. HF diet consumption notably drives increased Firmicutes to Bacteroidetes phyla ratio and increased members of the Actinobacteria phylum. Firmicutes and Actinobacteria are largely gram positive (GP). In this study, we aimed to determine if byproducts from GP bacteria can induce an inflammatory response in cultured NG and to characterize the mechanism and cell types involved in the response. NG were collected from male Wistar rats and cultured for a total of 72 hours. At 48-68 hours after plating, cultures were treated with neuronal culture media in which Serinicoccus chungangensis had been grown and removed (SUP), lipoteichoic acid (LTA), or meso-diaminopimelic acid (meso-DAP). Some treatments included the glial inhibitors minocycline (MINO) and/or fluorocitrate (FC). The responses were evaluated using immunocytochemistry, qPCR, and electrochemiluminescence. We found that SUP induced an inflammatory response characterized by increased interleukin (IL)-6 staining and increased expression of genes for IL-6, interferon (IFN)γ, and tumor necrosis factor (TNF)α along with genes associated with cell-to-cell communication such as C-C motif chemokine ligand-2 (CCL2). Inclusion of inhibitors attenuated some responses but failed to completely normalize all indications of response, highlighting the role of immunocompetent cellular crosstalk in regulating the inflammatory response. LTA and meso-DAP produced responses that shared characteristics with SUP but were not identical. Our results support a role for HF associated GP bacterial byproducts' ability to contribute to vagal inflammation and to engage signaling from nonneuronal cells.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Rebecca A Kirkland
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Shreya Pandya
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Nigel A Brinson
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Claire B de La Serre
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States.
| |
Collapse
|
31
|
Quaresma PGF, Teixeira PDS, Wasinski F, Campos AMP, List EO, Kopchick JJ, Donato J. Cholinergic neurons in the hypothalamus and dorsal motor nucleus of the vagus are directly responsive to growth hormone. Life Sci 2020; 259:118229. [PMID: 32781065 DOI: 10.1016/j.lfs.2020.118229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
AIMS Cholinergic neurons are distributed in brain areas containing growth hormone (GH)-responsive cells. We determined if cholinergic neurons are directly responsive to GH and the metabolic consequences of deleting the GH receptor (GHR) specifically in choline acetyltransferase (ChAT)-expressing cells. MAIN METHODS Mice received an acute injection of GH to detect neurons co-expressing ChAT and phosphorylated STAT5 (pSTAT5), a well-established marker of GH-responsive cells. For the physiological studies, mice carrying ablation of GHR exclusively in ChAT-expressing cells were produced and possible changes in energy and glucose homeostasis were determined when consuming regular chow or high-fat diet (HFD). KEY FINDINGS The majority of cholinergic neurons in the arcuate nucleus (60%) and dorsomedial nucleus (84%) of the hypothalamus are directly responsive to GH. Approximately 34% of pre-ganglionic parasympathetic neurons in the dorsal motor nucleus of the vagus also exhibited GH-induced pSTAT5. GH-induced pSTAT5 in these ChAT neurons was absent in GHR ChAT knockout mice. Mice carrying ChAT-specific GHR deletion, either in chow or HFD, did not exhibit significant changes in body weight, body adiposity, lean body mass, food intake, energy expenditure, respiratory quotient, ambulatory activity, serum leptin levels, glucose tolerance, insulin sensitivity and metabolic responses to 2-deoxy-d-glucose. However, GHR deletion in ChAT neurons caused decreased hypothalamic Pomc mRNA levels in HFD mice. SIGNIFICANCE Cholinergic neurons that regulate the metabolism are directly responsive to GH, although GHR signaling in these cells is not required for energy and glucose homeostasis. Thus, the physiological importance of GH action on cholinergic neurons still needs to be identified.
Collapse
Affiliation(s)
- Paula G F Quaresma
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Pryscila D S Teixeira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Ana M P Campos
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
32
|
Holland J, Sorrell J, Yates E, Smith K, Arbabi S, Arnold M, Rivir M, Morano R, Chen J, Zhang X, Dimarchi R, Woods SC, Sanchez-Gurmaches J, Wohleb E, Perez-Tilve D. A Brain-Melanocortin-Vagus Axis Mediates Adipose Tissue Expansion Independently of Energy Intake. Cell Rep 2020; 27:2399-2410.e6. [PMID: 31116984 PMCID: PMC6550338 DOI: 10.1016/j.celrep.2019.04.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
The melanocortin system is a brain circuit that influences energy balance by regulating energy intake and expenditure. In addition, the brain-melanocortin system controls adipose tissue metabolism to optimize fuel mobilization and storage. Specifically, increased brain-melanocortin signaling or negative energy balance promotes lipid mobilization by increasing sympathetic nervous system input to adipose tissue. In contrast, calorie-independent mechanisms favoring energy storage are less understood. Here, we demonstrate that reduction of brain-melanocortin signaling actively promotes fat mass gain by activating the lipogenic program and adipocyte and endothelial cell proliferation in white fat depots independently of caloric intake via efferent nerve fibers conveyed by the common hepatic branch of the vagus nerve. Those vagally regulated obesogenic signals also contribute to the fat mass gain following chronic high-fat diet feeding. These data reveal a physiological mechanism whereby the brain controls energy stores that may contribute to increased susceptibility to obesity. Brain-melanocortin signaling controls fat mass indirectly by regulating energy balance and by direct control of lipid mobilization from adipose tissue via sympathetic nervous system activity. Holland et al. show that reduced brain-melanocortin signaling promotes white adipose tissue expansion via signals conveyed by efferent innervation of the vagus nerve.
Collapse
Affiliation(s)
- Jenna Holland
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joyce Sorrell
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Emily Yates
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathleen Smith
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shahriar Arbabi
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Marita Rivir
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rachel Morano
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jenny Chen
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard Dimarchi
- Novo Nordisk Research Center Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Endocrinology and Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
33
|
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
|
34
|
Bookout AL, Gautron L. Characterization of a cell bridge variant connecting the nodose and superior cervical ganglia in the mouse: Prevalence, anatomical features, and practical implications. J Comp Neurol 2020; 529:111-128. [PMID: 32356570 DOI: 10.1002/cne.24936] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/08/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022]
Abstract
While autonomic ganglia have been extensively studied in rats instead of mice, there is renewed interest in the anatomy of the mouse autonomic nervous system. This study examined the prevalence and anatomical features of a cell bridge linking two autonomic ganglia of the neck, namely, the nodose ganglion (NG) and the superior cervical ganglion (SCG) in a cohort of C57BL/6J mice. We identified a cell bridge between the NG and the cranial pole of the SCG. This cell bridge was tubular shaped with an average length and width of 700 and 240 μm, respectively. The cell bridge was frequently unilateral and significantly more prevalent in the ganglionic masses from males (38%) than females (21%). On each of its extremities, it contained a mixed of vagal afferents and postganglionic sympathetic neurons. The two populations of neurons abruptly replaced each other in the middle of the cell bridge. We examined the mRNA expression for selected autonomic markers in samples of the NG with or without cell bridge. Our results indicated that the cell bridge was enriched in both markers of postganglionic sympathetic and vagal afferents neurons. Lastly, using FluoroGold microinjection into the NG, we found that the existence of a cell bridge may occasionally lead to the inadvertent contamination of the SCG. In summary, this study describes the anatomy of a cell bridge variant consisting of the fusion of the mouse NG and SCG. The practical implications of our observations are discussed with respect to studies of the mouse vagal afferents, an area of research of increasing popularity.
Collapse
Affiliation(s)
- Angie L Bookout
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
35
|
Bassi GS, Kanashiro A, Coimbra NC, Terrando N, Maixner W, Ulloa L. Anatomical and clinical implications of vagal modulation of the spleen. Neurosci Biobehav Rev 2020; 112:363-373. [PMID: 32061636 DOI: 10.1016/j.neubiorev.2020.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023]
Abstract
The vagus nerve coordinates most physiologic functions including the cardiovascular and immune systems. This mechanism has significant clinical implications because electrical stimulation of the vagus nerve can control inflammation and organ injury in infectious and inflammatory disorders. The complex mechanisms that mediate vagal modulation of systemic inflammation are mainly regulated via the spleen. More specifically, vagal stimulation prevents organ injury and systemic inflammation by inhibiting the production of cytokines in the spleen. However, the neuronal regulation of the spleen is controversial suggesting that it can be mediated by either monosynaptic innervation of the splenic parenchyma or secondary neurons from the celiac ganglion depending on the experimental conditions. Recent physiologic and anatomic studies suggest that inflammation is regulated by neuro-immune multi-synaptic interactions between the vagus and the splanchnic nerves to modulate the spleen. Here, we review the current knowledge on these interactions, and discuss their experimental and clinical implications in infectious and inflammatory disorders.
Collapse
Affiliation(s)
- Gabriel S Bassi
- Center for Perioperative Organ Protection, Department of Anesthesiology. Duke University Medical Center, Durham, NC 27710, USA.
| | - Alexandre Kanashiro
- Department of Pharmacology and Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Norberto C Coimbra
- Department of Pharmacology and Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Niccolò Terrando
- Center for Perioperative Organ Protection, Department of Anesthesiology. Duke University Medical Center, Durham, NC 27710, USA
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology. Duke University, Durham, NC 27710, USA
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology. Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
36
|
Pro-inflammatory cytokines in the paraventricular nucleus mediate the adipose afferent reflex in rats. Pflugers Arch 2020; 472:343-354. [PMID: 32086614 DOI: 10.1007/s00424-020-02356-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 01/01/2023]
Abstract
Our previous study showed that the adipose afferent reflex (AAR) induced by chemical stimulation of white adipose tissue (WAT) increased sympathetic outflow and blood pressure. We also found that pro-inflammatory cytokines (PICs) in the hypothalamic paraventricular nucleus (PVN) potentiate the cardiac sympathetic afferent reflex in rats. However, the role of PICs in the PVN in regulating the AAR is still not clear. This study determined whether PICs in the PVN mediate the AAR in rats. The AAR was evaluated based on renal sympathetic nerve activity and mean arterial blood pressure in response to capsaicin injection into inguinal WAT (iWAT). PIC levels were measured by ELISA. PVN microinjection with the PICs tumor necrosis factor (TNF)-α or interleukin (IL)-1β enhanced the AAR in a dose-dependent manner. Furthermore, pretreatment via the bilateral microinjection of the TNF-α-blocker etanercept or IL-1β blocker IL-1ra into the PVN attenuated the AAR. In rats pretreated with TNF-α or IL-1β, a sub-response dose of angiotensin II (Ang II) significantly enhanced the AAR. Moreover, delivery of the angiotensin II type 1(AT1) receptor antagonist losartan into the PVN attenuated the effects of TNF-α or IL-1β on the AAR. In addition, stimulating either iWAT or retroperitoneal WAT with capsaicin increased TNF-α or IL-1β levels in the PVN, but the injection of capsaicin into the jugular vein, skeletal muscle, and skin had no effects on TNF-α or IL-1β levels in the PVN. These results suggest that TNF-α or IL-1β and Ang II in the PVN synergistically enhance the AAR in rats.
Collapse
|
37
|
Jarczyk J, Yard BA, Hoeger S. The Cholinergic Anti-Inflammatory Pathway as a Conceptual Framework to Treat Inflammation-Mediated Renal Injury. Kidney Blood Press Res 2020; 44:435-448. [PMID: 31307039 DOI: 10.1159/000500920] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/12/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway, positioned at the interface of the nervous and immune systems, is the efferent limb of the "inflammatory reflex" which mainly signals through the vagus nerve. As such, the brain can modulate peripheral inflammatory responses by the activation of vagal efferent fibers. Importantly, immune cells in the spleen express most cholinergic system components such as acetylcholine (ACh), choline acetyltransferase, acetylcholinesterase, and both muscarinic and nicotinic ACh receptors, making communication between both systems possible. In general, this communication down-regulates the inflammation, achieved through different mechanisms and depending on the cells involved. SUMMARY With the awareness that the cholinergic anti-inflammatory pathway serves to prevent or limit inflammation in peripheral organs, vagus nerve stimulation has become a promising strategy in the treatment of several inflammatory conditions. Both pharmacological and non-pharmacological methods have been used in many studies to limit organ injury as a consequence of inflammation. Key Messages: In this review, we will highlight our current knowledge of the cholinergic anti-inflammatory pathway, with emphasis on its potential clinical use in the treatment of inflammation-triggered kidney injury.
Collapse
Affiliation(s)
- Jonas Jarczyk
- Department of Urology, University Medical Center Mannheim, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| | - Benito A Yard
- Vth Medical Department, University Medical Center Mannheim, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| | - Simone Hoeger
- Vth Medical Department, University Medical Center Mannheim, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, Mannheim, Germany, .,Bioassay GmbH, Heidelberg, Germany,
| |
Collapse
|
38
|
Zhou L, Lin X, Ma X, Liu Y, Ma L, Chen Z, Chen H, Si L, Chen X. Acetylcholine regulates the development of experimental autoimmune encephalomyelitis via the CD4+ cells proliferation and differentiation. Int J Neurosci 2020; 130:788-803. [PMID: 31906749 DOI: 10.1080/00207454.2019.1706504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Linli Zhou
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
- Epidemiology and Infection Control Section, Medical Affairs Department, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiuli Lin
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Xiaomeng Ma
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Yingying Liu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Lili Ma
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Zhaoyu Chen
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Hao Chen
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Lei Si
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Xiaohong Chen
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong China
| |
Collapse
|
39
|
Tanaka S, Okusa MD. Crosstalk between the nervous system and the kidney. Kidney Int 2019; 97:466-476. [PMID: 32001065 DOI: 10.1016/j.kint.2019.10.032] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
Under physiological states, the nervous system and the kidneys communicate with each other to maintain normal body homeostasis. However, pathological states disrupt this interaction as seen in hypertension, and kidney damage can cause impaired renorenal reflex and sodium handling. In acute kidney injury (AKI) and chronic kidney disease (CKD), damaged kidneys can have a detrimental effect on the central nervous system. CKD is an independent risk factor for cerebrovascular disease and cognitive impairment, and many factors, including retention of uremic toxins and phosphate, have been proposed as CKD-specific factors responsible for structural and functional cerebral changes in patients with CKD. However, more studies are needed to determine the precise pathogenesis. Epidemiological studies have shown that AKI is associated with a subsequent risk for developing stroke and dementia. However, recent animal studies have shown that the renal nerve contributes to kidney inflammation and fibrosis, whereas activation of the cholinergic anti-inflammatory pathway, which involves the vagus nerve, the splenic nerve, and immune cells in the spleen, has a significant renoprotective effect. Therefore, elucidating mechanisms of communication between the nervous system and the kidney enables us not only to develop new strategies to ameliorate neurological conditions associated with kidney disease but also to design safe and effective clinical interventions for kidney disease, using the neural and neuroimmune control of kidney injury and disease.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
40
|
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
|
41
|
Linares R, Rosas G, Vieyra E, Ramírez DA, Velázquez DR, Espinoza JA, Morán C, Domínguez R, Morales-Ledesma L. In Adult Rats With Polycystic Ovarian Syndrome, Unilateral or Bilateral Vagotomy Modifies the Noradrenergic Concentration in the Ovaries and the Celiac Superior Mesenteric Ganglia in Different Ways. Front Physiol 2019; 10:1309. [PMID: 31695622 PMCID: PMC6817458 DOI: 10.3389/fphys.2019.01309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
In rats with polycystic ovarian syndrome (PCOS) induced by estradiol valerate (EV) injection, sectioning of the vagus nerve in the juvenile stage restores ovulatory function, suggesting that the vagus nerve stimulates the onset and development of PCOS. We analyzed whether in adult rats, the role played by the vagus nerve in PCOS development is associated with the nerve’s regulation of noradrenergic activity in the celiac superior mesenteric ganglion (CSMG). Ten-day-old rats were injected with corn oil [vehicle (Vh)] or EV (2 mg). At 76 days of age, rats injected with Vh or EV were subjected to sham surgery or the sectioning of one or both vagus nerves (vagotomy). The animals were sacrificed at 80–82 days of age at vaginal estrus smear. Compared to Vh-treated animals, EV-induced PCOS rats showed a lack of ovulation, the presence of follicular cysts, and a high concentration of testosterone, without changes in noradrenaline concentrations in the CSMG or ovaries. In PCOS rats, sham surgery lowered serum testosterone and noradrenaline concentrations in the CSMG but did not restore ovulation. In animals with PCOS, vagotomy lowered testosterone concentrations to a larger degree than in sham-surgery animals. The ovaries of rats with PCOS and vagotomy showed fresh corpora lutea, indicating ovulation. In EV-treated rats with unilateral vagotomy, the concentration of noradrenaline in the CSMG was similar to that in rats with PCOS and sham surgery, which did not ovulate, while in the ovaries of PCOS rats with left or bilateral vagotomy, the noradrenaline concentration was lower than that in sham-surgery-treated animals. Our results suggest that the vagus nerve regulates PCOS development through a different mechanism than the increase in the noradrenergic activity in the CSMG; however, in ovaries, the restoration of ovulation is associated with a decrease in ovarian noradrenaline.
Collapse
Affiliation(s)
- Rosa Linares
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Gabriela Rosas
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Elizabeth Vieyra
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Deyra A Ramírez
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Daniel R Velázquez
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Julieta A Espinoza
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Roberto Domínguez
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| | - Leticia Morales-Ledesma
- Laboratorio de Fisiología Reproductiva, de la Unidad de Investigación en Biología de la Reproducción, Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
| |
Collapse
|
42
|
Thompson N, Mastitskaya S, Holder D. Avoiding off-target effects in electrical stimulation of the cervical vagus nerve: Neuroanatomical tracing techniques to study fascicular anatomy of the vagus nerve. J Neurosci Methods 2019; 325:108325. [PMID: 31260728 PMCID: PMC6698726 DOI: 10.1016/j.jneumeth.2019.108325] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022]
Abstract
Vagus nerve stimulation (VNS) is a promising therapy for treatment of various conditions that are resistant to standard medication, such as heart failure, epilepsy, and depression. The vagus nerve is a complex nerve providing afferent and efferent innervation of the pharynx, larynx, heart, tracheobronchial tree and lungs, oesophagus, stomach, liver, pancreas, small intestine and proximal colon. It is therefore a prime target for intervention for VNS. Surprisingly, the fascicular organisation of the vagus nerve at the cervical level is still not well understood. This, along with the current stimulation techniques, results in the entire nerve being stimulated, which leads to unwanted off-target effects. Neuronal tracing is a promising method to delineate the organ-specific innervation by the vagus nerve, thereby providing valuable insight into the fascicular anatomy. In this review we discuss the current knowledge of vagus nerve anatomy and neuronal tracers used for mapping of its organ-specific projections in various species. Efferent vagal projections are a chain of two neurones (pre- and postganglionic), while afferent projections consist of only one pseudounipolar neurone with one branch terminating in the target organ/tissue directly and another in the brainstem. It would be feasible to retrogradely trace the afferent fibres from their respective visceral targets and identify them at the cervical level using non-transsynaptic neuronal tracers. Using this to create a map of the functional anatomical organisation of the vagus nerve will enable selective VNS ultimately allowing for the avoidance of the off-target effects and improving overall efficacy.
Collapse
Affiliation(s)
- Nicole Thompson
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom.
| | - Svetlana Mastitskaya
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - David Holder
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| |
Collapse
|
43
|
Tanaka S, Hammond B, Rosin DL, Okusa MD. Neuroimmunomodulation of tissue injury and disease: an expanding view of the inflammatory reflex pathway. Bioelectron Med 2019; 5:13. [PMID: 32232102 PMCID: PMC7098254 DOI: 10.1186/s42234-019-0029-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroimmunomodulation through peripheral nerve activation is an important therapeutic approach to various disorders. Central to this approach is the inflammatory reflex pathway in which the cholinergic anti-inflammatory pathway represents the efferent limb. Recent studies provide a framework for understanding this control pathway, however our understanding remains incomplete. Genetically modified mice, using optogenetics and pharmacogenomics, have been invaluable resources that will allow investigators to disentangle neural pathways that provide a unifying mechanism by which vagal nerve stimulation (and other means of stimulating the pathway) leads to an anti-inflammatory and tissue protective effect. In this review we describe disease models that contribute to our understanding of how vagal nerve stimulation attenuates inflammation and organ injury: acute kidney injury, rheumatoid arthritis, and inflammatory gastrointestinal disease. The gut microbiota contributes to health and disease and the potential role of the vagus nerve in affecting the relationship between gut microbiota and the immune system and modifying diseases remains an intriguing opportunity to attenuate local and systemic inflammation that undergird disease processes.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia USA
| | | | - Diane L. Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia USA
| | - Mark D. Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia USA
| |
Collapse
|
44
|
Berthoud HR, Neuhuber WL. Vagal mechanisms as neuromodulatory targets for the treatment of metabolic disease. Ann N Y Acad Sci 2019; 1454:42-55. [PMID: 31268181 PMCID: PMC6810744 DOI: 10.1111/nyas.14182] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/23/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
With few effective treatments available, the global rise of metabolic diseases, including obesity, type 2 diabetes mellitus, and cardiovascular disease, seems unstoppable. Likely caused by an obesogenic environment interacting with genetic susceptibility, the pathophysiology of obesity and metabolic diseases is highly complex and involves crosstalk between many organs and systems, including the brain. The vagus nerve is in a key position to bidirectionally link several peripheral metabolic organs with the brain and is increasingly targeted for neuromodulation therapy to treat metabolic disease. Here, we review the basics of vagal functional anatomy and its implications for vagal neuromodulation therapies. We find that most existing vagal neuromodulation techniques either ignore or misinterpret the rich functional specificity of both vagal efferents and afferents as demonstrated by a large body of literature. This lack of specificity of manipulating vagal fibers is likely the reason for the relatively poor beneficial long‐term effects of such therapies. For these therapies to become more effective, rigorous validation of all physiological endpoints and optimization of stimulation parameters as well as electrode placements will be necessary. However, given the large number of function‐specific fibers in any vagal branch, genetically guided neuromodulation techniques are more likely to succeed.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Winfried L Neuhuber
- Institut fur Anatomie und Zellbiologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
45
|
Abstract
Neuroimmune interaction is an emerging concept, wherein the nervous system modulates the immune system and vice versa. This concept is gaining attention as a novel therapeutic target in various inflammatory diseases including acute kidney injury (AKI). Vagus nerve stimulation or treatment with pulsed ultrasound activates the cholinergic anti-inflammatory pathway to prevent AKI in mice. The kidneys are innervated by sympathetic efferent and sensory afferent neurons, and these neurons also may play a role in the modulation of inflammation in AKI. In this review, we discuss several neural circuits with respect to the control of renal inflammation and AKI as well as optogenetics as a novel tool for understanding these complex neural circuits.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA.
| |
Collapse
|
46
|
Elkhatib SK, Case AJ. Autonomic regulation of T-lymphocytes: Implications in cardiovascular disease. Pharmacol Res 2019; 146:104293. [PMID: 31176794 DOI: 10.1016/j.phrs.2019.104293] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022]
Abstract
The nervous and immune systems both serve as essential assessors and regulators of physiological function. Recently, there has been a great interest in how the nervous and immune systems interact to modulate both physiological and pathological states. In particular, the autonomic nervous system has a direct line of communication with immune cells anatomically, and moreover, immune cells possess receptors for autonomic neurotransmitters. This circumstantial evidence is suggestive of a functional interplay between the two systems, and extensive research over the past few decades has demonstrated neurotransmitters such as the catecholamines (i.e. dopamine, norepinephrine, and epinephrine) and acetylcholine have potent immunomodulating properties. Furthermore, immune cells, particularly T-lymphocytes, have now been found to express the cellular machinery for both the synthesis and degradation of neurotransmitters, which suggests the ability for both autocrine and paracrine signaling from these cells independent of the nervous system. The details underlying the functional interplay of this complex network of neuroimmune communication are still unclear, but this crosstalk is suggestive of significant implications on the pathogenesis of a number of autonomic-dysregulated and inflammation-mediated diseases. In particular, it is widely accepted that numerous forms of cardiovascular diseases possess imbalanced autonomic tone as well as altered T-lymphocyte function, but a paucity of literature exists discussing the direct role of neurotransmitters in shaping the inflammatory microenvironment during the progression or therapeutic management of these diseases. This review seeks to provide a fundamental framework for this autonomic neuroimmune interaction within T-lymphocytes, as well as the implications this may have in cardiovascular diseases.
Collapse
Affiliation(s)
- Safwan K Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
47
|
Kaestner CL, Smith EH, Peirce SG, Hoover DB. Immunohistochemical analysis of the mouse celiac ganglion: An integrative relay station of the peripheral nervous system. J Comp Neurol 2019; 527:2742-2760. [PMID: 31021409 DOI: 10.1002/cne.24705] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022]
Abstract
Celiac ganglia are important sites of signal integration and transduction. Their complex neurochemical anatomy has been studied extensively in guinea pigs but not in mice. The goal of this study was to provide detailed neurochemical characterization of mouse celiac ganglia and noradrenergic nerves in two target tissues, spleen and stomach. A vast majority of mouse celiac neurons express a noradrenergic phenotype, which includes tyrosine hydroxylase (TH), vesicular monoamine transporter 2, and the norepinephrine transporter. Over 80% of these neuron also express neuropeptide Y (NPY), and this coexpression is maintained by dissociated neurons in culture. Likewise, TH and NPY were colocalized in noradrenergic nerves throughout the spleen and in stomach blood vessels. Somatostatin was not detected in principal neurons but did occur in small, TH-negative cells presumed to be interneurons and in a few varicose nerve fibers. Cholinergic nerves provided the most abundant input to the ganglia, and small percentages of these also contained nitric oxide synthase or vasoactive intestinal polypeptide. A low-to-moderate density of nerves also stained separately for the latter markers. Additionally, nerve bundles and varicose nerve fibers containing the sensory neuropeptides, calcitonin gene-related polypeptide, and substance P, occurred at variable density throughout the ganglia. Collectively, these findings demonstrate that principal neurons of mouse celiac ganglia have less neurochemical diversity than reported for guinea pig and other species but receive input from nerves expressing an array of neurochemical markers. This profile suggests celiac neurons integrate input from many sources to influence target tissues by releasing primarily norepinephrine and NPY.
Collapse
Affiliation(s)
- Charlotte L Kaestner
- Department of Biomedical Sciences, Quillen College of Medicine, Johnson City, Tennessee
| | - Elizabeth H Smith
- Department of Biomedical Sciences, Quillen College of Medicine, Johnson City, Tennessee
| | - Stanley G Peirce
- Department of Biomedical Sciences, Quillen College of Medicine, Johnson City, Tennessee
| | - Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, Johnson City, Tennessee.,Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
48
|
Characterization of plasma cytokine response to intraperitoneally administered LPS & subdiaphragmatic branch vagus nerve stimulation in rat model. PLoS One 2019; 14:e0214317. [PMID: 30921373 PMCID: PMC6438475 DOI: 10.1371/journal.pone.0214317] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 03/11/2019] [Indexed: 01/17/2023] Open
Abstract
Vagus nerve stimulation (VNS) has been on the forefront of inflammatory disorder research and has yielded many promising results. Questions remain, however, about the biological mechanisms of such treatments and the inconsistencies in the methods used in research efforts. Here, we aimed to clarify the inflammatory response to intraperitoneal (IP) injections of lipopolysaccharide (LPS) in rats, while analyzing corresponding effects of electrical stimulation to subdiaphragmatic branches (anterior gastric, accessory celiac, and hepatic) of the left vagus nerve. We accomplished an in-depth characterization of the time-varying cytokine cascade response in the serum of 58 rats to an acute IP LPS challenge over a 330-minute period by utilizing curve-fitting and starting point-alignment methods. We then explored the post-LPS neuromodulation effects of electrically stimulating individually cuffed subdiaphragmatic branches. Through our analysis, we found there to be a consistent order of IP LPS cytokine response (IL-10, TNF-α, GM-CSF, IL-17F, IL-6, IL-22, INF-γ). Apart from IL-10, the IP cytokine cascade was more variable in starting time and occurred later than in previously recorded intravenous (IV) challenges. We also found distinct regulatory effects on multiple cytokine levels by each of the three subdiaphragmatic stimulation subsets. While the time-variability of IP LPS use in rats complicates its utility, we have shown it to be a practical, arguably more physiologically relevant method than IV in rats when our methods are used. More importantly, we have shown that selective subdiaphragmatic neurostimulation can be utilized to selectively induce specific effects on inflammation in the body.
Collapse
|
49
|
Soyalp C, Kocak MN, Ahiskalioglu A, Aksoy M, Atalay C, Aydin MD, Cakir M, Calikoglu C, Ozmen S. New determinants for casual peripheral mechanism of neurogenic lung edema in subarachnoid hemorrhage due to ischemic degeneration of vagal nerve, kidney and lung circuitry. Experimental study1. Acta Cir Bras 2019; 34:e201900303. [PMID: 30892389 PMCID: PMC6585894 DOI: 10.1590/s0102-865020190030000003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/10/2019] [Indexed: 01/13/2023] Open
Abstract
Purpose To evaluate whether there is a relationship between renal artery vasospasm
related low glomerular density or degeneration and neurogenic lung edema
(NLE) following subarachnoid hemorrhage. Methods This study was conducted on 26 rabbits. A control group was formed of five
animals, a SHAM group of 5 to which saline and a study group (n=16) injected
with homologous blood into the sylvian cisterna. Numbers of degenerated
axons of renal branches of vagal nerves, atrophic glomerulus numbers and NLE
scores were recorded. Results Important vagal degeneration, severe renal artery vasospasm, intrarenal
hemorrhage and glomerular atrophy observed in high score NLE detected
animals. The mean degenerated axon density of vagal nerves
(n/mm2), atrophic glomerulus density (n/mm3) and NLE
scores of control, SHAM and study groups were estimated as 2.40±1.82,
2.20±1.30, 1.80±1.10, 8.00±2.24, 8.80±2.39, 4.40±1.14 and 154.38±13.61,
34.69±2.68 and 12.19±1.97 consecutively. Degenerated vagal axon, atrophic
glomerulus and NLE scores are higher in study group than other groups and
the differences are statistically meaningful (p<0.001). Conclusion Vagal complex degeneration based glomerular atrophy have important roles on
NLE following SAH which has not been extensively mentioned in the
literature.
Collapse
Affiliation(s)
- Celaleddin Soyalp
- MD, Department of Anesthesiology, Medical Faculty, Yil University, Van, Turkey. Conception and design of the study, acquisition of data, manuscript writing
| | - Mehmet Nuri Kocak
- MD, Department of Neurology, Medical Faculty, Ataturk University, Erzurum, Turkey. Technical procedures, manuscript preparation
| | - Ali Ahiskalioglu
- Assistant Prof., Department of Anesthesiology and Reanimation, Medical Faculty, Ataturk University, Erzurum, Turkey. Acquisition, analysis and interpretation of data; manuscript preparation; critical revision
| | - Mehmet Aksoy
- Assistant Prof., Department of Anesthesiology and Reanimation, Medical Faculty, Ataturk University, Erzurum, Turkey. Acquisition, analysis and interpretation of data; manuscript preparation; critical revision
| | - Canan Atalay
- Assistant Prof., Department of Anesthesiology and Reanimation, Medical Faculty, Ataturk University, Erzurum, Turkey. Acquisition, analysis and interpretation of data; manuscript preparation; critical revision
| | - Mehmet Dumlu Aydin
- Prof., Department of Neurosurgery, Medical Faculty, Ataturk University, Erzurum, Turkey. Histopathological examinations, manuscript writing, critical revision, final approval
| | - Murteza Cakir
- Prof., Department of Neurosurgery, Medical Faculty, Ataturk University, Erzurum, Turkey. Histopathological examinations, manuscript writing, critical revision, final approval
| | - Cagatay Calikoglu
- Prof., Department of Neurosurgery, Medical Faculty, Ataturk University, Erzurum, Turkey. Histopathological examinations, manuscript writing, critical revision, final approval
| | - Sevilay Ozmen
- MD, Department of Pathology, Medical Faculty, Ataturk University, Erzurum, Turkey. Histopathological examinations
| |
Collapse
|
50
|
Wasilczuk KM, Bayer KC, Somann JP, Albors GO, Sturgis J, Lyle LT, Robinson JP, Irazoqui PP. Modulating the Inflammatory Reflex in Rats Using Low-Intensity Focused Ultrasound Stimulation of the Vagus Nerve. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:481-489. [PMID: 30396599 DOI: 10.1016/j.ultrasmedbio.2018.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 06/08/2023]
Abstract
Tumor necrosis factor α (TNF-α) is linked to several chronic inflammatory diseases. Electrical vagus nerve stimulation reduces serum TNF-α levels but may cause chronic nerve damage and requires surgery. Alternatively, we proposed focused ultrasound stimulation of the vagus nerve (uVNS), which can be applied non-invasively. In this study, we induced an inflammatory response in rats using lipopolysaccharides (LPS) and collected blood to analyze the effects of uVNS on cytokine concentrations. We applied one or three 5-min pulsed focused ultrasound stimulation treatments to the vagus nerve (250 kHz, ISPPA = 3 W/cm2). Animals receiving a single ultrasound application had an average reduction in TNF-α levels of 19%, similar to the 16% reduction observed in electrically stimulated animals. With multiple applications, uVNS therapy statistically reduced serum TNF-α levels by 73% compared with control animals without any observed damage to the nerve. These findings suggest that uVNS is a suitable way to attenuate TNF-α levels.
Collapse
Affiliation(s)
- Kelsey M Wasilczuk
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.
| | - Kelsey C Bayer
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Jesse P Somann
- Department of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Gabriel O Albors
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Jennifer Sturgis
- Purdue University Cytometry Laboratories, Purdue University, West Lafayette, Indiana, USA
| | - L Tiffany Lyle
- College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - J Paul Robinson
- Purdue University Cytometry Laboratories, Purdue University, West Lafayette, Indiana, USA
| | - Pedro P Irazoqui
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA; Department of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|