1
|
Janssen HJB, Geraedts TCM, Fransen LFC, van Ark I, Leusink-Muis T, Folkerts G, Garssen J, Ruurda JP, Nieuwenhuijzen GAP, van Hillegersberg R, Luyer MDP. Electrical vagus nerve stimulation is a promising approach to reducing pulmonary complications after an esophagectomy: an experimental rodent model. Immunol Res 2024; 72:1247-1258. [PMID: 39083131 PMCID: PMC11618150 DOI: 10.1007/s12026-024-09523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/22/2024] [Indexed: 12/08/2024]
Abstract
After esophagectomy, an imbalanced inflammatory response increases the risk of postoperative morbidity. The vagus nerve modulates local and systemic inflammatory responses, but its pulmonary branches are transected during esophagectomy as part of the oncological resection, which may account for the high incidence of postoperative (pulmonary) complications. This study investigated the effect of electrical vagus nerve stimulation (VNS) on lipopolysaccharide (LPS)-induced lung injury in rats. Rats (n = 60) were randomly assigned to a non-vagotomy or cervical vagotomy group, with VNS or without (NOSTIM). There were four non-vagotomy groups: NOSTIM and bilateral VNS with 100, 50, or 10 µA. The four vagotomy groups were NOSTIM and VNS with fixed amplitude (50 µA) bilaterally before (VNS-50-before) or after bilateral vagotomy (VNS-50-after), or unilaterally (left) before ipsilateral vagotomy (VNS-50-unilaterally). LPS was administered intratracheally after surgery. Pulmonary function, pro-inflammatory cytokines in serum, broncho-alveolar lavage fluid (BALF), and histopathological lung injury (LIS) were assessed 180 min post-procedure. In non-vagotomized rats, neutrophil influx in BALF following intra-tracheal LPS (mean 30 [± 23]; P = 0.075) and LIS (mean 0.342 [± 0.067]; P = 0.142) were similar after VNS-100, compared with NOSTIM. VNS-50 reduced neutrophil influx (23 [± 19]; P = 0.024) and LIS (0.316 [± 0.093]; P = 0.043). VNS-10 reduced neutrophil influx (15 [± 6]; P = 0.009), while LIS (0.331 [± 0.053]; P = 0.088) was similar. In vagotomized rats, neutrophil influx (52 [± 37]; P = 0.818) and LIS (0.407 [SD ± 0.037]; P = 0.895) in VNS-50-before were similar compared with NOSTIM, as well as in VNS-50-after (neutrophils 30 [± 26]; P = 0.090 and LIS 0.344 [± 0.053]; P = 0.073). In contrast, VNS-50-unilaterally reduced neutrophil influx (26 [± 10]; P = 0.050) and LIS (0.296 [± 0.065]; P = 0.005). Systemic levels of cytokines TNF-α and IL-6 were undetectable in all groups. Pulmonary function was not statistically significantly affected. In conclusion, VNS limited influx of neutrophils in lungs in non-vagotomized rats and may attenuate LIS. Unilateral VNS attenuated lung injury even after ipsilateral vagotomy. This effect was absent for bilateral VNS before and after bilateral vagotomy. It is suggested that the effect of VNS is dependent on (partially) intact vagus nerves and that the level of the vagotomy during esophagectomy may influence postoperative pulmonary outcomes.
Collapse
Affiliation(s)
- Henricus J B Janssen
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands.
| | - Tessa C M Geraedts
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
| | - Laura F C Fransen
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
| | - Ingrid van Ark
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Nutricia Research, Immunology, Utrecht, The Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Grard A P Nieuwenhuijzen
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
| | - Richard van Hillegersberg
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands.
- Department of Electrical Engineering, University of Technology Eindhoven, Eindhoven, The Netherlands.
| |
Collapse
|
2
|
Khodadadi F, Punait S, Ketabchi F, Khodabandeh Z, Bahaoddini A, Lewis GF. Comparison of heart rate variability, hemodynamic, metabolic and inflammatory parameters in various phases of decompansatory hemorrhagic shock of normal and vagotomized conscious male rats. BMC Cardiovasc Disord 2024; 24:661. [PMID: 39567879 PMCID: PMC11577762 DOI: 10.1186/s12872-024-04342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Heart rate variability (HRV) analysis has shown promise as a valuable complementary tool for clinical assessment in trauma cases. This study aims to evaluate the utility of HRV in monitoring different severities of hemorrhagic shock (HS) and its correlation with traditional hemodynamic and metabolic parameters. METHODS Male Sprague-Dawley rats were divided into different experimental groups, including those with and without vagotomy, and were exposed to different classes of decompensatory HS. To induce varying severities of HS, volume resuscitation was delayed by gradually returning 0%, 20%, or 50% of the shed blood volume at the end of the compensation phase, referred to as 0% DFR, 20% DFR, and 50% DFR class, respectively. Hemodynamic parameters were monitored, and HRV was calculated. Levels of TNF-α and IL-10 were determined in lung tissue at the end of the experiments. Correlations between HRV, hemodynamic parameters, inflammatory gene expression and arterial blood gas variables were evaluated. RESULTS HRV showed increased power of the low-frequency (LF) and respiratory sinus arrhythmia (RSA) in all groups during the hypotension phase of HS (Nadir 1). Subdiaphragmatic vagotomy blunted the increase in the LF component in the Nadir 1. After volume resuscitation, systolic blood pressure (SBP), RSA and LF returned to baseline in the 0% DFR and 20% DFR classes. However, animals in 50% DFR class exhibited a reduced SBP and LF and lower pH. Notably, strong correlations were found between LF and SBP as well as tissue hypoperfusion markers. The expression of TNF-α in the lung was increased in all HS groups, while this gene expression was significantly higher in the vagotomized animals. CONCLUSION The alterations in HRV components were found to be significantly correlated with the hemodynamic and metabolic status of the animals, while showing no association with inflammatory responses. Additionally, the intervention of subdiaphragmatic vagotomy significantly impacted both HRV components and inflammatory responses. Collectively, these findings suggest the potential of HRV components for the assessment of the presence and severity of HS.
Collapse
Affiliation(s)
- Fateme Khodadadi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
- Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Sujata Punait
- Intelligent Systems Engineering, Indiana University, The Traumatic Stress Research Consortium at the Kinsey Institute, Indiana University, Bloomington, IN, United States
| | - Farzaneh Ketabchi
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Gregory F Lewis
- Kinsey Institute, Indiana University, Bloomington, IN, United States.
| |
Collapse
|
3
|
Berthon A, Wernisch L, Stoukidi M, Thornton M, Tessier-Lariviere O, Fortier-Poisson P, Mamen J, Pinkney M, Lee S, Sarkans E, Annecchino L, Appleton B, Garsed P, Patterson B, Gonshaw S, Jakopec M, Shunmugam S, Edwards T, Tukiainen A, Jennings J, Lajoie G, Hewage E, Armitage O. Using neural biomarkers to personalize dosing of vagus nerve stimulation. Bioelectron Med 2024; 10:15. [PMID: 38880906 PMCID: PMC11181600 DOI: 10.1186/s42234-024-00147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) is an established therapy for treating a variety of chronic diseases, such as epilepsy, depression, obesity, and for stroke rehabilitation. However, lack of precision and side-effects have hindered its efficacy and extension to new conditions. Achieving a better understanding of the relationship between VNS parameters and neural and physiological responses is therefore necessary to enable the design of personalized dosing procedures and improve precision and efficacy of VNS therapies. METHODS We used biomarkers from recorded evoked fiber activity and short-term physiological responses (throat muscle, cardiac and respiratory activity) to understand the response to a wide range of VNS parameters in anaesthetised pigs. Using signal processing, Gaussian processes (GP) and parametric regression models we analyse the relationship between VNS parameters and neural and physiological responses. RESULTS Firstly, we illustrate how considering multiple stimulation parameters in VNS dosing can improve the efficacy and precision of VNS therapies. Secondly, we describe the relationship between different VNS parameters and the evoked fiber activity and show how spatially selective electrodes can be used to improve fiber recruitment. Thirdly, we provide a detailed exploration of the relationship between the activations of neural fiber types and different physiological effects. Finally, based on these results, we discuss how recordings of evoked fiber activity can help design VNS dosing procedures that optimize short-term physiological effects safely and efficiently. CONCLUSION Understanding of evoked fiber activity during VNS provide powerful biomarkers that could improve the precision, safety and efficacy of VNS therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Guillaume Lajoie
- Université de Montréal and Mila-Quebec AI Institute, Montréal, Canada
| | | | | |
Collapse
|
4
|
Alvarado-Peña N, Galeana-Cadena D, Gómez-García IA, Mainero XS, Silva-Herzog E. The microbiome and the gut-lung axis in tuberculosis: interplay in the course of disease and treatment. Front Microbiol 2023; 14:1237998. [PMID: 38029121 PMCID: PMC10643882 DOI: 10.3389/fmicb.2023.1237998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Tuberculosis is a chronic infectious disease caused by Mycobacterium tuberculosis (MTB) that remains a significant global health challenge. The extensive use of antibiotics in tuberculosis treatment, disrupts the delicate balance of the microbiota in various organs, including the gastrointestinal and respiratory systems. This gut-lung axis involves dynamic interactions among immune cells, microbiota, and signaling molecules from both organs. The alterations of the microbiome resulting from anti-TB treatment can significantly influence the course of tuberculosis, impacting aspects such as complete healing, reinfection, and relapse. This review aims to provide a comprehensive understanding of the gut-lung axis in the context of tuberculosis, with a specific focus on the impact of anti-TB treatment on the microbiome.
Collapse
Affiliation(s)
- Néstor Alvarado-Peña
- Clínica de Tuberculosis, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, México City, Mexico
| | - David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias, México City, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, Mexico
| | - Xavier Soberón Mainero
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Eugenia Silva-Herzog
- Laboratorio de Vinculación Científica, Facultad de Medicina-Universidad Nacional Autonoma de México-Instituto Nacional de Medicina Genomica, México City, Mexico
| |
Collapse
|
5
|
Sant'Anna FM, Resende RCL, Sant'Anna LB, Couceiro SLM, Pinto RBS, Sant'Anna MB, Chao LW, Szeles JC, Kaniusas E. Auricular vagus nerve stimulation: a new option to treat inflammation in COVID-19? REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230345. [PMID: 37283364 DOI: 10.1590/1806-9282.20230345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/19/2023] [Indexed: 06/08/2023]
Affiliation(s)
- Fernando Mendes Sant'Anna
- Universidade Federal do Rio de Janeiro - Macaé (RJ), Brazil
- Hospital Santa Izabel - Cabo Frio (RJ), Brazil
| | | | | | | | | | | | - Liaw Wen Chao
- Universidade de São Paulo, Hospital das Clínicas - São Paulo (SP), Brazil
| | - Jozsef Constantin Szeles
- Medical University of Vienna, Department of Surgery, Division of Vascular Surgery - Vienna, Austria
| | - Eugenijus Kaniusas
- Vienna Universit y of Technology, Institute of Biomedical Electronics, Faculty of Electrical Engineering and Information Technology - Vienna, Austria
| |
Collapse
|
6
|
Seesing MFJ, Janssen HJB, Geraedts TCM, Weijs TJ, van Ark I, Leusink-Muis T, Folkerts G, Garssen J, Ruurda JP, Nieuwenhuijzen GAP, van Hillegersberg R, Luyer MDP. Exploring the Modulatory Effect of High-Fat Nutrition on Lipopolysaccharide-Induced Acute Lung Injury in Vagotomized Rats and the Role of the Vagus Nerve. Nutrients 2023; 15:2327. [PMID: 37242210 PMCID: PMC10220714 DOI: 10.3390/nu15102327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
During esophagectomy, the vagus nerve is transected, which may add to the development of postoperative complications. The vagus nerve has been shown to attenuate inflammation and can be activated by a high-fat nutrition via the release of acetylcholine. This binds to α7 nicotinic acetylcholine receptors (α7nAChR) and inhibits α7nAChR-expressing inflammatory cells. This study investigates the role of the vagus nerve and the effect of high-fat nutrition on lipopolysaccharide (LPS)-induced lung injury in rats. Firstly, 48 rats were randomized in 4 groups as follows: sham (sparing vagus nerve), abdominal (selective) vagotomy, cervical vagotomy and cervical vagotomy with an α7nAChR-agonist. Secondly, 24 rats were randomized in 3 groups as follows: sham, sham with an α7nAChR-antagonist and cervical vagotomy with an α7nAChR-antagonist. Finally, 24 rats were randomized in 3 groups as follows: fasting, high-fat nutrition before sham and high-fat nutrition before selective vagotomy. Abdominal (selective) vagotomy did not impact histopathological lung injury (LIS) compared with the control (sham) group (p > 0.999). There was a trend in aggravation of LIS after cervical vagotomy (p = 0.051), even after an α7nAChR-agonist (p = 0.090). Cervical vagotomy with an α7nAChR-antagonist aggravated lung injury (p = 0.004). Furthermore, cervical vagotomy increased macrophages in bronchoalveolar lavage (BAL) fluid and negatively impacted pulmonary function. Other inflammatory cells, TNF-α and IL-6, in the BALF and serum were unaffected. High-fat nutrition reduced LIS after sham (p = 0.012) and selective vagotomy (p = 0.002) compared to fasting. vagotomy. This study underlines the role of the vagus nerve in lung injury and shows that vagus nerve stimulation using high-fat nutrition is effective in reducing lung injury, even after selective vagotomy.
Collapse
Affiliation(s)
- Maarten F. J. Seesing
- Department of Surgery, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, The Netherlands
| | | | - Tessa C. M. Geraedts
- Department of Surgery, Catharina Hospital, 5623 Eindhoven, The Netherlands; (H.J.B.J.)
| | - Teus J. Weijs
- Department of Surgery, Catharina Hospital, 5623 Eindhoven, The Netherlands; (H.J.B.J.)
| | - Ingrid van Ark
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands
- Danone Nutricia Research & Innovation, Immunology, 3584 Utrecht, The Netherlands
| | - Jelle P. Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, The Netherlands
| | | | - Richard van Hillegersberg
- Department of Surgery, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, The Netherlands
| | - Misha D. P. Luyer
- Department of Surgery, Catharina Hospital, 5623 Eindhoven, The Netherlands; (H.J.B.J.)
| |
Collapse
|
7
|
Zhang LM, Feng NN, Du HB, Zhang H, Guo R, Zhai JY, Zhang YP, Zhao ZG. Omega-3 polyunsaturated fatty acids alleviates lung injury mediated by post-hemorrhagic shock mesenteric lymph. Respir Physiol Neurobiol 2023; 310:104003. [PMID: 36566003 DOI: 10.1016/j.resp.2022.104003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Severe hemorrhage-induced acute lung injury (ALI) remains the major contributor to critical patient mortality and is associated with posthemorrhagic shock mesenteric lymph (PHSML) return. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) play overall protection on acute hemorrhage, but a reliable mechanism needs to be identified. The aims of this study were to investigate the role of ω-3 PUFAs in alleviating ALI and whether is related to the endotoxin contained in PHSML. Mesenteric lymph was harvested from rats subjected to hemorrhagic shock (hemorrhage-induced hypotension of 40 ± 2 mmHg for 90 min plus by resuscitation) or sham shock. The effect of ω-3 PUFAs on pulmonary function, water content, morphology, and LBP, CD14, TNF-α, and IL-6 levels were observed in rats subjected to hemorrhagic shock, while the effect of PHSML intravenous infusion on the beneficial effect of ω-3 PUFAs also was investigated. In addition, the effect of ω-3 PUFAs on the endotoxin contents in mesenteric lymph were detected. Hemorrhagic shock-induced ALI was characterized by increased functional residual capacity (FRC), lung resistance (RI), inspiratory capacity (IC), respiratory frequency, water contents and structural damage, along with increases in LBP, IL-6, and TNF-α. ω-3 PUFAs treatment reduced FRC, RI, IC, frequency, water contents, LBP, IL-6, TNF-α, and alleviated morphological damage. In contrast, PHSML infusion abolished the advantageous effects of ω-3 PUFAs on the above indices and CD14. Furthermore, the endotoxin level of PHSML was significantly enhanced, but declined following ω-3 PUFAs administration. These findings together suggested that treatment with ω-3 PUFAs ameliorates hemorrhagic shock-induced ALI, which is associated with reduced endotoxin contained in PHSML.
Collapse
Affiliation(s)
- Li-Min Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Niu-Niu Feng
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Hui-Bo Du
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Hong Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Rui Guo
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Jia-Yi Zhai
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China
| | - Yu-Ping Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei, PR China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei, PR China; Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, Hebei, PR China.
| |
Collapse
|
8
|
Huang Q, Gao S, Yao Y, Wang Y, Li J, Chen J, guo C, Zhao D, Li X. Innate immunity and immunotherapy for hemorrhagic shock. Front Immunol 2022; 13:918380. [PMID: 36091025 PMCID: PMC9453212 DOI: 10.3389/fimmu.2022.918380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022] Open
Abstract
Hemorrhagic shock (HS) is a shock result of hypovolemic injury, in which the innate immune response plays a central role in the pathophysiology ofthe severe complications and organ injury in surviving patients. During the development of HS, innate immunity acts as the first line of defense, mediating a rapid response to pathogens or danger signals through pattern recognition receptors. The early and exaggerated activation of innate immunity, which is widespread in patients with HS, results in systemic inflammation, cytokine storm, and excessive activation of complement factors and innate immune cells, comprised of type II innate lymphoid cells, CD4+ T cells, natural killer cells, eosinophils, basophils, macrophages, neutrophils, and dendritic cells. Recently, compelling evidence focusing on the innate immune regulation in preclinical and clinical studies promises new treatment avenues to reverse or minimize HS-induced tissue injury, organ dysfunction, and ultimately mortality. In this review, we first discuss the innate immune response involved in HS injury, and then systematically detail the cutting-edge therapeutic strategies in the past decade regarding the innate immune regulation in this field; these strategies include the use of mesenchymal stem cells, exosomes, genetic approaches, antibody therapy, small molecule inhibitors, natural medicine, mesenteric lymph drainage, vagus nerve stimulation, hormones, glycoproteins, and others. We also reviewed the available clinical studies on immune regulation for treating HS and assessed the potential of immune regulation concerning a translation from basic research to clinical practice. Combining therapeutic strategies with an improved understanding of how the innate immune system responds to HS could help to identify and develop targeted therapeutic modalities that mitigate severe organ dysfunction, improve patient outcomes, and reduce mortality due to HS injury.
Collapse
Affiliation(s)
- Qingxia Huang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Song Gao
- Jilin Xiuzheng Pharmaceutical New Drug Development Co., Ltd., Changchun, China
| | - Yao Yao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yisa Wang
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jinjin Chen
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chen guo
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Daqing Zhao, ; Xiangyan Li,
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Daqing Zhao, ; Xiangyan Li,
| |
Collapse
|
9
|
Costantini TW, Coimbra R, Weaver JL, Eliceiri BP. Precision targeting of the vagal anti-inflammatory pathway attenuates the systemic inflammatory response to burn injury. J Trauma Acute Care Surg 2022; 92:323-329. [PMID: 34789702 PMCID: PMC8792272 DOI: 10.1097/ta.0000000000003470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND The systemic inflammatory response (SIRS) drives late morbidity and mortality after injury. The α7 nicotinic acetylcholine receptor (α7nAchR) expressed on immune cells regulates the vagal anti-inflammatory pathway that prevents an overwhelming SIRS response to injury. Nonspecific pharmacologic stimulation of the vagus nerve has been evaluated as a potential therapeutic to limit SIRS. Unfortunately, the results of clinical trials have been underwhelming. We hypothesized that directly targeting the α7nAchR would more precisely stimulate the vagal anti-inflammatory pathway on immune cells and decrease gut and lung injury after severe burn. METHODS C57BL/6 mice underwent 30% total body surface area steam burn. Mice were treated with an intraperitoneal injection of a selective agonist of the α7nAchR (AR-R17779) at 30 minutes postburn. Intestinal permeability to 4 kDa FITC-dextran was measured at multiple time points postinjury. Lung vascular permeability was measured 6 hours after burn injury. Serial behavioral assessments were performed to quantify activity levels. RESULTS Intestinal permeability peaked at 6 hours postburn. AR-R17779 decreased burn-induced intestinal permeability in a dose-dependent fashion (p < 0.001). There was no difference in gut permeability to 4 kDa FITC-dextran between sham and burn-injured animals treated with 5 mg/kg of AR-R17779. While burn injury increased lung permeability 10-fold, AR-R17779 prevented burn-induced lung permeability with no difference compared with sham (p < 0.01). Postinjury activity levels were significantly improved in burned animals treated with AR-R17779. CONCLUSION Directly stimulating the α7nAchR prevents burn-induced gut and lung injury. Directly targeting the α7nAChR that mediates the cholinergic anti-inflammatory response may be an improved strategy compared with nonspecific vagal agonists.
Collapse
Affiliation(s)
- Todd W. Costantini
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| | - Raul Coimbra
- Comparative Effectiveness and Clinical Outcomes Research Center, Riverside University Health System, Loma Linda University School of Medicine, Riverside, CA
| | - Jessica L. Weaver
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| | - Brian P. Eliceiri
- Department of Surgery, Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, UC San Diego School of Medicine, San Diego, CA
| |
Collapse
|
10
|
Wu J, Yin Y, Qin M, Li K, Liu F, Zhou X, Song X, Li B. Vagus Nerve Stimulation Protects Enterocyte Glycocalyx After Hemorrhagic Shock Via the Cholinergic Anti-Inflammatory Pathway. Shock 2021; 56:832-839. [PMID: 33927140 PMCID: PMC8519159 DOI: 10.1097/shk.0000000000001791] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/08/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Electrical vagal nerve stimulation is known to decrease gut permeability and alleviate gut injury caused by traumatic hemorrhagic shock. However, the specific mechanism of action remains unclear. Glycocalyx, located on the surface of the intestinal epithelium, is associated with the buildup of the intestinal barrier. Therefore, the goal of our study was to explore whether vagal nerve stimulation affects enterocyte glycocalyx, gut permeability, gut injury, and remote lung injury. MATERIALS AND METHODS Male Sprague Dawley rats were anesthetized and their cervical nerves were exposed. The rats underwent traumatic hemorrhagic shock (with maintenance of mean arterial pressure of 30-35 mmHg for 60 min) with fluid resuscitation. Vagal nerve stimulation was added to two cohorts of animals before fluid resuscitation, and one of them was injected with methyllycaconitine to block the cholinergic anti-inflammatory pathway. Intestinal epithelial glycocalyx was detected using immunofluorescence. Intestinal permeability, the degree of gut and lung injury, and inflammation factors were also assessed. RESULTS Vagal nerve stimulation alleviated the damage to the intestinal epithelial glycocalyx and decreased intestinal permeability by 43% compared with the shock/resuscitation phase (P < 0.05). Methyllycaconitine partly eliminated the effects of vagal nerve stimulation on the intestinal epithelial glycocalyx (P < 0.05). Vagal nerve stimulation protected against traumatic hemorrhagic shock/fluid resuscitation-induced gut and lung injury, and some inflammatory factor levels in the gut and lung tissue were downregulated after vagal nerve stimulation (P < 0.05). CONCLUSIONS Vagal nerve stimulation could relieve traumatic hemorrhagic shock/fluid resuscitation-induced intestinal epithelial glycocalyx damage via the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Juan Wu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yushuang Yin
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Mingzhe Qin
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Kun Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Fang Liu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Xiang Zhou
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Xiaoyang Song
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Bixi Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Nakatsutsumi K, Morishita K, Yagi M, Doki S, Watanabe A, Ikegami N, Kobayashi T, Kojima M, Senda A, Yamamoto K, Aiboshi J, Coimbra R, Otomo Y. Vagus nerve stimulation modulates arachidonic acid production in the mesenteric lymph following intestinal ischemia-reperfusion injury. J Trauma Acute Care Surg 2021; 91:700-707. [PMID: 34238858 DOI: 10.1097/ta.0000000000003345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Inflammatory lipid mediators in mesenteric lymph (ML), including arachidonic acid (AA), are considered to play an important role in the pathogenesis of multiple-organ dysfunction after hemorrhagic shock. A previous study suggested that vagus nerve stimulation (VNS) could relieve shock-induced gut injury and abrogate ML toxicity, resulting in the prevention of multiple-organ dysfunction. However, the detailed mechanism of VNS in lymph toxicity remains unclear. The study aimed to investigate the relationship between VNS and inflammatory lipid mediators in ML. METHODS Male Sprague-Dawley rats underwent laparotomy and superior mesenteric artery obstruction (SMAO) for 60 minutes to induce intestinal ischemia followed by reperfusion and observation. The ML duct was cannulated, and ML samples were obtained both before and after SMAO. The distal ileum was removed at the end of the observation period. In one group of animals, VNS was performed from 10 minutes before 10 minutes after SMAO (5 V, 0.5 Hz). Liquid chromatography-electrospray ionization-tandem mass spectrometry analysis of AA was performed for each ML sample. The biological activity of ML was examined using a monocyte nuclear factor κ-light-chain-enhancer of activated B cells activation assay. Western blotting of phospholipase A2 group IIA (PLA2-IIA) was also performed for ML and ileum samples. RESULTS Vagus nerve stimulation relieved the SMAO-induced histological gut injury. The concentration of AA and level of nuclear factor κ-light-chain-enhancer of activated B cells activation in ML increased significantly after SMAO, whereas VNS prevented these responses. Western blotting showed PLA2-IIA expression in the ML and ileum after SMAO; however, the appearance of PLA2-IIA band was remarkably decreased in the samples from VNS-treated animals. CONCLUSION The results suggested that VNS could relieve gut injury induced by SMAO and decrease the production of AA in ML by altering PLA2-IIA expression in the gut and ML.
Collapse
Affiliation(s)
- Keita Nakatsutsumi
- From the Department of Acute Critical Care and Disaster Medicine (K.N., Y.O.), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University; Department of Acute Critical Care and Disaster Medicine (K.N., K.M., A.S., J.A., Y.O.), Tokyo Medical and Dental University Hospital of Medicine; Department of Biological Sciences (S.D., A.W., N.I., T.K.), Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo; Emergency Medicine and Acute Care Surgery (M.Y.), Matsudo City General Hospital, Chiba; Emergency and Critical Care Center (M.K.), Tokyo Women's Medical University Medical Center East; Department of Comprehensive Pathology (K.Y.), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; and Department of Surgery (R.C.), Riverside University Health System Medical Center, Loma Linda University School of Medicine, Loma Linda, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Stewart RH. A Modern View of the Interstitial Space in Health and Disease. Front Vet Sci 2020; 7:609583. [PMID: 33251275 PMCID: PMC7674635 DOI: 10.3389/fvets.2020.609583] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Increases in the volume of the interstitial space are readily recognized clinically as interstitial edema formation in the loose connective tissue of skin, mucosa, and lung. However, the contents and the hydrostatic pressure of this interstitial fluid can be very difficult to determine even in experimental settings. These difficulties have long obscured what we are beginning to appreciate is a dynamic milieu that is subject to both intrinsic and extrinsic regulation. This review examines current concepts regarding regulation of interstitial volume, pressure, and flow and utilizes that background to address three major topics of interest that impact IV fluid administration. The first of these started with the discovery that excess dietary salt can be stored non-osmotically in the interstitial space with minimal impact on vascular volume and pressures. This led to the hypothesis that, along with the kidney, the interstitial space plays an active role in the long-term regulation of blood pressure. Second, it now appears that hypovolemic shock leads to systemic inflammatory response syndrome principally through the entry of digestive enzymes into the intestinal interstitial space and the subsequent progression of enzymes and inflammatory agents through the mesenteric lymphatic system to the general circulation. Lastly, current evidence strongly supports the non-intuitive view that the primary factor leading to inflammatory edema formation is a decrease in interstitial hydrostatic pressure that dramatically increases microvascular filtration.
Collapse
Affiliation(s)
- Randolph H Stewart
- Department of Veterinary Physiology and Pharmacology, Michael E. DeBakey Institute, Texas A&M University, College Station, TX, United States
| |
Collapse
|
13
|
Kaniusas E, Szeles JC, Kampusch S, Alfageme-Lopez N, Yucuma-Conde D, Li X, Mayol J, Neumayer C, Papa M, Panetsos F. Non-invasive Auricular Vagus Nerve Stimulation as a Potential Treatment for Covid19-Originated Acute Respiratory Distress Syndrome. Front Physiol 2020; 11:890. [PMID: 32848845 PMCID: PMC7399203 DOI: 10.3389/fphys.2020.00890] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/30/2020] [Indexed: 01/08/2023] Open
Abstract
Background: Covid-19 is an infectious disease caused by an invasion of the alveolar epithelial cells by coronavirus 19. The most severe outcome of the disease is the Acute Respiratory Distress Syndrome (ARDS) combined with hypoxemia and cardiovascular damage. ARDS and co-morbidities are associated with inflammatory cytokine storms, sympathetic hyperactivity, and respiratory dysfunction. Hypothesis: In the present paper, we present and justify a novel potential treatment for Covid19-originated ARDS and associated co-morbidities, based on the non-invasive stimulation of the auricular branch of the vagus nerve. Methods: Auricular vagus nerve stimulation activates the parasympathetic system including anti-inflammatory pathways (the cholinergic anti-inflammatory pathway and the hypothalamic pituitary adrenal axis) while regulating the abnormal sympatho-vagal balance and improving respiratory control. Results: Along the paper (1) we expose the role of the parasympathetic system and the vagus nerve in the control of inflammatory processes (2) we formulate our physiological and methodological hypotheses (3) we provide a large body of clinical and preclinical data that support the favorable effects of auricular vagus nerve stimulation in inflammation, sympatho-vagal balance as well as in respiratory and cardiac ailments, and (4) we list the (few) possible collateral effects of the treatment. Finally, we discuss auricular vagus nerve stimulation protective potential, especially in the elderly and co-morbid population with already reduced parasympathetic response. Conclusions: Auricular vagus nerve stimulation is a safe clinical procedure and it could be either an effective treatment for ARDS originated by Covid-19 and similar viruses or a supplementary treatment to actual ARDS therapeutic approaches.
Collapse
Affiliation(s)
- Eugenijus Kaniusas
- Faculty of Electrical Engineering and Information Technology, Institute of Electrodynamics, Microwave and Circuit Engineering, Vienna University of Technology, Vienna, Austria
- SzeleSTIM GmbH, Vienna, Austria
| | - Jozsef C. Szeles
- General Hospital of the City of Vienna, Vienna, Austria
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Nuria Alfageme-Lopez
- Faculty of Biology and Faculty of Optics, Complutense University of Madrid, Madrid, Spain
| | - Daniela Yucuma-Conde
- Department of Clinical Epidemiology and Biostatistics, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Xie Li
- The Pediatric Department, Women and Children's Hospital of Hunan, Changsha, China
| | - Julio Mayol
- San Carlos Clinical Hospital, Madrid, Spain
- Institute for Health Research, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
- Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Christoph Neumayer
- General Hospital of the City of Vienna, Vienna, Austria
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Michele Papa
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fivos Panetsos
- Faculty of Biology and Faculty of Optics, Complutense University of Madrid, Madrid, Spain
- Institute for Health Research, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| |
Collapse
|
14
|
Powell K, Shah K, Hao C, Wu YC, John A, Narayan RK, Li C. Neuromodulation as a new avenue for resuscitation in hemorrhagic shock. Bioelectron Med 2019; 5:17. [PMID: 32232106 PMCID: PMC7098257 DOI: 10.1186/s42234-019-0033-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hemorrhagic shock (HS), a major cause of early death from trauma, accounts for around 40% of mortality, with 33–56% of these deaths occurring before the patient reaches a medical facility. Intravenous fluid therapy and blood transfusions are the cornerstone of treating HS. However, these options may not be available soon after the injury, resulting in death or a poorer quality of survival. Therefore, new strategies are needed to manage HS patients before they can receive definitive care. Recently, various forms of neuromodulation have been investigated as possible supplementary treatments for HS in the prehospital phase of care. Here, we provide an overview of neuromodulation methods that show promise to treat HS, such as vagus nerve stimulation, electroacupuncture, trigeminal nerve stimulation, and phrenic nerve stimulation and outline their possible mechanisms in the treatment of HS. Although all of these approaches are only validated in the preclinical models of HS and are yet to be translated to clinical settings, they clearly represent a paradigm shift in the way that this deadly condition is managed in the future.
Collapse
Affiliation(s)
- Keren Powell
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA
| | - Kevin Shah
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Caleb Hao
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA
| | - Yi-Chen Wu
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA
| | - Aashish John
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA
| | - Raj K Narayan
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA
| | - Chunyan Li
- Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY USA.,Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA.,Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030 USA
| |
Collapse
|
15
|
Abstract
In this review, we provide an overview of the US Food and Drug Administration (FDA)-approved clinical uses of vagus nerve stimulation (VNS) as well as information about the ongoing studies and preclinical research to expand the use of VNS to additional applications. VNS is currently FDA approved for therapeutic use in patients aged >12 years with drug-resistant epilepsy and depression. Recent studies of VNS in in vivo systems have shown that it has anti-inflammatory properties which has led to more preclinical research aimed at expanding VNS treatment across a wider range of inflammatory disorders. Although the signaling pathway and mechanism by which VNS affects inflammation remain unknown, VNS has shown promising results in treating chronic inflammatory disorders such as sepsis, lung injury, rheumatoid arthritis (RA), and diabetes. It is also being used to control pain in fibromyalgia and migraines. This new preclinical research shows that VNS bears the promise of being applied to a wider range of therapeutic applications.
Collapse
Affiliation(s)
- Rhaya L Johnson
- Lawrence D Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Christopher G Wilson
- Lawrence D Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA.,Department of Pediatrics, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
16
|
Vagus nerve stimulation in pregnant rats and effects on inflammatory markers in the brainstem of neonates. Pediatr Res 2018; 83:514-519. [PMID: 29053705 PMCID: PMC5866172 DOI: 10.1038/pr.2017.265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/25/2017] [Indexed: 01/29/2023]
Abstract
BackgroundVagus nerve stimulation (VNS) is an Food and Drug Administration-approved method delivering electrical impulses for treatment of depression and epilepsy in adults. The vagus nerve innervates the majority of visceral organs and cervix, but potential impacts of VNS on the progress of pregnancy and the fetus are not well studied.MethodsWe tested the hypothesis that VNS in pregnant dams does not induce inflammatory changes in the cardio-respiratory control regions of the pups' brainstem, potentially impacting the morbidity and mortality of offspring. Pregnant dams were implanted with stimulators providing intermittent low or high frequency electrical stimulation of the sub-diaphragmatic esophageal segment of the vagus nerve for 6-7 days until delivery. After birth, we collected pup brainstems that included cardio-respiratory control regions and counted the cells labeled for pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, tumor necrosis factor-α) and high mobility group box 1.ResultsNeither pup viability nor number of cells labeled for pro-inflammatory cytokines in nucleus tractus solitarii or hypoglossal motor nucleus was impaired by VNS. We provide evidence suggesting that chronic VNS of pregnant mothers does not impede the progress or outcome of pregnancy.ConclusionVNS does not cause preterm birth, affect well-being of progeny, or impact central inflammatory processes that are critical for normal cardiovascular and respiratory function in newborns.
Collapse
|
17
|
Song X, Chen G, Luo X, You G, Yin Y, Wang Y, Zhao L, Zhou H. Early resuscitation with exendin-4 alleviates acute lung injury after hemorrhagic shock in rats. J Surg Res 2017; 216:73-79. [PMID: 28807216 DOI: 10.1016/j.jss.2017.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/16/2017] [Accepted: 04/11/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Oxidative stress induced by hemorrhagic shock (HS) is known to initiate a systemic inflammatory response, which leads to subsequent acute lung injury. This study is aimed to assess the efficacy of exendin-4 (Ex-4) in attenuating lung injury in a rat model of HS and resuscitation (HS/R). METHODS HS was induced in sodium pentobarbital-anesthetized adult male Wistar rats by withdrawing blood to maintain a mean arterial pressure of 30-35 mm Hg for 50 min. Then, the animals received Ex-4 (5 μg/kg) or vehicle (saline) intravenously and were resuscitated with a volume of normal saline 1.5 times that of the shed blood volume. Mean arterial pressure was measured throughout the experiment, and acid-base status, oxidative stress, inflammation, and lung injury were evaluated at 2 h after resuscitation. RESULTS Ex-4 infusion reduced the methemoglobin content, the malondialdehyde content, the myeloperoxidase activity, and the expression of tumor necrosis factor-α and interleukin-6 in the lungs. The histologic injury was also markedly decreased in the Ex-4 group compared with the vehicle group. CONCLUSIONS Ex-4 ameliorates the oxidative stress, inflammatory response, and subsequent acute lung injury occurring after HS/R. Although future studies are required to elucidate the underlying mechanism, our results indicate that Ex-4 infusion may be a promising strategy for improving lung injury in the treatment of HS.
Collapse
Affiliation(s)
- Xiang Song
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Gan Chen
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Xin Luo
- Department of Blood Transfusion, 163 Hospital of PLA, Changsha, P.R. China
| | - Guoxing You
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Yujing Yin
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Ying Wang
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Lian Zhao
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China.
| | - Hong Zhou
- Department of Blood Products and Substitutes, Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing, P.R. China.
| |
Collapse
|
18
|
Langness S, Kojima M, Coimbra R, Eliceiri BP, Costantini TW. Enteric glia cells are critical to limiting the intestinal inflammatory response after injury. Am J Physiol Gastrointest Liver Physiol 2017; 312:G274-G282. [PMID: 28082286 DOI: 10.1152/ajpgi.00371.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
Abstract
Vagal nerve stimulation (VNS) has been shown to limit intestinal inflammation following injury; however, a direct connection between vagal terminals and resident intestinal immune cells has yet to be identified. We have previously shown that enteric glia cell (EGC) expression is increased after injury through a vagal-mediated pathway to help restore gut barrier function. We hypothesize that EGCs modulate immune cell recruitment following injury and relay vagal anti-inflammatory signals to resident immune cells in the gut. EGCs were selectively ablated from an isolated segment of distal bowel with topical application of benzalkonium chloride (BAC) in male mice. Three days following BAC application, mice were subjected to an ischemia-reperfusion injury (I/R) by superior mesenteric artery occlusion for 30 min. VNS was performed in a separate cohort of animals. EGC+ and EGC- segments were compared utilizing histology, flow cytometry, immunohistochemistry, and intestinal permeability. VNS significantly reduced immune cell recruitment after I/R injury in EGC+ segments with cell percentages similar to sham. VNS failed to limit immune cell recruitment in EGC- segments. Histologic evidence of gut injury was diminished with VNS application in EGC+ segments, whereas EGC- segments showed features of more severe injury. Intestinal permeability increased following I/R injury in both EGC+ and EGC- segments. Permeability was significantly lower after VNS application compared with injury alone in EGC+ segments only (95.1 ± 30.0 vs. 217.6 ± 21.7 μg/ml, P < 0.05). Therefore, EGC ablation uncouples the protective effects of VNS, suggesting that vagal-mediated signals are translated to effector cells through EGCs.NEW & NOTEWORTHY Intestinal inflammation is initiated by local immune cell activation and epithelial barrier breakdown, resulting in the production of proinflammatory mediators with subsequent leukocyte recruitment. Vagal nerve stimulation (VNS) has been shown to limit intestinal inflammation following injury; however, direct connection between vagal terminals and resident intestinal immune cells has yet to be identified. Here, we demonstrate that intact enteric glia cells are required to transmit the gut anti-inflammatory effects of VNS.
Collapse
Affiliation(s)
- Simone Langness
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Mitsuaki Kojima
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| |
Collapse
|
19
|
Ren C, Tong YL, Li JC, Lu ZQ, Yao YM. The Protective Effect of Alpha 7 Nicotinic Acetylcholine Receptor Activation on Critical Illness and Its Mechanism. Int J Biol Sci 2017; 13:46-56. [PMID: 28123345 PMCID: PMC5264260 DOI: 10.7150/ijbs.16404] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022] Open
Abstract
Critical illnesses and injuries are recognized as major threats to human health, and they are usually accompanied by uncontrolled inflammation and dysfunction of immune response. The alpha 7 nicotinic acetylcholine receptor (α7nAchR), which is a primary receptor of cholinergic anti-inflammatory pathway (CAP), exhibits great benefits for critical ill conditions. It is composed of 5 identical α7 subunits that form a central pore with high permeability for calcium. This putative structure is closely associated with its functional states. Activated α7nAChR exhibits extensive anti-inflammatory and immune modulatory reactions, including lowered pro-inflammatory cytokines levels, decreased expressions of chemokines as well as adhesion molecules, and altered differentiation and activation of immune cells, which are important in maintaining immune homeostasis. Well understanding of the effects and mechanisms of α7nAChR will be of great value in exploring effective targets for treating critical diseases.
Collapse
Affiliation(s)
- Chao Ren
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Ya-Lin Tong
- Department of Burns and Plastic Surgery, the 181st Hospital of Chinese PLA, Guilin 541002, People's Republic of China
| | - Jun-Cong Li
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Zhong-Qiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.; State Key Laboratory of Kidney Disease, the Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| |
Collapse
|
20
|
Langness S, Costantini TW, Morishita K, Eliceiri BP, Coimbra R. Modulating the Biologic Activity of Mesenteric Lymph after Traumatic Shock Decreases Systemic Inflammation and End Organ Injury. PLoS One 2016; 11:e0168322. [PMID: 27977787 PMCID: PMC5158049 DOI: 10.1371/journal.pone.0168322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 11/29/2016] [Indexed: 12/18/2022] Open
Abstract
Introduction Trauma/hemorrhagic shock (T/HS) causes the release of pro-inflammatory mediators into the mesenteric lymph (ML), triggering a systemic inflammatory response and acute lung injury (ALI). Direct and pharmacologic vagal nerve stimulation prevents gut barrier failure and alters the biologic activity of ML after injury. We hypothesize that treatment with a pharmacologic vagal agonist after T/HS would attenuate the biologic activity of ML and prevent ALI. Methods ML was collected from male Sprague-Dawley rats after T/HS, trauma-sham shock (T/SS) or T/HS with administration of the pharmacologic vagal agonist CPSI-121. ML samples from each experimental group were injected into naïve mice to assess biologic activity. Blood samples were analyzed for changes in STAT3 phosphorylation (pSTAT3). Lung injury was characterized by histology, permeability and immune cell recruitment. Results T/HS lymph injected in naïve mice caused a systemic inflammatory response characterized by hypotension and increased circulating monocyte pSTAT3 activity. Injection of T/HS lymph also resulted in ALI, confirmed by histology, lung permeability and increased recruitment of pulmonary macrophages and neutrophils to lung parenchyma. CPSI-121 attenuated T/HS lymph-induced systemic inflammatory response and ALI with stable hemodynamics and similar monocyte pSTAT3 levels, lung histology, lung permeability and lung immune cell recruitment compared to animals injected with lymph from T/SS. Conclusion Treatment with CPSI-121 after T/HS attenuated the biologic activity of the ML and decreased ALI. Given the superior clinical feasibility of utilizing a pharmacologic approach to vagal nerve stimulation, CPSI-121 is a potential treatment strategy to limit end organ dysfunction after injury.
Collapse
MESH Headings
- Acute Lung Injury/metabolism
- Acute Lung Injury/pathology
- Acute Lung Injury/prevention & control
- Animals
- Disease Models, Animal
- Hydrazones/therapeutic use
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Inflammation Mediators/metabolism
- Lymph/drug effects
- Lymph/immunology
- Lymph/metabolism
- Lymphatic Vessels/drug effects
- Lymphatic Vessels/metabolism
- Male
- Mesentery/drug effects
- Mesentery/immunology
- Mesentery/metabolism
- Mesentery/pathology
- Mice
- Mice, Inbred C57BL
- Rats
- Rats, Sprague-Dawley
- Shock, Hemorrhagic/complications
- Shock, Hemorrhagic/drug therapy
- Shock, Hemorrhagic/immunology
- Shock, Hemorrhagic/metabolism
- Shock, Traumatic/complications
- Shock, Traumatic/drug therapy
- Shock, Traumatic/immunology
- Shock, Traumatic/metabolism
Collapse
Affiliation(s)
- Simone Langness
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California, United States of America
| | - Todd W. Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California, United States of America
| | - Koji Morishita
- Division of Acute Critical Care and Disaster Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Brian P. Eliceiri
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California, United States of America
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
21
|
Abstract
Inflammation and immunity are regulated by neural reflexes. Recent basic science research has demonstrated that a neural reflex, termed the inflammatory reflex, modulates systemic and regional inflammation in a multiplicity of clinical conditions encountered in perioperative medicine and critical care. In this review, the authors describe the anatomic and physiologic basis of the inflammatory reflex and review the evidence implicating this pathway in the modulation of sepsis, ventilator-induced lung injury, postoperative cognitive dysfunction, myocardial ischemia-reperfusion injury, and traumatic hemorrhage. The authors conclude with a discussion of how these new insights might spawn novel therapeutic strategies for the treatment of inflammatory diseases in the context of perioperative and critical care medicine.
Collapse
|
22
|
Wang L, Zhao B, Chen Y, Ma L, Chen EZ, Mao EQ. Biliary tract external drainage protects against intestinal barrier injury in hemorrhagic shock rats. World J Gastroenterol 2015; 21:12800-12813. [PMID: 26668504 PMCID: PMC4671035 DOI: 10.3748/wjg.v21.i45.12800] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/20/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effects of biliary tract external drainage (BTED) on intestinal barrier injury in rats with hemorrhagic shock (HS). METHODS BTED was performed via cannula insertion into the bile duct of rats. HS was induced by drawing blood from the femoral artery at a rate of 1 mL/min until a mean arterial pressure (MAP) of 40 ± 5 mmHg was achieved. That MAP was maintained for 60 min. A total of 99 Sprague-Dawley rats were randomized into a sham group, an HS group and an HS + BTED group. Nine rats in the sham group were sacrificed 0.5 h after surgery. Nine rats in each of the HS and HS + BTED groups were sacrificed 0.5 h, 1 h, 2 h, 4 h and 6 h after resuscitation. Plasma tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and lipopolysaccharide (LPS) levels were analyzed using enzyme-linked immunosorbent assay. Plasma D-lactate levels were analyzed using colorimetry. The expression levels of occludin and claudin-1 in the ileum were analyzed using Western blot and immunohistochemistry. Histology of the ileum was evaluated by hematoxylin and eosin staining. RESULTS Plasma TNF-α levels in the HS + BTED group decreased significantly compared with the HS group at 1 h and 6 h after resuscitation (P < 0.05). Plasma IL-6 levels in the HS + BTED group decreased significantly compared with the HS group at 0.5 h, 1 h and 2 h after resuscitation (P < 0.05). Plasma D-lactate and LPS levels in the HS + BTED group decreased significantly compared with the HS group at 6 h after resuscitation (P < 0.05). The expression levels of occludin in the HS + BTED group increased significantly compared with the HS group at 4 h and 6 h after resuscitation (P < 0.05). The expression levels of claudin-1 in the HS + BTED group increased significantly compared with the HS group at 6 h after resuscitation (P < 0.05). Phenomena of putrescence and desquamation of epithelial cells in the ileal mucosa were attenuated in the HS + BTED group. Ileal histopathologic scores in the HS + BTED group decreased significantly compared with the HS group at 2 h, 4 h and 6 h after resuscitation (P < 0.05). CONCLUSION BTED protects against intestinal barrier injury in HS rats.
Collapse
|
23
|
Morishita K, Coimbra R, Langness S, Eliceiri BP, Costantini TW. Neuroenteric axis modulates the balance of regulatory T cells and T-helper 17 cells in the mesenteric lymph node following trauma/hemorrhagic shock. Am J Physiol Gastrointest Liver Physiol 2015; 309:G202-8. [PMID: 26045612 DOI: 10.1152/ajpgi.00097.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/27/2015] [Indexed: 01/31/2023]
Abstract
CD103(+) dendritic cells (DCs) continuously migrate from the intestine to the mesenteric lymph nodes (MLNs) and maintain tolerance by driving the development of regulatory T cells (Treg) in the gut. The relative expression of Treg and T-helper 17 (Th17) cells determines the balance between tolerance and immunity in the gut. We hypothesized that trauma/hemorrhagic shock (T/HS) would decrease the CD103(+) DC population in the mesenteric lymph and alter the Treg-to-Th17 ratio in the MLN. We further hypothesized that vagus nerve stimulation (VNS) would promote tolerance to inflammation by increasing the Treg-to-Th17 ratio in the MLN after injury. Male rats were assigned to sham shock (SS), trauma/sham shock (T/SS), or T/HS. T/HS was induced by laparotomy and 60 min of HS (blood pressure 35 mmHg) followed by fluid resuscitation. A separate cohort of animals underwent cervical VNS after the HS phase. MLN samples were collected 24 h after resuscitation. The CD103(+) DC population and Treg-to-Th17 cell ratio in the MLN were decreased after T/HS compared with SS and T/SS, suggesting a shift to an inflammatory response. VNS prevented the T/HS-induced decrease in the CD103(+) DC population and increased the Treg-to-Th17 ratio compared with T/HS alone. VNS alters the gut inflammatory response to injury by modulating the Treg-Th17 cell balance in the MLN. VNS promotes tolerance to inflammation in the gut, further supporting its ability to modulate the inflammatory set point and alter the response to injury.
Collapse
Affiliation(s)
- Koji Morishita
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Simone Langness
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| |
Collapse
|
24
|
Langness S, Coimbra R, Eliceiri BP, Costantini TW. Vagus Nerve Mediates the Neural Stem Cell Response to Intestinal Injury. J Am Coll Surg 2015. [PMID: 26209457 DOI: 10.1016/j.jamcollsurg.2015.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intestinal ischemia and reperfusion injury results in damage to elements critical to maintaining intestinal barrier function, including neurons and glia cells, which are part of the enteric nervous system (ENS). To limit inflammation, the ENS must be restored or replaced, yet the process by which this occurs is poorly understood. Multipotent progenitor cells called enteric nervous stem cells (ENSC) can differentiate into neurons or glia when stimulated. The ability of this cell population to respond to intestinal injury is unknown. In this study, we hypothesized that resolution of intestinal barrier injury would be associated with vagus nerve-mediated expansion of ENSCs. STUDY DESIGN Ischemia and reperfusion injury was reproduced in male mice by occluding the superior mesenteric artery for 30 minutes. Abdominal vagotomy was performed in a separate cohort to study the effects of the vagus nerve. Terminal ileum was harvested at various time points after reperfusion and analyzed with histology, flow cytometry, and immunohistochemistry. RESULTS Enteric nervous stem cell expansion occurs at 2, 4, and 8 hours after injury compared with sham (4.6% vs 2.1%; p < 0.001) and correlated with increased glial fibrillary acidic protein on immunohistochemistry. Vagotomy prevented both ENSC expansion and increased glial fibrillary acidic protein staining after injury. Intestinal permeability was restored to baseline by 48 hours after injury, but remained elevated in the vagotomy group compared with sham and injury alone at 48 hours (3.25 mg/mL vs 0.57 mg/mL and 0.26 mg/mL, respectively; p < 0.05). CONCLUSIONS Vagal-mediated expansion of ENSCs occurs after ischemia and reperfusion injury and results in improved kinetics of injury resolution.
Collapse
Affiliation(s)
- Simone Langness
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, CA
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, CA
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, CA
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, CA.
| |
Collapse
|
25
|
A pharmacologic approach to vagal nerve stimulation prevents mesenteric lymph toxicity after hemorrhagic shock. J Trauma Acute Care Surg 2015; 78:52-8; discussion 58-9. [PMID: 25539203 DOI: 10.1097/ta.0000000000000489] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Electrical stimulation of the vagus nerve (VN) prevents gut and lung inflammation and mesenteric lymph (ML) toxicity in animal models of injury. We have previously shown that treatment with CPSI-121, a guanylhydrazone-derived compound, prevents gut barrier failure after burn injury. While the structure of CPSI-121 predicts that it will activate parasympathetic signaling, its ability to stimulate the VN is unknown. The aims of this study were to (1) measure the ability of CPSI-121 to induce VN activity, (2) determine whether CPSI-121 causes significant hemodynamic effects, and (3) further define the potential for CPSI-121 to limit the systemic inflammatory response to injury. METHODS Male Sprague-Dawley rats were given 1-mg/kg CPSI-121 intravenously while blood pressure, heart rate, and efferent VN electrical activity were recorded. Rats were also assigned to sham or trauma/hemorrhagic shock (T/HS). T/HS was induced by laparotomy and 60 minutes of HS (mean arterial pressure, 35 mm Hg) followed by fluid resuscitation. A separate cohort of animals received CPSI-121 after the HS phase. Gut and lung tissues were harvested for histologic analysis. Lung wet-dry ratios were also evaluated. The ability of ML to prime neutrophils was assessed by measuring in vitro oxidative burst using flow cytometry. RESULTS Blood pressure was not altered after treatment with CPSI-121, while heart rate decreased only slightly. Recording of efferent VN electrical activity revealed an increase in discharge rate after administration of CPSI-121. T/HS caused gut and lung injury, which were prevented in animals treated with CPSI-121 (p < 0.05). Treatment with CPSI-121 following T/HS attenuated neutrophil priming after exposure to ML (p < 0.05). CONCLUSION CPSI-121 causes efferent VN output and limits shock-induced gut and lung injury as well as ML toxicity. CPSI-121 is a candidate pharmacologic approach to VN stimulation aimed at limiting the inflammatory response in patients following T/HS.
Collapse
|
26
|
Lowry DM, Morishita K, Eliceiri BP, Bansal V, Coimbra R, Costantini TW. The vagus nerve alters the pulmonary dendritic cell response to injury. J Surg Res 2014; 192:12-8. [PMID: 25005822 DOI: 10.1016/j.jss.2014.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/29/2014] [Accepted: 06/04/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND We have shown previously that vagal nerve stimulation (VNS) protects against burn-induced acute lung injury (ALI). Although the mobilization and activation of immune cells is central to tissue injury caused by the systemic inflammatory response, the specific inflammatory cell populations that are modulated by VNS have yet to be fully defined. The purpose of this study was to assess whether VNS alters inflammatory cell recruitment to the lung after severe burn injury. MATERIALS AND METHODS Male C57BL/6 mice were subjected to 30% total body surface area steam burn with and without electrical stimulation of the right cervical vagus nerve. The relative levels of pulmonary dendritic cells (DC) and macrophages were compared at 4 h versus 24 h after burn injury. Lung tissue injury was characterized by histology to assess changes in lung architecture, and measure the protein levels of interleukin 6 and transforming growth factor-β1. RESULTS Severe burn caused an increase in pulmonary DC recruitment at 4 h after injury that persisted at 24 h after severe burn, whereas there was no change in the number of pulmonary macrophages. In contrast, VNS limited the burn-induced recruitment of pulmonary DC. VNS prevented histologic lung injury and attenuated the release of interleukin 6 and transforming growth factor-β1 in the lung after burn injury. CONCLUSIONS VNS is an effective method to limit pulmonary DC recruitment to the lung and prevent ALI after burn injury. Identifying strategies to limit inflammatory cell recruitment to the lung may have clinical utility in preventing ALI in severely burned patients.
Collapse
Affiliation(s)
- Debra M Lowry
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Koji Morishita
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Vishal Bansal
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego Health Sciences, San Diego, California.
| |
Collapse
|
27
|
Vagal nerve stimulation modulates the dendritic cell profile in posthemorrhagic shock mesenteric lymph. J Trauma Acute Care Surg 2014; 76:610-7; discussion 617-8. [PMID: 24553526 DOI: 10.1097/ta.0000000000000137] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Previous studies have established that posthemorrhagic shock mesenteric lymph (PHSML) contains proinflammatory mediators, while the cellular basis of PHSML is less well characterized in acute models of injury. CD103 dendritic cells (DCs) have been identified in the mesenteric lymph (ML) in models of chronic intestinal inflammation, suggesting an important role in the gut response to injury. We have previously demonstrated the ability of vagal nerve stimulation (VNS) to prevent gut barrier failure after trauma/hemorrhagic shock (T/HS); however, the ability of VNS to alter ML DCs is unknown. We hypothesized that the CD103 MHC-II DC population would change in PHSML and that VNS would prevent injury-induced changes in this population in PHSML. METHODS Male Sprague-Dawley rats were randomly assigned to trauma/sham shock or T/HS. T/HS was induced by midline laparotomy and 60 minutes of HS (blood pressure, 35 mm Hg), followed by fluid resuscitation. A separate cohort of animals underwent cervical VNS after the HS phase. Gut tissue was harvested at 2 hours after injury for histologic analysis. ML was collected during the pre-HS, HS, and post-HS phase. For flow cytometric analysis, ML cells were subjected to staining with CD103 and MHC-II antibodies, and this cell population was compared in the pre-HS and post-HS phase from the same animal. The CD4Foxp3 cell (T reg) population in the ML node (MLN) was also tested to determine effects of CD103 DC modulation in the ML. RESULTS VNS reduced histologic gut injury and ML flow seen after injury. The CD103 MHC-II DC population in the PHSML was significantly decreased compared with pre-HS and was associated with decreased T reg expression in the MLN. VNS prevented the injury-induced decrease in the CD103 MHC-II+ DC population in the ML and restored the T reg population in the MLN. CONCLUSION These findings suggest that VNS mediates the inflammatory responses in ML DCs and MLN T reg cells by affecting the set point of T/HS responsiveness.
Collapse
|
28
|
Short SS, Wang J, Castle SL, Fernandez GE, Smiley N, Zobel M, Pontarelli EM, Papillon SC, Grishin AV, Ford HR. Low doses of celecoxib attenuate gut barrier failure during experimental peritonitis. J Transl Med 2013; 93:1265-75. [PMID: 24126890 PMCID: PMC3966546 DOI: 10.1038/labinvest.2013.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier becomes compromised during systemic inflammation, leading to the entry of luminal bacteria into the host and gut origin sepsis. Pathogenesis and treatment of inflammatory gut barrier failure is an important problem in critical care. In this study, we examined the role of cyclooxygenase-2 (COX-2), a key enzyme in the production of inflammatory prostanoids, in gut barrier failure during experimental peritonitis in mice. I.p. injection of LPS or cecal ligation and puncture (CLP) increased the levels of COX-2 and its product prostaglandin E2 (PGE2) in the ileal mucosa, caused pathologic sloughing of the intestinal epithelium, increased passage of FITC-dextran and bacterial translocation across the barrier, and increased internalization of the tight junction (TJ)-associated proteins junction-associated molecule-A and zonula occludens-1. Luminal instillation of PGE2 in an isolated ileal loop increased transepithelial passage of FITC-dextran. Low doses (0.5-1 mg/kg), but not a higher dose (5 mg/kg) of the specific COX-2 inhibitor Celecoxib partially ameliorated the inflammatory gut barrier failure. These results demonstrate that high levels of COX-2-derived PGE2 seen in the mucosa during peritonitis contribute to gut barrier failure, presumably by compromising TJs. Low doses of specific COX-2 inhibitors may blunt this effect while preserving the homeostatic function of COX-2-derived prostanoids. Low doses of COX-2 inhibitors may find use as an adjunct barrier-protecting therapy in critically ill patients.
Collapse
Affiliation(s)
- Scott S. Short
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Jin Wang
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Shannon L. Castle
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | | | - Nancy Smiley
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Michael Zobel
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Elizabeth M. Pontarelli
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Stephanie C. Papillon
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Anatoly V. Grishin
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| | - Henri R. Ford
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, CA,Department of Surgery, University of Southern California, Los Angeles, CA
| |
Collapse
|