51
|
Possible Mechanisms for the Effects of Sound Vibration on Human Health. Healthcare (Basel) 2021; 9:healthcare9050597. [PMID: 34069792 PMCID: PMC8157227 DOI: 10.3390/healthcare9050597] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
This paper presents a narrative review of research literature to “map the landscape” of the mechanisms of the effect of sound vibration on humans including the physiological, neurological, and biochemical. It begins by narrowing music to sound and sound to vibration. The focus is on low frequency sound (up to 250 Hz) including infrasound (1–16 Hz). Types of application are described and include whole body vibration, vibroacoustics, and focal applications of vibration. Literature on mechanisms of response to vibration is categorized into hemodynamic, neurological, and musculoskeletal. Basic mechanisms of hemodynamic effects including stimulation of endothelial cells and vibropercussion; of neurological effects including protein kinases activation, nerve stimulation with a specific look at vibratory analgesia, and oscillatory coherence; of musculoskeletal effects including muscle stretch reflex, bone cell progenitor fate, vibration effects on bone ossification and resorption, and anabolic effects on spine and intervertebral discs. In every category research on clinical applications are described. The conclusion points to the complexity of the field of vibrational medicine and calls for specific comparative research on type of vibration delivery, amount of body or surface being stimulated, effect of specific frequencies and intensities to specific mechanisms, and to greater interdisciplinary cooperation and focus.
Collapse
|
52
|
Donegà M, Fjordbakk CT, Kirk J, Sokal DM, Gupta I, Hunsberger GE, Crawford A, Cook S, Viscasillas J, Stathopoulou TR, Miranda JA, Dopson WJ, Goodwin D, Rowles A, McGill P, McSloy A, Werling D, Witherington J, Chew DJ, Perkins JD. Human-relevant near-organ neuromodulation of the immune system via the splenic nerve. Proc Natl Acad Sci U S A 2021; 118:e2025428118. [PMID: 33972441 PMCID: PMC8157920 DOI: 10.1073/pnas.2025428118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuromodulation of immune function by stimulating the autonomic connections to the spleen has been demonstrated in rodent models. Consequently, neuroimmune modulation has been proposed as a new therapeutic strategy for the treatment of inflammatory conditions. However, demonstration of the translation of these immunomodulatory mechanisms in anatomically and physiologically relevant models is still lacking. Additionally, translational models are required to identify stimulation parameters that can be transferred to clinical applications of bioelectronic medicines. Here, we performed neuroanatomical and functional comparison of the mouse, rat, pig, and human splenic nerve using in vivo and ex vivo preparations. The pig was identified as a more suitable model of the human splenic innervation. Using functional electrophysiology, we developed a clinically relevant marker of splenic nerve engagement through stimulation-dependent reversible reduction in local blood flow. Translation of immunomodulatory mechanisms were then assessed using pig splenocytes and two models of acute inflammation in anesthetized pigs. The pig splenic nerve was shown to locally release noradrenaline upon stimulation, which was able to modulate cytokine production by pig splenocytes. Splenic nerve stimulation was found to promote cardiovascular protection as well as cytokine modulation in a high- and a low-dose lipopolysaccharide model, respectively. Importantly, splenic nerve-induced cytokine modulation was reproduced by stimulating the efferent trunk of the cervical vagus nerve. This work demonstrates that immune responses can be modulated by stimulation of spleen-targeted autonomic nerves in translational species and identifies splenic nerve stimulation parameters and biomarkers that are directly applicable to humans due to anatomical and electrophysiological similarities.
Collapse
Affiliation(s)
- Matteo Donegà
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom;
| | - Cathrine T Fjordbakk
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Joseph Kirk
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - David M Sokal
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Isha Gupta
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Gerald E Hunsberger
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Abbe Crawford
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Simon Cook
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Jaime Viscasillas
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | | | - Jason A Miranda
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Wesley J Dopson
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - David Goodwin
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Alison Rowles
- Non-Clinical Safety, GlaxoSmithKline, Ware SG12 0DP, United Kingdom
| | - Paul McGill
- Bioimaging, GlaxoSmithKline, Ware SG12 0DP, United Kingdom
| | - Alex McSloy
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, United Kingdom
| | - Jason Witherington
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Daniel J Chew
- Translation and Engineering, Galvani Bioelectronics, Stevenage SG1 2NY, United Kingdom
| | - Justin D Perkins
- Clinical Sciences and Services, The Royal Veterinary College, Hatfield AL9 7TA, United Kingdom;
| |
Collapse
|
53
|
Brognara F, Castania JA, Kanashiro A, Dias DPM, Salgado HC. Physiological Sympathetic Activation Reduces Systemic Inflammation: Role of Baroreflex and Chemoreflex. Front Immunol 2021; 12:637845. [PMID: 33995355 PMCID: PMC8117744 DOI: 10.3389/fimmu.2021.637845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Baroreflex and chemoreflex act through the autonomic nervous system, which is involved with the neural regulation of inflammation. The present study reports the effects of reflex physiological sympathetic activation in endotoxemic rats using bilateral carotid occlusion (BCO), a physiological approach involving the baroreflex and chemoreflex mechanisms and the influence of the baroreceptors and peripheral chemoreceptors in the cardiovascular and systemic inflammatory responses. After lipopolysaccharide (LPS) administration, the arterial pressure was recorded during 360 min in unanesthetized rats, and serial blood samples were collected to analyze the plasma cytokine levels. BCO elicited the reflex activation of the sympathetic nervous system, providing the following outcomes: (I) increased the power of the low-frequency band in the spectrum of the systolic arterial pressure during the BCO period; (II) reduced the levels of pro-inflammatory cytokines in plasma, including the tumor necrosis factor (TNF) and the interleukin (IL)-1β; (III) increased the plasma levels of anti-inflammatory cytokine IL-10, 90 min after LPS administration. Moreover, selective baroreceptor or chemoreceptor denervation deactivated mechanosensitive and chemical sensors, respectively, and decreased the release of the LPS-induced cytokine but did not alter the BCO modulatory effects. These results show, for the first time, that physiological reflex activation of the sympathetic circuit decreases the inflammatory response in endotoxemic rats and suggest a novel function for the baroreceptors as immunosensors during the systemic inflammation.
Collapse
Affiliation(s)
- Fernanda Brognara
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jaci Airton Castania
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Kanashiro
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Helio Cesar Salgado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
54
|
Ueno M. Restoring neuro-immune circuitry after brain and spinal cord injuries. Int Immunol 2021; 33:311-325. [PMID: 33851981 DOI: 10.1093/intimm/dxab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Neuro-immune interactions are essential for our body's defense and homeostasis. Anatomical and physiological analyses have shown that the nervous system comprises multiple pathways that regulate the dynamics and functions of immune cells, which are mainly mediated by the autonomic nervous system and adrenal signals. These are disturbed when the neurons and circuits are damaged by diseases of the central nervous system (CNS). Injuries caused by stroke or trauma often cause immune dysfunction by abrogation of the immune-regulating neural pathways, which leads to an increased risk of infections. Here, I review the structures and functions of the neural pathways connecting the brain and the immune system, and the neurogenic mechanisms of immune dysfunction that emerge after CNS injuries. Recent technological advances in manipulating specific neural circuits have added mechanistic aspects of neuro-immune interactions and their dysfunctions. Understanding the neural bases of immune control and their pathological processes will deepen our knowledge of homeostasis and lead to the development of strategies to cure immune deficiencies observed in various CNS disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan
| |
Collapse
|
55
|
Mastitskaya S, Thompson N, Holder D. Selective Vagus Nerve Stimulation as a Therapeutic Approach for the Treatment of ARDS: A Rationale for Neuro-Immunomodulation in COVID-19 Disease. Front Neurosci 2021; 15:667036. [PMID: 33927594 PMCID: PMC8076564 DOI: 10.3389/fnins.2021.667036] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the most severe form of acute lung injury. It is induced by sepsis, aspiration, and pneumonia, including that caused by SARS coronavirus and human influenza viruses. The main pathophysiological mechanism of ARDS is a systemic inflammatory response. Vagus nerve stimulation (VNS) can limit cytokine production in the spleen and thereby dampen any systemic inflammation and inflammation-induced tissue damage in the lungs and other organs. However, the effects of increased parasympathetic outflow to the lungs when non-selective VNS is applied may result in bronchoconstriction, increased mucus secretion and enhance local pulmonary inflammatory activity; this may outweigh the beneficial systemic anti-inflammatory action of VNS. Organ/function-specific therapy can be achieved by imaging of localized fascicle activity within the vagus nerve and selective stimulation of identified organ-specific fascicles. This may be able to provide selective neuromodulation of different pathways within the vagus nerve and offer a novel means to improve outcome in ARDS. This has motivated this review in which we discuss the mechanisms of anti-inflammatory effects of VNS, progress in selective VNS techniques, and a possible application for ARDS.
Collapse
Affiliation(s)
- Svetlana Mastitskaya
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | | | | |
Collapse
|
56
|
Vagus nerve stimulation enhances the cholinergic anti-inflammatory pathway to reduce lung injury in acute respiratory distress syndrome via STAT3. Cell Death Discov 2021; 7:63. [PMID: 33782389 PMCID: PMC8005666 DOI: 10.1038/s41420-021-00431-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/11/2021] [Accepted: 02/14/2021] [Indexed: 01/02/2023] Open
Abstract
The cholinergic anti-inflammatory pathway (CAIP) is important for antagonizing inflammation and treating several diseases, including acute respiratory distress syndrome (ARDS), and is related to vagus nerve integrity. However, its underlying pathophysiological mechanism is still unclear. We hypothesized that CAIP regulates lung injury repair after ARDS through the STAT3 signaling pathway, which is an important downstream effector of α7nAchR. We enhanced CAIP activity by subjecting rats to vagus nerve stimulation (VNS), and administered the α-7 acetylcholine receptor (α7nAchR) agonist and antagonist to determine whether VNS can reduce lung injury by regulating the pulmonary inflammatory response through CAIP. After being subjected to VNS, the secretion of TNF-α and IL-1β was decreased, while the level of IL-10 was increased in the rat model of ARDS. Moreover, VNS treatment reduced lung mRNA levels of M1 macrophage markers, while increased those of M2 macrophage markers. The expression of Caspase-1 decreased, while that of STAT3 increased in lung tissue after VNS treatment. The aforementioned effects of VNS were reversed by cutting the cervical vagus efferent branch and blocking α7nAchR. These findings suggest that VNS inhibits the ARDS inflammatory response by promoting CAIP activity. Next, we used lentivirus knockdown of STAT3 expression to explore the mechanism of VNS through CAIP on lung inflammation in ARDS model rats. VNS activates α7nAchR, increases STAT3 expression, reduces Caspase-1 expression, suppresses inflammation by inhibiting inflammatory pyroptosis and M1 to M2 macrophage transformation, which may constitute the main mechanism of VNS action in ARDS.
Collapse
|
57
|
Sokal DM, McSloy A, Donegà M, Kirk J, Colas RA, Dolezalova N, Gomez EA, Gupta I, Fjordbakk CT, Ouchouche S, Matteucci PB, Schlegel K, Bashirullah R, Werling D, Harman K, Rowles A, Yazicioglu RF, Dalli J, Chew DJ, Perkins JD. Splenic Nerve Neuromodulation Reduces Inflammation and Promotes Resolution in Chronically Implanted Pigs. Front Immunol 2021; 12:649786. [PMID: 33859641 PMCID: PMC8043071 DOI: 10.3389/fimmu.2021.649786] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/11/2021] [Indexed: 11/28/2022] Open
Abstract
Neuromodulation of the immune system has been proposed as a novel therapeutic strategy for the treatment of inflammatory conditions. We recently demonstrated that stimulation of near-organ autonomic nerves to the spleen can be harnessed to modulate the inflammatory response in an anesthetized pig model. The development of neuromodulation therapy for the clinic requires chronic efficacy and safety testing in a large animal model. This manuscript describes the effects of longitudinal conscious splenic nerve neuromodulation in chronically-implanted pigs. Firstly, clinically-relevant stimulation parameters were refined to efficiently activate the splenic nerve while reducing changes in cardiovascular parameters. Subsequently, pigs were implanted with a circumferential cuff electrode around the splenic neurovascular bundle connected to an implantable pulse generator, using a minimally-invasive laparoscopic procedure. Tolerability of stimulation was demonstrated in freely-behaving pigs using the refined stimulation parameters. Longitudinal stimulation significantly reduced circulating tumor necrosis factor alpha levels induced by systemic endotoxemia. This effect was accompanied by reduced peripheral monocytopenia as well as a lower systemic accumulation of CD16+CD14high pro-inflammatory monocytes. Further, lipid mediator profiling analysis demonstrated an increased concentration of specialized pro-resolving mediators in peripheral plasma of stimulated animals, with a concomitant reduction of pro-inflammatory eicosanoids including prostaglandins. Terminal electrophysiological and physiological measurements and histopathological assessment demonstrated integrity of the splenic nerves up to 70 days post implantation. These chronic translational experiments demonstrate that daily splenic nerve neuromodulation, via implanted electronics and clinically-relevant stimulation parameters, is well tolerated and is able to prime the immune system toward a less inflammatory, pro-resolving phenotype.
Collapse
Affiliation(s)
- David M. Sokal
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Alex McSloy
- Clinical Science & Services, The Royal Veterinary College, Hatfield, United Kingdom
| | - Matteo Donegà
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Joseph Kirk
- Clinical Science & Services, The Royal Veterinary College, Hatfield, United Kingdom
| | - Romain A. Colas
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Nikola Dolezalova
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Esteban A. Gomez
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Isha Gupta
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | | | - Sebastien Ouchouche
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Paul B. Matteucci
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Kristina Schlegel
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Rizwan Bashirullah
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Kim Harman
- Clinical Science & Services, The Royal Veterinary College, Hatfield, United Kingdom
| | - Alison Rowles
- Non-Clinical Safety, GlaxoSmithKline, Ware, United Kingdom
| | | | - Jesmond Dalli
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Daniel J. Chew
- Translation and Engineering, Galvani Bioelectronics, Stevenage, United Kingdom
| | - Justin D. Perkins
- Clinical Science & Services, The Royal Veterinary College, Hatfield, United Kingdom
| |
Collapse
|
58
|
Gaidhani N, Tucci FC, Kem WR, Beaton G, Uteshev VV. Therapeutic efficacy of α7 ligands after acute ischaemic stroke is linked to conductive states of α7 nicotinic ACh receptors. Br J Pharmacol 2021; 178:1684-1704. [PMID: 33496352 DOI: 10.1111/bph.15392] [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: 07/29/2020] [Revised: 12/22/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Targeting α7 nicotinic ACh receptors (nAChRs) in neuroinflammatory disorders including acute ischaemic stroke holds significant therapeutic promise. However, therapeutically relevant signalling mechanisms remain unidentified. Activation of neuronal α7 nAChRs triggers ionotropic signalling, but there is limited evidence for it in immunoglial tissues. The α7 ligands which are effective in reducing acute ischaemic stroke damage promote α7 ionotropic activity, suggesting a link between their therapeutic effects for treating acute ischaemic stroke and activation of α7 conductive states. EXPERIMENTAL APPROACH This hypothesis was tested using a transient middle cerebral artery occlusion (MCAO) model of acute ischaemic stroke, NS6740, a known selective non-ionotropic agonist of α7 nAChRs and 4OH-GTS-21, a partial α7 agonist. NS6740-like ligands exhibiting low efficacy/potency for ionotropic activity will be referred to as non-ionotropic agonists or "metagonists". KEY RESULTS 4OH-GTS-21, used as a positive control, significantly reduced neurological deficits and brain injury after MCAO as compared to vehicle and NS6740. By contrast, NS6740 was ineffective in identical assays and reversed the effects of 4OH-GTS-21 when these compounds were co-applied. Electrophysiological recordings from acute hippocampal slices obtained from NS6740-injected animals demonstrated its remarkable brain availability and protracted effects on α7 nAChRs as evidenced by sustained (>8 h) alterations in α7 ionotropic responsiveness. CONCLUSION AND IMPLICATIONS These results suggest that α7 ionotropic activity may be obligatory for therapeutic efficacy of α7 ligands after acute ischaemic stroke yet, highlight the potential for selective application of α7 ligands to disease states based on their mode of receptor activation.
Collapse
Affiliation(s)
- Nikhil Gaidhani
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Fabio C Tucci
- Epigen Biosciences, Inc., San Diego, California, USA
| | - William R Kem
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Graham Beaton
- Epigen Biosciences, Inc., San Diego, California, USA
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
59
|
Yang NN, Yang JW, Ye Y, Huang J, Wang L, Wang Y, Su XT, Lin Y, Yu FT, Ma SM, Qi LY, Lin LL, Wang LQ, Shi GX, Li HP, Liu CZ. Electroacupuncture ameliorates intestinal inflammation by activating α7nAChR-mediated JAK2/STAT3 signaling pathway in postoperative ileus. Am J Cancer Res 2021; 11:4078-4089. [PMID: 33754049 PMCID: PMC7977469 DOI: 10.7150/thno.52574] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/24/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory cytokines produced by muscularis macrophages largely contribute to the pathological signs of postoperative ileus (POI). Electroacupuncture (EA) can suppress inflammation, mainly or partly via activation of vagal efferent. The goal of this study was to investigate the mechanisms by which EA stimulation at an hindlimb region ameliorates inflammation in POI. Methods: Intestinal motility and inflammation were examined after 24 h after intestinal manipulation (IM)-induced POI in mice. Local immune response in the intestinal muscularis, expression of macrophages, α7 nicotinic acetylcholine receptor (α7nAChR), Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were determined by flow cytometry, Western Blot, qPCR and immunofluorescence. The effects of α7nAChR antagonists (methyllycaconitine and α-bungarotoxin) and JAK2/STAT3 inhibitors (AG490 and WP1066) were also administered in a subset of mice prior to EA. In the parasympathetic pathways, intestinal motility and inflammation were determined after cervical vagotomy and sub-diaphragmatic vagotomy. The expression of gamma absorptiometry aminobutyric acid (GABAA) receptor in dorsal motor nucleus of vagal (DMV) cholinergic neurons was assessed by immunofluorescence and the response to DMV microinjection of bicuculine (antagonist of GABAA receptor) or muscimol (agonist of GABAA receptor) were assessed. Results: EA suppressed intestinal inflammation and promoted gastrointestinal motility. Mechanistically, EA activated the α7nAChR-mediated JAK2/STAT3 signaling pathway in macrophages which reduced the production of inflammatory cytokines. Furthermore, we also demonstrated that hindlimb region stimulation drove vagal efferent output by inhibiting the expression of GABAA receptor in DMV to ameliorate inflammation. Conclusions: The present study revealed that EA of hindlimb regions inhibited the expression of GABAA receptor in DMV neurons, whose excited vagal nerve, in turn suppressed IM-induced inflammation via activation of α7nAChR-mediated JAK2/STAT3 signaling pathway.
Collapse
|
60
|
Murray K, Rude KM, Sladek J, Reardon C. Divergence of neuroimmune circuits activated by afferent and efferent vagal nerve stimulation in the regulation of inflammation. J Physiol 2021; 599:2075-2084. [PMID: 33491187 DOI: 10.1113/jp281189] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS It has previously been shown that afferent and efferent vagal nerve stimulation potently inhibits lipopolysaccharide (LPS)-induced inflammation Our data show inhibition of inflammation by efferent but not afferent vagal nerve stimulation requires T-cell derived acetylcholine We show that afferent and efferent neuroimmune circuits require β2 -adrenergic receptor signalling ABSTRACT: Chronic inflammation due to inappropriate immune cell activation can have significant effects on a variety of organ systems, reducing lifespan and quality of life. As such, highly targeted control of immune cell activation is a major therapeutic goal. Vagus nerve stimulation (VNS) has emerged as a therapeutic modality that exploits neuroimmune communication to reduce immune cell activation and consequently inflammation. Although vagal efferent fibres were originally identified as the primary driver of anti-inflammatory actions, the vagus nerve in most species of animals predominantly comprises afferent fibres. Stimulation of vagal afferent fibres can also reduce inflammation; it is, however, uncertain how these two neuroimmune circuits diverge. Here we show that afferent VNS induces a mechanism distinct from efferent VNS, ameliorating lipopolysaccharide (LPS)-induced inflammation independently of T-cell derived acetylcholine (ACh) which is required by efferent VNS. Using a β2 -adrenergic receptor antagonist (β2 -AR), we find that immune regulation induced by intact, afferent, or efferent VNS occurs in a β2- AR-dependent manner. Together, our findings indicate that intact VNS activates at least two distinct neuroimmune circuits each with unique mechanisms of action. Selective targeting of either the vagal efferent or afferent fibres may provide more personalized, robust and effective control over inappropriate immune responses.
Collapse
Affiliation(s)
- Kaitlin Murray
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - Kavi M Rude
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - Jessica Sladek
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, CA, USA
| |
Collapse
|
61
|
Zhang L, Wu Z, Tong Z, Yao Q, Wang Z, Li W. Vagus Nerve Stimulation Decreases Pancreatitis Severity in Mice. Front Immunol 2021; 11:595957. [PMID: 33519809 PMCID: PMC7840568 DOI: 10.3389/fimmu.2020.595957] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Background Vagus nerve stimulation (VNS) is effective in reducing inflammation in various diseases, such as rheumatoid arthritis, colitis and acute kidney injury. The anti-inflammatory effect of vagus nerve in these diseases necessitates the interactions of neural activation and α7 nicotinic acetylcholine receptors (α7nAChRs) on splenic macrophages. In this study, we aimed to investigate the effect of VNS on severity in experimental acute pancreatitis (AP). Methods Two independent AP models were used, which induced in ICR mice with caerulein or pancreatic duct ligation (PDL). Thirty minutes after modeling, the left cervical carotid sheath containing the vagus nerve was electrically stimulated for 2 min. Plasma lipase and amylase activities, TNF-α levels and pancreas histologic damage were evaluated. In caerulein mice, the percentages of α7nAChR+ macrophage in pancreas and spleen were assessed by flow cytometry. Furthermore, splenectomy and adoptive transfer of VNS-conditioned α7nAChR splenocytes were performed in caerulein mice to evaluate the role of spleen in the protective effect of VNS. Results VNS reduced plasma lipase and amylase activities, blunted the concentrations of TNF-α and protected against pancreas histologic damage in two AP models. Survival rates were improved in the PDL model after VNS. In caerulein AP mice, VNS increased the percentages of α7nAChR+ macrophages in pancreas and spleen. Adoptive transfer of VNS-treated α7nAChR splenocytes provided protection against pancreatitis in recipient mice. However, splenectomy did not abolish the protective effect of VNS. Conclusions VNS reduces disease severity and attenuates inflammation in AP mice. This effect is independent of spleen and is probably related to α7nAChR on macrophage.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Pathology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyang Wu
- Department of Critical Care Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Zhihui Tong
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qi Yao
- Department of Pathology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziyu Wang
- Department of Pathology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiqin Li
- Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
62
|
Fudim M, Qadri YJ, Ghadimi K, MacLeod DB, Molinger J, Piccini JP, Whittle J, Wischmeyer PE, Patel MR, Ulloa L. Implications for Neuromodulation Therapy to Control Inflammation and Related Organ Dysfunction in COVID-19. J Cardiovasc Transl Res 2020; 13:894-899. [PMID: 32458400 PMCID: PMC7250255 DOI: 10.1007/s12265-020-10031-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022]
Abstract
COVID-19 is a syndrome that includes more than just isolated respiratory disease, as severe acute respiratory syndrome-coronavirus 2 (SARS-CoV2) also interacts with the cardiovascular, nervous, renal, and immune system at multiple levels, increasing morbidity in patients with underlying cardiometabolic conditions and inducing myocardial injury or dysfunction. Emerging evidence suggests that patients with the highest rate of morbidity and mortality following SARS-CoV2 infection have also developed a hyperinflammatory syndrome (also termed cytokine release syndrome). We lay out the potential contribution of a dysfunction in autonomic tone to the cytokine release syndrome and related multiorgan damage in COVID-19. We hypothesize that a cholinergic anti-inflammatory pathway could be targeted as a therapeutic avenue. Graphical Abstract .
Collapse
Affiliation(s)
- Marat Fudim
- Department of Medicine, Division of Cardiology, Duke University, 2301 Erwin Road, Durham, NC, 27710, USA.
| | - Yawar J Qadri
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - Kamrouz Ghadimi
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - David B MacLeod
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - Jeroen Molinger
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - Jonathan P Piccini
- Department of Medicine, Division of Cardiology, Duke University, 2301 Erwin Road, Durham, NC, 27710, USA
- Duke Center for Atrial Fibrillation, Duke University Medical Center, Duke University, Durham, NC, USA
| | - John Whittle
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - Paul E Wischmeyer
- Department of Anesthesiology & Critical Care, Duke University, Durham, NC, USA
| | - Manesh R Patel
- Department of Medicine, Division of Cardiology, Duke University, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, USA
| |
Collapse
|
63
|
Gaidhani N, Kem WR, Uteshev VV. Spleen is not required for therapeutic effects of 4OH-GTS-21, a selective α7 nAChR agonist, in the sub-acute phase of ischemic stroke in rats. Brain Res 2020; 1751:147196. [PMID: 33159972 DOI: 10.1016/j.brainres.2020.147196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 11/15/2022]
Abstract
Acute ischemic stroke (AIS) causes both central and peripheral inflammation, while activation of α7 nicotinic acetylcholine receptors (nAChRs) provides both central and peripheral anti-inflammatory and anti-apoptotic effects. Here, we provide evidence that 4OH-GTS-21, a selective α7 agonist, produces its therapeutic effects via primarily central sites of action because 4OH-GTS-21 was found equally effective in splenectomized and non-spenectomized rats in the sub-acute phase of ischemic stroke (≤1 week). However, the spleen may boost the therapeutic efficacy of 4OH-GTS-21 in certain behavioral tasks as our data also indicated. In our tests, AIS was modeled by transient middle cerebral artery occlusion (tMCAO). Splenectomy was done 2 weeks before tMCAO. We determined that: 1) Daily 4OH-GTS-21 treatments for 7 days after tMCAO significantly reduced neurological deficits and brain injury in both splenectomized and non-spelenectomized rats demonstrating that the spleen is not required for therapeutic benefits of 4OH-GTS-21; 2) The effects of 4OH-GTS-21 in the adhesive sticker removal test were significantly weaker in splenectomized animals suggesting that the spleen boosts the efficacy of 4OH-GTS-21 in the first week after tMCAO; and 3) Ischemic brain injury was not significantly affected by splenectomy in both vehicle-treated and 4OH-GTS-21-treated animals. These data support the hypothesis that the therapeutic efficacy of sub-chronic (≤1 week) 4OH-GTS-21 primarily originates from central sites of action. These results validate brain availability as a critical factor for developing novel α7 ligands for AIS.
Collapse
Affiliation(s)
- Nikhil Gaidhani
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - William R Kem
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, 1200 Newell Drive, Gainesville, FL 32610, United States
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States.
| |
Collapse
|
64
|
Chen W, Shu Q, Fan J. Neural Regulation of Interactions Between Group 2 Innate Lymphoid Cells and Pulmonary Immune Cells. Front Immunol 2020; 11:576929. [PMID: 33193374 PMCID: PMC7658006 DOI: 10.3389/fimmu.2020.576929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence supports the involvement of nervous system in the regulation of immune responses. Group 2 innate lymphoid cells (ILC2), which function as a crucial bridge between innate and adaptive immunity, are present in large numbers in barrier tissues. Neuropeptides and neurotransmitters have been found to participate in the regulation of ILC2, adding a new dimension to neuroimmunity. However, a comprehensive and detailed overview of the mechanisms of neural regulation of ILC2, associated with previous findings and prospects for future research, is still lacking. In this review, we compile existing information that supports neurons as yet poorly understood regulators of ILC2 in the field of lung innate and adaptive immunity, focusing on neural regulation of the interaction between ILC2 and pulmonary immune cells.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiang Shu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
65
|
Morphometric analysis of the splenic artery using contrast-enhanced computed tomography (CT). Surg Radiol Anat 2020; 43:377-384. [PMID: 33104863 PMCID: PMC7897610 DOI: 10.1007/s00276-020-02598-1] [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: 08/13/2020] [Accepted: 10/10/2020] [Indexed: 10/30/2022]
Abstract
PURPOSE To evaluate the morphology and course of the splenic artery, which might impact the surgical implantation of systems that stimulate the nerves surrounding the splenic artery. Experimental studies indicate that these nerves play an important part in immune modulation, and might be a potential target in the treatment of autoimmune diseases. METHODS This retrospective cohort study made use of contrast-enhanced CT images from 40 male and 40 female patients (age 30-69) that underwent a CT examination of the aorta, kidneys or pancreas. Anatomic features were described including total splenic artery length, calibers, tortuosity, the presence of arterial loops and the branching pattern of the splenic artery. RESULTS No age-gender-related differences could be found related to tortuosity or branching pattern. The length of splenic artery in contact with pancreatic tissue decreased with increasing age, but was not different between genders. Artery diameters were wider in male compared to female subjects. Loops of variable directions, that represent a part of the artery that curls out of the pancreatic tissue, were identified in each age-gender category and were present in nearly all subjects (86%). CONCLUSION This study suggests that although some anatomic features of the splenic artery are subject to factors as age and gender, the tortuosity of the splenic artery is not age dependent. Most subjects had one or multiple loops, which can serve as a target for neuromodulatory devices. Future studies should investigate whether splenic nerve stimulation is safe and feasible.
Collapse
|
66
|
Chung CH, Bretherton B, Zainalabidin S, Deuchars SA, Deuchars J, Mahadi MK. Mediation of Cardiac Macrophage Activity via Auricular Vagal Nerve Stimulation Ameliorates Cardiac Ischemia/Reperfusion Injury. Front Neurosci 2020; 14:906. [PMID: 33013299 PMCID: PMC7506070 DOI: 10.3389/fnins.2020.00906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022] Open
Abstract
Background Myocardial infarction (MI) reperfusion therapy causes paradoxical cardiac complications. Following restoration of blood flow to infarcted regions, a multitude of inflammatory cells are recruited to the site of injury for tissue repair. Continual progression of cardiac inflammatory responses does, however, lead to adverse cardiac remodeling, inevitably causing heart failure. Main Body Increasing evidence of the cardioprotective effects of both invasive and non-invasive vagal nerve stimulation (VNS) suggests that these may be feasible methods to treat myocardial ischemia/reperfusion injury via anti-inflammatory regulation. The mechanisms through which auricular VNS controls inflammation are yet to be explored. In this review, we discuss the potential of autonomic nervous system modulation, particularly via the parasympathetic branch, in ameliorating MI. Novel insights are provided about the activation of the cholinergic anti-inflammatory pathway on cardiac macrophages. Acetylcholine binding to the α7 nicotinic acetylcholine receptor (α7nAChR) expressed on macrophages polarizes the pro-inflammatory into anti-inflammatory subtypes. Activation of the α7nAChR stimulates the signal transducer and activator of transcription 3 (STAT3) signaling pathway. This inhibits the secretion of pro-inflammatory cytokines, limiting ischemic injury in the myocardium and initiating efficient reparative mechanisms. We highlight recent developments in the controversial auricular vagal neuro-circuitry and how they may relate to activation of the cholinergic anti-inflammatory pathway. Conclusion Emerging published data suggest that auricular VNS is an inexpensive healthcare modality, mediating the dynamic balance between pro- and anti-inflammatory responses in cardiac macrophages and ameliorating cardiac ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Chee Hooi Chung
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Beatrice Bretherton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Center for Toxicology and Health Risk Study (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Susan A Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jim Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Mohd Kaisan Mahadi
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
67
|
Stakenborg N, Gomez‐Pinilla PJ, Verlinden TJM, Wolthuis AM, D’Hoore A, Farré R, Herijgers P, Matteoli G, Boeckxstaens GE. Comparison between the cervical and abdominal vagus nerves in mice, pigs, and humans. Neurogastroenterol Motil 2020; 32:e13889. [PMID: 32476229 PMCID: PMC7507132 DOI: 10.1111/nmo.13889] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/25/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Vagus nerve (VN) stimulation is currently evaluated as a novel approach to treat immune-mediated disorders. The optimal stimulation parameters, however, largely depend on the VN composition potentially impacting on its clinical translation. Hence, we evaluated whether morphological differences exist between the cervical and abdominal VNs across different species. MATERIALS AND METHODS The cervical and abdominal VNs of mouse, pig, and humans were stained for major basic protein and neurofilament F to identify the percentage and size of myelinated and non-myelinated fibers. RESULTS The percentage of myelinated fibers was comparable between species, but was higher in the cervical VN compared with the abdominal VN. The cervical VN contained 54 ± 4%, 47 ± 7%, and 54 ± 7% myelinated fibers in mouse, pig, and humans, respectively. The myelinated fibers consisted of small-diameter (mouse: 71%, pig: 80%, and humans: 63%), medium-diameter (mouse: 21%, pig: 18%, and humans: 33%), and large-diameter fibers (mouse: 7%, pig: 2%, and humans: 4%). The abdominal VN predominantly contained unmyelinated fibers (mouse: 93%, pig: 90%, and humans: 94%). The myelinated fibers mainly consisted of small-diameter fibers (mouse: 99%, pig: 85%, and humans: 74%) and fewer medium-diameter (mouse: 1%, pig: 13%, and humans: 23%) and large-diameter fibers (mouse: 0%, pig: 2%, and humans: 3%). CONCLUSION The VN composition was largely similar with respect to myelinated and unmyelinated fibers in the species studied. Human and porcine VNs had a comparable diameter and similar amounts of fibrous tissue and contained multiple fascicles, implying that the porcine VN may be suitable to optimize stimulation parameters for clinical trials.
Collapse
Affiliation(s)
- Nathalie Stakenborg
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Translational Research Center for Gastrointestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| | - Pedro J. Gomez‐Pinilla
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Translational Research Center for Gastrointestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| | - Thomas J. M. Verlinden
- Department of Anatomy & EmbryologyFaculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Albert M. Wolthuis
- Department of Abdominal SurgeryUniversity Hospital of LeuvenLeuvenBelgium
| | - Andre D’Hoore
- Department of Abdominal SurgeryUniversity Hospital of LeuvenLeuvenBelgium
| | - Ricard Farré
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Translational Research Center for Gastrointestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| | - Paul Herijgers
- Department of Cardiovascular ScienceKU LeuvenLeuvenBelgium
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Translational Research Center for Gastrointestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| | - Guy E. Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Translational Research Center for Gastrointestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| |
Collapse
|
68
|
Ramirez V, Swain S, Murray K, Reardon C. Neural Immune Communication in the Control of Host-Bacterial Pathogen Interactions in the Gastrointestinal Tract. Infect Immun 2020; 88:e00928-19. [PMID: 32341116 PMCID: PMC7440759 DOI: 10.1128/iai.00928-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The orchestration of host immune responses to enteric bacterial pathogens is a complex process involving the integration of numerous signals, including from the nervous system. Despite the recent progress in understanding the contribution of neuroimmune interactions in the regulation of inflammation, the mechanisms and effects of this communication during enteric bacterial infection are only beginning to be characterized. As part of this neuroimmune communication, neurons specialized to detect painful or otherwise noxious stimuli can respond to bacterial pathogens. Highlighting the complexity of these systems, the immunological consequences of sensory neuron activation can be either host adaptive or maladaptive, depending on the pathogen and organ system. These are but one of many types of neuroimmune circuits, with the vagus nerve and sympathetic innervation of numerous organs now known to modulate immune cell function and therefore dictate immunological outcomes during health and disease. Here, we review the evidence for neuroimmune communication in response to bacterial pathogens, and then discuss the consequences to host morbidity and mortality during infection of the gastrointestinal tract.
Collapse
Affiliation(s)
- Valerie Ramirez
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Samantha Swain
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Kaitlin Murray
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Colin Reardon
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
69
|
Yamawaki H, Futagami S, Sakasegawa N, Murakami M, Agawa S, Ikeda G, Noda H, Kirita K, Gudis K, Higuchi K, Kodaka Y, Ueki N, Iwakiri K. Acotiamide attenuates central urocortin 2-induced intestinal inflammatory responses, and urocortin 2 treatment reduces TNF-α productions in LPS-stimulated macrophage cell lines. Neurogastroenterol Motil 2020; 32:e13813. [PMID: 32030855 DOI: 10.1111/nmo.13813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/21/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND To determine whether central and in vitro administration of urocortin 2 (Ucn 2) affected intestinal inflammatory responses in LPS-stimulated rat models and macrophage cell lines and acotiamide modified mucosal inflammation in this model. METHODS Rats were divided into four groups. LPS-stimulated group (n = 4); LPS- and urocortin 2-treated group (n = 4); LPS- and acotiamide-treated group (n = 4); and LPS-, urocortin 2-, and acotiamide-treated group (n = 4). CD68-, CCR2-, and corticotropin-releasing hormone receptor type 2 (CRHR2)-positive cells were assessed by immunostaining. Myeloperoxidase (MPO) activity was measured. TNF-α, IL-6, and IL-4 levels were measured by ELISA method. Gastric emptying and small intestinal transit time were determined using Evans blue. KEY RESULTS Central administration of Ucn 2 significantly aggravated infiltrations of CD68- and CCR2-positive cells in the intestinal mucosa of LPS-stimulated rat models compared to those in LPS treatment alone. Interestingly, acotiamide treatment significantly reduced the migrations of both CD68- and CCR2-positive cells in the jejunum of central Ucn 2-treated LPS-stimulated rat models. Acotiamide significantly reduced the expression levels of IkB-α phosphorylation in LPS- and MCP-1-stimulated NR8383 cells. Central administration of Ucn 2 significantly delayed gastric emptying. In contrast, Ucn 2 stimulation significantly reduced TNF-α and IL-6 productions in LPS-stimulated NR8383 cells and astressin B reversed the inhibition of TNF-α production in stimulated NR8383 cells. Acotiamide (30 μmol/L) significantly reduced TNF-α and IL-6 productions in LPS- and MCP-1-stimulated NR8383 cells. CONCLUSIONS AND INFERENCES Central and in vitro treatments of Ucn 2 affected intestinal inflammatory responses, respectively, and acotiamide improved them.
Collapse
Affiliation(s)
- Hiroshi Yamawaki
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Seiji Futagami
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | | | - Makoto Murakami
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Shuhei Agawa
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Go Ikeda
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Hiroto Noda
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Kumiko Kirita
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Katya Gudis
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | | | - Yasuhiro Kodaka
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Nobue Ueki
- Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
70
|
Passaglia P, Faim FDL, Batalhão ME, Bendhack LM, Antunes-Rodrigues J, Ulloa L, Kanashiro A, Carnio EC. Central angiotensin-(1-7) attenuates systemic inflammation via activation of sympathetic signaling in endotoxemic rats. Brain Behav Immun 2020; 88:606-618. [PMID: 32335195 PMCID: PMC7643008 DOI: 10.1016/j.bbi.2020.04.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 01/21/2023] Open
Abstract
Angiotensin-(1-7) [Ang-(1-7)] is an angiotensin-derived neuropeptide with potential anti-hypertensive and anti-inflammatory properties. However, a possible action of Ang-(1-7) in neuroimmune interactions to regulate inflammatory response has not been explored. Thus, the aim of this study was to determine whether the intracerebroventricular (i.c.v.) administration of Ang-(1-7) can modulate systemic inflammation via sympathetic efferent circuits. Wistar male rats received systemic administration of lipopolysaccharide (LPS) (1.5 mg/Kg). Ang-(1-7) (0.3 nmol in 2 µL) promoted the release of splenic norepinephrine and attenuated tumor necrosis factor (TNF) and nitric oxide (NO), but increased interleukin-10 (IL-10), levels in the serum, spleen, and liver in endotoxemic rats. Furthermore, 6-hydroxydopamine-induced chemical sympathectomy (100 mg/Kg, intravenous) or i.c.v. administration of Mas receptor antagonist A779 (3 nmol in 2 µL) abolished the anti-inflammatory effects of central Ang-(1-7) injection. Moreover, this treatment did not alter the plasmatic LPS-induced corticosterone and vasopressin. The administration of Ang-(1-7) reverted the low resistance in response to catecholamines of rings of thoracic aorta isolated from endotoxemic rats, treated or not, with this peptide by a mechanism dependent on the regulation of NO released from perivascular adipose tissue. Together, our results indicate that Ang-(1-7) regulates systemic inflammation and vascular hyporesponsiveness in endotoxemia via activation of a central Mas receptors/sympathetic circuits/norepinephrine axis and provide novel mechanistic insights into the anti-inflammatory Ang-(1-7) properties.
Collapse
Affiliation(s)
- Patrícia Passaglia
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Felipe de Lima Faim
- Department of Physiology, Ribeirão Preto Medical School – University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marcelo Eduardo Batalhão
- Department of General and Specialized Nursing Ribeirão Preto, College of Nursing – University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lusiane Maria Bendhack
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José Antunes-Rodrigues
- Department of Physiology, Ribeirão Preto Medical School – University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alexandre Kanashiro
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Evelin Capellari Carnio
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil; Department of General and Specialized Nursing Ribeirão Preto, College of Nursing - University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
71
|
Cleypool CGJ, Lotgerink Bruinenberg D, Roeling T, Irwin E, Bleys RLAW. Splenic artery loops: Potential splenic plexus stimulation sites for neuroimmunomodulatory-based anti-inflammatory therapy? Clin Anat 2020; 34:371-380. [PMID: 32583891 PMCID: PMC7984037 DOI: 10.1002/ca.23643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 06/15/2020] [Accepted: 06/20/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The splenic plexus might represent a novel neuroimmunomodulatory therapeutic target as electrical stimulation of this tissue has been shown to have beneficial anti-inflammatory effects. Tortuous splenic artery segments (splenic artery loops), including their surrounding nerve plexus, have been evaluated as potential stimulation sites in humans. At present, however, our understanding of these loops and their surrounding nerve plexus is incomplete. This study aims to characterize the dimensions of these loops and their surrounding nerve tissue. MATERIALS AND METHODS Six formaldehyde fixed human cadavers were dissected and qualitative and quantitative macro- and microscopic data on splenic artery loops and their surrounding nerve plexus were collected. RESULTS One or multiple loops were observed in 83% of the studied specimens. These loops, including their surrounding nerve plexus could be easily dissected free circumferentially thereby providing sufficient space for further surgical intervention. The splenic plexus surrounding the loops contained a significant amount of nerves that contained predominantly sympathetic fibers. CONCLUSION The results of this study support that splenic artery loops could represent suitable electrical splenic plexus stimulation sites in humans. Dimensions with respect to loop height and width, provide sufficient space for introduction of surgical instruments and electrode implantation, and, the dissected neurovascular bundles contain a substantial amount of sympathetic nerve tissue. This knowledge may contribute to further development of surgical techniques and neuroelectrode interface design.
Collapse
Affiliation(s)
- Cindy G J Cleypool
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Dyonne Lotgerink Bruinenberg
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Tom Roeling
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Eric Irwin
- Galvani Bioelectronics, Stevenage, UK.,Department of Surgery, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Ronald L A W Bleys
- Department of Anatomy, Division of Surgical Specialties, University Medical Center Utrecht, Utrecht University, The Netherlands
| |
Collapse
|
72
|
Li Y, Xu G, Hu S, Wu H, Dai Y, Zhang W, Tang F, Luo H, Shi X. Electroacupuncture alleviates intestinal inflammation and barrier dysfunction by activating dopamine in a rat model of intestinal ischaemia. Acupunct Med 2020; 39:208-216. [PMID: 32517478 DOI: 10.1177/0964528420922232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND To investigate whether the mechanism underlying the anti-inflammatory effects of electroacupuncture (EA) at ST36 involves dopamine (DA) and its receptor and whether it is mediated by the vagus nerve in a rat model of intestinal ischaemia-reperfusion (I/R) injury. METHODS Rats were subjected to gut ischaemia for 30 min and then received EA for 30 min with or without abdominal vagotomy or intraperitoneal administration of butaclamol (D1 receptor antagonist) or spiperone (D2 receptor antagonist). Plasma levels of DA and tumour necrosis factor (TNF)-α were assessed 1 or 4 h after reperfusion. Myeloperoxidase (MPO) activity and malondialdehyde (MDA) content in intestinal tissues were assessed using enzyme-linked immunosorbent assay (ELISA) kits. Intestinal tissue injury was assessed by observation of the pathological lesions and permeability to 4 kDa fluorescein isothiocyanate (FITC)-dextran. RESULTS EA significantly increased levels of DA and lowered levels of TNF-α. EA also inhibited intestinal levels of MPO and MDA and intestinal tissue injury and decreased intestinal permeability to FITC-dextran. Abdominal vagotomy and intraperitoneal administration of butaclamol (but not spiperone) inhibited the effects of EA. CONCLUSION These findings suggest that EA at ST36 could attenuate intestinal I/R-induced inflammatory injury and that the underlying mechanism may involve EA-induced increases in levels of DA, mediated by the vagus nerve and D1 receptors.
Collapse
Affiliation(s)
- Yumeng Li
- Department of Encephalopathy, Anyang Hospital of Traditional Chinese Medicine, Anyang, China
| | - Guochen Xu
- Out-Patient Department, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Sen Hu
- Laboratory of Shock and Multiple Organ Dysfunction, Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China.,Research Center of Trauma Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing, China
| | - Hong Wu
- Department of Encephalopathy, Anyang Hospital of Traditional Chinese Medicine, Anyang, China
| | - Yuelong Dai
- Chinese People's Armed Police Force Academy, Langfang, China
| | - Wenhua Zhang
- Laboratory of Shock and Multiple Organ Dysfunction, Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Fubo Tang
- Chengdu Hospital of Sichuan Provincial Corps, Chinese People's Armed Police Force, Chengdu, China
| | - Hongmin Luo
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xian Shi
- Department of Acupuncture and Moxibustion, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
73
|
Arriaga-Pizano L, Gómez-Jiménez DC, Flores-Mejía LA, Pérez-Cervera Y, Solórzano-Mata CJ, López-Macías C, Isibasi A, Torres-Rosas R. Low back pain in athletes can be controlled with acupuncture by a catecholaminergic pathway: clinical trial. Acupunct Med 2020; 38:388-395. [PMID: 32429680 DOI: 10.1177/0964528420912251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Activation of the sympathetic nervous system attenuates inflammation via catecholamines. Recent evidence has shown that electroacupuncture (EA) activates neuronal networks involved in the release of dopamine and norepinephrine that control systemic inflammation. In muscle, catecholamines are related to cyclic adenosine monophosphate (cAMP). This signaling molecule has been implicated in recovery from sustained contractile activity, which may induce muscular pain, such as that which occurs during low back pain (LBP). OBJECTIVE Our aim was to evaluate the effects of EA used for the control of LBP on the activation of the sympathetic nervous system in a randomized controlled clinical trial in athletes. METHODS Two groups of athletes with acute or chronic low back pain were studied. EA, sham EA and pharmacological treatment (diclofenac sodium) were evaluated. The outcome measures included a pain score represented by a visual analogue scale (VAS) and serum levels of catecholamines quantified by enzyme-linked immunosorbent assay. In addition, blood was collected into chilled heparin tubes, placed in 96-well cell culture plates and incubated with an equal volume of Roswell Park Memorial Institute (RPMI) medium, with lipopolysaccharide (LPS) alone or with catecholamines. Tumor necrosis factor (TNF)-α levels in the supernatants were analyzed. RESULTS The results indicated that the initial pain ratings did not differ between the groups analyzed. EA induced epinephrine secretion but not norepinephrine or dopamine secretion. Although EA and pharmacological treatment did not differ in terms of pain relief, in vitro epinephrine and norepinephrine reduced TNF-α production in response to LPS stimuli. CONCLUSION EA activates the sympathetic nervous system and induces the release of epinephrine, which could ameliorate inflammation and protect muscular tissue in addition to relieving pain.
Collapse
Affiliation(s)
- Lourdes Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | | | - Luis Angel Flores-Mejía
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Yobana Pérez-Cervera
- "Laboratorio de Inmunología, Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México.,Centro de Investigación UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México
| | - Carlos Josué Solórzano-Mata
- "Laboratorio de Inmunología, Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México.,Centro de Investigación UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Armando Isibasi
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Rafael Torres-Rosas
- "Laboratorio de Inmunología, Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México
| |
Collapse
|
74
|
Rosso P, Iannitelli A, Pacitti F, Quartini A, Fico E, Fiore M, Greco A, Ralli M, Tirassa P. Vagus nerve stimulation and Neurotrophins: a biological psychiatric perspective. Neurosci Biobehav Rev 2020; 113:338-353. [PMID: 32278791 DOI: 10.1016/j.neubiorev.2020.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Since 2004, vagus nerve stimulation (VNS) has been used in treatment-resistant or treatment-intolerant depressive episodes. Today, VNS is suggested as possible therapy for a larger spectrum of psychiatric disorders, including schizophrenia, obsessive compulsive disorders, and panic disorders. Despite a large body of literature supports the application of VNS in patients' treatment, the exact mechanism of action of VNS remains not fully understood. In the present study, the major knowledges on the brain areas and neuronal pathways regulating neuroimmune and autonomic response subserving VNS effects are reviewed. Furthermore, the involvement of the neurotrophins (NTs) Nerve Growth Factor (NGF) and Brain Derived Neurotrophic Factor (BDNF) in vagus nerve (VN) physiology and stimulation is revised. The data on brain NGF/BDNF synthesis and in turn on the activity-dependent plasticity, connectivity rearrangement and neurogenesis, are presented and discussed as potential biomarkers for optimizing stimulatory parameters for VNS. A vagus nerve-neurotrophin interaction model in the brain is finally proposed as a working hypothesis for future studies addressed to understand pathophysiology of psychiatric disturbance.
Collapse
Affiliation(s)
- Pamela Rosso
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Angela Iannitelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca Pacitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy; Psychiatry Unit San Salvatore Hospital, L'Aquila, Italy
| | - Adele Quartini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elena Fico
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Marco Fiore
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Paola Tirassa
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy.
| |
Collapse
|
75
|
Chu C, Artis D, Chiu IM. Neuro-immune Interactions in the Tissues. Immunity 2020; 52:464-474. [PMID: 32187517 PMCID: PMC10710744 DOI: 10.1016/j.immuni.2020.02.017] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022]
Abstract
The ability of the nervous system to sense environmental stimuli and to relay these signals to immune cells via neurotransmitters and neuropeptides is indispensable for effective immunity and tissue homeostasis. Depending on the tissue microenvironment and distinct drivers of a certain immune response, the same neuronal populations and neuro-mediators can exert opposing effects, promoting or inhibiting tissue immunity. Here, we review the current understanding of the mechanisms that underlie the complex interactions between the immune and the nervous systems in different tissues and contexts. We outline current gaps in knowledge and argue for the importance of considering infectious and inflammatory disease within a conceptual framework that integrates neuro-immune circuits both local and systemic, so as to better understand effective immunity to develop improved approaches to treat inflammation and disease.
Collapse
Affiliation(s)
- Coco Chu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
76
|
Maturo MG, Soligo M, Gibson G, Manni L, Nardini C. The greater inflammatory pathway-high clinical potential by innovative predictive, preventive, and personalized medical approach. EPMA J 2020; 11:1-16. [PMID: 32140182 PMCID: PMC7028895 DOI: 10.1007/s13167-019-00195-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND LIMITATIONS Impaired wound healing (WH) and chronic inflammation are hallmarks of non-communicable diseases (NCDs). However, despite WH being a recognized player in NCDs, mainstream therapies focus on (un)targeted damping of the inflammatory response, leaving WH largely unaddressed, owing to three main factors. The first is the complexity of the pathway that links inflammation and wound healing; the second is the dual nature, local and systemic, of WH; and the third is the limited acknowledgement of genetic and contingent causes that disrupt physiologic progression of WH. PROPOSED APPROACH Here, in the frame of Predictive, Preventive, and Personalized Medicine (PPPM), we integrate and revisit current literature to offer a novel systemic view on the cues that can impact on the fate (acute or chronic inflammation) of WH, beyond the compartmentalization of medical disciplines and with the support of advanced computational biology. CONCLUSIONS This shall open to a broader understanding of the causes for WH going awry, offering new operational criteria for patients' stratification (prediction and personalization). While this may also offer improved options for targeted prevention, we will envisage new therapeutic strategies to reboot and/or boost WH, to enable its progression across its physiological phases, the first of which is a transient acute inflammatory response versus the chronic low-grade inflammation characteristic of NCDs.
Collapse
Affiliation(s)
- Maria Giovanna Maturo
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Greg Gibson
- Center for Integrative Genomics, School of Biological Sciences, Georgia Tech, Atlanta, GA USA
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Christine Nardini
- IAC Institute for Applied Computing, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
- Bio Unit, Scientific and Medical Direction, SOL Group, Monza, Italy
| |
Collapse
|
77
|
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: 8.0] [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
|
78
|
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: 3.4] [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
|
79
|
Levine YA, Faltys M, Chernoff D. Harnessing the Inflammatory Reflex for the Treatment of Inflammation-Mediated Diseases. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a034330. [PMID: 30833463 DOI: 10.1101/cshperspect.a034330] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Treating diseases nonpharmacologically, using targeted neurostimulation instead of systemic drugs, is a hallmark of the burgeoning field of bioelectronic medicine. In this review, we provide a brief overview of the discovery and function of the prototypical neuroimmune reflex, the "inflammatory reflex." We discuss various biomarkers developed and used to translate early physiological discoveries into dosing parameters used in experimental settings, from the treatment of animal models of disease through a proof-of-concept clinical study in rheumatoid arthritis (RA). Finally, we relate how unique aspects of this form of therapy enabled the design of a next-generation implanted pulse generator using integrated electrodes, currently under evaluation in a U.S.-based clinical study for patients with drug refractory RA.
Collapse
|
80
|
Uhlich R, Pierce V, Kerby J, Bosarge P, Hu P. Splenectomy does not affect the development of pneumonia following severe traumatic brain injury. Brain Behav Immun Health 2020; 1:100007. [PMID: 38377417 PMCID: PMC8474628 DOI: 10.1016/j.bbih.2019.100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 01/01/2023] Open
Abstract
The cholinergic anti-inflammatory pathway offers a proposed mechanism to describe the increased risk of pneumonia following severe traumatic brain injury (sTBI). Vagal activity transmitted to the spleen results in decreased inflammatory cytokine production and immunosuppression. However, no clinical evidence exists. We sought to compare pneumonia rates among patients with TBI and splenectomy using a retrospective analysis of all trauma patients with splenic injury requiring splenectomy or TBI admitted to an ACS verified level one trauma center from 2011 to 2016. Admission Glasgow Coma Score (GCS) ≤ 8 was used to identify sTBI. Pneumonia was defined by respiratory culture obtained by bronchoalveolar lavage. Analysis included χ2 and one-way analysis of variance followed by multivariate logistic regression to determine the association of sTBI and splenectomy of development of pneumonia. Four hundred and twenty-seven patients were included for primary analysis, 247 with sTBI, 180 with splenectomy, and 14 with both sTBI and splenectomy. Rates of pneumonia were increased, although not significant among patients with sTBI and splenectomy and both sTBI alone (71.4 vs. 49.4%, p = 0.11). On multivariate regression, the risk of pneumonia was increased with both splenectomy and sTBI (OR 3.18; 95% CI, 0.75-13.45) and sTBI alone, although significant in the latter only (OR 3.56; 95% CI, 2.12-5.97). Based on these results, splenectomy does not appear to influence the development of pulmonary immunosuppression and pneumonia following sTBI.
Collapse
Affiliation(s)
- Rindi Uhlich
- Department of Surgery, University of Alabama at Birmingham, 701 19th Street South, LHRB 112, Birmingham, AL, 35294, USA
| | - Virginia Pierce
- Division of Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, 701 19th Street South, LHRB 112, Birmingham, AL, 35294, USA
| | - Jeffrey Kerby
- Division of Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, 1922 7th Avenue South, KB 120, Birmingham, AL, 35294, USA
| | - Patrick Bosarge
- Division of Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, 701 19th Street South, LHRB 112, Birmingham, AL, 35294, USA
| | - Parker Hu
- Division of Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, 701 19th Street South, LHRB 112, Birmingham, AL, 35294, USA
| |
Collapse
|
81
|
Cleypool CGJ, Schurink B, van der Horst DEM, Bleys RLAW. Sympathetic nerve tissue in milky spots of the human greater omentum. J Anat 2020; 236:156-164. [PMID: 31498441 PMCID: PMC6904595 DOI: 10.1111/joa.13077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2019] [Indexed: 12/02/2022] Open
Abstract
Omental milky spots (OMSs), small lymphoid structures positioned in the greater omentum, are involved in peritoneal immune homeostasis and the formation of omental metastases. Sympathetic nerve activity is known to regulate immune function in other lymphoid organs (e.g. spleen and lymph nodes) and to create a favourable microenvironment for various tumour types. However, it is still unknown whether OMSs receive sympathetic innervation. Therefore, the aim of this study was to establish whether OMSs of the adult human greater omentum receive sympathetic innervation. A total of 18 OMSs were isolated from five omenta, which were removed from 3% formaldehyde-perfused cadavers (with a median age of 84 years, ranging from 64 to 94). OMSs were embedded in paraffin, cut and stained with a general (PGP9.5) and sympathetic nerve marker (TH and DBH), and evaluated by bright field microscopy. A T-cell, B-cell, and macrophage staining was performed to confirm OMS identity. In 50% of the studied OMSs, sympathetic nerve fibres were observed at multiple levels of the same OMS. Nerve fibres were represented as dots or elongated structures and often observed in relation to small vessels and occasionally as individual structures residing between lymphoid cells. The current study shows that 50% of the investigated OMSs contain sympathetic nerve fibres. These findings may contribute to our understanding of neural regulation of peritoneal immune response and the involvement of OMSs in omental metastases.
Collapse
Affiliation(s)
- Cindy G. J. Cleypool
- Department of AnatomyDivision of Surgical SpecialtiesUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Bernadette Schurink
- Department of AnatomyDivision of Surgical SpecialtiesUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Dorinde E. M. van der Horst
- Department of AnatomyDivision of Surgical SpecialtiesUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Ronald L. A. W. Bleys
- Department of AnatomyDivision of Surgical SpecialtiesUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
82
|
Chemical sympathectomy attenuates lipopolysaccharide-induced increase of plasma cytokine levels in rats pretreated by ACTH. J Neuroimmunol 2019; 337:577086. [DOI: 10.1016/j.jneuroim.2019.577086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/15/2022]
|
83
|
Kusuda R, Carreira EU, Ulloa L, Cunha FQ, Kanashiro A, Cunha TM. Choline attenuates inflammatory hyperalgesia activating nitric oxide/cGMP/ATP-sensitive potassium channels pathway. Brain Res 2019; 1727:146567. [PMID: 31783002 DOI: 10.1016/j.brainres.2019.146567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/18/2019] [Accepted: 11/23/2019] [Indexed: 12/25/2022]
Abstract
New findings on neural regulation of immunity are allowing the design of novel pharmacological strategies to control inflammation and nociception. Herein, we report that choline, a 7-nicotinic acetylcholine receptor (α7nAChRs) agonist, prevents carrageenan-induced hyperalgesia without affecting inflammatory parameters (neutrophil migration or cytokine/chemokines production) or inducing sedation or even motor impairment. Choline also attenuates prostaglandin-E2 (PGE2)-induced hyperalgesia via α7nAChR activation and this antinociceptive effect was abrogated by administration of LNMMA (a nitric oxide synthase inhibitor), ODQ (an inhibitor of soluble guanylate cyclase; cGMP), andglibenclamide(an inhibitor of ATP-sensitive potassium channels). Furthermore, choline attenuates long-lasting Complete Freund's Adjuvant and incision-induced hyperalgesia suggesting its therapeutic potential to treat pain in rheumatoid arthritis or post-operative recovery, respectively. Our results suggest that choline modulates inflammatory hyperalgesia by activating the nitric oxide/cGMP/ATP-sensitive potassium channels without interfering in inflammatory events, and could be used in persistent pain conditions.
Collapse
Affiliation(s)
- Ricardo Kusuda
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eleonora Uchôa Carreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC 27710, USA
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Kanashiro
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
84
|
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: 37] [Impact Index Per Article: 6.2] [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
|
85
|
Nogueira JE, Agostinho BK, Mota CM, Branco LG. Splenic anti-inflammatory reflex in immune tolerance. J Therm Biol 2019; 85:102411. [DOI: 10.1016/j.jtherbio.2019.102411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/27/2022]
|
86
|
Neuroimmune Interactions in the Gut and Their Significance for Intestinal Immunity. Cells 2019; 8:cells8070670. [PMID: 31269754 PMCID: PMC6679154 DOI: 10.3390/cells8070670] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBD) have a complex, multifactorial pathophysiology with an unmet need for effective treatment. This calls for novel strategies to improve disease outcome and quality of life for patients. Increasing evidence suggests that autonomic nerves and neurotransmitters, as well as neuropeptides, modulate the intestinal immune system, and thereby regulate the intestinal inflammatory processes. Although the autonomic nervous system is classically divided in a sympathetic and parasympathetic branch, both play a pivotal role in the crosstalk with the immune system, with the enteric nervous system acting as a potential interface. Pilot clinical trials that employ vagus nerve stimulation to reduce inflammation are met with promising results. In this paper, we review current knowledge on the innervation of the gut, the potential of cholinergic and adrenergic systems to modulate intestinal immunity, and comment on ongoing developments in clinical trials.
Collapse
|
87
|
Fertan E, Rodrigues GJ, Wheeler RV, Goguen D, Wong AA, James H, Stadnyk A, Brown RE, Weaver IC. Cognitive Decline, Cerebral-Spleen Tryptophan Metabolism, Oxidative Stress, Cytokine Production, and Regulation of the Txnip Gene in a Triple Transgenic Mouse Model of Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1435-1450. [DOI: 10.1016/j.ajpath.2019.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/01/2019] [Accepted: 03/07/2019] [Indexed: 02/06/2023]
|
88
|
Gao Y, Kang K, Zhang X, Han Q, Liu H, Kong W, Zhang X, Huang R, Yang Z, Qi Z, Zheng J, Li M, Li J, Liu R, Liu Y, Wang S, Zhang W, Wang H, Yu K. Effect of splenectomy on attenuation of LPS-induced AKI through GTS-21-induced cholinergic anti-inflammatory pathway. Am J Transl Res 2019; 11:2540-2549. [PMID: 31105861 PMCID: PMC6511767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
This work was undertaken to explore the role of splenectomy on attenuation of lipopolysaccharide (LPS)-induced acute kidney injury (AKI) through GTS-21-induced cholinergic anti-inflammatory pathway. C57BL/6 mice were used to construct models of sepsis-induced renal injury. HE, Tunel and blood assays were used to determine the success of the model. The animals were examined after splenectomy with or without LPS and GTS-21+LPS treatments. The pathological changes and apoptosis in the renal tissue were detected using HE and Tunel assays. The contents of creatinine (Cr) and cystatin-C (Cys-C) were measured using ELISA. The expression of IL-6, NF-kB p65, Caspase-3, anti-apoptotic protein Bcl-2, apoptotic protein Bax and α7nAChR was quantified using qRT-PCR. The expression of Bcl-2, Bax, Caspase-3, IL-6, NF-kB p65, α7nAChR and p-STAT3 was using assessed using Western blot analysis. HE, Tunel, BUN and serum creatinine (SC) assay showed that renal injury models were successfully established. Compared with the control, the apoptosis in the LPS group was significantly increased and decreased after GTS-21 treatment. However, splenectomy combined with GTS-21 increased the apoptosis, indicating that splenectomy could partially offset the anti-apoptosis effect of GTS-21. In animals treated with LPS, the contents of Cr and Cys-C increased significantly. These contents reduced following GTS-21 treatment, but increased after splenectomy. After LPS treatment, the expression of IL-6, NF-kB p65, p-STAT3, Caspase-3 and Bax was significantly up-regulated, while the expression of α7nAChR and Bcl-2 significantly down-regulated. Compared with LPS treated mice, splenectomy reduced the expression of IL-6, NF-kB p65 and p-STAT3, suggesting that splenectomy inhibits the activation of α7nAChR pathway by the GTS-21. It is clear that GTS-21 effectively attenuates LPS-induced renal injury; splenectomy suppresses the anti-inflammatory and anti-apoptosis activity and renal protective effect of GTS-21. On other hand, splenectomy reduces the production of inflammatory cytokines in the circulation, and has certain protective effect on the kidney. Therefore, the impact of splenectomy on LPS-induced AKI depends on the strength of the two aspects.
Collapse
Affiliation(s)
- Yang Gao
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Xinyu Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Qiuyuan Han
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Haitao Liu
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Weilan Kong
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
| | - Xing Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Rui Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Zhenyu Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Zhidong Qi
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Junbo Zheng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Ming Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Jiayu Li
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Ruijin Liu
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Yansong Liu
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Sicong Wang
- Department of Critical Care Medicine, The Cancer Hospital of Harbin Medical UniversityHarbin 150081, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical UniversityHarbin 150086, China
- Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University Ministry of EducationHarbin 150086, China
| | - Hongliang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150086, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, China
- The Centre for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical UniversityHarbin 150086, China
| |
Collapse
|
89
|
Cholinergic Stimulation by Pyridostigmine Bromide Before Myocardial Infarction Prevent Cardiac and Autonomic Dysfunction. Sci Rep 2019; 9:2481. [PMID: 30792425 PMCID: PMC6385301 DOI: 10.1038/s41598-019-38841-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Inflammatory processes and cardiovascular autonomic imbalance are very relevant characteristic of the enormous dynamic process that is a myocardial infarction (MI). In this sense, some studies are investigating pharmacological therapies using acetylcholinesterase inhibitors, such as pyridostigmine bromide (PYR), aiming to increase parasympathetic tone after MI. Here we hypothesized that the use of PYR before the MI might bring an additional positive effect to the autonomic function, and consequently, in the inflammatory response and cardiac function. The present study aimed to evaluate left ventricular function, baroreflex sensitivity, autonomic modulation, and inflammatory profile in PYR-treated rats previously to MI. Methods: Male Wistar rats (250–300 g) were treated for 60 days with PYR. After treatment, they were submitted to the MI. After the MI, the autonomic and ventricular function were evaluated, as well as the systemic, left ventricle, and adipose tissue inflammatory profile. Results: PYR, performed before MI, prevented HR increase, systolic function impairment, baroreflex sensitivity drop, as well as pulse interval variance, RMSSD, blood pressure and parasympathetic modulation reduction in treated rats compared to untreated rats. Also, this positive functional changes may have been a result of the reduced inflammatory parameters in the left ventricle (IFN-γ, IL-6, and IL-1β), as well as increased IL-10 expression and IL-10/TNF-α ratio in treated animals before MI. Conclusion: Prior treatment with PYR prevents impairment of the autonomic nervous system after MI, which may be associated with the attenuated expression of inflammatory factors and heart dysfunction.
Collapse
|
90
|
Liu L, Zhao M, Yu X, Zang W. Pharmacological Modulation of Vagal Nerve Activity in Cardiovascular Diseases. Neurosci Bull 2019; 35:156-166. [PMID: 30218283 PMCID: PMC6357265 DOI: 10.1007/s12264-018-0286-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/13/2018] [Indexed: 01/17/2023] Open
Abstract
Cardiovascular diseases are life-threatening illnesses with high morbidity and mortality. Suppressed vagal (parasympathetic) activity and increased sympathetic activity are involved in these diseases. Currently, pharmacological interventions primarily aim to inhibit over-excitation of sympathetic nerves, while vagal modulation has been largely neglected. Many studies have demonstrated that increased vagal activity reduces cardiovascular risk factors in both animal models and human patients. Therefore, the improvement of vagal activity may be an alternate approach for the treatment of cardiovascular diseases. However, drugs used for vagus nerve activation in cardiovascular diseases are limited in the clinic. In this review, we provide an overview of the potential drug targets for modulating vagal nerve activation, including muscarinic, and β-adrenergic receptors. In addition, vagomimetic drugs (such as choline, acetylcholine, and pyridostigmine) and the mechanism underlying their cardiovascular protective effects are also discussed.
Collapse
Affiliation(s)
- Longzhu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiaojiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Weijin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
91
|
Wang Z, He D, Zeng YY, Zhu L, Yang C, Lu YJ, Huang JQ, Cheng XY, Huang XH, Tan XJ. The spleen may be an important target of stem cell therapy for stroke. J Neuroinflammation 2019; 16:20. [PMID: 30700305 PMCID: PMC6352449 DOI: 10.1186/s12974-019-1400-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Stroke is the most common cerebrovascular disease, the second leading cause of death behind heart disease and is a major cause of long-term disability worldwide. Currently, systemic immunomodulatory therapy based on intravenous cells is attracting attention. The immune response to acute stroke is a major factor in cerebral ischaemia (CI) pathobiology and outcomes. Over the past decade, the significant contribution of the spleen to ischaemic stroke has gained considerable attention in stroke research. The changes in the spleen after stroke are mainly reflected in morphology, immune cells and cytokines, and these changes are closely related to the stroke outcomes. Autonomic nervous system (ANS) activation, release of central nervous system (CNS) antigens and chemokine/chemokine receptor interactions have been documented to be essential for efficient brain-spleen cross-talk after stroke. In various experimental models, human umbilical cord blood cells (hUCBs), haematopoietic stem cells (HSCs), bone marrow stem cells (BMSCs), human amnion epithelial cells (hAECs), neural stem cells (NSCs) and multipotent adult progenitor cells (MAPCs) have been shown to reduce the neurological damage caused by stroke. The different effects of these cell types on the interleukin (IL)-10, interferon (IFN), and cholinergic anti-inflammatory pathways in the spleen after stroke may promote the development of new cell therapy targets and strategies. The spleen will become a potential target of various stem cell therapies for stroke represented by MAPC treatment.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.,Institute of Reproductive and Stem Cell Research, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Da He
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Ya-Yue Zeng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Li Zhu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Chao Yang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Yong-Juan Lu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Jie-Qiong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Yan Cheng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiang-Hong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Jun Tan
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.
| |
Collapse
|
92
|
Joseph B, Shimojo G, Li Z, Thompson-Bonilla MDR, Shah R, Kanashiro A, Salgado HC, Ulloa L. Glucose Activates Vagal Control of Hyperglycemia and Inflammation in Fasted Mice. Sci Rep 2019; 9:1012. [PMID: 30700738 PMCID: PMC6354016 DOI: 10.1038/s41598-018-36298-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/14/2018] [Indexed: 11/18/2022] Open
Abstract
Sepsis is a leading cause of death in hospitalized patients. Many experimental treatments may have failed in clinical trials for sepsis, in part, because they focused on immune responses of healthy animals that did not mimic the metabolic settings of septic patients. Epidemiological studies show an association between metabolic and immune alterations and over 1/3 of septic patients are diabetic, but the mechanism linking these systems is unknown. Here, we report that metabolic fasting increased systemic inflammation and worsened survival in experimental sepsis. Feeding and administration of glucose in fasted mice activated the vagal tone without affecting blood pressure. Vagal stimulation attenuated hyperglycemia and serum TNF levels in sham but only hyperglycemia in splenectomized mice. Vagal stimulation induced the production of dopamine from the adrenal glands. Experimental diabetes increased hyperglycemia and systemic inflammation in experimental sepsis. Fenoldopam, a specific dopaminergic type-1 agonist, attenuated hyperglycemia and systemic inflammation in diabetic endotoxemic mice. These results indicate that glucose activates vagal control of hyperglycemia and inflammation in fasted septic mice via dopamine.
Collapse
Affiliation(s)
- Biju Joseph
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Guilherme Shimojo
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Zhifeng Li
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Maria Del Rocio Thompson-Bonilla
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
- Hospital "October 1st", ISSSTE", 1669 National Polytechnic Institute Ave, Mexico City, Mexico
| | - Roshan Shah
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alexandre Kanashiro
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Physiology, Medical School - University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Helio C Salgado
- Department of Physiology, Medical School - University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luis Ulloa
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
- Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
93
|
Non-canonical cholinergic anti-inflammatory pathway-mediated activation of peritoneal macrophages induces Hes1 and blocks ischemia/reperfusion injury in the kidney. Kidney Int 2019; 95:563-576. [PMID: 30670317 DOI: 10.1016/j.kint.2018.09.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 09/11/2018] [Accepted: 09/20/2018] [Indexed: 11/22/2022]
Abstract
The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic β2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including β2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.
Collapse
|
94
|
Shimojo G, Joseph B, Shah R, Consolim-Colombo FM, De Angelis K, Ulloa L. Exercise activates vagal induction of dopamine and attenuates systemic inflammation. Brain Behav Immun 2019; 75:181-191. [PMID: 30394312 PMCID: PMC6334665 DOI: 10.1016/j.bbi.2018.10.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/26/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
Physical exercise is one of the most important factors improving quality of life, but it is not feasible for patients with morbidity or limited mobility. Most previous studies focused on high-intensity or long-term exercise that causes metabolic stress or physiological adaption, respectively. Here, we studied how moderate-intensity swimming affects systemic inflammation in 6-8 week old C57BL/6J male mice during endotoxemia. One-hour swimming prevented hypokalemia, hypocalcemia, attenuated serum levels of inflammatory cytokines, increased anti-inflammatory cytokines but affected neither IL6 nor glycemia before or after the endotoxic challenge. Exercise attenuated serum TNF levels by inhibiting its production in the spleen through a mechanism mediated by the subdiaphragmatic vagus nerve but independent of the splenic nerve. Exercise increased serum levels of dopamine, and adrenalectomy prevented the potential of exercise to induce dopamine and to attenuate serum TNF levels. Dopaminergic agonist type-1, fenoldopam, inhibited TNF production in splenocytes. Conversely, dopaminergic antagonist type-1, butaclamol, attenuated exercise control of serum TNF levels. These results suggest that vagal induction of dopamine may contribute to the anti-inflammatory potential of physical exercise.
Collapse
Affiliation(s)
- Guilherme Shimojo
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA; Nove de Julho University (UNINOVE), Sao Paulo, Brazil
| | - Biju Joseph
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Roshan Shah
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Fernanda M Consolim-Colombo
- Nove de Julho University (UNINOVE), Sao Paulo, Brazil; Hypertension Unit, Heart Institute (INCOR) School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Kátia De Angelis
- Nove de Julho University (UNINOVE), Sao Paulo, Brazil; Department of Physiology, Federal University of Sao Paulo (UNIFESP), Brazil
| | - Luis Ulloa
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA; Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ 07103, USA.
| |
Collapse
|
95
|
Martelli D, Farmer DGS, McKinley MJ, Yao ST, McAllen RM. Anti-inflammatory reflex action of splanchnic sympathetic nerves is distributed across abdominal organs. Am J Physiol Regul Integr Comp Physiol 2018; 316:R235-R242. [PMID: 30576218 DOI: 10.1152/ajpregu.00298.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The splanchnic anti-inflammatory pathway has been proposed as the efferent arm of the inflammatory reflex. Although much evidence points to the spleen as the principal target organ where sympathetic nerves inhibit immune function, a systematic study to locate the target organ(s) of the splanchnic anti-inflammatory pathway has not yet been made. In anesthetized rats made endotoxemic with lipopolysaccharide (LPS, 60 µg/kg iv), plasma levels of tumor necrosis factor-α (TNF-α) were measured in animals with cut (SplancX) or sham-cut (Sham) splanchnic nerves. We confirm here that disengagement of the splanchnic anti-inflammatory pathway in SplancX rats (17.01 ± 0.95 ng/ml, mean ± SE) strongly enhances LPS-induced plasma TNF-α levels compared with Sham rats (3.76 ± 0.95 ng/ml). In paired experiments, the responses of SplancX and Sham animals were compared after the single or combined removal of organs innervated by the splanchnic nerves. Removal of target organ(s) where the splanchnic nerves inhibit systemic inflammation should abolish any difference in LPS-induced plasma TNF-α levels between Sham and SplancX rats. Any secondary effects of extirpating organs should apply to both groups. Surprisingly, removal of the spleen and/or the adrenal glands did not prevent the reflex splanchnic anti-inflammatory action nor did the following removals: spleen + adrenals + intestine; spleen + intestine + stomach and pancreas; or spleen + intestine + stomach and pancreas + liver. Only when spleen, adrenals, intestine, stomach, pancreas, and liver were all removed did the difference between SplancX and Sham animals disappear. We conclude that the reflex anti-inflammatory action of the splanchnic nerves is distributed widely across abdominal organs.
Collapse
Affiliation(s)
- Davide Martelli
- Florey Institute of Neuroscience and Mental Health , Parkville, Victoria , Australia.,Department of Biomedical and Neuromotor Science, University of Bologna , Bologna , Italy
| | - David G S Farmer
- Florey Institute of Neuroscience and Mental Health , Parkville, Victoria , Australia
| | - Michael J McKinley
- Florey Institute of Neuroscience and Mental Health , Parkville, Victoria , Australia.,Department of Biomedical and Neuromotor Science, University of Bologna , Bologna , Italy.,Department of Physiology, University of Melbourne , Melbourne, Victoria , Australia
| | - Song T Yao
- Florey Institute of Neuroscience and Mental Health , Parkville, Victoria , Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria , Australia
| | - Robin M McAllen
- Florey Institute of Neuroscience and Mental Health , Parkville, Victoria , Australia
| |
Collapse
|
96
|
Chen SH, Chen HC, Hsieh CL, Chao PM. Electric stimulation of ears accelerates body weight loss mediated by high-fat to low-fat diet switch accompanied by increased white adipose tissue browning in C57BL/6 J mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:323. [PMID: 30518367 PMCID: PMC6282328 DOI: 10.1186/s12906-018-2388-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/27/2018] [Indexed: 01/09/2023]
Abstract
Background Weight reduction frequently occurs in patients receiving vagus nerve stimulation (VNS) therapy. Therefore, we hypothesized that during dietary intervention for weight loss, auricular electric stimulation (AES), an alternative of VNS, accelerates weight loss by increasing white adipose tissue (WAT) browning and increases energy expenditure. Methods C57BL/6J male mice were fed a high-fat diet for 5 wk. to induce obesity, then switched to a low-fat diet for 5 wk. and allocated into 3 groups to receive 2 Hz electric stimulation on ears, electrode clamps only, or nothing (AES, Sham and Ctrl, respectively). Results Switching to a low-fat diet reduced body weight progressively in all 3 groups, with the greatest reduction in the AES group. In accordance with a mild decrease in feed intake, hypothalamus mRNA levels of Npy, AgRP tended to be reduced, while Pomc tended to be increased by AES. Mice in the AES group had the highest concentrations of norepinephrine in serum and inguinal WAT, and expression levels of uncoupling protein-1 (UCP-1) and tyrosine hydroxylase in inguinal WAT. Furthermore, their subcutaneous adipocytes had multilocular and UCP-1+ characteristics, along with a smaller cell size. Conclusion AES, by increasing WAT browning, could be used in conjunction with a low-fat diet to augment weight loss in addition to suppressing appetite.
Collapse
|
97
|
Role of C1 neurons in anti-inflammatory reflex: Mediation between afferents and efferents. Neurosci Res 2018; 136:6-12. [DOI: 10.1016/j.neures.2018.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/24/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
|
98
|
Abstract
The interplay between the immune and nervous systems has been acknowledged in the past, but only more recent studies have started to unravel the cellular and molecular players of such interactions. Mounting evidence indicates that environmental signals are sensed by discrete neuro-immune cell units (NICUs), which represent defined anatomical locations in which immune and neuronal cells colocalize and functionally interact to steer tissue physiology and protection. These units have now been described in multiple tissues throughout the body, including lymphoid organs, adipose tissue, and mucosal barriers. As such, NICUs are emerging as important orchestrators of multiple physiological processes, including hematopoiesis, organogenesis, inflammation, tissue repair, and thermogenesis. In this review we focus on the impact of NICUs in tissue physiology and how this fast-evolving field is driving a paradigm shift in our understanding of immunoregulation and organismal physiology.
Collapse
Affiliation(s)
- Cristina Godinho-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal; , ,
| | - Filipa Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal; , ,
| | | |
Collapse
|
99
|
Gaidhani N, Uteshev VV. Treatment duration affects cytoprotective efficacy of positive allosteric modulation of α7 nAChRs after focal ischemia in rats. Pharmacol Res 2018; 136:121-132. [PMID: 30205140 PMCID: PMC6218269 DOI: 10.1016/j.phrs.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/22/2018] [Accepted: 09/02/2018] [Indexed: 12/30/2022]
Abstract
To minimize irreversible brain injury after acute ischemic stroke (AIS), the time to treatment (i.e., treatment delay) should be minimized. However, thus far, all cytoprotective clinical trials have failed. Analysis of literature identified short treatment durations (≤72 h) as a common motif among completed cytoprotective clinical trials. Here, we argue that short cytoprotective regimens even if given early after AIS may only slow down the evolution of ischemic brain injury and fail to deliver sustained long-term solutions leading to relapses that may be misinterpreted for conceptual failure of cytoprotection. In this randomized blinded study, we used young adult male rats subjected to transient 90 min suture middle cerebral artery occlusion (MCAO) and treated with acute vs. sub-chronic regimens of PNU120596, a prototypical positive allosteric modulator of α7 nicotinic acetylcholine receptors with anti-inflammatory cytoprotective properties to test the hypothesis that insufficient treatment durations may reduce therapeutic benefits of otherwise efficacious cytoprotectants after AIS. A single acute treatment 90 min after MCAO significantly reduced brain injury and neurological deficits 24 h later, but these effects vanished 72 h after MCAO. These relapses were avoided by utilizing sub-chronic treatments. Thus, extending treatment duration augments therapeutic efficacy of PNU120596 after MCAO. Furthermore, sub-chronic treatments could offset the negative effects of prolonged treatment delays in cases where the acute treatment window after MCAO was left unexploited. We conclude that a combination of short treatment delays and prolonged treatment durations may be required to maximize therapeutic effects of PNU120596, reduce relapses and ensure sustained therapeutic efficacy after AIS. Similar concepts may hold for other cytoprotectants including those that failed in clinical trials.
Collapse
Affiliation(s)
- Nikhil Gaidhani
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States.
| |
Collapse
|
100
|
Komegae EN, Farmer DGS, Brooks VL, McKinley MJ, McAllen RM, Martelli D. Vagal afferent activation suppresses systemic inflammation via the splanchnic anti-inflammatory pathway. Brain Behav Immun 2018; 73:441-449. [PMID: 29883598 PMCID: PMC6319822 DOI: 10.1016/j.bbi.2018.06.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/11/2018] [Accepted: 06/04/2018] [Indexed: 01/11/2023] Open
Abstract
Electrical stimulation of the vagus nerve (VNS) is a novel strategy used to treat inflammatory conditions. Therapeutic VNS activates both efferent and afferent fibers; however, the effects attributable to vagal afferent stimulation are unclear. Here, we tested if selective activation of afferent fibers in the abdominal vagus suppresses systemic inflammation. In urethane-anesthetized rats challenged with lipopolysaccharide (LPS, 60 µg/kg, i.v.), abdominal afferent VNS (2 Hz for 20 min) reduced plasma tumor necrosis factor alpha (TNF) levels 90 min later by 88% compared with unmanipulated animals. Pre-cutting the cervical vagi blocked this anti-inflammatory action. Interestingly, the surgical procedure to expose and prepare the abdominal vagus for afferent stimulation ('vagal manipulation') also had an anti-inflammatory action. Levels of the anti-inflammatory cytokine IL-10 were inversely related to those of TNF. Prior bilateral section of the splanchnic sympathetic nerves reversed the anti-inflammatory actions of afferent VNS and vagal manipulation. Sympathetic efferent activity in the splanchnic nerve was shown to respond reflexly to abdominal vagal afferent stimulation. These data demonstrate that experimentally activating abdominal vagal afferent fibers suppresses systemic inflammation, and that the efferent neural pathway for this action is in the splanchnic sympathetic nerves.
Collapse
Affiliation(s)
- Evilin Naname Komegae
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Virginia Leah Brooks
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, USA
| | - Michael Joseph McKinley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia,Department of Physiology, University of Melbourne, Australia
| | - Robin Michael McAllen
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia.
| | - Davide Martelli
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Department of Biomedical and Neuromotor Science (DIBINEM), University of Bologna, Bologna, Italy.
| |
Collapse
|