1
|
Chang Y, Chen J, Peng Y, Zhang K, Zhang Y, Zhao X, Wang D, Li L, Zhu J, Liu K, Li Z, Pan S, Huang K. Gut-derived macrophages link intestinal damage to brain injury after cardiac arrest through TREM1 signaling. Cell Mol Immunol 2025; 22:437-455. [PMID: 39984674 PMCID: PMC11955566 DOI: 10.1038/s41423-025-01263-0] [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: 08/28/2024] [Revised: 12/19/2024] [Accepted: 01/20/2025] [Indexed: 02/23/2025] Open
Abstract
Brain injury is the leading cause of death and disability in survivors of cardiac arrest, where neuroinflammation triggered by infiltrating macrophages plays a pivotal role. Here, we seek to elucidate the origin of macrophages infiltrating the brain and their mechanism of action after cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Wild-type or photoconvertible Cd68-Cre:R26-LSL-KikGR mice were subjected to 10-min CA/CPR, and the migration of gut-derived macrophages into brain was assessed. Transcriptome sequencing was performed to identify the key proinflammatory signal of macrophages infiltrating the brain, triggering receptor expressed on myeloid cells 1 (TREM1). Upon drug intervention, the effects of TREM1 on post-CA/CPR brain injury were further evaluated. 16S rRNA sequencing was used to detect gut dysbiosis after CA/CPR. Through photoconversion experiments, we found that small intestine-derived macrophages infiltrated the brain and played a crucial role in triggering secondary brain injury after CA/CPR. The infiltrating peripheral macrophages showed upregulated TREM1 levels, and we further revealed the crucial role of gut-derived TREM1+ macrophages in post-CA/CPR brain injury through a drug intervention targeting TREM1. Moreover, a close correlation between upregulated TREM1 expression and poor neurological outcomes was observed in CA survivors. Mechanistically, CA/CPR caused a substantial expansion of Enterobacter at the early stage, which ignited intestinal TREM1 signaling via the activation of Toll-like receptor 4 on macrophages through the release of lipopolysaccharide. Our findings reveal essential crosstalk between the gut and brain after CA/CPR and underscore the potential of targeting TREM1+ small intestine-derived macrophages as a novel therapeutic strategy for mitigating post-CA/CPR brain injury.
Collapse
Affiliation(s)
- Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiancong Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqin Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolin Zhao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Di Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Lei Li
- Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juan Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kewei Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhentong Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China.
| |
Collapse
|
2
|
Tamura T, Cheng C, Villaseñor-Altamirano A, Yamada K, Ikeda K, Hayashida K, Menon JA, Chen XD, Chung H, Varon J, Chen J, Choi J, Cullen AM, Guo J, Lin X, Olenchock BA, Pinilla-Vera MA, Manandhar R, Sheikh MDA, Hou PC, Lawler PR, Oldham WM, Seethala RR, Baron RM, Bohula EA, Morrow DA, Blumberg RS, Chen F, Merriam LT, Weissman AJ, Brenner MB, Chen X, Ichinose F, Kim EY. Diverse NKT cells regulate early inflammation and neurological outcomes after cardiac arrest and resuscitation. Sci Transl Med 2024; 16:eadq5796. [PMID: 39630883 PMCID: PMC11792709 DOI: 10.1126/scitranslmed.adq5796] [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: 06/23/2024] [Accepted: 09/30/2024] [Indexed: 12/07/2024]
Abstract
Neurological injury drives most deaths and morbidity among patients hospitalized for out-of-hospital cardiac arrest (OHCA). Despite its clinical importance, there are no effective pharmacological therapies targeting post-cardiac arrest (CA) neurological injury. Here, we analyzed circulating immune cells from a large cohort of patients with OHCA, finding that lymphopenia independently associated with poor neurological outcomes. Single-cell RNA sequencing of immune cells showed that T cells with features of both innate T cells and natural killer (NK) cells were increased in patients with favorable neurological outcomes. We more specifically identified an early increase in circulating diverse NKT (dNKT) cells in a separate cohort of patients with OHCA who had good neurological outcomes. These cells harbored a diverse T cell receptor repertoire but were consistently specific for sulfatide antigen. In mice, we found that sulfatide-specific dNKT cells trafficked to the brain after CA and resuscitation. In the brains of mice lacking NKT cells (Cd1d-/-), we observed increased inflammatory chemokine and cytokine expression and accumulation of macrophages when compared with wild-type mice. Cd1d-/- mice also had increased neuronal injury, neurological dysfunction, and worse mortality after CA. To therapeutically enhance dNKT cell activity, we treated mice with sulfatide lipid after CA, showing that it improved neurological function. Together, these data show that sulfatide-specific dNKT cells are associated with good neurological outcomes after clinical OHCA and are neuroprotective in mice after CA. Strategies to enhance the number or function of dNKT cells may thus represent a treatment approach for CA.
Collapse
Affiliation(s)
- Tomoyoshi Tamura
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Changde Cheng
- Department of Computational Biology, St Jude Children’s Research Hospital, Nashville 38105, TN
- Department of Medicine, Division of Hematology and Oncology, Stem Cell Biology Program, University of Alabama at Birmingham, Birmingham 35233, AL
| | - Ana Villaseñor-Altamirano
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Kohei Yamada
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Kohei Ikeda
- Harvard Medical School; Boston, MA 02115
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston 02114, MA
| | - Kei Hayashida
- Harvard Medical School; Boston, MA 02115
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston 02114, MA
| | - Jaivardhan A Menon
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Xi Dawn Chen
- Broad Institute of Harvard and MIT, Cambridge 02138, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Hattie Chung
- Broad Institute of Harvard and MIT, Cambridge 02138, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Jack Varon
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Jiani Chen
- Department of Computational Biology, St Jude Children’s Research Hospital, Nashville 38105, TN
| | - Jiyoung Choi
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston 02115, MA
| | - Aidan M. Cullen
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston 02115, MA
| | - Jingyu Guo
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston 02115, MA
| | - Xi Lin
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Gastroenterology, Brigham and Women’s Hospital, Boston, MA 02142, USA
| | - Benjamin A. Olenchock
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Cardiovascular Division, Brigham and Women’s Hospital, Boston 02115, MA
| | - Mayra A. Pinilla-Vera
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
| | - Reshmi Manandhar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Muhammad Dawood Amir Sheikh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
| | - Peter C. Hou
- Harvard Medical School; Boston, MA 02115
- Department of Emergency Medicine, Division of Emergency and Critical Care Medicine, Brigham and Women’s Hospital, Boston 02115, MA
| | - Patrick R. Lawler
- McGill University Health Centre, Montreal, Quebec H3A 2B4, Canada
- University of Toronto, Toronto, Ontario M5R 0A3, Canada
| | - William M. Oldham
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Raghu R. Seethala
- Harvard Medical School; Boston, MA 02115
- Department of Emergency Medicine, Division of Emergency and Critical Care Medicine, Brigham and Women’s Hospital, Boston 02115, MA
| | | | - Rebecca M. Baron
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| | - Erin A. Bohula
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Cardiovascular Division, Brigham and Women’s Hospital, Boston 02115, MA
| | - David A. Morrow
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Cardiovascular Division, Brigham and Women’s Hospital, Boston 02115, MA
| | - Richard S. Blumberg
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Gastroenterology, Brigham and Women’s Hospital, Boston, MA 02142, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge 02138, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Louis T. Merriam
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
| | - Alexandra J. Weissman
- Department of Emergency Medicine, University of Pittsburgh School of Medicine; Pittsburgh 15261, PA
| | - Michael B. Brenner
- Harvard Medical School; Boston, MA 02115
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston 02115, MA
| | - Xiang Chen
- Department of Computational Biology, St Jude Children’s Research Hospital, Nashville 38105, TN
| | - Fumito Ichinose
- Harvard Medical School; Boston, MA 02115
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston 02114, MA
| | - Edy Y. Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School; Boston, MA 02115
| |
Collapse
|
3
|
Abi Zeid Daou Y, Lidouren F, Bois A, Watanabe N, Jendoubi A, Faucher E, Surenaud M, Chateau-Joubert S, Hue S, Ghaleh B, Kohlhauer M, Tissier R. Investigation of fingolimod-induced lymphocyte sequestration on inflammatory response and neurological damages after cardiac arrest. Intensive Care Med Exp 2024; 12:57. [PMID: 38954057 PMCID: PMC11219599 DOI: 10.1186/s40635-024-00645-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND A sepsis-like syndrome is known to occur after cardiac arrest, leading to cerebral infiltration by white blood cells (WBC). We hypothesized that pharmacological sequestration of WBC, and more specifically lymphocytes within lymphoid tissues, could reduce the cerebral infiltration by these inflammatory cells and subsequent acute brain injury in a porcine model of cardiac arrest. Lymphocyte sequestration was induced by the sphingosine-1 phosphate receptors agonist fingolimod. METHODS In a first set of experiments, anesthetized pigs underwent a sham instrumentation with no cardiac arrest (n = 4). They received an administration of fingolimod (1 mg/kg, i.v.) in order to confirm its effect on WBC. In a second set of experiments, animals randomly received fingolimod or saline two hours prior to an episode of ventricular fibrillation (14 min) with subsequent resuscitation (n = 6 in each group). Neurological injury was assessed 24 h after resuscitation. RESULTS In the first set of experiments, WBC and blood lymphocyte counts were significantly reduced by - 61 ± 10% and - 75 ± 6% two hours after fingolimod administration. In the second set of experiments, blood lymphocyte counts, but not WBC, were also significantly reduced after cardiac arrest in Fingolimod vs Control group. However, most cytokine blood levels were not different among groups, including Interleukin (IL)-1ra, IL-8 or IL-18 blood levels. A difference was only observed for IL-6, which decreased in Fingolimod vs Control (e.g., 5.6 ± 4.8 vs 59.4 ± 20.6 pg/ml at 2 h after cardiac arrest, respectively; p = 0.126). Neurofilament light chain (NFL) blood levels were not different among groups (57 ± 25 vs 84 ± 41 pg/ml in Fingolimod vs Control at 6 h after resuscitation, respectively). After awakening, 3 and 2 animals were prematurely euthanized for ethical reasons due to recurrent seizures in Fingolimod and Control groups, respectively. At Day 1, neurological dysfunction score was not different between groups (87 ± 7 vs 87 ± 5% in Fingolimod vs Control, respectively). Conversely, a decrease in the number of CD3 + cells was observed in the brain of surviving animals in Fingolimod vs Control group (3.10 ± 0.50 vs 7.53 ± 0.57 CD3 + cells/field, respectively; p = 0.0286). CONCLUSION Fingolimod-induced WBC sequestration, and more specifically lymphocytes sequestration, did not improve clinical neurological dysfunction following cardiac arrest although it reduced cerebral infiltration by lymphocytes.
Collapse
Affiliation(s)
- Yara Abi Zeid Daou
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Fanny Lidouren
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Antoine Bois
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
- Service de Médecine Intensive-Réanimation, Hôpitaux Universitaires Paris Centre, Hopital Cochin, Paris, France
| | - Naoto Watanabe
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Ali Jendoubi
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Estelle Faucher
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Mathieu Surenaud
- Vaccine Research Institute, Univ Paris Est-Creteil, 94000, Creteil, France
| | - Sophie Chateau-Joubert
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Sophie Hue
- Vaccine Research Institute, Univ Paris Est-Creteil, 94000, Creteil, France
| | - Bijan Ghaleh
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Matthias Kohlhauer
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Renaud Tissier
- Univ Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France.
- Ecole Nationale Vétérinaire d'Alfort, IMRB, AfterROSC Network, 7 avenue du Général de Gaulle, 94700, Maisons-Alfort, France.
| |
Collapse
|
4
|
Aoki T, Wong V, Yin T, Nakamura E, Endo Y, Hayashida K, Robson SC, Nandurkar H, Diamond B, Kim SJ, Murao A, Wang P, Becker LB, Shinozaki K. Immune cell expression patterns of CD39/CD73 ectonucleotidases in rodent models of cardiac arrest and resuscitation. Front Immunol 2024; 15:1362858. [PMID: 38545102 PMCID: PMC10967020 DOI: 10.3389/fimmu.2024.1362858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Background Cardiac arrest (CA) is a significant public health concern. There is the high imminent mortality and survival in those who are resuscitated is substantively compromised by the post-CA syndrome (PCAS), characterized by multiorgan ischemia-reperfusion injury (IRI). The inflammatory response in PCAS is complex and involves various immune cell types, including lymphocytes and myeloid cells that have been shown to exacerbate organ IRI, such as myocardial infarction. Purinergic signaling, as regulated by CD39 and CD73, has emerged as centrally important in the context of organ-specific IRI. Hence, comprehensive understanding of such purinergic responses may be likewise imperative for improving outcomes in PCAS. Methods We have investigated alterations of immune cell populations after CA by utilizing rodent models of PCAS. Blood and spleen were collected after CA and resuscitation and underwent flow cytometry analysis to evaluate shifts in CD3+CD4+ helper T cells, CD3+CD8a+ cytotoxic T cells, and CD4/CD8a ratios. We then examined the expression of CD39 and CD73 across diverse cell types, including myeloid cells, T lymphocytes, and B lymphocytes. Results In both rat and mouse models, there were significant increases in the frequency of CD3+CD4+ T lymphocytes in PCAS (rat, P < 0.01; mouse, P < 0.001), with consequently elevated CD4/CD8a ratios in whole blood (both, P < 0.001). Moreover, CD39 and CD73 expression on blood leukocytes were markedly increased (rat, P < 0.05; mouse, P < 0.01 at 24h). Further analysis in the experimental mouse model revealed that CD11b+ myeloid cells, with significant increase in their population (P < 0.01), had high level of CD39 (88.80 ± 2.05 %) and increased expression of CD73 (P < 0.05). CD19+ B lymphocytes showed slight increases of CD39 (P < 0.05 at 2h) and CD73 (P < 0.05 at 2h), while, CD3+ T lymphocytes had decreased levels of them. These findings suggested a distinct patterns of expression of CD39 and CD73 in these specific immune cell populations after CA. Conclusions These data have provided comprehensive insights into the immune response after CA, highlighting high-level expressions of CD39 and CD73 in myeloid cells.
Collapse
Affiliation(s)
- Tomoaki Aoki
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Vanessa Wong
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- State University of New York Downstate Medical Center, NY, United States
| | - Tai Yin
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Eriko Nakamura
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Yusuke Endo
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Kei Hayashida
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Betty Diamond
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Sun Jung Kim
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Lance B. Becker
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY, United States
| | - Koichiro Shinozaki
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY, United States
- Department of Emergency & Critical Care Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
5
|
Aoki T, Endo Y, Nakamura E, Kuschner CE, Kazmi J, Singh P, Yin T, Becker LB, Hayashida K. Therapeutic potential of mitochondrial transplantation in modulating immune responses post-cardiac arrest: a narrative review. J Transl Med 2024; 22:230. [PMID: 38433198 PMCID: PMC10909283 DOI: 10.1186/s12967-024-05003-2] [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: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Mitochondrial transplantation (MTx) has emerged as a novel therapeutic strategy, particularly effective in diseases characterized by mitochondrial dysfunction. This review synthesizes current knowledge on MTx, focusing on its role in modulating immune responses and explores its potential in treating post-cardiac arrest syndrome (PCAS). METHODS We conducted a comprehensive narrative review of animal and human studies that have investigated the effects of MTx in the context of immunomodulation. This included a review of the immune responses following critical condition such as ischemia reperfusion injury, the impact of MTx on these responses, and the therapeutic potential of MTx in various conditions. RESULTS Recent studies indicate that MTx can modulate complex immune responses and reduce ischemia-reperfusion injury post-CA, suggesting MTx as a novel, potentially more effective approach. The review highlights the role of MTx in immune modulation, its potential synergistic effects with existing treatments such as therapeutic hypothermia, and the need for further research to optimize its application in PCAS. The safety and efficacy of autologous versus allogeneic MTx, particularly in the context of immune reactions, are critical areas for future investigation. CONCLUSION MTx represents a promising frontier in the treatment of PCAS, offering a novel approach to modulate immune responses and restore cellular energetics. Future research should focus on long-term effects, combination therapies, and personalized medicine approaches to fully harness the potential of MTx in improving patient outcomes in PCAS.
Collapse
Affiliation(s)
- Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jacob Kazmi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Parmeshar Singh
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA.
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
6
|
Curran C, Vaitaitis G, Waid D, Volmer T, Alverez E, Wagner DH. Ocrevus reduces TH40 cells, a biomarker of systemic inflammation, in relapsing multiple sclerosis (RMS) and in progressive multiple sclerosis (PMS). J Neuroimmunol 2023; 374:578008. [PMID: 36535240 PMCID: PMC9868100 DOI: 10.1016/j.jneuroim.2022.578008] [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: 06/21/2022] [Revised: 08/16/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Treating MS has been difficult. One successful drug is Ocrelizumab (anti-CD20), used for the chronic relapsing MS (RMS) and the progressive MS (PMS) forms. TH40 cells are pathogenic effector T cells that increase in percentage and numbers during chronic inflammation. Here we show that in the earliest MS course, clinically isolated syndrome (CIS), TH40 cells expand in number. In PMS TH40 cell numbers remain expanded demonstrating sustained chronic inflammation. In RMS TH40 cells were found in CSF and express CD20. Ocrelizumab reduced TH40 cells to healthy control levels in patients. During treatment inflammatory cytokine producing TH40 cells were decreased.
Collapse
Affiliation(s)
- Christian Curran
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Gisela Vaitaitis
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Dan Waid
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Timothy Volmer
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Enrique Alverez
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - David H Wagner
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America.
| |
Collapse
|
7
|
Dietz RM, Dingman AL, Herson PS. Cerebral ischemia in the developing brain. J Cereb Blood Flow Metab 2022; 42:1777-1796. [PMID: 35765984 PMCID: PMC9536116 DOI: 10.1177/0271678x221111600] [Citation(s) in RCA: 24] [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] [Received: 03/13/2021] [Revised: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
Brain ischemia affects all ages, from neonates to the elderly population, and is a leading cause of mortality and morbidity. Multiple preclinical rodent models involving different ages have been developed to investigate the effect of ischemia during different times of key brain maturation events. Traditional models of developmental brain ischemia have focused on rodents at postnatal day 7-10, though emerging models in juvenile rodents (postnatal days 17-25) indicate that there may be fundamental differences in neuronal injury and functional outcomes following focal or global cerebral ischemia at different developmental ages, as well as in adults. Here, we consider the timing of injury in terms of excitation/inhibition balance, oxidative stress, inflammatory responses, blood brain barrier integrity, and white matter injury. Finally, we review translational strategies to improve function after ischemic brain injury, including new ideas regarding neurorestoration, or neural repair strategies that restore plasticity, at delayed time points after ischemia.
Collapse
Affiliation(s)
- Robert M Dietz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
- Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andra L Dingman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paco S Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
8
|
Cunningham CA, Coppler PJ, Skolnik AB. The immunology of the post-cardiac arrest syndrome. Resuscitation 2022; 179:116-123. [PMID: 36028143 DOI: 10.1016/j.resuscitation.2022.08.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
Abstract
Patients successfully resuscitated from cardiac arrest often have brain injury, myocardial dysfunction, and systemic ischemia-reperfusion injury, collectively termed the post-cardiac arrest syndrome (PCAS). To improve outcomes, potential therapies must be able to be administered early in the post-arrest course and provide broad cytoprotection, as ischemia-reperfusion injury affects all organ systems. Our understanding of the immune system contributions to the PCAS has expanded, with animal models detailing biologically plausible mechanisms of secondary injury, the protective effects of available immunomodulatory drugs, and how immune dysregulation underlies infection susceptibility after arrest. In this narrative review, we discuss the dysregulated immune response in PCAS, human trials of targeted immunomodulation therapies, and future directions for immunomodulation following cardiac arrest.
Collapse
Affiliation(s)
- Cody A Cunningham
- Mayo Clinic School of Graduate Medical Education, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ, USA.
| | - Patrick J Coppler
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron B Skolnik
- Department of Critical Care Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| |
Collapse
|
9
|
Inhaled nitric oxide improves post-cardiac arrest outcomes via guanylate cyclase-1 in bone marrow-derived cells. Nitric Oxide 2022; 125-126:47-56. [PMID: 35716999 DOI: 10.1016/j.niox.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
RATIONALE Nitric oxide (NO) exerts its biological effects primarily via activation of guanylate cyclase (GC) and production of cyclic guanosine monophosphate. Inhaled NO improves outcomes after cardiac arrest and cardiopulmonary resuscitation (CPR). However, mechanisms of the protective effects of breathing NO after cardiac arrest are incompletely understood. OBJECTIVE To elucidate the mechanisms of beneficial effects of inhaled NO on outcomes after cardiac arrest. METHODS Adult male C57BL/6J wild-type (WT) mice, GC-1 knockout mice, and chimeric WT mice with WT or GC-1 knockout bone marrow were subjected to 8 min of potassium-induced cardiac arrest to determine the role of GC-1 in bone marrow-derived cells. Mice breathed air or 40 parts per million NO for 23 h starting at 1 h after CPR. RESULTS Breathing NO after CPR prevented hypercoagulability, cerebral microvascular occlusion, an increase in circulating polymorphonuclear neutrophils and neutrophil-to-lymphocyte ratio, and right ventricular dysfunction in WT mice, but not in GC-1 knockout mice, after cardiac arrest. The lack of GC-1 in bone marrow-derived cells diminished the beneficial effects of NO breathing after CPR. CONCLUSIONS GC-dependent signaling in bone marrow-derived cells is essential for the beneficial effects of inhaled NO after cardiac arrest and CPR.
Collapse
|
10
|
He F, Zheng G, Hu J, Ge W, Ji X, Bradley JL, Peberdy MA, Ornato JP, Tang W. Necrosulfonamide improves post-resuscitation myocardial dysfunction via inhibiting pyroptosis and necroptosis in a rat model of cardiac arrest. Eur J Pharmacol 2022; 926:175037. [PMID: 35588872 DOI: 10.1016/j.ejphar.2022.175037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 11/24/2022]
Abstract
The systemic inflammatory response following global myocardial ischemia/reperfusion (I/R) injury is a critical driver of poor outcomes. Both pyroptosis and necroptosis are involved in the systemic inflammatory response and contribute to regional myocardial I/R injury. This study aimed to explore the effect of necrosulfonamide (NSA) on post-resuscitation myocardial dysfunction in a rat model of cardiac arrest. Sprague-Dawley rats were randomly categorized to Sham, CPR and CPR-NSA groups. For rats in the latter two groups, ventricular fibrillation was induced without treatment for 6 min, with cardiopulmonary resuscitation (CPR) being sustained for 8 min. Rats were injected with NSA (10 mg/kg in DMSO) or vehicle at 5 min following return of spontaneous circulation. Myocardial function was measured by echocardiography, survival and neurological deficit score (NDS) were recorded at 24, 48, and 72 h after ROSC. Western blotting was used to assess pyroptosis- and necroptosis-related protein expression. ELISAs were used to measure levels of inflammatory cytokine. Rats in the CPR-NSA group were found to exhibit superior post-resuscitation myocardial function, and better NDS values in the group of CPR-NSA. Rats in the group of CPR-NSA exhibited median survival duration of 68 ± 8 h as compared to 34 ± 21 h in the CPR group. After treatment with NSA, NOD-like receptor 3 (NLRP3), GSDMD-N, phosphorylated-MLKL, and phosphorylated-RIP3 levels in cardiac tissue were reduced with corresponding reductions in inflammatory cytokine levels. Administration of NSA significantly improved myocardial dysfunction succeeding global myocardial I/R injury and enhanced survival outcomes through protective mechanisms potentially related to inhibition of pyroptosis and necroptosis pathways.
Collapse
Affiliation(s)
- Fenglian He
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Guanghui Zheng
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Juntao Hu
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Weiwei Ge
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Xianfei Ji
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Jennifer L Bradley
- Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA.
| | - Mary Ann Peberdy
- Departments of Internal Medicine and Emergency Medicine, Virginia Commonwealth University Health System, Richmond, VA, USA.
| | - Joseph P Ornato
- Department of Emergency Medicine, Virginia Commonwealth University Health System, Richmond, VA, USA.
| | - Wanchun Tang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Weil Institute of Emergency and Critical Care Research, Virginia Commonwealth University, Richmond, VA, USA; Department of Emergency Medicine, Virginia Commonwealth University Health System, Richmond, VA, USA.
| |
Collapse
|
11
|
Abstract
The susceptibility of the brain to ischaemic injury dramatically limits its viability following interruptions in blood flow. However, data from studies of dissociated cells, tissue specimens, isolated organs and whole bodies have brought into question the temporal limits within which the brain is capable of tolerating prolonged circulatory arrest. This Review assesses cell type-specific mechanisms of global cerebral ischaemia, and examines the circumstances in which the brain exhibits heightened resilience to injury. We suggest strategies for expanding such discoveries to fuel translational research into novel cytoprotective therapies, and describe emerging technologies and experimental concepts. By doing so, we propose a new multimodal framework to investigate brain resuscitation following extended periods of circulatory arrest.
Collapse
|
12
|
Zhao Q, Shen Y, Li R, Wu J, Lyu J, Jiang M, Lu L, Zhu M, Wang W, Wang Z, Liu Q, Hoffmann U, Karhausen J, Sheng H, Zhang W, Yang W. Cardiac arrest and resuscitation activates the hypothalamic-pituitary-adrenal axis and results in severe immunosuppression. J Cereb Blood Flow Metab 2021; 41:1091-1102. [PMID: 32787543 PMCID: PMC8054717 DOI: 10.1177/0271678x20948612] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In patients who are successfully resuscitated after initial cardiac arrest (CA), mortality and morbidity rates are high, due to ischemia/reperfusion injury to the whole body including the nervous and immune systems. How the interactions between these two critical systems contribute to post-CA outcome remains largely unknown. Using a mouse model of CA and cardiopulmonary resuscitation (CA/CPR), we demonstrate that CA/CPR induced neuroinflammation in the brain, in particular, a marked increase in pro-inflammatory cytokines, which subsequently activated the hypothalamic-pituitary-adrenal (HPA) axis. Importantly, this activation was associated with a severe immunosuppression phenotype after CA. The phenotype was characterized by a striking reduction in size of lymphoid organs accompanied by a massive loss of immune cells and reduced immune function of splenic lymphocytes. The mechanistic link between post-CA immunosuppression and the HPA axis was substantiated, as we discovered that glucocorticoid treatment, which mimics effects of the activated HPA axis, exacerbated post-CA immunosuppression, while RU486 treatment, which suppresses its effects, significantly mitigated lymphopenia and lymphoid organ atrophy and improved CA outcome. Taken together, targeting the HPA axis could be a viable immunomodulatory therapeutic to preserve immune homeostasis after CA/CPR and thus improve prognosis of post-resuscitation CA patients.
Collapse
Affiliation(s)
- Qiang Zhao
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Yuntian Shen
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ran Li
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jiangbo Wu
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jingjun Lyu
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Department of Emergency Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Maorong Jiang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Liping Lu
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Minghua Zhu
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Wei Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Zhuoran Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Qiang Liu
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Ulrike Hoffmann
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jörn Karhausen
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Huaxin Sheng
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Weiguo Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
13
|
Smida T, Koller AC, Menegazzi JJ, Salcido DD. Early cytotoxic lymphocyte localization to the brain following resuscitation in a porcine model of asphyxial cardiac arrest: A pilot study. Resusc Plus 2021; 6:100125. [PMID: 34223383 PMCID: PMC8244478 DOI: 10.1016/j.resplu.2021.100125] [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: 02/05/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Out-of-hospital cardiac arrest (OHCA) is a major cause of morbidity and mortality in the US. Of major concern is a lack of therapies to mitigate associated brain injury. Immune cell infiltration (ICI) into the brain, which may exacerbate injury post-resuscitation, is one possible therapeutic target, although the post-OHCA immune response has not been fully characterized. Objective In this pilot study, we aimed to detect early post-resuscitation cytotoxic lymphocyte ICI in porcine brain using a model of opioid-mediated asphyxial OHCA. Methods Ten young, healthy swine (26.7+/-3.4 kg) were sedated, anaesthetized and paralyzed. In eight of the animals, this was followed by induction of asphyxial OHCA via fentanyl bolus and concurrent airway occlusion. The remaining two ‘sham’ animals were instrumented but did not undergo asphyxia. After nine minutes of asphyxia, mechanical CPR and manual ventilations were started, in an initial BLS followed by ALS configuration. At termination of resuscitation or euthanasia, the whole brain was removed. Immune cells were extracted and analyzed via flow cytometry. Results 304 +/− 62.2 cells/g were discovered to be CD8 single positive cells in animals that achieved ROSC, 481 +/− 274.4 cells/g in animals that did not achieve ROSC, and 40 +/− 11.31 cells/g in sham animals. CD8 single positive cells made up 0.473 +/− 0.24% of detected cells in animals that achieved ROSC, 0.395 +/− 0.062% in animals that did not achieve ROSC, and 0.19 +/− 0.014% in sham animals (No ROSC vs Sham, p = 0.012). Conclusions These data suggest that cytotoxic lymphocytes may be localizing to the brain during cardiac arrest resuscitation.
Collapse
Affiliation(s)
- Tanner Smida
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Allison C Koller
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - James J Menegazzi
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - David D Salcido
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Lei TY, Ye YZ, Zhu XQ, Smerin D, Gu LJ, Xiong XX, Zhang HF, Jian ZH. The immune response of T cells and therapeutic targets related to regulating the levels of T helper cells after ischaemic stroke. J Neuroinflammation 2021; 18:25. [PMID: 33461586 PMCID: PMC7814595 DOI: 10.1186/s12974-020-02057-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Through considerable effort in research and clinical studies, the immune system has been identified as a participant in the onset and progression of brain injury after ischaemic stroke. Due to the involvement of all types of immune cells, the roles of the immune system in stroke pathology and associated effects are complicated. Past research concentrated on the functions of monocytes and neutrophils in the pathogenesis of ischaemic stroke and tried to demonstrate the mechanisms of tissue injury and protection involving these immune cells. Within the past several years, an increasing number of studies have elucidated the vital functions of T cells in the innate and adaptive immune responses in both the acute and chronic phases of ischaemic stroke. Recently, the phenotypes of T cells with proinflammatory or anti-inflammatory function have been demonstrated in detail. T cells with distinctive phenotypes can also influence cerebral inflammation through various pathways, such as regulating the immune response, interacting with brain-resident immune cells and modulating neurogenesis and angiogenesis during different phases following stroke. In view of the limited treatment options available following stroke other than tissue plasminogen activator therapy, understanding the function of immune responses, especially T cell responses, in the post-stroke recovery period can provide a new therapeutic direction. Here, we discuss the different functions and temporal evolution of T cells with different phenotypes during the acute and chronic phases of ischaemic stroke. We suggest that modulating the balance between the proinflammatory and anti-inflammatory functions of T cells with distinct phenotypes may become a potential therapeutic approach that reduces the mortality and improves the functional outcomes and prognosis of patients suffering from ischaemic stroke.
Collapse
Affiliation(s)
- Tian-Yu Lei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ying-Ze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xi-Qun Zhu
- Department of Head and Neck and Neurosurgery, Hubei Cancer Hospital, Wuhan, 430079, Hubei Province, People's Republic of China
| | - Daniel Smerin
- University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Li-Juan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xiao-Xing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Zhi-Hong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
15
|
Hayashida K, Miyara SJ, Shinozaki K, Takegawa R, Yin T, Rolston DM, Choudhary RC, Guevara S, Molmenti EP, Becker LB. Inhaled Gases as Therapies for Post-Cardiac Arrest Syndrome: A Narrative Review of Recent Developments. Front Med (Lausanne) 2021; 7:586229. [PMID: 33585501 PMCID: PMC7873953 DOI: 10.3389/fmed.2020.586229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/04/2020] [Indexed: 01/22/2023] Open
Abstract
Despite recent advances in the management of post-cardiac arrest syndrome (PCAS), the survival rate, without neurologic sequelae after resuscitation, remains very low. Whole-body ischemia, followed by reperfusion after cardiac arrest (CA), contributes to PCAS, for which established pharmaceutical interventions are still lacking. It has been shown that a number of different processes can ultimately lead to neuronal injury and cell death in the pathology of PCAS, including vasoconstriction, protein modification, impaired mitochondrial respiration, cell death signaling, inflammation, and excessive oxidative stress. Recently, the pathophysiological effects of inhaled gases including nitric oxide (NO), molecular hydrogen (H2), and xenon (Xe) have attracted much attention. Herein, we summarize recent literature on the application of NO, H2, and Xe for treating PCAS. Recent basic and clinical research has shown that these gases have cytoprotective effects against PCAS. Nevertheless, there are likely differences in the mechanisms by which these gases modulate reperfusion injury after CA. Further preclinical and clinical studies examining the combinations of standard post-CA care and inhaled gas treatment to prevent ischemia-reperfusion injury are warranted to improve outcomes in patients who are being failed by our current therapies.
Collapse
Affiliation(s)
- Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Ryosuke Takegawa
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Daniel M Rolston
- Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Department of Surgery, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Rishabh C Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Sara Guevara
- Department of Surgery, Northwell Health, Manhasset, NY, United States
| | - Ernesto P Molmenti
- Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| |
Collapse
|
16
|
Abstract
Cardiac arrest is a catastrophic event with high morbidity and mortality. Despite advances over time in cardiac arrest management and postresuscitation care, the neurologic consequences of cardiac arrest are frequently devastating to patients and their families. Targeted temperature management is an intervention aimed at limiting postanoxic injury and improving neurologic outcomes following cardiac arrest. Recovery of neurologic function governs long-term outcome after cardiac arrest and prognosticating on the potential for recovery is a heavy burden for physicians. An early and accurate estimate of the potential for recovery can establish realistic expectations and avoid futile care in those destined for a poor outcome. This chapter reviews the epidemiology, pathophysiology, therapeutic interventions, prognostication, and neurologic sequelae of cardiac arrest.
Collapse
Affiliation(s)
- Rick Gill
- Department of Neurology, Loyola University Chicago, Chicago, Stritch School of Medicine, Maywood, IL, United States
| | - Michael Teitcher
- Department of Neurology, Loyola University Chicago, Chicago, Stritch School of Medicine, Maywood, IL, United States
| | - Sean Ruland
- Department of Neurology, Loyola University Chicago, Chicago, Stritch School of Medicine, Maywood, IL, United States.
| |
Collapse
|
17
|
Tao T, Liu M, Chen M, Luo Y, Wang C, Xu T, Jiang Y, Guo Y, Zhang JH. Natural medicine in neuroprotection for ischemic stroke: Challenges and prospective. Pharmacol Ther 2020; 216:107695. [DOI: 10.1016/j.pharmthera.2020.107695] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
|
18
|
Hosseini M, Wilson RH, Crouzet C, Amirhekmat A, Wei KS, Akbari Y. Resuscitating the Globally Ischemic Brain: TTM and Beyond. Neurotherapeutics 2020; 17:539-562. [PMID: 32367476 PMCID: PMC7283450 DOI: 10.1007/s13311-020-00856-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrest (CA) afflicts ~ 550,000 people each year in the USA. A small fraction of CA sufferers survive with a majority of these survivors emerging in a comatose state. Many CA survivors suffer devastating global brain injury with some remaining indefinitely in a comatose state. The pathogenesis of global brain injury secondary to CA is complex. Mechanisms of CA-induced brain injury include ischemia, hypoxia, cytotoxicity, inflammation, and ultimately, irreversible neuronal damage. Due to this complexity, it is critical for clinicians to have access as early as possible to quantitative metrics for diagnosing injury severity, accurately predicting outcome, and informing patient care. Current recommendations involve using multiple modalities including clinical exam, electrophysiology, brain imaging, and molecular biomarkers. This multi-faceted approach is designed to improve prognostication to avoid "self-fulfilling" prophecy and early withdrawal of life-sustaining treatments. Incorporation of emerging dynamic monitoring tools such as diffuse optical technologies may provide improved diagnosis and early prognostication to better inform treatment. Currently, targeted temperature management (TTM) is the leading treatment, with the number of patients needed to treat being ~ 6 in order to improve outcome for one patient. Future avenues of treatment, which may potentially be combined with TTM, include pharmacotherapy, perfusion/oxygenation targets, and pre/postconditioning. In this review, we provide a bench to bedside approach to delineate the pathophysiology, prognostication methods, current targeted therapies, and future directions of research surrounding hypoxic-ischemic brain injury (HIBI) secondary to CA.
Collapse
Affiliation(s)
- Melika Hosseini
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Robert H Wilson
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Christian Crouzet
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Arya Amirhekmat
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Kevin S Wei
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Yama Akbari
- Department of Neurology, School of Medicine, University of California, Irvine, USA.
- Beckman Laser Institute, University of California, Irvine, USA.
| |
Collapse
|
19
|
Lin QM, Tang XH, Lin SR, Chen BD, Chen F. Bone marrow-derived mesenchymal stem cell transplantation attenuates overexpression of inflammatory mediators in rat brain after cardiopulmonary resuscitation. Neural Regen Res 2020; 15:324-331. [PMID: 31552906 PMCID: PMC6905325 DOI: 10.4103/1673-5374.265563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Emerging evidence suggests that bone marrow-derived mesenchymal stem cell transplantation improves neurological function after cardiac arrest and cardiopulmonary resuscitation; however, the precise mechanisms remain unclear. This study aimed to investigate the effect of bone marrow-derived mesenchymal stem cell treatment on expression profiles of multiple cytokines in the brain after cardiac arrest and cardiopulmonary resuscitation. Cardiac arrest was induced in rats by asphyxia and cardiopulmonary resuscitation was initiated 6 minutes after cardiac arrest. One hour after successful cardiopulmonary resuscitation, rats were injected with either phosphate-buffered saline (control) or 1 × 106 bone marrow-derived mesenchymal stem cells via the tail vein. Serum S100B levels were measured by enzyme-linked immunosorbent assay and neurological deficit scores were evaluated to assess brain damage at 3 days after cardiopulmonary resuscitation. Serum S100B levels were remarkably decreased and neurological deficit scores were obviously improved in the mesenchymal stem cell group compared with the phosphate-buffered saline group. Brains were isolated from the rats and expression levels of 90 proteins were determined using a RayBio Rat Antibody Array, to investigate the cytokine profiles. Brain levels of the inflammatory mediators tumor necrosis factor-α, interferon-γ, macrophage inflammatory protein-1α, macrophage inflammatory protein-2, macrophage inflammatory protein-3α, macrophage-derived chemokine, and matrix metalloproteinase-2 were decreased ≥ 1.5-fold, while levels of the anti-inflammatory factor interleukin-10 were increased ≥ 1.5-fold in the mesenchymal stem cell group compared with the control group. Donor mesenchymal stem cells were detected by immunofluorescence to determine their distribution in the damaged brain, and were primarily observed in the cerebral cortex. These results indicate that bone marrow-derived mesenchymal stem cell transplantation attenuates brain damage induced by cardiac arrest and cardiopulmonary resuscitation, possibly via regulation of inflammatory mediators. This experimental protocol was approved by the Institutional Animal Care and Use Committee of Fujian Medical University, China in January 2016 (approval No. 2016079).
Collapse
Affiliation(s)
- Qing-Ming Lin
- Institute of Fujian Emergency Medicine, Clinical College of Fujian Medical University; Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Emergency Center, Fuzhou, Fujian Province, China
| | - Xia-Hong Tang
- Institute of Fujian Emergency Medicine, Clinical College of Fujian Medical University; Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Emergency Center, Fuzhou, Fujian Province, China
| | - Shi-Rong Lin
- Institute of Fujian Emergency Medicine, Clinical College of Fujian Medical University; Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Emergency Center, Fuzhou, Fujian Province, China
| | - Ben-Dun Chen
- Institute of Fujian Emergency Medicine, Clinical College of Fujian Medical University; Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Emergency Center, Fuzhou, Fujian Province, China
| | - Feng Chen
- Institute of Fujian Emergency Medicine, Clinical College of Fujian Medical University; Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Emergency Center, Fuzhou, Fujian Province, China
| |
Collapse
|
20
|
Dietz RM, Cruz-Torres I, Orfila JE, Patsos OP, Shimizu K, Chalmers N, Deng G, Tiemeier E, Quillinan N, Herson PS. Reversal of Global Ischemia-Induced Cognitive Dysfunction by Delayed Inhibition of TRPM2 Ion Channels. Transl Stroke Res 2019; 11:254-266. [PMID: 31250378 DOI: 10.1007/s12975-019-00712-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/23/2022]
Abstract
Hippocampal injury and cognitive impairments are common after cardiac arrest and stroke and do not have an effective intervention despite much effort. Therefore, we developed a new approach aimed at reversing synaptic dysfunction by targeting TRPM2 channels. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in mice was used to investigate cognitive deficits and the role of the calcium-permeable ion channel transient receptor potential-M2 (TRPM2) in ischemia-induced synaptic dysfunction. Our data indicates that absence (TRPM2-/-) or acute inhibition of TRPM2 channels with tatM2NX reduced hippocampal cell death in males only, but prevented synaptic plasticity deficits in both sexes. Remarkably, administration of tatM2NX weeks after injury reversed hippocampal plasticity and memory deficits. Finally, TRPM2-dependent activation of calcineurin-GSK3β pathway contributes to synaptic plasticity impairments. These data suggest persistent TRPM2 activity following ischemia contributes to impairments of the surviving hippocampal network and that inhibition of TRPM2 channels at chronic time points may represent a novel strategy to improve functional recovery following cerebral ischemia that is independent of neuroprotection.
Collapse
Affiliation(s)
- Robert M Dietz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ivelisse Cruz-Torres
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - James E Orfila
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Olivia P Patsos
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kaori Shimizu
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicholas Chalmers
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Guiying Deng
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erika Tiemeier
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nidia Quillinan
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paco S Herson
- Neuronal Injury & Plasticity Program, University of Colorado School of Medicine, Aurora, CO, USA. .,Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA. .,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
21
|
Ryzhov S, May T, Dziodzio J, Emery IF, Lucas FL, Leclerc A, McCrum B, Lord C, Eldridge A, Robich MP, Ichinose F, Sawyer DB, Riker R, Seder DB. Number of Circulating CD 73-Expressing Lymphocytes Correlates With Survival After Cardiac Arrest. J Am Heart Assoc 2019; 8:e010874. [PMID: 31237169 PMCID: PMC6662342 DOI: 10.1161/jaha.118.010874] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Patients resuscitated from cardiac arrest (CA) have highly variable neurological, circulatory, and systemic ischemia‐reperfusion injuries. After the initial hypoxic‐ischemic insult, a cascade of immune and inflammatory responses develops and is often fatal. The role of the immune response in pathophysiological characteristics and recovery is not well understood. We studied immune cell activity and its association with outcomes in a cohort of CA survivors. Methods and Results After informed consent, we collected blood samples at intervals over a week after resuscitation from CA. We examined the expression of CD39 and CD73 (alias 5′‐nucleotidase), production of tumor necrosis factor‐α, generation of reactive oxygen species, and secretion of vascular endothelial growth factor by circulating myeloid and lymphoid cells, in comparison to cells obtained from control subjects before coronary artery bypass grafting surgery. The number of circulating total and CD73‐expressing lymphocytes correlated with survival after CA. Incubation of immune cells, obtained from post‐CA subjects, with AMP, a substrate for CD73, resulted in inhibition of tumor necrosis factor‐α production and generation of reactive oxygen species. This effect was blocked by adenosine 5′‐(α, β‐methylene) diphosphate, a specific inhibitor of CD73 and ZM 241385, an A2 adenosine receptor antagonist. We also found that AMP‐dependent activation of CD73 induces production of vascular endothelial growth factor. Conclusions CD73‐expressing lymphocytes mediate cellular protection from inflammation after CA through inhibition of proinflammatory activation of myeloid cells and promotion of vascular endothelial growth factor secretion. The contribution of CD73 lymphocytes in the regulation of acute inflammation and tissue injury after CA warrants further study.
Collapse
Affiliation(s)
- Sergey Ryzhov
- 1 Maine Medical Center Research Institute Scarborough ME
| | - Teresa May
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| | - John Dziodzio
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Ivette F Emery
- 1 Maine Medical Center Research Institute Scarborough ME
| | - F L Lucas
- 3 Center for Outcomes Research and Evaluation Maine Medical Center Portland ME
| | - Angela Leclerc
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Barbara McCrum
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Christine Lord
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Ashley Eldridge
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Michel P Robich
- 1 Maine Medical Center Research Institute Scarborough ME.,4 Maine Medical Center Cardiovascular Institute Portland ME
| | - Fumito Ichinose
- 5 Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston MA
| | - Douglas B Sawyer
- 1 Maine Medical Center Research Institute Scarborough ME.,4 Maine Medical Center Cardiovascular Institute Portland ME
| | - Richard Riker
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| | - David B Seder
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| |
Collapse
|
22
|
Don-Doncow N, Vanherle L, Zhang Y, Meissner A. T-Cell Accumulation in the Hypertensive Brain: A Role for Sphingosine-1-Phosphate-Mediated Chemotaxis. Int J Mol Sci 2019; 20:ijms20030537. [PMID: 30695999 PMCID: PMC6386943 DOI: 10.3390/ijms20030537] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
Hypertension is considered the major modifiable risk factor for the development of cognitive impairment. Because increased blood pressure is often accompanied by an activation of the immune system, the concept of neuro-inflammation gained increasing attention in the field of hypertension-associated neurodegeneration. Particularly, hypertension-associated elevated circulating T-lymphocyte populations and target organ damage spurred the interest to understanding mechanisms leading to inflammation-associated brain damage during hypertension. The present study describes sphingosine-1-phosphate (S1P) as major contributor to T-cell chemotaxis to the brain during hypertension-associated neuro-inflammation and cognitive impairment. Using Western blotting, flow cytometry and mass spectrometry approaches, we show that hypertension stimulates a sphingosine kinase 1 (SphK1)-dependent increase of cerebral S1P concentrations in a mouse model of angiotensin II (AngII)-induced hypertension. The development of a distinct S1P gradient between circulating blood and brain tissue associates to elevated CD3+ T-cell numbers in the brain. Inhibition of S1P1-guided T-cell chemotaxis with the S1P receptor modulator FTY720 protects from augmentation of brain CD3 expression and the development of memory deficits in hypertensive WT mice. In conclusion, our data highlight a new approach to the understanding of hypertension-associated inflammation in degenerative processes of the brain during disease progression.
Collapse
Affiliation(s)
| | - Lotte Vanherle
- Department of Experimental Science, Lund University, 22 184 Lund, Sweden.
| | - Yun Zhang
- Department of Experimental Science, Lund University, 22 184 Lund, Sweden.
| | - Anja Meissner
- Department of Experimental Science, Lund University, 22 184 Lund, Sweden.
- Wallenberg Center for Molecular Medicine, Lund University, 22 184 Lund, Sweden.
| |
Collapse
|
23
|
Nguyen PL, Alreshaid L, Poblete RA, Konye G, Marehbian J, Sung G. Targeted Temperature Management and Multimodality Monitoring of Comatose Patients After Cardiac Arrest. Front Neurol 2018; 9:768. [PMID: 30254606 PMCID: PMC6141756 DOI: 10.3389/fneur.2018.00768] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/24/2018] [Indexed: 01/14/2023] Open
Abstract
Out-of-hospital cardiac arrest (CA) remains a leading cause of sudden morbidity and mortality; however, outcomes have continued to improve in the era of targeted temperature management (TTM). In this review, we highlight the clinical use of TTM, and provide an updated summary of multimodality monitoring possible in a modern ICU. TTM is neuroprotective for survivors of CA by inhibiting multiple pathophysiologic processes caused by anoxic brain injury, with a final common pathway of neuronal death. Current guidelines recommend the use of TTM for out-of-hospital CA survivors who present with a shockable rhythm. Further studies are being completed to determine the optimal timing, depth and duration of hypothermia to optimize patient outcomes. Although a multidisciplinary approach is necessary in the CA population, neurologists and neurointensivists are central in selecting TTM candidates and guiding patient care and prognostic evaluation. Established prognostic tools include clinal exam, SSEP, EEG and MR imaging, while functional MRI and invasive monitoring is not validated to improve outcomes in CA or aid in prognosis. We recommend that an evidence-based TTM and prognostication algorithm be locally implemented, based on each institution's resources and limitations. Given the high incidence of CA and difficulty in predicting outcomes, further study is urgently needed to determine the utility of more recent multimodality devices and studies.
Collapse
Affiliation(s)
- Peggy L Nguyen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Laith Alreshaid
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Roy A Poblete
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Geoffrey Konye
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jonathan Marehbian
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Gene Sung
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
24
|
Zhang C, Brandon NR, Koper K, Tang P, Xu Y, Dou H. Invasion of Peripheral Immune Cells into Brain Parenchyma after Cardiac Arrest and Resuscitation. Aging Dis 2018; 9:412-425. [PMID: 29896429 PMCID: PMC5988596 DOI: 10.14336/ad.2017.0926] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
Although a direct link has long been suspected between systemic immune responses and neuronal injuries after stroke, it is unclear which immune cells play an important role. A question remains as to whether the blood brain barrier (BBB) is transiently disrupted after circulatory arrest to allow peripheral immune cells to enter brain parenchyma. Here, we developed a clinically relevant cardiac arrest and resuscitation model in mice to investigate the BBB integrity using noninvasive magnetic resonance imaging. Changes in immune signals in the brain and periphery were assayed by immunohistochemistry and flow cytometry. Quantitative variance maps from T1-weighted difference images before and after blood-pool contrast clearance revealed BBB disruptions immediately after resuscitation and one day after reperfusion. Time profiles of hippocampal CA1 neuronal injuries correlated with the morphological changes of microglia activation. Cytotoxic T cells, CD11b+CD11c+ dendritic cells, and CD11b+CD45+hi monocytes and macrophages were significantly increased in the brain three days after cardiac arrest and resuscitation, suggesting direct infiltration of these cells following the BBB disruption. Importantly, these immune cell changes were coupled with a parallel increase in the same subset of immune cell populations in the bone marrow and blood. We conclude that neurovascular breakdown during the initial reperfusion phase contributes to the systemic immune cell invasion and subsequent neuropathogenesis affecting the long-term outcome after cardiac arrest and resuscitation.
Collapse
Affiliation(s)
| | | | | | - Pei Tang
- 1Departments of Anesthesiology.,2Pharmacology and Chemical Biology.,3Computational and Systems Biology
| | - Yan Xu
- 1Departments of Anesthesiology.,2Pharmacology and Chemical Biology.,4Physics and Astronomy, and.,5Structural Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huanyu Dou
- 6Department of Biomedical Sciences, Paul L. Foster School of Medicine, and.,7Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| |
Collapse
|
25
|
Tahsili-Fahadan P, Farrokh S, Geocadin RG. Hypothermia and brain inflammation after cardiac arrest. Brain Circ 2018; 4:1-13. [PMID: 30276330 PMCID: PMC6057700 DOI: 10.4103/bc.bc_4_18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/17/2018] [Accepted: 03/18/2018] [Indexed: 12/14/2022] Open
Abstract
The cessation (ischemia) and restoration (reperfusion) of cerebral blood flow after cardiac arrest (CA) induce inflammatory processes that can result in additional brain injury. Therapeutic hypothermia (TH) has been proven as a brain protective strategy after CA. In this article, the underlying pathophysiology of ischemia-reperfusion brain injury with emphasis on the role of inflammatory mechanisms is reviewed. Potential targets for immunomodulatory treatments and relevant effects of TH are also discussed. Further studies are needed to delineate the complex pathophysiology and interactions among different components of immune response after CA and identify appropriate targets for clinical investigations.
Collapse
Affiliation(s)
- Pouya Tahsili-Fahadan
- Department of Medicine, Virginia Commonwealth University, Falls Church, Virginia, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Salia Farrokh
- Department of Pharmacy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Romergryko G Geocadin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Kragsterman B, Bergqvist D, Siegbahn A, Parsson H. Carotid Endarterectomy Induces the Release of Inflammatory Markers and the Activation of Coagulation as Measured in the Jugular Bulb. J Stroke Cerebrovasc Dis 2017; 26:2320-2328. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/07/2017] [Accepted: 05/13/2017] [Indexed: 12/12/2022] Open
|
27
|
Villois P, Grimaldi D, Spadaro S, Shinotsuka CR, Fontana V, Scolletta S, Franchi F, Vincent JL, Creteur J, Taccone FS. Lymphopaenia in cardiac arrest patients. Ann Intensive Care 2017; 7:85. [PMID: 28808927 PMCID: PMC5555958 DOI: 10.1186/s13613-017-0308-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/06/2017] [Indexed: 12/29/2022] Open
Abstract
Background A decrease in circulating lymphocytes has been described as a marker of poor prognosis after septic shock; however, scarce data are available after cardiac arrest (CA). The aim of this study was to evaluate the impact of lymphopaenia after successful cardiopulmonary resuscitation. Methods This is a retrospective analysis of an institutional database including all adult CA patients admitted to the intensive care unit (ICU) between January 2007 and December 2014 who survived for at least 24 h. Demographic, CA-related data and ICU mortality were recorded as was lymphocyte count on admission and for the first 48 h. A cerebral performance category score of 3–5 at 3 months was considered as an unfavourable neurological outcome. Results Data from 377 patients were analysed (median age: 62 [IQRs: 52–75] years). Median time to return of spontaneous circulation (ROSC) was 15 [8–25] min and 232 (62%) had a non-shockable initial rhythm. ICU mortality was 58% (n = 217) and 246 (65%) patients had an unfavourable outcome at 3 months. The median lymphocyte count on admission was 1208 [700–2350]/mm3 and 151 (40%) patients had lymphopaenia (lymphocyte count <1000/mm3). Predictors of lymphopaenia on admission were older age, a shorter time to ROSC, prior use of corticosteroid therapy and high C-reactive protein levels on admission. ICU non-survivors had lower lymphocyte counts on admission than survivors (1100 [613–2317] vs. 1316 [891–2395]/mm3; p = 0.05) as did patients with unfavourable compared to those with favourable neurological outcomes (1100 [600–2013] vs. 1350 [919–2614]/mm3; p = 0.003). However, lymphopaenia on admission was not an independent predictor of poor outcomes in the entire population, but only among OHCA patients. Conclusions A low lymphocyte count is common in CA survivors and is associated with poor outcome after OHCA. Electronic supplementary material The online version of this article (doi:10.1186/s13613-017-0308-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola Villois
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - David Grimaldi
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Savino Spadaro
- Department of Morphological Surgery and Experimental Medicine, Arcispedale Sant'Anna, Università di Ferrara, Via AldoMoro, 8, 44121, Ferrara, Italy
| | - Claudia Righy Shinotsuka
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Vito Fontana
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Sabino Scolletta
- Department of Anesthesia and Intensive Care, Policlinico Santa Maria alle Scotte, Universitá di Siena, Viale Bracci, 14, 53100, Siena, Italy
| | - Federico Franchi
- Department of Anesthesia and Intensive Care, Policlinico Santa Maria alle Scotte, Universitá di Siena, Viale Bracci, 14, 53100, Siena, Italy
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium.
| |
Collapse
|
28
|
Vaitaitis GM, Waid DM, Yussman MG, Wagner DH. CD40-mediated signalling influences trafficking, T-cell receptor expression, and T-cell pathogenesis, in the NOD model of type 1 diabetes. Immunology 2017; 152:243-254. [PMID: 28542921 DOI: 10.1111/imm.12761] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 12/23/2022] Open
Abstract
CD40 plays a critical role in the pathogenesis of type 1 diabetes (T1D). The mechanism of action, however, is undetermined, probably because CD40 expression has been grossly underestimated. CD40 is expressed on numerous cell types that now include T cells and pancreatic β cells. CD40+ CD4+ cells [T helper type 40 (TH40)] prove highly pathogenic in NOD mice and in translational human T1D studies. We generated BDC2.5.CD40-/- and re-derived NOD.CD154-/- mice to better understand the CD40 mechanism of action. Fully functional CD40 expression is required not only for T1D development but also for insulitis. In NOD mice, TH40 cell expansion in pancreatic lymph nodes occurs before insulitis and demonstrates an activated phenotype compared with conventional CD4+ cells, apparently regardless of antigen specificity. TH40 T-cell receptor (TCR) usage demonstrates increases in several Vα and Vβ species, particularly Vα3.2+ that arise early and are sustained throughout disease development. TH40 cells isolated from diabetic pancreas demonstrate a relatively broad TCR repertoire rather than restricted clonal expansions. The expansion of the Vα/Vβ species associated with diabetes depends upon CD40 signalling; NOD.CD154-/- mice do not expand the same TCR species. Finally, CD40-mediated signals significantly increase pro-inflammatory Th1- and Th17-associated cytokines whereas CD28 co-stimulus alternatively promotes regulatory cytokines.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dan M Waid
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Martin G Yussman
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David H Wagner
- The Webb-Waring Center, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Division of Pulmonary Sciences and Critical Care, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
29
|
Neurologic Recovery After Cardiac Arrest: a Multifaceted Puzzle Requiring Comprehensive Coordinated Care. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:52. [PMID: 28536893 DOI: 10.1007/s11936-017-0548-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Surviving cardiac arrest (CA) requires a longitudinal approach with multiple levels of responsibility, including fostering a culture of action by increasing public awareness and training, optimization of resuscitation measures including frequent updates of guidelines and their timely implementation into practice, and optimization of post-CA care. This clearly goes beyond resuscitation and targeted temperature management. Brain-directed physiologic goals should dictate the post-CA management, as accumulating evidence suggests that the degree of hypoxic brain injury is the main determinant of survival, regardless of the etiology of arrest. Early assessment of the need for further hemodynamic and electrophysiologic cardiac interventions, adjusting ventilator settings to avoid hyperoxia/hypoxia while targeting high-normal to mildly elevated PaCO2, maintaining mean arterial blood pressures >65 mmHg, evaluating for and treating seizures, maintaining euglycemia, and aggressively pursuing normothermia are key steps in reducing the bioenergetic failure that underlies secondary brain injury. Accurate neuroprognostication requires a multimodal approach with standardized assessments accounting for confounders while recognizing the importance of a delayed prognostication when there is any uncertainty regarding outcome. The concept of a highly specialized post-CA team with expertise in the management of post-CA syndrome (mindful of the brain-directed physiologic goals during the early post-resuscitation phase), TTM, and neuroprognostication, guiding the comprehensive care to the CA survivor, is likely cost-effective and should be explored by institutions that frequently care for these patients. Finally, providing tailored rehabilitation care with systematic reassessment of the needs and overall goals is key for increasing independence and improving quality-of-life in survivors, thereby also alleviating the burden on families. Emerging evidence from multicenter collaborations advances the field of resuscitation at an incredible pace, challenging previously well-established paradigms. There is no more room for "conventional wisdom" in saving the survivors of cardiac arrest.
Collapse
|
30
|
Vaitaitis GM, Yussman MG, Waid DM, Wagner DH. Th40 cells (CD4+CD40+ Tcells) drive a more severe form of Experimental Autoimmune Encephalomyelitis than conventional CD4 T cells. PLoS One 2017; 12:e0172037. [PMID: 28192476 PMCID: PMC5305068 DOI: 10.1371/journal.pone.0172037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/30/2017] [Indexed: 12/31/2022] Open
Abstract
CD40-CD154 interaction is critically involved in autoimmune diseases, and CD4 T cells play a dominant role in the Experimental Autoimmune Encephalomyelitis (EAE) model of Multiple Sclerosis (MS). CD4 T cells expressing CD40 (Th40) are pathogenic in type I diabetes but have not been evaluated in EAE. We demonstrate here that Th40 cells drive a rapid, more severe EAE disease course than conventional CD4 T cells. Adoptively transferred Th40 cells are present in lesions in the CNS and are associated with wide spread demyelination. Primary Th40 cells from EAE-induced donors adoptively transfer EAE without further in-vitro expansion and without requiring the administration of the EAE induction regimen to the recipient animals. This has not been accomplished with primary, non-TCR-transgenic donor cells previously. If co-injection of Th40 donor cells with Freund's adjuvant (CFA) in the recipient animals is done, the disease course is more severe. The CFA component of the EAE induction regimen causes generalized inflammation, promoting expansion of Th40 cells and infiltration of the CNS, while MOG-antigen shapes the antigen-specific TCR repertoire. Those events are both necessary to precipitate disease. In MS, viral infections or trauma may induce generalized inflammation in susceptible individuals with subsequent disease onset. It will be important to further understand the events leading up to disease onset and to elucidate the contributions of the Th40 T cell subset. Also, evaluating Th40 levels as predictors of disease onset would be highly useful because if either the generalized inflammation event or the TCR-honing can be interrupted, disease onset may be prevented.
Collapse
Affiliation(s)
- Gisela M. Vaitaitis
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Martin G. Yussman
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Dan M. Waid
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - David H. Wagner
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
31
|
Reis C, Akyol O, Araujo C, Huang L, Enkhjargal B, Malaguit J, Gospodarev V, Zhang JH. Pathophysiology and the Monitoring Methods for Cardiac Arrest Associated Brain Injury. Int J Mol Sci 2017; 18:ijms18010129. [PMID: 28085069 PMCID: PMC5297763 DOI: 10.3390/ijms18010129] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/31/2016] [Accepted: 01/04/2017] [Indexed: 12/23/2022] Open
Abstract
Cardiac arrest (CA) is a well-known cause of global brain ischemia. After CA and subsequent loss of consciousness, oxygen tension starts to decline and leads to a series of cellular changes that will lead to cellular death, if not reversed immediately, with brain edema as a result. The electroencephalographic activity starts to change as well. Although increased intracranial pressure (ICP) is not a direct result of cardiac arrest, it can still occur due to hypoxic-ischemic encephalopathy induced changes in brain tissue, and is a measure of brain edema after CA and ischemic brain injury. In this review, we will discuss the pathophysiology of brain edema after CA, some available techniques, and methods to monitor brain oxygen, electroencephalography (EEG), ICP (intracranial pressure), and microdialysis on its measurement of cerebral metabolism and its usefulness both in clinical practice and possible basic science research in development. With this review, we hope to gain knowledge of the more personalized information about patient status and specifics of their brain injury, and thus facilitating the physicians’ decision making in terms of which treatments to pursue.
Collapse
Affiliation(s)
- Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Onat Akyol
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Camila Araujo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Jay Malaguit
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Vadim Gospodarev
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
32
|
Wagner DH. Overlooked Mechanisms in Type 1 Diabetes Etiology: How Unique Costimulatory Molecules Contribute to Diabetogenesis. Front Endocrinol (Lausanne) 2017; 8:208. [PMID: 28878738 PMCID: PMC5572340 DOI: 10.3389/fendo.2017.00208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/08/2017] [Indexed: 01/16/2023] Open
Abstract
Type 1 Diabetes (T1D) develops when immune cells invade the pancreatic islets resulting in loss of insulin production in beta cells. T cells have been proven to be central players in that process. What is surprising, however, is that classic mechanisms of tolerance cannot explain diabetogenesis; alternate mechanisms must now be considered. T cell receptor (TCR) revision is the process whereby T cells in the periphery alter TCR expression, outside the safety-net of thymic selection pressures. This process results in an expanded T cell repertoire, capable of responding to a universe of pathogens, but limitations are that increased risk for autoimmune disease development occurs. Classic T cell costimulators including the CD28 family have long been thought to be the major drivers for full T cell activation. In actuality, CD28 and its family member counterparts, ICOS and CTLA-4, all drive regulatory responses. Inflammation is driven by CD40, not CD28. CD40 as a costimulus has been largely overlooked. When naïve T cells interact with antigen presenting cell CD154, the major ligand for CD40, is induced. This creates a milieu for T cell (CD40)-T cell (CD154) interaction, leading to inflammation. Finally, defined pathogenic effector cells including TH40 (CD4+CD40+) cells can express FOXP3 but are not Tregs. The cells loose FOXP3 to become pathogenic effector cells. Each of these mechanisms creates novel options to better understand diabetogenesis and create new therapeutic targets for T1D.
Collapse
Affiliation(s)
- David H. Wagner
- The Program in Integrated Immunology, Department of Medicine, Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: David H. Wagner Jr.,
| |
Collapse
|
33
|
Wagner DH. Of the multiple mechanisms leading to type 1 diabetes, T cell receptor revision may play a prominent role (is type 1 diabetes more than a single disease?). Clin Exp Immunol 2016; 185:271-80. [PMID: 27271348 DOI: 10.1111/cei.12819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/20/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022] Open
Abstract
A single determinant factor for autoimmunity does not exist; disease development probably involves contributions from genetics, the environment and immune dysfunction. Type 1 diabetes is no exception. Genomewide-associated studies (GWAS) analysis in T1D has proved disappointing in revealing contributors to disease prediction; the only reliable marker has been human leucocyte antigen (HLA). Specific HLAs include DR3/DR4/DQ2/DQ8, for example. Because HLA molecules present antigen to T cells, it is reasonable that certain HLA molecules have a higher affinity to present self-antigen. Recent studies have shown that additional polymorphisms in HLA that are restricted to autoimmune conditions are further contributory. A caveat is that not all individuals with the appropriate 'pro-autoimmune' HLA develop an autoimmune disease. Another crucial component is autoaggressive T cells. Finding a biomarker to discriminate autoaggressive T cells has been elusive. However, a subset of CD4 helper cells that express the CD40 receptor have been described as becoming pathogenic. An interesting function of CD40 on T cells is to induce the recombination-activating gene (RAG)1/RAG2 T cell receptor recombination machinery. This observation is contrary to immunology paradigms that changes in TCR molecules cannot take place outside the thymic microenvironment. Alteration in TCR, called TCR revision, not only occurs, but may help to account for the development of autoaggressive T cells. Another interesting facet is that type 1 diabetes (T1D) may be more than a single disease; that is, multiple cellular components contribute uniquely, but result ultimately in the same clinical outcome, T1D. This review considers the process of T cell maturation and how that could favor auto-aggressive T cell development in T1D. The potential contribution of TCR revision to autoimmunity is also considered.
Collapse
Affiliation(s)
- D H Wagner
- Department of Medicine, Department of Neurology, Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
34
|
Xiang Y, Zhao H, Wang J, Zhang L, Liu A, Chen Y. Inflammatory mechanisms involved in brain injury following cardiac arrest and cardiopulmonary resuscitation. Biomed Rep 2016; 5:11-17. [PMID: 27330748 DOI: 10.3892/br.2016.677] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/25/2016] [Indexed: 12/24/2022] Open
Abstract
Cardiac arrest (CA) is a leading cause of fatality and long-term disability worldwide. Recent advances in cardiopulmonary resuscitation (CPR) have improved survival rates; however, the survivors are prone to severe neurological injury subsequent to successful CPR following CA. Effective therapeutic options to protect the brain from CA remain limited, due to the complexities of the injury cascades caused by global cerebral ischemia/reperfusion (I/R). Although the precise mechanisms of neurological impairment following CA-initiated I/R injury require further clarification, evidence supports that one of the key cellular pathways of cerebral injury is inflammation. The inflammatory response is orchestrated by activated glial cells in response to I/R injury. Increased release of danger-associated molecular pattern molecules and cellular dysfunction in activated microglia and astrocytes contribute to ischemia-induced cytotoxic and pro-inflammatory cytokines generation, and ultimately to delayed death of neurons. Furthermore, cytokines and adhesion molecules generated within activated microglia, as well as astrocytes, are involved in the innate immune response; modulate influx of peripheral immune and inflammatory cells into the brain, resulting in neurological injury. The present review discusses the molecular aspects of immune and inflammatory mechanisms in global cerebral I/R injury following CA and CPR, and the potential therapeutic strategies that target neuroinflammation and the innate immune system.
Collapse
Affiliation(s)
- Yanxiao Xiang
- Department of Clinical Pharmacy, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Emergency, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hua Zhao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jiali Wang
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Luetao Zhang
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Anchang Liu
- Department of Clinical Pharmacy, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuguo Chen
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
35
|
Secher N, Østergaard L, Iversen NK, Lambertsen KL, Clausen BH, Tønnesen E, Granfeldt A. Preserved Cerebral Microcirculation After Cardiac Arrest in a Rat Model. Microcirculation 2015; 22:464-74. [DOI: 10.1111/micc.12217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/23/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Niels Secher
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience; Aarhus University; Aarhus C Denmark
| | - Nina K. Iversen
- Center of Functionally Integrative Neuroscience; Aarhus University; Aarhus C Denmark
| | - Kate L. Lambertsen
- Department of Neurobiology Research; Institute of Molecular Medicine; University of Southern Denmark; Odense C Denmark
| | - Bettina H. Clausen
- Department of Neurobiology Research; Institute of Molecular Medicine; University of Southern Denmark; Odense C Denmark
| | - Else Tønnesen
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| | - Asger Granfeldt
- Department of Anaesthesiology and Intensive Care Medicine; Aarhus University Hospital; Aarhus C Denmark
| |
Collapse
|
36
|
Grace PM, Shimizu K, Strand KA, Rice KC, Traystman RJ, Watkins LR, Herson PS. (+)-Naltrexone is neuroprotective and promotes alternative activation in the mouse hippocampus after cardiac arrest/cardiopulmonary resuscitation. Brain Behav Immun 2015; 48:115-22. [PMID: 25774010 PMCID: PMC5548128 DOI: 10.1016/j.bbi.2015.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 10/23/2022] Open
Abstract
Despite dramatic improvement in cardiopulmonary resuscitation (CPR) and other techniques for cardiac arrest (CA), the majority of survivors continue to show signs of decreased memory or executive cognitive function. Such memory impairment may be due to hippocampal CA1 neuronal death, which is delayed by several days after CA/CPR. Classical microgliosis in the CA1 region may contribute to neuronal death, yet the role of a key activation receptor Toll Like Receptor 4 (TLR4) has not been previously investigated for such neuronal death after CA/CPR. We show that (+)-naltrexone was neuroprotective after CA/CPR. TLR4 blockade was associated with decreased expression of markers for microglial/macrophage activation and T cell and B cell infiltration, as well as decreased pro-inflammatory cytokine levels. Notably, IL-10 expression was elevated in response to CA/CPR, but was not attenuated by (+)-naltrexone, suggesting that the local monocyte/microglial phenotype had shifted towards alternative activation. This was confirmed by elevated expression of Arginase-1, and decreased expression of NFκB p65 subunit. Thus, (+)-naltrexone and other TLR4 antagonists may represent a novel therapeutic strategy to alleviate the substantial burden of memory or executive cognitive function impairment after CA/CPR.
Collapse
Affiliation(s)
- Peter M. Grace
- Department of Psychology and The Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Kaori Shimizu
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Keith A. Strand
- Department of Psychology and The Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Kenner C. Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - Richard J. Traystman
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Linda R. Watkins
- Department of Psychology and The Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Paco S. Herson
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
37
|
Abstract
Stroke results in cerebral inflammation that causes brain injury and triggers immunodepression, resulting in an increased incidence of morbidity and mortality secondary to remote infection. It is well known that T cells modulate brain inflammation after ischemic stroke, and targeting T cells may be an innovative therapeutic strategy for stroke treatment. T cell deficiency is neuro-protective, but the observed protective effects differ between ischemic models. Recent studies suggest different T cell subsets may have distinct effects on the injured brain. In addition to their role in cerebral inflammation, T cells also play a role in stroke-induced immunodepression. Therefore, T cell-targeted therapies designed to provide protection against brain inflammation might paradoxically contribute to remote organ infection and mortality. Further investigations are required to determine the role of specific T cell subsets in cerebral inflammation and stroke-induced immunodepression, the optimal therapeutic window for treatment, and the appropriate dose of anti-T cell therapy.
Collapse
Affiliation(s)
- Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | | | | | | |
Collapse
|