1
|
Iaia N, Noviello C, Muscaritoli M, Costelli P. Inflammation in cancer cachexia: still the central tenet or just another player? Am J Physiol Cell Physiol 2025; 328:C1837-C1852. [PMID: 40250836 DOI: 10.1152/ajpcell.00808.2024] [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: 11/06/2024] [Revised: 11/23/2024] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
Cancer cachexia, a multifactorial syndrome characterized by body weight loss, muscle, and adipose tissue wasting, affects patients with cancer. Over time, the definition of cachexia has been modified, including inflammation as one of the main causal factors. Evidence has suggested that a range of proinflammatory mediators may be involved in the regulation of intracellular signaling, resulting in enhanced resting energy expenditure, metabolic changes, and muscle atrophy, all of which are typical features of cachexia. Physiologically speaking, however, inflammation is a response aimed at facing potentially damaging events. Along this line, its induction in the cancer hosts could be an attempt to restore the physiological homeostasis. Interesting observations have shown that cytokines such as interleukins 4 and 6 could improve muscle wasting, supporting the view that the same mediator may exert pro- or anti-inflammatory activity depending on the immune cells involved as well as on the tissue metabolic demand. In conclusion, whether inflammation is crucial to the occurrence of cachexia or just one contributor among others, is still unclear. Indeed, while inflammation is a trigger of cachexia, the alterations of energy and protein metabolism and of the hormonal homeostasis occurring in cachexia likely act as inflammatory stimuli on their own. Whether the causative role prevails over the compensatory one likely depends on the tumor type and stage, patient lifestyle, the presence of comorbidities, and the response to anticancer treatments paving the way to a holistic, personalized approach to cancer cachexia.
Collapse
Affiliation(s)
- Noemi Iaia
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Chiara Noviello
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | | | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
2
|
Han C, Zhu X, Sokol CL. Neuroimmune Circuits in Allergic Diseases. Annu Rev Immunol 2025; 43:367-394. [PMID: 39977604 DOI: 10.1146/annurev-immunol-082423-032154] [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] [Indexed: 02/22/2025]
Abstract
Communication between the nervous and immune systems is evolutionarily conserved. From primitive eukaryotes to higher mammals, neuroimmune communication utilizes multiple complex and complementary mechanisms to trigger effective but balanced responses to environmental dangers such as allergens and tissue damage. These responses result from a tight integration of the nervous and immune systems, and accumulating evidence suggests that this bidirectional communication is crucial in modulating the initiation and development of allergic inflammation. In this review, we discuss the basic mechanisms of neuroimmune communication, with a focus on the recent advances underlying the importance of such communication in the allergic immune response. We examine neuronal sensing of allergens, how neuropeptides and neurotransmitters regulate allergic immune cell functions, and how inflammatory factors derived from immune cells coordinate complex peripheral and central nervous system responses. Furthermore, we highlight how fundamental aspects of host biology, from aging to circadian rhythm, might affect these pathways. Appreciating neuroimmune communications as an evolutionarily conserved and functionally integrated system that is fundamentally involved in type 2 immunity will provide new insights into allergic inflammation and reveal exciting opportunities for the management of acute and chronic allergic diseases.
Collapse
Affiliation(s)
- Cai Han
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Xueping Zhu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
3
|
Kayyal H, Cruciani F, Chandran SK, Edry E, Schif-Zuck S, Koren T, Yiannakas A, Rolls A, Ariel A, Rosenblum K. Retrieval of conditioned immune response in male mice is mediated by an anterior-posterior insula circuit. Nat Neurosci 2025; 28:589-601. [PMID: 39870921 DOI: 10.1038/s41593-024-01864-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/05/2024] [Indexed: 01/29/2025]
Abstract
To protect the body from infections, the brain has evolved the ability to coordinate behavioral and immunological responses. The conditioned immune response (CIR) is a form of Pavlovian conditioning wherein a sensory (for example, taste) stimulus, when paired with an immunomodulatory agent, evokes aversive behavior and an anticipatory immune response after re-experiencing the taste. Although taste and its valence are represented in the anterior insular cortex and immune response in the posterior insula and although the insula is pivotal for CIRs, the precise circuitry underlying CIRs remains unknown. Here, we demonstrated that a bidirectional circuit connecting the anterior and posterior (aIC-pIC) insula mediates the CIR in male mice. Retrieving the behavioral dimension of the association requires activity of aIC-to-pIC neurons, whereas modulating the anticipatory immunological dimension requires bidirectional projections. These results illuminate a mechanism by which experience shapes interactions between sensory internal representations and the immune system. Moreover, this newly described intrainsular circuit contributes to the preservation of brain-dependent immune homeostasis.
Collapse
Affiliation(s)
- Haneen Kayyal
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
| | - Federica Cruciani
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
| | | | - Efrat Edry
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| | - Sagie Schif-Zuck
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Departments of Human Biology, University of Haifa, Haifa, Israel
| | - Tamar Koren
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Adonis Yiannakas
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel
- European University of Cyprus Medical School, Frankfurt am Main, Germany
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amiram Ariel
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Departments of Human Biology, University of Haifa, Haifa, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel.
| |
Collapse
|
4
|
Huang Y, Dong S, Zhang Y, Zhang Y, Guo Y, Shi J, Li X, Liu S, Chen Y, Yu J. Electroacupuncture promotes resolution of inflammation by modulating SPMs via vagus nerve activation in LPS-induced ALI. Int Immunopharmacol 2025; 147:113941. [PMID: 39746272 DOI: 10.1016/j.intimp.2024.113941] [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: 09/04/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025]
Abstract
During the process of acute lung injury (ALI) associated with sepsis, the α7nAChR in the cholinergic anti-inflammatory pathway (CAP) plays a crucial role. However, the roles of electroacupuncture (EA) and specialized pro-resolving mediators (SPMs) in this context remain unclear. In this study, we demonstrated that EA activates CAP via α7nAChR, reducing lung permeability and inflammatory cytokine release. Our results highlighted lipoxin A4 (LXA4) as a crucial SPM in this process. EA was shown to enhance LXA4 synthesis and alleviate symptoms in patients with sepsis-related acute respiratory distress syndrome (ARDS). Studies using α7nAChR-deficient mice confirmed its essential role in LXA4 regulation. Macrophages in bronchoalveolar lavage fluid (BALF) were identified as key contributors to the protective effects of LXA4, further supported by experiments involving pulmonary macrophage depletion. In summary, we discovered a novel anti-inflammatory pathway where EA activates α7nAChR, leading to increased LXA4 production and lung protection.
Collapse
Affiliation(s)
- Yan Huang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Ye Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Yan Guo
- Department of Anesthesiology, The Heji Affiliated Hospital of Changzhi Medical College, Changzhi Medical College, Shanxi, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Shasha Liu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Yong Chen
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
| |
Collapse
|
5
|
Serhan CN, Levy BD. Proresolving Lipid Mediators in the Respiratory System. Annu Rev Physiol 2025; 87:491-512. [PMID: 39303274 PMCID: PMC11810588 DOI: 10.1146/annurev-physiol-020924-033209] [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] [Indexed: 09/22/2024]
Abstract
Lung inflammation, infection, and injury can lead to critical illness and death. The current means to pharmacologically treat excessive uncontrolled lung inflammation needs improvement because many treatments are or will become immunosuppressive. The inflammatory response evolved to protect the host from microbes, injury, and environmental insults. This response brings phagocytes from the bloodstream to the tissue site to phagocytize and neutralize bacterial invaders and enables airway antimicrobial functions. This physiologic response is ideally self-limited with initiation and resolution phases. Polyunsaturated essential fatty acids are precursors to potent molecules that govern both phases. In the initiation phase, arachidonic acid is converted to prostaglandins and leukotrienes that activate leukocytes to transmigrate from postcapillary venules. The omega-3 fatty acids (e.g., DHA and EPA) are precursors to resolvins, protectins, and maresins, which are families of chemically distinct mediators with potent functions in resolution of acute and chronic inflammation in the respiratory system.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
6
|
Hajivalili M, Nikkhoo N, Salahi S, Hosseini M. Traumatic brain Injury: Comprehensive overview from pathophysiology to Mesenchymal stem Cell-Based therapies. Int Immunopharmacol 2025; 146:113816. [PMID: 39708488 DOI: 10.1016/j.intimp.2024.113816] [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: 09/23/2024] [Revised: 11/16/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Traumatic brain injury (TBI) is a disastrous phenomenon which is considered to cause high mortality and morbidity rate. Regarding the importance of TBI due to its prevalence and its effects on the brain and other organs, finding new therapeutic methods and improvement of conventional therapies seems to be vital. TBI involves a complex physiological mechanism, with inflammation being a key component among various contributing factors. After incidence of TBI, inflammation can act as a double-edged sword in the process. Inflammation actually plays its role both as initiator and progressive index during TBI which can accumulate myeloid and lymphoid immune cells and trigger cell death pathways. Through this study we made this concept bold that that besides conventional therapies that could be used for traumatic brain injury, treatments based on mesenchyme stem cells (MSCs) and their derivatives including secretomes and exosomes demonstrate more efficacies particularly in preventing secondary injuries caused by TBI. Of note, we highlighted the valuable features of MSC-based therapies such as self-direction toward inflamed tissues and amplifying neuro-regenerative aspects. We listed possible challenges in the way of reaching this therapy to clinic to provide a clear and updated of the field.
Collapse
Affiliation(s)
- Mahsa Hajivalili
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Nikkhoo
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Serhan CN, Chiang N, Nshimiyimana R. Low-dose pro-resolving mediators temporally reset the resolution response to microbial inflammation. Mol Med 2024; 30:153. [PMID: 39294573 PMCID: PMC11411770 DOI: 10.1186/s10020-024-00877-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/15/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Specialized pro-resolving mediators (SPMs) promote resolution of inflammation, clear infections and stimulate tissue regeneration. These include resolvins, protectins, and maresins. During self-resolving acute inflammation, SPMs are produced and have key functions activating endogenous resolution response for returning to homeostasis. Herein, we addressed whether infections initiated with ongoing inflammation alter resolution programs, and if low-dose repetitive SPM regimen re-programs the resolution response. METHODS Inflammation was initiated with zymosan (1 mg/mouse) followed by E. coli (105 CFU/mouse) infections carried out in murine peritonitis, and exudates collected at 4-72 h. Leukocytes were enumerated using light microscopy, percentages of PMN, monocytes and macrophages were determined using flow cytometry, and resolution indices calculated. Lipid mediators and SPM profiles were established using mass spectrometry-based metabololipidomics. Repetitive dosing with a SPM panel consisting of RvD1, RvD2, RvD5, MaR1 and RvE2 (0.1 ng/mouse each, i.p.) was given to mice, followed by zymosan challenge. Leukocyte composition, resolution indices and RNA-sequencing were carried out for the repetitive SPM treatments. RESULTS E. coli infections initiated acute inflammation-resolution programs with temporal SPM production in the infectious exudates. Zymosan-induced inflammation prior to E. coli peritonitis shifted exudate resolution indices and delayed E. coli clearance. Lipid mediator metabololipidomics demonstrated that E. coli infection with ongoing zymosan-induced inflammation shifted the time course of exudate SPMs, activating a SPM cluster that included RvD1, RvD5 and MaR1 during the initiation phase of infectious inflammation (0-4 h); RvD5 and MaR1 were present also in the resolution phase (24-48 h). To emulate daily SPM regimens used in humans, a repetitive subthreshold dosing of the SPM panel RvD1, RvD2, RvD5, MaR1 and RvE2 each at 0.1 ng per mouse was administered. This low-dose SPM regimen accelerated exudate PMN clearance following zymosan-induced inflammation, and shortened the resolution interval by > 70%. These low-dose SPMs regulated genes and pathways related to immune response, chemokine clearance and tissue repair, as demonstrated by using RNA-sequencing. CONCLUSIONS Infections encountered during ongoing inflammation in mice reset the resolution mechanisms of inflammation via SPM clusters. Low-dose SPMs activate innate immune responses and pathways towards the resolution response that can be reprogrammed.
Collapse
Affiliation(s)
- Charles N Serhan
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA.
| | - Nan Chiang
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA
| | - Robert Nshimiyimana
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA
| |
Collapse
|
8
|
Huang Y, Dong S, Li X, Shi J, Zhang Y, Liu S, Zhang Y, Yu J. VNS-mediated α7nAChR signaling promotes SPM synthesis via regulation of netrin-1 expression during LPS-induced ALI. FASEB J 2024; 38:e9664. [PMID: 38038805 DOI: 10.1096/fj.202301623r] [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: 08/09/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023]
Abstract
The α7 nicotinic acetylcholine receptor (α7nAChR) plays a crucial role in the cholinergic anti-inflammatory pathway (CAP) during sepsis-associated acute lung injury (ALI). Increasing evidence suggests that specialized pro-resolving mediators (SPMs) are important in resolving α7nAChR-mediated ALI resolution. Our study aims to elucidate the pivotal role of α7nAChR in the CAP during LPS-associated acute lung injury (ALI). By employing vagus nerve stimulation (VNS), we identified α7nAChR as the key CAP subunit in ALI mice, effectively reducing lung permeability and the release of inflammatory cytokines. We further investigated the alterations in SPMs regulated by α7nAChR, revealing a predominant synthesis of lipoxin A4 (LXA4). The significance of α7nAChR-netrin-1 pathway in governing SPM synthesis was confirmed through the use of netrin-1 knockout mice and siRNA-transfected macrophages. Additionally, our evaluation identified a synchronous alteration of LXA4 synthesis in the α7nAChR-netrin-1 pathway accompanied by 5-lipoxygenase (5-LOX), thereby confirming an ameliorative effect of LXA4 on lung injury and macrophage inflammatory response. Concurrently, inhibiting the function of LXA4 annulled the lung-protective effect of VNS. As a result, our findings reveal a novel anti-inflammatory pathway wherein VNS modulates netrin-1 expression via α7nAChR, ultimately leading to LXA4 synthesis and subsequent lung protection.
Collapse
Affiliation(s)
- Yan Huang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shasha Liu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Ye Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
9
|
D’Haens G, Eberhardson M, Cabrijan Z, Danese S, van den Berg R, Löwenberg M, Fiorino G, Schuurman PR, Lind G, Almqvist P, Olofsson PS, Tracey KJ, Hanauer SB, Zitnik R, Chernoff D, Levine YA. Neuroimmune Modulation Through Vagus Nerve Stimulation Reduces Inflammatory Activity in Crohn's Disease Patients: A Prospective Open-label Study. J Crohns Colitis 2023; 17:1897-1909. [PMID: 37738465 PMCID: PMC10798868 DOI: 10.1093/ecco-jcc/jjad151] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] is a debilitating, inflammatory condition affecting the gastrointestinal tract. There is no cure and sustained clinical and endoscopic remission is achieved by fewer than half of patients with current therapies. The immunoregulatory function of the vagus nerve, the 'inflammatory reflex', has been established in patients with rheumatoid arthritis and biologic-naive CD. The aim of this study was to explore the safety and efficacy of vagus nerve stimulation in patients with treatment-refractory CD, in a 16-week, open-label, multicentre, clinical trial. METHODS A vagus nerve stimulator was implanted in 17 biologic drug-refractory patients with moderately to severely active CD. One patient exited the study pre-treatment, and 16 patients were treated with vagus nerve stimulation [4/16 receiving concomitant biologics] during 16 weeks of induction and 24 months of maintenance treatment. Endpoints included clinical improvement, patient-reported outcomes, objective measures of inflammation [endoscopic/molecular], and safety. RESULTS There was a statistically significant and clinically meaningful decrease in CD Activity Index at Week 16 [mean ± SD: -86.2 ± 92.8, p = 0.003], a significant decrease in faecal calprotectin [-2923 ± 4104, p = 0.015], a decrease in mucosal inflammation in 11/15 patients with paired endoscopies [-2.1 ± 1.7, p = 0.23], and a decrease in serum tumour necrosis factor and interferon-γ [46-52%]. Two quality-of-life indices improved in 7/11 patients treated without biologics. There was one study-related severe adverse event: a postoperative infection requiring device explantation. CONCLUSIONS Neuroimmune modulation via vagus nerve stimulation was generally safe and well tolerated, with a clinically meaningful reduction in clinical disease activity associated with endoscopic improvement, reduced levels of faecal calprotectin and serum cytokines, and improved quality of life.
Collapse
Affiliation(s)
- Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michael Eberhardson
- Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Zeljko Cabrijan
- Division of Gastroenterology, Hepatology and Clinical Nutrition, University Hospital Dubrava, Zagreb, Croatia
- Division of Gastroenterology, University of Applied Health Sciences, Zagreb, Croatia
- Josip Juraj Strossmayer University of Osijek School of Medicine, Osijek, Croatia
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Italy
- Department of Gastroenterology and Endoscopy, University Vita-Salute San Raffaele, Milano, Italy
| | - Remco van den Berg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gionata Fiorino
- Department of Gastroenterology and Digestive Endoscopy, VIta-Salute San Raffaele Hospital, Milan, Italy
- IBD Unit, Department of Gastroenterology and Digestive Endoscopy, San Camillo-Forlanini Hospital, Rome, Italy
| | | | - Göran Lind
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Per Almqvist
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
- Neurosurgery Stockholm AB, Stockholm, Sweden
| | - Peder S Olofsson
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Feinstein Institutes for Medical Research, Manhasset, New York
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, New York
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Stephen B Hanauer
- Division of Gastroenterology and Hepatology, Northwestern University–Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ralph Zitnik
- SetPoint Medical, Valencia, California, USA
- Valerio Consulting, Santa Barbara, California, USA
| | | | - Yaakov A Levine
- Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- SetPoint Medical, Valencia, California, USA
| |
Collapse
|
10
|
Salm DC, Horewicz VV, Tanaka F, Ferreira JK, de Oliveira BH, Maio JMB, Donatello NN, Ludtke DD, Mazzardo-Martins L, Dutra AR, Mack JM, de C H Kunzler D, Cargnin-Ferreira E, Salgado ASI, Bittencourt EB, Bianco G, Piovezan AP, Bobinski F, Moré AOO, Martins DF. Electrical Stimulation of the Auricular Branch Vagus Nerve Using Random and Alternating Frequencies Triggers a Rapid Onset and Pronounced Antihyperalgesia via Peripheral Annexin A1-Formyl Peptide Receptor 2/ALX Pathway in a Mouse Model of Persistent Inflammatory Pain. Mol Neurobiol 2023; 60:2889-2909. [PMID: 36745336 DOI: 10.1007/s12035-023-03237-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 01/13/2023] [Indexed: 02/07/2023]
Abstract
This study evaluated the antihyperalgesic and anti-inflammatory effects of percutaneous vagus nerve electrical stimulation (pVNS) by comparing the effects of alternating and random frequencies in an animal model of persistent inflammatory hyperalgesia. The model was induced by Freund's complete adjuvant (CFA) intraplantar (i.pl.) injection. Mice were treated with different protocols of time (10, 20, or 30 min), ear laterality (right, left or both), and frequency (alternating or random). Mechanical hyperalgesia was evaluated, and some groups received i.pl. WRW4 (FPR2/ALX antagonist) to determine the involvement. Edema, paw surface temperature, and spontaneous locomotor activity were evaluated. Interleukin-1β, IL-6, IL-10, and IL4 levels were verified by enzyme-linked immunosorbent assay. AnxA1, FPR2/ALX, neutrophil, M1 and M2 phenotype macrophage, and apoptotic cells markers were identified using western blotting. The antihyperalgesic effect pVNS with alternating and random frequency effect is depending on the type of frequency, time, and ear treated. The pVNS random frequency in the left ear for 10 min had a longer lasting antihyperalgesic effect, superior to classical stimulation using alternating frequency and the FPR2/ALX receptor was involved in this effect. There was a reduction in the levels of pro-inflammatory cytokines and an increase in the immunocontent of AnxA1 and CD86 in mice paw. pVNS with a random frequency in the left ear for 10 min showed to be optimal for inducing an antihyperalgesic effect. Thus, the random frequency was more effective than the alternating frequency. Therefore, pVNS may be an important adjunctive treatment for persistent inflammatory pain.
Collapse
Affiliation(s)
- Daiana C Salm
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Verônica V Horewicz
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Fernanda Tanaka
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Júlia K Ferreira
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Bruna H de Oliveira
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Julia Maria Batista Maio
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Nathalia N Donatello
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Daniela D Ludtke
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Leidiane Mazzardo-Martins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Aline R Dutra
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Josiel M Mack
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Deborah de C H Kunzler
- Department of Physiotherapy, State University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | | | | | - Gianluca Bianco
- Research Laboratory of Posturology and Neuromodulation RELPON, Department of Human Neuroscience, Sapienza University, Rome, Italy
- Istituto Di Formazione in Agopuntura E Neuromodulazione IFAN, Rome, Italy
| | - Anna Paula Piovezan
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Ari O O Moré
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
- Integrative Medicine and Acupuncture Division, University Hospital, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniel F Martins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil.
- Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil.
| |
Collapse
|
11
|
Hughes FM, Odom MR, Cervantes A, Livingston AJ, Purves JT. Why Are Some People with Lower Urinary Tract Symptoms (LUTS) Depressed? New Evidence That Peripheral Inflammation in the Bladder Causes Central Inflammation and Mood Disorders. Int J Mol Sci 2023; 24:2821. [PMID: 36769140 PMCID: PMC9917564 DOI: 10.3390/ijms24032821] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Anecdotal evidence has long suggested that patients with lower urinary tract symptoms (LUTS) develop mood disorders, such as depression and anxiety, at a higher rate than the general population and recent prospective studies have confirmed this link. Breakthroughs in our understanding of the diseases underlying LUTS have shown that many have a substantial inflammatory component and great strides have been made recently in our understanding of how this inflammation is triggered. Meanwhile, studies on mood disorders have found that many are associated with central neuroinflammation, most notably in the hippocampus. Excitingly, work on other diseases characterized by peripheral inflammation has shown that they can trigger central neuroinflammation and mood disorders. In this review, we discuss the current evidence tying LUTS to mood disorders, its possible bidirectionally, and inflammation as a common mechanism. We also review modern theories of inflammation and depression. Finally, we discuss exciting new animal studies that directly tie two bladder conditions characterized by extensive bladder inflammation (cyclophosphamide-induced hemorrhagic cystitis and bladder outlet obstruction) to neuroinflammation and depression. We conclude with a discussion of possible mechanisms by which peripheral inflammation is translated into central neuroinflammation with the resulting psychiatric concerns.
Collapse
Affiliation(s)
- Francis M. Hughes
- Department Urology, Duke University Medical Center, P.O. Box 3831, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
12
|
Ji RR. Specialized Pro-Resolving Mediators as Resolution Pharmacology for the Control of Pain and Itch. Annu Rev Pharmacol Toxicol 2023; 63:273-293. [PMID: 36100219 DOI: 10.1146/annurev-pharmtox-051921-084047] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Specialized pro-resolving mediators (SPMs), including resolvins, protectins, and maresins, are endogenous lipid mediators that are synthesized from omega-3 polyunsaturated fatty acids during the acute phase or resolution phase of inflammation. Synthetic SPMs possess broad safety profiles and exhibit potent actions in resolving inflammation in preclinical models. Accumulating evidence in the past decade has demonstrated powerful analgesia of exogenous SPMs in rodent models of inflammatory, neuropathic, and cancer pain. Furthermore, endogenous SPMs are produced by sham surgery and neuromodulation (e.g., vagus nerve stimulation). SPMs produce their beneficial actions through multiple G protein-coupled receptors, expressed by immune cells, glial cells, and neurons. Notably, loss of SPM receptors impairs the resolution of pain. I also highlight the emerging role of SPMs in the control of itch. Pharmacological targeting of SPMs or SPM receptors has the potential to lead to novel therapeutics for pain and itch as emerging approaches in resolution pharmacology.
Collapse
Affiliation(s)
- Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, and Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, North Carolina, USA;
| |
Collapse
|
13
|
Barreto Chang OL, Possin KL, Maze M. Age-Related Perioperative Neurocognitive Disorders: Experimental Models and Druggable Targets. Annu Rev Pharmacol Toxicol 2023; 63:321-340. [PMID: 36100220 DOI: 10.1146/annurev-pharmtox-051921-112525] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
With the worldwide increase in life span, surgical patients are becoming older and have a greater propensity for postoperative cognitive impairment, either new onset or through deterioration of an existing condition; in both conditions, knowledge of the patient's preoperative cognitive function and postoperative cognitive trajectory is imperative. We describe the clinical utility of a tablet-based technique for rapid assessment of the memory and attentiveness domains required for executive function. The pathogenic mechanisms for perioperative neurocognitive disorders have been investigated in animal models in which excessive and/or prolonged postoperative neuroinflammation has emerged as a likely contender. The cellular and molecular species involved in postoperative neuroinflammation are the putative targets for future therapeutic interventions that are efficacious and do not interfere with the surgical patient's healing process.
Collapse
Affiliation(s)
- Odmara L Barreto Chang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA;
| | - Katherine L Possin
- Memory and Aging Center, Department of Neurology, and Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Mervyn Maze
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA; .,Center for Cerebrovascular Research, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Flak MB, Koenis DS, Gonzalez-Nunez M, Chopo-Pizarro A, Dalli J. Deletion of macrophage Gpr101 disrupts their phenotype and function dysregulating host immune responses in sterile and infectious inflammation. Biochem Pharmacol 2023; 207:115348. [PMID: 36400250 DOI: 10.1016/j.bcp.2022.115348] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
We recently found that the G protein coupled receptor GPR101 mediates the phagocyte-directed pro-resolving activities of RvD5n-3 DPA (n-3 docosapentaenoic acid-derived Resolvin D5). Herein, we investigated the endogenous role of this pro-resolving receptor in modulating macrophage biology using a novel mouse line where the expression of Gpr101 was conditionally deleted in macrophages (MacGpr101KO). Peritoneal macrophages obtained from naïve MacGpr101KO mice displayed a marked shift in the expression of phenotypic and activation markers, including the Interleukin (IL)-10 and IL-23 receptors. Loss of Gpr101 on macrophages was also associated with a significant disruption in their cellular metabolism and a decreased ability to migrate towards the chemoattractant Mcp-1. The alterations in macrophage phenotype observed in Gpr101 deficient macrophages were maintained following inflammatory challenge. This was linked with an increased inflammatory response in the Gpr101 deficient animals and a reduced ability of phagocytes, including macrophages, to clear bacteria. Loss of Gpr101 on macrophages disrupted host pro-resolving responses to zymosan challenge with MacGpr101KO mice exhibiting significantly higher neutrophil numbers and a delay in the resolution interval when compared with control mice. These observations were linked with a marked dysregulation in peritoneal lipid mediator concentrations in Gpr101 deficient mice, with a downregulation of pro-resolving mediators including MaR2n-3 DPA, Resolvin (Rv) D3 and RvE3. Together these findings identify Gpr101 as a novel regulator of both macrophage phenotype and function, modulating key biological activities in both limiting the propagation of inflammation and expediting its resolution.
Collapse
Affiliation(s)
- Magdalena B Flak
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Duco S Koenis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ UK
| | - Maria Gonzalez-Nunez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ UK
| | - Ana Chopo-Pizarro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ UK
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ UK; Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
15
|
Heck-Swain KL, Li J, Ruan W, Yuan X, Wang Y, Koeppen M, Eltzschig HK. Myeloid hypoxia-inducible factor HIF1A provides cardio-protection during ischemia and reperfusion via induction of netrin-1. Front Cardiovasc Med 2022; 9:970415. [PMID: 36247475 PMCID: PMC9554136 DOI: 10.3389/fcvm.2022.970415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
The transcription factor hypoxia-inducible factor HIF1A induces cardioprotection from ischemia and reperfusion injury. Here, we investigate tissue-specific pathways that are critical for HIF1A-elicited tissue protection. Initial studies showed that mice with induced global Hif1a deletion (Hif1aloxP/loxP UbiquitinCre+) have exaggerated myocardial injury during in situ ischemia and reperfusion. Surprisingly, this phenotype was mirrored only in mice with myeloid-specific Hif1a deletion (Hif1a loxP/loxP LysM Cre+). In contrast, mice with myocardial specific (Hif1aloxP/loxP Myosin Cre+), or vascular Hif1a deletion (Hif1aloxP/loxP VEcadherin Cre+) experienced similar levels of injury as controls. Subsequent studies using adoptive transfer of Hif1a-deficient polymorphonuclear neutrophils (PMNs) prior to myocardial injury demonstrated increased reperfusion injury. On the contrary, the adoptive transfer of PMNs treated ex vivo with the hypoxia inducible factor (HIF) stabilizer dimethyloxalylglycine (DMOG) was associated with attenuated myocardial injury. Furthermore, DMOG-mediated cardioprotection was abolished in Hif1aloxP/loxP LysM Cre+ mice, but not in Hif2aloxP/loxP LysM Cre+ mice. Finally, studies of PMN-dependent HIF1A target genes implicated the neuronal guidance molecule netrin-1 in mediating the cardioprotective effects of myeloid HIF1A. Taken together, the present studies identified a functional role for myeloid-expressed HIF1A in providing cardioprotection during ischemia and reperfusion injury, which is mediated, at least in part, by the induction of the netrin-1 neuronal guidance molecule in neutrophils.
Collapse
Affiliation(s)
- Ka Lin Heck-Swain
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Ruan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yanyu Wang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Michael Koeppen
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
16
|
Barthelemy JC, Pichot V, Hupin D, Berger M, Celle S, Mouhli L, Bäck M, Lacour JR, Roche F. Targeting autonomic nervous system as a biomarker of well-ageing in the prevention of stroke. Front Aging Neurosci 2022; 14:969352. [PMID: 36185479 PMCID: PMC9521604 DOI: 10.3389/fnagi.2022.969352] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke prediction is a key health issue for preventive medicine. Atrial fibrillation (AF) detection is well established and the importance of obstructive sleep apneas (OSA) has emerged in recent years. Although autonomic nervous system (ANS) appears strongly implicated in stroke occurrence, this factor is more rarely considered. However, the consequences of decreased parasympathetic activity explored in large cohort studies through measurement of ANS activity indicate that an ability to improve its activity level and equilibrium may prevent stroke. In support of these observations, a compensatory neurostimulation has already proved beneficial on endothelium function. The available data on stroke predictions from ANS is based on many long-term stroke cohorts. These data underline the need of repeated ANS evaluation for the general population, in a medical environment, and remotely by emerging telemedicine digital tools. This would help uncovering the reasons behind the ANS imbalance that would need to be medically adjusted to decrease the risk of stroke. This ANS unbalance help to draw attention on clinical or non-clinical evidence, disclosing the vascular risk, as ANS activity integrates the cumulated risk from many factors of which most are modifiable, such as metabolic inadaptation in diabetes and obesity, sleep ventilatory disorders, hypertension, inflammation, and lack of physical activity. Treating these factors may determine ANS recovery through the appropriate management of these conditions. Natural aging also decreases ANS activity. ANS recovery will decrease global circulating inflammation, which will reinforce endothelial function and thus protect the vessels and the associated organs. ANS is the whistle-blower of vascular risk and the actor of vascular health. Such as, ANS should be regularly checked to help draw attention on vascular risk and help follow the improvements in response to our interventions. While today prediction of stroke relies on classical cardiovascular risk factors, adding autonomic biomarkers as HRV parameters may significantly increase the prediction of stroke.
Collapse
Affiliation(s)
- Jean-Claude Barthelemy
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
- *Correspondence: Jean-Claude Barthelemy,
| | - Vincent Pichot
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
| | - David Hupin
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
- Section of Translational Cardiology, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mathieu Berger
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
- Centre d’Investigation et de Recherche sur le Sommeil, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sébastien Celle
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
| | - Lytissia Mouhli
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- Département de Neurologie, Hôpital Universitaire Nord, Saint-Étienne, France
| | - Magnus Bäck
- Section of Translational Cardiology, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Jean-René Lacour
- Laboratoire de Physiologie, Faculté de Médecine Lyon-Sud, Oullins, France
| | - Frederic Roche
- Physical Exercise and Clinical Physiology Department, CHU Nord, Saint-Étienne, France
- INSERM U1059 Santé Ingénierie Biologie, Université Jean Monnet, Saint-Étienne, France
| |
Collapse
|
17
|
Shivshankar P, Karmouty-Quintana H, Mills T, Doursout MF, Wang Y, Czopik AK, Evans SE, Eltzschig HK, Yuan X. SARS-CoV-2 Infection: Host Response, Immunity, and Therapeutic Targets. Inflammation 2022; 45:1430-1449. [PMID: 35320469 PMCID: PMC8940980 DOI: 10.1007/s10753-022-01656-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/27/2022] [Accepted: 02/25/2022] [Indexed: 02/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in a global pandemic with severe socioeconomic effects. Immunopathogenesis of COVID-19 leads to acute respiratory distress syndrome (ARDS) and organ failure. Binding of SARS-CoV-2 spike protein to human angiotensin-converting enzyme 2 (hACE2) on bronchiolar and alveolar epithelial cells triggers host inflammatory pathways that lead to pathophysiological changes. Proinflammatory cytokines and type I interferon (IFN) signaling in alveolar epithelial cells counter barrier disruption, modulate host innate immune response to induce chemotaxis, and initiate the resolution of inflammation. Here, we discuss experimental models to study SARS-CoV-2 infection, molecular pathways involved in SARS-CoV-2-induced inflammation, and viral hijacking of anti-inflammatory pathways, such as delayed type-I IFN response. Mechanisms of alveolar adaptation to hypoxia, adenosinergic signaling, and regulatory microRNAs are discussed as potential therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Pooja Shivshankar
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Internal Medicine, Divisions of Critical Care, Pulmonary and Sleep Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Marie-Francoise Doursout
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Yanyu Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Agnieszka K Czopik
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Scott E Evans
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Yuan X, Mills T, Doursout MF, Evans SE, Vidal Melo MF, Eltzschig HK. Alternative adenosine Receptor activation: The netrin-Adora2b link. Front Pharmacol 2022; 13:944994. [PMID: 35910389 PMCID: PMC9334855 DOI: 10.3389/fphar.2022.944994] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
During hypoxia or inflammation, extracellular adenosine levels are elevated. Studies using pharmacologic approaches or genetic animal models pertinent to extracellular adenosine signaling implicate this pathway in attenuating hypoxia-associated inflammation. There are four distinct adenosine receptors. Of these, it is not surprising that the Adora2b adenosine receptor functions as an endogenous feedback loop to control hypoxia-associated inflammation. First, Adora2b activation requires higher adenosine concentrations compared to other adenosine receptors, similar to those achieved during hypoxic inflammation. Second, Adora2b is transcriptionally induced during hypoxia or inflammation by hypoxia-inducible transcription factor HIF1A. Studies seeking an alternative adenosine receptor activation mechanism have linked netrin-1 with Adora2b. Netrin-1 was originally discovered as a neuronal guidance molecule but also functions as an immune-modulatory signaling molecule. Similar to Adora2b, netrin-1 is induced by HIF1A, and has been shown to enhance Adora2b signaling. Studies of acute respiratory distress syndrome (ARDS), intestinal inflammation, myocardial or hepatic ischemia and reperfusion implicate the netrin-Adora2b link in tissue protection. In this review, we will discuss the potential molecular linkage between netrin-1 and Adora2b, and explore studies demonstrating interactions between netrin-1 and Adora2b in attenuating tissue inflammation.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Marie-Francoise Doursout
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott E. Evans
- Department of Pulmonology, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
19
|
Liotti F, Marotta M, Sorriento D, Pagliuca C, Caturano V, Mantova G, Scaglione E, Salvatore P, Melillo RM, Prevete N. The probiotic Lactobacillus rhamnosus GG (LGG) restrains the angiogenic potential of colorectal carcinoma cells by activating a pro-resolving program via formyl peptide receptor 1. Mol Oncol 2022; 16:2959-2980. [PMID: 35808840 PMCID: PMC9394235 DOI: 10.1002/1878-0261.13280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/31/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Formyl peptide receptors (FPR1, FPR2 and FPR3) are innate immune sensors of pathogen and commensal bacteria and have a role in colonic mucosa homeostasis. We identified FPR1 as a tumour suppressor in gastric cancer cells due to its ability to sustain an inflammation resolution response with antiangiogenic potential. Here, we investigate whether FPR1 exerts similar functions in colorectal carcinoma (CRC) cells. Since it has been shown that the commensal bacterium Lactobacillus rhamnosus GG (LGG) can promote intestinal epithelial homeostasis through FPR1, we explored the possibility that it could induce proresolving and antiangiogenic effects in CRC cells. We demonstrated that pharmacologic inhibition or genetic deletion of FPR1 in CRC cells caused a reduction of proresolving mediators and a consequent upregulation of angiogenic factors. The activation of FPR1 mediates opposite effects. Proresolving, antiangiogenic and homeostatic functions were also observed upon treatment of CRC cells with supernatant of LGG culture, but not of other lactic acid or nonprobiotic bacteria (i.e. Bifidobacterium bifidum or Escherichia coli). These activities of LGG are dependent on FPR1 expression and on the subsequent MAPK signalling activation. Thus, the innate immune receptor FPR1 could be a regulator of the balance between microbiota, inflammation and cancer in CRC models.
Collapse
Affiliation(s)
- Federica Liotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology (IEOS), CNR, Naples, Italy
| | - Maria Marotta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Valeria Caturano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Giuseppe Mantova
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,CEINGE, Biotecnologie Avanzate s.c.ar.l., Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology (IEOS), CNR, Naples, Italy
| | - Nella Prevete
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.,Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
Liotti F, Marotta M, Melillo RM, Prevete N. The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression. Cancers (Basel) 2022; 14:3333. [PMID: 35884394 PMCID: PMC9316558 DOI: 10.3390/cancers14143333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/23/2022] Open
Abstract
Non-resolving inflammation is an enabling feature of cancer. A novel super-family of lipid mediators termed Specialized Pro-resolving Mediators (SPMs) have a role as bioactive molecules mediating the resolution of inflammation in cancer biology. SPMs are derived from ω-3 and ω-6 polyunsaturated fatty acids through the activity of lipoxygenases. SPMs have been described to directly modulate cancer progression by interfering with the epithelial to mesenchymal transition and invasion of cancer cells. SPMs have also been demonstrated to act on several components of the tumor microenvironment (TME). Consistently with their natural immunomodulatory and anti-inflammatory properties, SPMs are able to reprogram macrophages to favor phagocytosis of cell debris, which are an important source of pro-inflammatory and pro-angiogenic signals; sustain a direct cytotoxic immune response against cancer cells; stimulate neutrophils anti-tumor activities; and inhibit the development of regulatory T and B cells, thus indirectly leading to enhanced anti-tumor immunity. Furthermore, the resolution pathways exert crucial anti-angiogenic functions in lung, liver, and gastrointestinal cancers, and inhibit cancer-associated fibroblast differentiation and functions in hepatocellular carcinoma and pancreatic cancer. The present review will be focused on the potential protective effects of resolution pathways against cancer, exerted by modulating different components of the TME.
Collapse
Affiliation(s)
- Federica Liotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
| | - Maria Marotta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
| | - Nella Prevete
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
21
|
Caravaca AS, Gallina AL, Tarnawski L, Shavva VS, Colas RA, Dalli J, Malin SG, Hult H, Arnardottir H, Olofsson PS. Vagus nerve stimulation promotes resolution of inflammation by a mechanism that involves Alox15 and requires the α7nAChR subunit. Proc Natl Acad Sci U S A 2022; 119:e2023285119. [PMID: 35622894 PMCID: PMC9295760 DOI: 10.1073/pnas.2023285119] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/09/2022] [Indexed: 12/31/2022] Open
Abstract
Nonresolving inflammation underlies a range of chronic inflammatory diseases, and therapeutic acceleration of resolution of inflammation may improve outcomes. Neural reflexes regulate the intensity of inflammation (for example, through signals in the vagus nerve), but whether activation of the vagus nerve promotes the resolution of inflammation in vivo has been unknown. To investigate this, mice were subjected to electrical vagus nerve stimulation (VNS) or sham surgery at the cervical level followed by zymosan-induced peritonitis. The duration of inflammation resolution was significantly reduced and efferocytosis was significantly increased in mice treated with VNS as compared with sham. Lipid mediator (LM) metabololipidomics revealed that mice treated with VNS had higher levels of specialized proresolving mediators (SPMs), particularly from the omega-3 docosahexaenoic (DHA) and docosapentaenoic (n-3 DPA) metabolomes, in peritoneal exudates. VNS also shifted the ratio between proinflammatory and proresolving LMs toward a proresolving profile, but this effect by VNS was inverted in mice deficient in 12/15-lipoxgenase (Alox15), a key enzyme in this SPM biosynthesis. The significant VNS-mediated reduction of neutrophil numbers in peritoneal exudates was absent in mice deficient in the cholinergic α7-nicotinic acetylcholine receptor subunit (α7nAChR), an essential component of the inflammatory reflex. Thus, VNS increased local levels of SPM and accelerated resolution of inflammation in zymosan-induced peritonitis by a mechanism that involves Alox15 and requires the α7nAChR.
Collapse
Affiliation(s)
- April S. Caravaca
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Alessandro L. Gallina
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Laura Tarnawski
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
| | - Vladimir S. Shavva
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Romain A. Colas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Stephen G. Malin
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Henrik Hult
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
- Department of Mathematics, KTH Royal Institute of Technology, Stockholm, 114 28, Sweden
| | - Hildur Arnardottir
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Peder S. Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, 171 76, Sweden
- Stockholm Center for Bioelectronic Medicine, MedTechLabs, Karolinska University Hospital, Solna, 171 76, Sweden
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, 11030
| |
Collapse
|
22
|
Resolution of inflammation: Intervention strategies and future applications. Toxicol Appl Pharmacol 2022; 449:116089. [DOI: 10.1016/j.taap.2022.116089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/23/2022]
|
23
|
Rudzki S, Praet S. Five-Year PTSD Symptom Remission in Two Patients Following Treatment With Rivastigmine. Mil Med 2022; 188:usac094. [PMID: 35446431 DOI: 10.1093/milmed/usac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/24/2022] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION The beneficial effect of rivastigmine, an acetylcholinesterase inhibitor (AChEi), which increases levels of acetylcholine (ACh), was first reported in 2013. This paper replicates those findings and reports sustained symptom remission. METHODS AND MATERIALS The high-frequency (HF) component of heart rate variability (HRV) is a measure of cholinergic withdrawal and was measured using a Zephyr Bioharness HR monitor, pre- and post-commencement of treatment. Data analysis was performed using Kubios HRV software. PTSD symptom severity was assessed using the Post-Traumatic Checklist-Civilian (PCL-C). RESULTS Low HF HRV was observed in both patients before rivastigmine treatment and reductions in PCL-C scores paralleled increases in HF HRV values. Follow-up revealed low HF HRV values in both patients despite PCL-C scores indicating remission. Sympathetic nervous system hyperactivity was observed in one patient, just before a suicide attempt. Following rivastigmine treatment, the patient had no further suicidal ideation or attempts. Another patient reported worsening of her PTSD symptoms in the peri-menstrual period, which was abolished by rivastigmine. She also experienced symptom relapse following prolonged infections. CONCLUSION Low HF HRV has been reported in PTSD patients, but findings have been inconsistent. Cholinergic withdrawal could explain the disturbances in sleep, learning, and memory seen in PTSD patients. The relapse of symptoms following prolonged infection implicates the immune system as a possible initiator of the disorder. ACh and estrogen have anti-inflammatory properties, supporting a possible role of inflammation in initiating PTSD. The effect of rivastigmine treatment should be tested in properly controlled clinical trials.
Collapse
Affiliation(s)
| | - Stephan Praet
- Ochre Health Medical Centre, University of Canberra, Bruce, ACT 2617, Australia
| |
Collapse
|
24
|
Falvey A, Metz CN, Tracey KJ, Pavlov VA. Peripheral nerve stimulation and immunity: the expanding opportunities for providing mechanistic insight and therapeutic intervention. Int Immunol 2022; 34:107-118. [PMID: 34498051 PMCID: PMC8783605 DOI: 10.1093/intimm/dxab068] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/07/2021] [Indexed: 12/29/2022] Open
Abstract
Pre-clinical research advances our understanding of the vagus nerve-mediated regulation of immunity and clinical trials successfully utilize electrical vagus nerve stimulation in the treatment of patients with inflammatory disorders. This symbiotic relationship between pre-clinical and clinical research exploring the vagus nerve-based 'inflammatory reflex' has substantially contributed to establishing the field of bioelectronic medicine. Recent studies identify a crosstalk between the vagus nerve and other neural circuitries in controlling inflammation and delineate new neural immunoregulatory pathways. Here we outline current mechanistic insights into the role of vagal and non-vagal neural pathways in neuro-immune communication and inflammatory regulation. We also provide a timely overview of expanding opportunities for bioelectronic neuromodulation in the treatment of various inflammatory disorders.
Collapse
Affiliation(s)
- Aidan Falvey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Christine N Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kevin J Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Valentin A Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
25
|
Forstenpointner J, Elman I, Freeman R, Borsook D. The Omnipresence of Autonomic Modulation in Health and Disease. Prog Neurobiol 2022; 210:102218. [PMID: 35033599 DOI: 10.1016/j.pneurobio.2022.102218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/13/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
Abstract
The Autonomic Nervous System (ANS) is a critical part of the homeostatic machinery with both central and peripheral components. However, little is known about the integration of these components and their joint role in the maintenance of health and in allostatic derailments leading to somatic and/or neuropsychiatric (co)morbidity. Based on a comprehensive literature search on the ANS neuroanatomy we dissect the complex integration of the ANS: (1) First we summarize Stress and Homeostatic Equilibrium - elucidating the responsivity of the ANS to stressors; (2) Second we describe the overall process of how the ANS is involved in Adaptation and Maladaptation to Stress; (3) In the third section the ANS is hierarchically partitioned into the peripheral/spinal, brainstem, subcortical and cortical components of the nervous system. We utilize this anatomical basis to define a model of autonomic integration. (4) Finally, we deploy the model to describe human ANS involvement in (a) Hypofunctional and (b) Hyperfunctional states providing examples in the healthy state and in clinical conditions.
Collapse
Affiliation(s)
- Julia Forstenpointner
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, SH, Germany.
| | - Igor Elman
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Departments of Psychiatry and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Molero-Chamizo A, Nitsche MA, Bolz A, Andújar Barroso RT, Alameda Bailén JR, García Palomeque JC, Rivera-Urbina GN. Non-Invasive Transcutaneous Vagus Nerve Stimulation for the Treatment of Fibromyalgia Symptoms: A Study Protocol. Brain Sci 2022; 12:brainsci12010095. [PMID: 35053839 PMCID: PMC8774206 DOI: 10.3390/brainsci12010095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/23/2022] Open
Abstract
Stimulation of the vagus nerve, a parasympathetic nerve that controls the neuro-digestive, vascular, and immune systems, induces pain relief, particularly in clinical conditions such as headache and rheumatoid arthritis. Transmission through vagal afferents towards the nucleus of the solitary tract (NST), the central relay nucleus of the vagus nerve, has been proposed as the main physiological mechanism that reduces pain intensity after vagal stimulation. Chronic pain symptoms of fibromyalgia patients might benefit from stimulation of the vagus nerve via normalization of altered autonomic and immune systems causing their respective symptoms. However, multi-session non-invasive vagal stimulation effects on fibromyalgia have not been evaluated in randomized clinical trials. We propose a parallel group, sham-controlled, randomized study to modulate the sympathetic–vagal balance and pain intensity in fibromyalgia patients by application of non-invasive transcutaneous vagus nerve stimulation (tVNS) over the vagal auricular and cervical branches. We will recruit 136 fibromyalgia patients with chronic moderate to high pain intensity. The primary outcome measure will be pain intensity, and secondary measures will be fatigue, health-related quality of life, sleep disorders, and depression. Heart rate variability and pro-inflammatory cytokine levels will be obtained as secondary physiological measures. We hypothesize that multiple tVNS sessions (five per week, for 4 weeks) will reduce pain intensity and improve quality of life as a result of normalization of the vagal control of nociception and immune–autonomic functions. Since both vagal branches project to the NST, we do not predict significantly different results between the two stimulation protocols.
Collapse
Affiliation(s)
- Andrés Molero-Chamizo
- Department of Clinical and Experimental Psychology, University of Huelva, 21007 Huelva, Spain; (R.T.A.B.); (J.R.A.B.)
- Correspondence: ; Tel.: +34-959218478
| | - Michael A. Nitsche
- Leibniz Research Centre for Working Environment and Human Factors, 44139 Dortmund, Germany;
- Department of Neurology, University Medical Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Armin Bolz
- tVNS Technologies GmbH, Ebrardstr. 31, 91052 Erlangen, Germany;
| | - Rafael Tomás Andújar Barroso
- Department of Clinical and Experimental Psychology, University of Huelva, 21007 Huelva, Spain; (R.T.A.B.); (J.R.A.B.)
| | - José R. Alameda Bailén
- Department of Clinical and Experimental Psychology, University of Huelva, 21007 Huelva, Spain; (R.T.A.B.); (J.R.A.B.)
| | - Jesús Carlos García Palomeque
- Department of the Histology, School of Medicine, Cadiz University and District Jerez Costa-N., Andalusian Health Service, 11003 Cádiz, Spain;
| | | |
Collapse
|
27
|
Somatosensory and autonomic neuronal regulation of the immune response. Nat Rev Neurosci 2022; 23:157-171. [PMID: 34997214 DOI: 10.1038/s41583-021-00555-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
Bidirectional communication between the peripheral nervous system (PNS) and the immune system is a crucial part of an effective but balanced mammalian response to invading pathogens, tissue damage and inflammatory stimuli. Here, we review how somatosensory and autonomic neurons regulate immune cellular responses at barrier tissues and in peripheral organs. Immune cells express receptors for neuronal mediators, including neuropeptides and neurotransmitters, allowing neurons to influence their function in acute and chronic inflammatory diseases. Distinct subsets of peripheral sensory, sympathetic, parasympathetic and enteric neurons are able to signal to innate and adaptive immune cells to modulate their cellular functions. In this Review, we highlight recent studies defining the molecular mechanisms by which neuroimmune signalling mediates tissue homeostasis and pathology. Understanding the neural circuitry that regulates immune responses can offer novel targets for the treatment of a wide array of diseases.
Collapse
|
28
|
Cecconello C, Clària Ribas P, Norling LV. Resolving acute inflammation; what happens when inflammation goes haywire? How can it get back in line? DIET, INFLAMMATION, AND HEALTH 2022:113-162. [DOI: 10.1016/b978-0-12-822130-3.00018-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Ziegon L, Schlegel M. Netrin-1: A Modulator of Macrophage Driven Acute and Chronic Inflammation. Int J Mol Sci 2021; 23:275. [PMID: 35008701 PMCID: PMC8745333 DOI: 10.3390/ijms23010275] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Netrins belong to the family of laminin-like secreted proteins, which guide axonal migration and neuronal growth in the developing central nervous system. Over the last 20 years, it has been established that netrin-1 acts as a chemoattractive or chemorepulsive cue in diverse biological processes far beyond neuronal development. Netrin-1 has been shown to play a central role in cell adhesion, cell migration, proliferation, and cell survival in neuronal and non-neuronal tissue. In this context, netrin-1 was found to orchestrate organogenesis, angiogenesis, tumorigenesis, and inflammation. In inflammation, as in neuronal development, netrin-1 plays a dichotomous role directing the migration of leukocytes, especially monocytes in the inflamed tissue. Monocyte-derived macrophages have long been known for a similar dual role in inflammation. In response to pathogen-induced acute injury, monocytes are rapidly recruited to damaged tissue as the first line of immune defense to phagocyte pathogens, present antigens to initiate the adaptive immune response, and promote wound healing in the resolution phase. On the other hand, dysregulated macrophages with impaired phagocytosis and egress capacity accumulate in chronic inflammation sites and foster the maintenance-and even the progression-of chronic inflammation. In this review article, we will highlight the dichotomous roles of netrin-1 and its impact on acute and chronic inflammation.
Collapse
Affiliation(s)
| | - Martin Schlegel
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany;
| |
Collapse
|
30
|
Ramos-Martínez IE, Rodríguez MC, Cerbón M, Ramos-Martínez JC, Ramos-Martínez EG. Role of the Cholinergic Anti-Inflammatory Reflex in Central Nervous System Diseases. Int J Mol Sci 2021; 22:ijms222413427. [PMID: 34948222 PMCID: PMC8705572 DOI: 10.3390/ijms222413427] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
In several central nervous system diseases, it has been reported that inflammation may be related to the etiologic process, therefore, therapeutic strategies are being implemented to control inflammation. As the nervous system and the immune system maintain close bidirectional communication in physiological and pathological conditions, the modulation of inflammation through the cholinergic anti-inflammatory reflex has been proposed. In this review, we summarized the evidence supporting chemical stimulation with cholinergic agonists and vagus nerve stimulation as therapeutic strategies in the treatment of various central nervous system pathologies, and their effect on inflammation.
Collapse
Affiliation(s)
- Ivan Emmanuel Ramos-Martínez
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris Est Créteil (UPEC), 94010 Créteil, France;
| | - María Carmen Rodríguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Morelos 62100, Mexico;
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Correspondence: (M.C.); (E.G.R.-M.)
| | - Juan Carlos Ramos-Martínez
- Cardiology Department, Hospital General Regional Lic. Ignacio Garcia Tellez IMSS, Yucatán 97150, Mexico;
| | - Edgar Gustavo Ramos-Martínez
- Escuela de Ciencias, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
- Instituto de Cómputo Aplicado en Ciencias, Oaxaca 68044, Mexico
- Correspondence: (M.C.); (E.G.R.-M.)
| |
Collapse
|
31
|
Camargo LF, Pinheiro GD, de Oliveira PB, Losada DM, Chagas EFB, Sperança MA, Chies AB, Spadella MA, Martins LPA. Influence of galantamine in the inflammatory process and tissular lesions caused by Trypanosoma cruzi QM2 strain. Rev Soc Bras Med Trop 2021; 54:e0201. [PMID: 34787259 PMCID: PMC8582970 DOI: 10.1590/0037-8682-0201-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/20/2021] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Trypanosoma cruzi infection triggers an inflammatory process with exacerbated production of cytokines that stimulate inflammatory and anti-inflammatory signals, including the efferent anti-inflammatory signal known as the anti-inflammatory cholinergic pathway. Thus, the use of anticholinesterase drugs, such as galantamine, could minimize the inflammatory process caused by this disease. METHODS For the study at 30, 60, and 90 days, 120 Swiss mice were divided into three groups. Each group was subdivided into four subgroups: uninfected/untreated (CTRL), uninfected/treated (GAL), infected/untreated (INF), and infected/treated (GAL/INF). The infected groups were inoculated intraperitoneally with 0.1 ml of mouse blood containing 5 × 104 trypomastigote forms of the T. cruzi QM2 strain. The galantamine-treated groups received 5 mg/kg of galantamine orally, through pipetting. From each subgroup, the parameters of parasitemia, histopathological analysis, butyrylcholinesterase activity (BuChE), and functional study of the colon were evaluated. RESULTS BuChE performance was observed when AChE was suppressed, with increased activity in the GAL/INF group similar to the INF group on the 30th day post infection, thus corroborating the absence of a significant difference in parasitic curves and histopathological analysis. CONCLUSIONS The presence of an inflammatory process and nests of amastigotes, as well as evidence of reactivity to ACh and NOR, suggest that galantamine did not interfere with the colonic inflammatory response or even in colonic tissue parasitism at this stage of Chagas disease.
Collapse
Affiliation(s)
| | | | | | - Daniele Moraes Losada
- Universidade Estadual de Campinas, Departamento de Anatomia Patológica, Campinas, SP, Brasil
| | | | - Márcia Aparecida Sperança
- Universidade Federal do ABC, Centro de Ciências Naturais e Humanas, São Bernardo do Campo, SP, Brasil
| | - Agnaldo Bruno Chies
- Faculdade de Medicina de Marília, Departamento de Farmacologia, Marília, SP, Brasil
| | | | | |
Collapse
|
32
|
Chen Z, He X, Yao MW, Li Z, Xu X. [Research advances on the cholinergic inflammatory reflex and inflammation resolution]. ZHONGHUA SHAO SHANG ZA ZHI = ZHONGHUA SHAOSHANG ZAZHI = CHINESE JOURNAL OF BURNS 2021; 37:885-889. [PMID: 34645156 PMCID: PMC11917218 DOI: 10.3760/cma.j.cn501120-20200609-00299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The vagus nerve plays an important role in regulating the homeostasis of inflammation. Inflammation signals in the body are passed to the vagus nerve efferent fibers via nerve reflexes, and the signals generated by efferent fibers will play an anti-inflammatory role in various inflammatory diseases through immune cells such as T cells that express choline acetyltransferase and macrophages. However, the resolution of inflammation is not only the interaction between pro-inflammatory and anti-inflammatory cytokines, but also an active process of biosynthesis, including the synthesis of various pro-resolving mediators and their physiological utility process. Moreover, the cholinergic inflammation reflex also plays a crucial role in inflammation resolution. This review reviews and summarizes the cholinergic inflammatory reflex and its key role in the process of inflammation resolution.
Collapse
Affiliation(s)
- Z Chen
- Basic Medical School of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - X He
- Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - M W Yao
- Department of Stem Cell and Regenerative Medicine, Army Medical Center of PLA, Chongqing 400042, China
| | - Z Li
- Department of Stem Cell and Regenerative Medicine, Army Medical Center of PLA, Chongqing 400042, China
| | - X Xu
- Department of Stem Cell and Regenerative Medicine, Army Medical Center of PLA, Chongqing 400042, China
| |
Collapse
|
33
|
Huang X, Ying J, Yang D, Fang P, Wang X, Zhou B, Zhang L, Fang Y, Yu W, Liu X, Zhen Q, Hua F. The Mechanisms of Sevoflurane-Induced Neuroinflammation. Front Aging Neurosci 2021; 13:717745. [PMID: 34421578 PMCID: PMC8375153 DOI: 10.3389/fnagi.2021.717745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Sevoflurane is one of the most commonly used inhaled anesthetics due to its low blood gas coefficient, fast onset, low airway irritation, and aromatic smell. However, recent studies have reported that sevoflurane exposure may have deleterious effects on cognitive function. Although neuroinflammation was most widely mentioned among the established mechanisms of sevoflurane-induced cognitive dysfunction, its upstream mechanisms have yet to be illustrated. Thus, we reviewed the relevant literature and discussed the most mentioned mechanisms, including the modulation of the microglial function, blood–brain barrier (BBB) breakdown, changes in gut microbiota, and ease of cholinergic neurotransmission to help us understand the properties of sevoflurane, providing us new perspectives for the prevention of sevoflurane-induced cognitive impairment.
Collapse
Affiliation(s)
- Xiangfei Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Pu Fang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Qingcui Zhen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
34
|
Schlegel M, Sharma M, Brown EJ, Newman AAC, Cyr Y, Afonso MS, Corr EM, Koelwyn GJ, van Solingen C, Guzman J, Farhat R, Nikain CA, Shanley LC, Peled D, Schmidt AM, Fisher EA, Moore KJ. Silencing Myeloid Netrin-1 Induces Inflammation Resolution and Plaque Regression. Circ Res 2021; 129:530-546. [PMID: 34289717 PMCID: PMC8529357 DOI: 10.1161/circresaha.121.319313] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rationale: Therapeutic efforts to decrease atherosclerotic cardiovascular disease risk have focused largely on reducing atherogenic lipoproteins, yet lipid-lowering therapies alone are insufficient to fully regress plaque burden. We postulate that arterial repair requires resolution of a maladaptive immune response and that targeting factors that hinder inflammation resolution will facilitate plaque regression. Objective: The guidance molecule Ntn1 (netrin-1) is secreted by macrophages in atherosclerotic plaques, where it sustains inflammation by enhancing macrophage survival and blocking macrophage emigration. We tested whether silencing Ntn1 in advanced atherosclerosis could resolve arterial inflammation and regress plaques. Methods and Results: To temporally silence Ntn1 in myeloid cells, we generated genetically modified mice in which Ntn1 could be selectively deleted in monocytes and macrophages using a tamoxifen-induced CX3CR1-driven cre recombinase (Ntn1fl/flCx3cr1creERT2+) and littermate control mice (Ntn1fl/flCx3cr1WT). Mice were fed Western diet in the setting of hepatic PCSK9 (proprotein convertase subtilisin/kexin type 9) overexpression to render them atherosclerotic and then treated with tamoxifen to initiate deletion of myeloid Ntn1 (MøΔNtn1) or not in controls (MøWT). Morphometric analyses performed 4 weeks later showed that myeloid Ntn1 silencing reduced plaque burden in the aorta (−50%) and plaque complexity in the aortic root. Monocyte-macrophage tracing experiments revealed lower monocyte recruitment, macrophage retention, and proliferation in MøΔNtn1 compared with MøWT plaques, indicating a restructuring of monocyte-macrophage dynamics in the artery wall upon Ntn1 silencing. Single-cell RNA sequencing of aortic immune cells before and after Ntn1 silencing revealed upregulation of gene pathways involved in macrophage phagocytosis and migration, including the Ccr7 chemokine receptor signaling pathway required for macrophage emigration from plaques and atherosclerosis regression. Additionally, plaques from MøΔNtn1 mice showed hallmarks of inflammation resolution, including higher levels of proresolving macrophages, IL (interleukin)-10, and efferocytosis, as compared to plaques from MøWT mice. Conclusion: Our data show that targeting Ntn1 in advanced atherosclerosis ameliorates atherosclerotic inflammation and promotes plaque regression.
Collapse
Affiliation(s)
- Martin Schlegel
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
- Department of Anesthesiology and Intensive Care, Technical University of Munich, School of Medicine, Germany (M. Schlegel)
| | - Monika Sharma
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Emily J Brown
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Alexandra A C Newman
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Yannick Cyr
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Milessa Silva Afonso
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Emma M Corr
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Graeme J Koelwyn
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Coen van Solingen
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Jonathan Guzman
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Rubab Farhat
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Cyrus A Nikain
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Lianne C Shanley
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Daniel Peled
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Ann Marie Schmidt
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University (A.M.S.). K.J. Moore, M. Schlegel, M. Sharma, A.M. Schmidt, and E.A. Fisher designed the study and performed data analysis and interpretation. M. Schlegel, M. Sharma, M.S. Afonso, E.J. Brown, E.M. Corr, C. van Solingen, G.J. Koelwyn, A.A.C. Newman, Y. Cyr, R. Farhat, J. Guzman, L.C. Shanley, and D. Peled conducted experiments, acquired data, and performed analyses. E.J. Brown analyzed the RNA-sequencing data. K.J. Moore and M. Schlegel wrote the article with input from all authors
| | - Edward A Fisher
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| |
Collapse
|
35
|
Abstract
Nonresolving inflammation, a hallmark of sepsis and/or multi-organ failure, still poses a challenge in medicine. The mortality rate is enormous, and so far no adequate curative therapy is available. Here we identify a previously unrecognized role of the neuronal guidance protein semaphorin 7A in the transition to resolution processes in severe systematic inflammation such as sepsis. Endogenous mediators regulating acute inflammatory responses in both the induction and resolution phases of inflammatory processes are pivotal in host defense and tissue homeostasis. Recent studies have identified neuronal guidance proteins characterized in axonal development that display immunomodulatory functions. Here, we identify the neuroimmune guidance cue Semaphorin 7A (Sema7A), which appears to link macrophage (MΦ) metabolic remodeling to inflammation resolution. Sema7A orchestrated MΦ chemotaxis and chemokinesis, activated MΦ differentiation and polarization toward the proresolving M2 phenotype, and promoted leukocyte clearance. Peritoneal MΦSema7A−/− displayed metabolic reprogramming, characterized by reductions in fatty acid oxidation and oxidative phosphorylation, increases in glycolysis and the pentose phosphate pathway, and truncation of the tricarboxylic acid cycle, which resulted in increased levels of the intermediates succinate and fumarate. The low accumulation of citrate in MΦSema7A−/− correlated with the decreased synthesis of prostaglandins, leading to a reduced impact on lipid-mediator class switching and the generation of specialized pro resolving lipid mediators. Signaling network analysis indicated that Sema7A induced the metabolic reprogramming of MΦ by activating the mTOR- and AKT2-signaling pathways. Administration of Sema7ASL4cd orchestrated the resolution response to tissue homeostasis by shortening the resolution interval, promoting tissue protection in murine peritonitis, and enhancing survival in polymicrobial sepsis.
Collapse
|
36
|
Kim B, Guaregua V, Chen X, Zhao C, Yeow W, Berg NK, Eltzschig HK, Yuan X. Characterization of a Murine Model System to Study MicroRNA-147 During Inflammatory Organ Injury. Inflammation 2021; 44:1426-1440. [PMID: 33566257 PMCID: PMC7873671 DOI: 10.1007/s10753-021-01427-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/15/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022]
Abstract
Inflammatory organ injury and sepsis have profound impacts on the morbidity and mortality of surgical and critical care patients. MicroRNAs are small RNAs composed of 20-25 nucleotides that have a significant contribution to gene regulation. MicroRNA-147 (miR-147), in particular, has been shown to have an emerging role in different physiological functions such as cell cycle regulation and inflammatory responses. However, animal model systems to study tissue-specific functions of miR-147 during inflammatory conditions in vivo are lacking. In the present study, we characterize miR-147 expression in different organs and cell types. Next, we generated a transgenic mouse line with a floxed miR-147 gene. Subsequently, we used this mouse line to generate mice with whole-body deletion of miR-147 (miR-147 -/-) by crossing "floxed" miR-147 mice with transgenic mice expressing Cre recombinase in all tissues (CMVcre mice). Systematic analysis of miR-147 -/- mice demonstrates normal growth, development, and off-spring. In addition, deletion of the target gene in different organs was successful at baseline or during inflammation, including the heart, intestine, stomach, liver, spleen, bone marrow, lungs, kidneys, or stomach. Moreover, miR-147 -/- mice have identical baseline inflammatory gene expression compared to C57BL/6 mice, except elevated IL-6 expression in the spleen (7.5 fold, p < 0.05). Taken together, our data show the successful development of a transgenic animal model for tissue and cell-specific deletion of miR-147 that can be used to study the functional roles of miR-147 during inflammatory organ injury.
Collapse
Affiliation(s)
- Boyun Kim
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Victor Guaregua
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Xuebo Chen
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Chad Zhao
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Wanyi Yeow
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Nathaniel K Berg
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
37
|
Berg NK, Li J, Kim B, Mills T, Pei G, Zhao Z, Li X, Zhang X, Ruan W, Eltzschig HK, Yuan X. Hypoxia-inducible factor-dependent induction of myeloid-derived netrin-1 attenuates natural killer cell infiltration during endotoxin-induced lung injury. FASEB J 2021; 35:e21334. [PMID: 33715200 PMCID: PMC8251729 DOI: 10.1096/fj.202002407r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022]
Abstract
Sepsis and sepsis‐associated lung inflammation significantly contribute to the morbidity and mortality of critical illness. Here, we examined the hypothesis that neuronal guidance proteins could orchestrate inflammatory events during endotoxin‐induced lung injury. Through a targeted array, we identified netrin‐1 as the top upregulated neuronal guidance protein in macrophages treated with lipopolysaccharide (LPS). Furthermore, we found that netrin‐1 is highly enriched in infiltrating myeloid cells, particularly in macrophages during LPS‐induced lung injury. Transcriptional studies implicate hypoxia‐inducible factor HIF‐1α in the transcriptional induction of netrin‐1 during LPS treatment. Subsequently, the deletion of netrin‐1 in the myeloid compartment (Ntn1loxp/loxp LysM Cre) resulted in exaggerated mortality and lung inflammation. Surprisingly, further studies revealed enhanced natural killer cells (NK cells) infiltration in Ntn1loxp/loxp LysM Cre mice, and neutralization of NK cell chemoattractant chemokine (C‐C motif) ligand 2 (CCL2) reversed the exaggerated lung inflammation. Together, these studies provide functional insight into myeloid cell‐derived netrin‐1 in controlling lung inflammation through the modulation of CCL2‐dependent infiltration of NK cells.
Collapse
Affiliation(s)
- Nathaniel K Berg
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.,Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Boyun Kim
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Tingting Mills
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Guangsheng Pei
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, TX, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, TX, USA
| | - Xiangyun Li
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.,Department of Anesthesiology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xu Zhang
- Department of Internal Medicine, The University of Texas Health Science Center, Houston, TX, USA.,Center for Clinical and Translational Sciences, The University of Texas Health Science Center, Houston, TX, USA
| | - Wei Ruan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.,Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
38
|
Liu Y, Forsythe P. Vagotomy and insights into the microbiota-gut-brain axis. Neurosci Res 2021; 168:20-27. [DOI: 10.1016/j.neures.2021.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
|
39
|
Tao X, Luo X, Zhang T, Hershey B, Esteller R, Ji RR. Spinal Cord Stimulation Attenuates Mechanical Allodynia and Increases Central Resolvin D1 Levels in Rats With Spared Nerve Injury. Front Physiol 2021; 12:687046. [PMID: 34248674 PMCID: PMC8267572 DOI: 10.3389/fphys.2021.687046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022] Open
Abstract
Mounting evidence from animal models of inflammatory and neuropathic pain suggests that inflammation regulates the resolution of pain by producing specialized pro-resolving mediators (SPMs), such as resolvin D1 (RvD1). However, it remains unclear how SPMs are induced in the central nervous system and whether these mechanisms can be reconciled with outcomes of neuromodulation therapies for pain, such as spinal cord stimulation. Here, we show that in a male rat model of neuropathic pain produced by spared nerve injury (SNI), 1 kHz spinal cord stimulation (1 kHz SCS) alone was sufficient to reduce mechanical allodynia and increase RvD1 in the cerebrospinal fluid (CSF). SNI resulted in robust and persistent mechanical allodynia and cold allodynia. Spinal cord electrode implantation was conducted at the T11-T13 vertebral level 1 week after SNI. The spinal locations of the implanted electrodes were validated by X-Ray radiography. 1 kHz SCS was applied for 6 h at 0.1 ms pulse-width, and this stimulation alone was sufficient to effectively reduce nerve injury-induced mechanical allodynia during stimulation without affecting SNI-induced cold allodynia. SCS alone significantly reduced interleukin-1β levels in both serum and CSF samples. Strikingly, SCS significantly increased RvD1 levels in the CSF but not serum. Finally, intrathecal injection of RvD1 (100 and 500 ng, i.t.) 4 weeks after nerve injury reduced SNI-induced mechanical allodynia in a dose-dependent manner. Our findings suggest that 1 kHz SCS may alleviate neuropathic pain via reduction of IL-1β and via production and/or release of RvD1 to control SNI-induced neuroinflammation.
Collapse
Affiliation(s)
- Xueshu Tao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States.,Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Tianhe Zhang
- Boston Scientific Neuromodulation Research and Advanced Concepts, Valencia, CA, United States
| | - Brad Hershey
- Boston Scientific Neuromodulation Research and Advanced Concepts, Valencia, CA, United States
| | - Rosana Esteller
- Boston Scientific Neuromodulation Research and Advanced Concepts, Valencia, CA, United States
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States.,Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.,Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
40
|
Li LC, Tian Y, Xiao J, Yang Y, Wu JN, Chen Y, Zhang PH, Gao-Smith F, Wang JG, Jin SW. Dexmedetomidine promotes inflammation resolving through TGF-β1 secreted by F4/80 +Ly6G + macrophage. Int Immunopharmacol 2021; 95:107480. [PMID: 33676148 DOI: 10.1016/j.intimp.2021.107480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist, which can regulate inflammatory responses. However, whether DEX interferes with the inflammation resolving remains unclear. Here, we reported the effects of DEX on zymosan-induced generalized inflammation in mice during resolution. Mice were administered intraperitoneally with DEX after the initiation of sepsis. The resolution interval (Ri), a vital resolution indice, decreased from twelve hours to eight hours after the administration of DEX. The induction of peritoneal pro-inflammatory interleukin [IL] - 1β and tumour necrosis factor-α (TNF-α) appeared to be inhibited. Of interest, the anti-inflammatory transforming growth factor-β1 (TGF-β1) but not IL-10 levels were up-regulated at twenty-four hours in the DEX group along with 1.0 mg/mice zymosan A (ZyA) treatment. The expression levels of multiple genes related to protective immune processes and clearance functions were detected and revealed the same trends. DEX markedly increased the F4/80+Ly6G+ macrophage population. Additionally, the adequate apoptotic neutrophil clearance from injury after DEX installation could be reverse by opsonization or co-instillation of TGF-β1 neutralizing antibody in vivo, promoting the inflammation-resolution programs. In conclusion, DEX post-treatment, via the increase of F4/80+Ly6G+ macrophages, provokes further secretion of TGF-β1, leading to the attenuated cytokine storm and accelerated inflammation resolving.
Collapse
Affiliation(s)
- Lin-Chao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yang Tian
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Jin-Ni Wu
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China
| | - Yan Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Fang Gao-Smith
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| | - Jian-Guang Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China.
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| |
Collapse
|
41
|
Datta-Chaudhuri T, Zanos T, Chang EH, Olofsson PS, Bickel S, Bouton C, Grande D, Rieth L, Aranow C, Bloom O, Mehta AD, Civillico G, Stevens MM, Głowacki E, Bettinger C, Schüettler M, Puleo C, Rennaker R, Mohanta S, Carnevale D, Conde SV, Bonaz B, Chernoff D, Kapa S, Berggren M, Ludwig K, Zanos S, Miller L, Weber D, Yoshor D, Steinman L, Chavan SS, Pavlov VA, Al-Abed Y, Tracey KJ. The Fourth Bioelectronic Medicine Summit "Technology Targeting Molecular Mechanisms": current progress, challenges, and charting the future. Bioelectron Med 2021; 7:7. [PMID: 34024277 PMCID: PMC8142479 DOI: 10.1186/s42234-021-00068-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023] Open
Abstract
There is a broad and growing interest in Bioelectronic Medicine, a dynamic field that continues to generate new approaches in disease treatment. The fourth bioelectronic medicine summit "Technology targeting molecular mechanisms" took place on September 23 and 24, 2020. This virtual meeting was hosted by the Feinstein Institutes for Medical Research, Northwell Health. The summit called international attention to Bioelectronic Medicine as a platform for new developments in science, technology, and healthcare. The meeting was an arena for exchanging new ideas and seeding potential collaborations involving teams in academia and industry. The summit provided a forum for leaders in the field to discuss current progress, challenges, and future developments in Bioelectronic Medicine. The main topics discussed at the summit are outlined here.
Collapse
Affiliation(s)
| | - Theodoros Zanos
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Eric H. Chang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | | | - Stephan Bickel
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Chad Bouton
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Daniel Grande
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Loren Rieth
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
- University of Utah, Salt Lake City, UT USA
| | - Cynthia Aranow
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Ona Bloom
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Ashesh D. Mehta
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | | | | | | | | | | | | | | | - Saroj Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Daniela Carnevale
- Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Silvia V. Conde
- CEDOC, Nova Medical School, Faculdade de Ciências Médicas, Lisbon, Portugal
| | - Bruno Bonaz
- University of Grenoble Alpes, INSERM, Grenoble, France
| | | | | | | | - Kip Ludwig
- University of Wisconsin, Madison, WI USA
| | - Stavros Zanos
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Larry Miller
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Doug Weber
- Carnegie Mellon University, Pittsburgh, PA USA
| | | | | | - Sangeeta S. Chavan
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Yousef Al-Abed
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| |
Collapse
|
42
|
Eberhardson M, Levine YA, Tarnawski L, Olofsson PS. The brain-gut axis, inflammatory bowel disease and bioelectronic medicine. Int Immunol 2021; 33:349-356. [PMID: 33912906 DOI: 10.1093/intimm/dxab018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
The hallmark of inflammatory bowel diseases (IBD) is chronic intestinal inflammation with typical onset in adolescents and young adults. An abundance of neutrophils is seen in the inflammatory lesions, but adaptive immunity is also an important player in the chronicity of the disease. There is an unmet need for new treatment options since modern medicines such as biological therapy with anti-cytokine antibodies still leave a substantial number of patients with persisting disease activity. The role of the central nervous system and its interaction with the gut in the pathophysiology of IBD have been brought to attention both in animal models and in humans after the discovery of the inflammatory reflex. The suggested control of gut immunity by the brain-gut axis represents a novel therapeutic target suitable for bioelectronic intervention. In this review, we discuss the role of the inflammatory reflex in gut inflammation and the recent advances in the treatment of IBD by intervening with the brain-gut axis through bioelectronic devices.
Collapse
Affiliation(s)
- Michael Eberhardson
- Department of Gastroenterology and Hepatology, University Hospital of Linköping, 581 91 Linköping, Sweden.,Department of Medicine, Center for Bioelectronic Medicine, Bioclinicum, Karolinska Institutet, 171 64 Stockholm, Sweden.,Department of Health, Medicine and Caring Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Yaakov A Levine
- Department of Medicine, Center for Bioelectronic Medicine, Bioclinicum, Karolinska Institutet, 171 64 Stockholm, Sweden.,SetPoint Medical, Valencia, CA 91355, USA
| | - Laura Tarnawski
- Department of Medicine, Center for Bioelectronic Medicine, Bioclinicum, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Peder S Olofsson
- Department of Medicine, Center for Bioelectronic Medicine, Bioclinicum, Karolinska Institutet, 171 64 Stockholm, Sweden.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| |
Collapse
|
43
|
MicroRNAs in shaping the resolution phase of inflammation. Semin Cell Dev Biol 2021; 124:48-62. [PMID: 33934990 DOI: 10.1016/j.semcdb.2021.03.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022]
Abstract
Inflammation is a host defense mechanism orchestrated through imperative factors - acute inflammatory responses mediated by cellular and molecular events leading to activation of defensive immune subsets - to marginalize detrimental injury, pathogenic agents and infected cells. These potent inflammatory events, if uncontrolled, may cause tissue damage by perturbing homeostasis towards immune dysregulation. A parallel host mechanism operates to contain inflammatory pathways and facilitate tissue regeneration. Thus, resolution of inflammation is an effective moratorium on the pro-inflammatory pathway to avoid the tissue damage inside the host and leads to reestablishment of tissue homeostasis. Dysregulation of the resolution pathway can have a detrimental impact on tissue functionality and contribute to the diseased state. Multiple reports have suggested peculiar dynamics of miRNA expression during various pro- and anti-inflammatory events. The roles of miRNAs in the regulation of immune responses are well-established. However, understanding of miRNA regulation of the resolution phase of events in infection or wound healing models, which is sometimes misconstrued as anti-inflammatory signaling, remains limited. Due to the deterministic role of miRNAs in pro-inflammatory and anti-inflammatory pathways, in this review we have provided a broad perspective on the putative role of miRNAs in the resolution of inflammation and explored their imminent role in therapeutics.
Collapse
|
44
|
Li J, Conrad C, Mills TW, Berg NK, Kim B, Ruan W, Lee JW, Zhang X, Yuan X, Eltzschig HK. PMN-derived netrin-1 attenuates cardiac ischemia-reperfusion injury via myeloid ADORA2B signaling. J Exp Med 2021; 218:212023. [PMID: 33891683 PMCID: PMC8077173 DOI: 10.1084/jem.20210008] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 01/03/2023] Open
Abstract
Previous studies implicated the neuronal guidance molecule netrin-1 in attenuating myocardial ischemia-reperfusion injury. However, the tissue-specific sources and receptor signaling events remain elusive. Neutrophils are among the first cells responding to an ischemic insult and can be associated with tissue injury or rescue. We found netrin-1 levels were elevated in the blood of patients with myocardial infarction, as well as in mice exposed to myocardial ischemia-reperfusion. Selectively increased infarct sizes and troponin levels were found in Ntn1loxP/loxP Lyz2 Cre+ mice, but not in mice with conditional netrin-1 deletion in other tissue compartments. In vivo studies using neutrophil depletion identified neutrophils as the main source for elevated blood netrin-1 during myocardial injury. Finally, pharmacologic studies using treatment with recombinant netrin-1 revealed a functional role for purinergic signaling events through the myeloid adenosine A2b receptor in mediating netrin-1-elicited cardioprotection. These findings suggest an autocrine signaling loop with a functional role for neutrophil-derived netrin-1 in attenuating myocardial ischemia-reperfusion injury through myeloid adenosine A2b signaling.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Catharina Conrad
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Tingting W Mills
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX
| | - Nathaniel K Berg
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Boyun Kim
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Wei Ruan
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX.,Department of Anesthesiology, Second Xiangya Hospital, Central South University, Hunan, China
| | - Jae W Lee
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT
| | - Xu Zhang
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX
| | - Xiaoyi Yuan
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| |
Collapse
|
45
|
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: 21] [Impact Index Per Article: 5.3] [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
|
46
|
Ansari J, Senchenkova EY, Vital SA, Al-Yafeai Z, Kaur G, Sparkenbaugh EM, Orr AW, Pawlinski R, Hebbel RP, Granger DN, Kubes P, Gavins FNE. Targeting the AnxA1/Fpr2/ALX pathway regulates neutrophil function, promoting thromboinflammation resolution in sickle cell disease. Blood 2021; 137:1538-1549. [PMID: 33512489 PMCID: PMC7976506 DOI: 10.1182/blood.2020009166] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play a crucial role in the intertwined processes of thrombosis and inflammation. An altered neutrophil phenotype may contribute to inadequate resolution, which is known to be a major pathophysiological contributor of thromboinflammatory conditions such as sickle cell disease (SCD). The endogenous protein annexin A1 (AnxA1) facilitates inflammation resolution via formyl peptide receptors (FPRs). We sought to comprehensively elucidate the functional significance of targeting the neutrophil-dependent AnxA1/FPR2/ALX pathway in SCD. Administration of AnxA1 mimetic peptide AnxA1Ac2-26 ameliorated cerebral thrombotic responses in Sickle transgenic mice via regulation of the FPR2/ALX (a fundamental receptor involved in resolution) pathway. We found direct evidence that neutrophils with SCD phenotype play a key role in contributing to thromboinflammation. In addition, AnxA1Ac2-26 regulated activated SCD neutrophils through protein kinase B (Akt) and extracellular signal-regulated kinases (ERK1/2) to enable resolution. We present compelling conceptual evidence that targeting the AnxA1/FPR2/ALX pathway may provide new therapeutic possibilities against thromboinflammatory conditions such as SCD.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
| | | | | | - Zaki Al-Yafeai
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | | | - Erica M Sparkenbaugh
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Rafal Pawlinski
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert P Hebbel
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | | | - Paul Kubes
- Departments of Physiology and Pharmacology, Microbiology and Immunology and Critical Care Medicine, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada; and
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology
- Department of Neurology, and
- Department of Life Sciences, Brunel University London, United Kingdom
| |
Collapse
|
47
|
Suarez-Roca H, Mamoun N, Sigurdson MI, Maixner W. Baroreceptor Modulation of the Cardiovascular System, Pain, Consciousness, and Cognition. Compr Physiol 2021; 11:1373-1423. [PMID: 33577130 DOI: 10.1002/cphy.c190038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Baroreceptors are mechanosensitive elements of the peripheral nervous system that maintain cardiovascular homeostasis by coordinating the responses to external and internal environmental stressors. While it is well known that carotid and cardiopulmonary baroreceptors modulate sympathetic vasomotor and parasympathetic cardiac neural autonomic drive, to avoid excessive fluctuations in vascular tone and maintain intravascular volume, there is increasing recognition that baroreceptors also modulate a wide range of non-cardiovascular physiological responses via projections from the nucleus of the solitary tract to regions of the central nervous system, including the spinal cord. These projections regulate pain perception, sleep, consciousness, and cognition. In this article, we summarize the physiology of baroreceptor pathways and responses to baroreceptor activation with an emphasis on the mechanisms influencing cardiovascular function, pain perception, consciousness, and cognition. Understanding baroreceptor-mediated effects on cardiac and extra-cardiac autonomic activities will further our understanding of the pathophysiology of multiple common clinical conditions, such as chronic pain, disorders of consciousness (e.g., abnormalities in sleep-wake), and cognitive impairment, which may result in the identification and implementation of novel treatment modalities. © 2021 American Physiological Society. Compr Physiol 11:1373-1423, 2021.
Collapse
Affiliation(s)
- Heberto Suarez-Roca
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University, Durham, North Carolina, USA
| | - Negmeldeen Mamoun
- Department of Anesthesiology, Division of Cardiothoracic Anesthesia and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Martin I Sigurdson
- Department of Anesthesiology and Critical Care Medicine, Landspitali, University Hospital, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - William Maixner
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University, Durham, North Carolina, USA
| |
Collapse
|
48
|
Faden AI, Barrett JP, Stoica BA, Henry RJ. Bidirectional Brain-Systemic Interactions and Outcomes After TBI. Trends Neurosci 2021; 44:406-418. [PMID: 33495023 DOI: 10.1016/j.tins.2020.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/23/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a debilitating disorder associated with chronic progressive neurodegeneration and long-term neurological decline. Importantly, there is now substantial and increasing evidence that TBI can negatively impact systemic organs, including the pulmonary, gastrointestinal (GI), cardiovascular, renal, and immune system. Less well appreciated, until recently, is that such functional changes can affect both the response to subsequent insults or diseases, as well as contribute to chronic neurodegenerative processes and long-term neurological outcomes. In this review, we summarize evidence showing bidirectional interactions between the brain and systemic organs following TBI and critically assess potential underlying mechanisms.
Collapse
Affiliation(s)
- Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
Gao R, Peng X, Perry C, Sun H, Ntokou A, Ryu C, Gomez JL, Reeves BC, Walia A, Kaminski N, Neumark N, Ishikawa G, Black KE, Hariri LP, Moore MW, Gulati M, Homer RJ, Greif DM, Eltzschig HK, Herzog EL. Macrophage-derived netrin-1 drives adrenergic nerve-associated lung fibrosis. J Clin Invest 2021; 131:136542. [PMID: 33393489 PMCID: PMC7773383 DOI: 10.1172/jci136542] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is a macrophage-driven process of uncontrolled extracellular matrix accumulation. Neuronal guidance proteins such as netrin-1 promote inflammatory scarring. We found that macrophage-derived netrin-1 stimulates fibrosis through its neuronal guidance functions. In mice, fibrosis due to inhaled bleomycin engendered netrin-1-expressing macrophages and fibroblasts, remodeled adrenergic nerves, and augmented noradrenaline. Cell-specific knockout mice showed that collagen accumulation, fibrotic histology, and nerve-associated endpoints required netrin-1 of macrophage but not fibroblast origin. Adrenergic denervation; haploinsufficiency of netrin-1's receptor, deleted in colorectal carcinoma; and therapeutic α1 adrenoreceptor antagonism improved collagen content and histology. An idiopathic pulmonary fibrosis (IPF) lung microarray data set showed increased netrin-1 expression. IPF lung tissues were enriched for netrin-1+ macrophages and noradrenaline. A longitudinal IPF cohort showed improved survival in patients prescribed α1 adrenoreceptor blockade. This work showed that macrophages stimulate lung fibrosis via netrin-1-driven adrenergic processes and introduced α1 blockers as a potentially new fibrotic therapy.
Collapse
Affiliation(s)
- Ruijuan Gao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xueyan Peng
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carrighan Perry
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Huanxing Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aglaia Ntokou
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jose L. Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Benjamin C. Reeves
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anjali Walia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nir Neumark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine, and
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meagan W. Moore
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J. Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| | - Daniel M. Greif
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, University of Texas at Houston Medical School, Houston, Texas, USA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| |
Collapse
|
50
|
Targeting inflammation in atherosclerosis - from experimental insights to the clinic. Nat Rev Drug Discov 2021; 20:589-610. [PMID: 33976384 PMCID: PMC8112476 DOI: 10.1038/s41573-021-00198-1] [Citation(s) in RCA: 613] [Impact Index Per Article: 153.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 02/03/2023]
Abstract
Atherosclerosis, a dominant and growing cause of death and disability worldwide, involves inflammation from its inception to the emergence of complications. Targeting inflammatory pathways could therefore provide a promising new avenue to prevent and treat atherosclerosis. Indeed, clinical studies have now demonstrated unequivocally that modulation of inflammation can forestall the clinical complications of atherosclerosis. This progress pinpoints the need for preclinical investigations to refine strategies for combatting inflammation in the human disease. In this Review, we consider a gamut of attractive possibilities for modifying inflammation in atherosclerosis, including targeting pivotal inflammatory pathways such as the inflammasomes, inhibiting cytokines, manipulating adaptive immunity and promoting pro-resolution mechanisms. Along with lifestyle measures, pharmacological interventions to mute inflammation could complement traditional targets, such as lipids and hypertension, to make new inroads into the management of atherosclerotic risk.
Collapse
|