1
|
Xu S, Gu L, Bao B, Liu Q, Jin Q, Ma Y, Zhou S, Li B, Xu L, Guo G, Zhu J, Su KP, Sun P. Mechanistic insights into the neuroprotective effects of low-intensity transcranial ultrasound stimulation in post-cardiac arrest brain injury: Modulation of the Piezo1-Dkk3/PI3K-Akt pathway. Brain Behav Immun 2025; 127:341-357. [PMID: 40118226 DOI: 10.1016/j.bbi.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Post-cardiac arrest brain injury (PCABI) remains a significant challenge, marked by high mortality and disability rates due to persistent neuroinflammation. This study explored the neuroprotective potential of low-intensity transcranial ultrasound stimulation (LITUS) in mitigating brain damage after cardiopulmonary resuscitation (CPR) using a murine model and in vitro assays. LITUS treatment improved 24-h survival rates and neurological recovery in cardiac arrest (CA) mice, as evidenced by behavioral assessments and reduced neurological deficit scores. Proteomic analyses revealed modulation of Piezo1-Dkk3/PI3K-Akt signaling pathway, characterized by decreased pro-inflammatory cytokines (IL-1β, IL-6, TNF-α). Mechanistic studies demonstrated that LITUS enhanced Piezo1 and Dkk3 activation, promoting calcium influx and anti-inflammatory responses. The Piezo1 antagonist GsMTx4 abrogated these effects, underscoring Piezo1's specific role. Additionally, in vitro experiments using oxygen/glucose deprivation and reoxygenation (OGD/R)-treated BV2 microglial cells confirmed that LITUS reduced inflammatory responses and enhanced cellular recovery via the Piezo1-Dkk3 axis. These findings highlight LITUS as a promising non-invasive therapeutic strategy to ameliorate PCABI by modulating neuroinflammation through the Piezo1-Dkk3/PI3K-Akt pathway. This work provides a basis for translational research and potential clinical applications in improving outcomes for CPR survivors.
Collapse
Affiliation(s)
- Shuang Xu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China; Department of Critical Care Medicine, Henan Key Laboratory for Critical Care Medicine, Zhengzhou Key Laboratory for Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, China
| | - Lulu Gu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Banghe Bao
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Qian Liu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yannan Ma
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Emergency Medicine, Beijing Chao-Yang Hospital Capital Medical University, Beijing, China
| | - Siyi Zhou
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Beibei Li
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Li Xu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Guangqi Guo
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinpiao Zhu
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Kuan-Pin Su
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| | - Peng Sun
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
2
|
Sun J, Lu L, Lian Y, Xu S, Zhu Y, Wu Y, Lin Q, Hou J, Li Y, Yu Z. Sodium butyrate attenuates microglia-mediated neuroinflammation by modulating the TLR4/MyD88/NF-κB pathway and microbiome-gut-brain axis in cardiac arrest mice. Mol Brain 2025; 18:13. [PMID: 39962509 PMCID: PMC11834616 DOI: 10.1186/s13041-025-01179-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Cardiac arrest (CA) is one of the most common illnesses worldwide. Post-CA brain injury (PCABI) is a major cause of death and poor recovery in CA patients and the current CA treatments are not very effective. The microbiome-gut-brain axis has been found to significantly affect brain ischemia injury. Furthermore, in ischemic stroke patients, short-chain fatty acids (SCFA), especially sodium butyrate (SB), have been observed to promote neuroprotective effects by modulating inflammatory response and microglial polarization in the cortex. However, the precise mechanism of SB on CA-induced injury remains elusive. Therefore, this research study established an oxygen-glucose deprivation and reoxygenation (OGD/R) model using BV-2 microglial and HT22 cells to simulate cerebral ischemia/reperfusion injury in vitro and a potassium chloride-induced CA mouse model to mimic CA in vivo. The data revealed that SB markedly improved neurological scores and reduced neuronal death and apoptosis. Moreover, it reduced M1 microglia and neuroinflammation in CA mice. In addition, SB increased intestinal integrity and alleviated systemic inflammation. The 16S rDNA sequencing analysis indicated that SB intervention mitigated CA-induced gut microbiota dysbiosis and SCFA depletion. It was also observed that CA mice's brain and OGD/R-exposed BV2 cells had substantially increased levels of MyD88, phosphorylated NF-κB p65, and TLR4 proteins, which were reduced after SB treatment. In summary, this study revealed that SB can protect against cerebral ischemia-reperfusion injury by controlling microglia polarization and microbiome-gut-brain axis to inhibit brain inflammation via the TLR4/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Jianfei Sun
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liping Lu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Yingtao Lian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Ying Zhu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanping Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianhui Lin
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Hou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yinping Li
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
3
|
Duan P, Li X, Bi Y, Feng W, Jin Z, Zhang X, He G, An D, Wen Z, Zhang B. GYY4137 ameliorates blood brain barrier damage by inhibiting autophagy mediated occludin degradation in cardiac arrest and resuscitation. Sci Rep 2025; 15:905. [PMID: 39762518 PMCID: PMC11704213 DOI: 10.1038/s41598-024-84948-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiopulmonary resuscitation (CPR) after cardiac arrest (CA) is an important cause of neurological impairment and leads to considerable morbidity and mortality. The stability of the blood-brain barrier (BBB) is crucial for minimizing secondary neurological damage and improving long-term prognosis. However, the precise mechanisms and regulatory pathways that contribute to BBB dysfunction after CPR remain elusive. GYY4137 is an innovative hydrogen sulfide slow-release agent with excellent properties as a hydrogen sulfide substitute. The aim of this study was to investigate the protective effects of GYY4137 on CA/CPR and the underlying mechanisms of BBB protection. The effects of GYY4137 on systemic inflammation, BBB integrity, and autophagy were evaluated using a mouse CA/CPR model. The underlying mechanisms of occludin changes associated with GYY4137 were investigated using oxygen-glucose deprivation / reoxygenation (OGD/R) model. ELISA, neurological function and other tests showed that GYY4137 ameliorates systemic inflammation and neurological prognosis. Western blotting, transwell migration and tube formation assays showed that GYY4137 improves BBB function both in vivo and in vitro. The detection of autophagy flow and protein degradation pathways showed the inhibition of occludin reduction by GYY4137 was mainly achieved by suppressing autophagy mediated degradation. Taken together, GYY4137 may improve BBB dysfunction following CPR by increasing occludin content. This effect was achieved by inhibiting autophagic degradation rather than promoting synthesis. GYY4137 also mitigated systemic inflammation and improved neurological outcomes after CA/CPR. In summary, our study provides valuable insights into protecting the integrity of BBB and improving neurological outcomes after CPR.
Collapse
Affiliation(s)
- Pengyu Duan
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Xiaoyan Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Yonghong Bi
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Weiyu Feng
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Zhehao Jin
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Xiaoqian Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Guanghui He
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China
| | - Da An
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhibin Wen
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Bing Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China.
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, China.
| |
Collapse
|
4
|
Zhao Y, Yao Z, Lu L, Xu S, Sun J, Zhu Y, Wu Y, Yu Z. Carbon monoxide-releasing molecule-3 exerts neuroprotection effects after cardiac arrest in mice: A randomized controlled study. Resusc Plus 2024; 19:100703. [PMID: 39040821 PMCID: PMC11260602 DOI: 10.1016/j.resplu.2024.100703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Background Post-cardiac arrest brain injury (PCABI) is the leading cause of death in survivors of cardiac arrest (CA). Carbon monoxide-releasing molecule (CORM-3) is a water-soluble exogenous carbon monoxide that has been shown to have neuroprotection benefits in several neurological disease models. However, the effects of CORM-3 on PCABI is still unclear. Methods A mice model combined asystole with hemorrhage was used. Mice were anesthetized and randomized into 4 groups (n = 12/group) and underwent either 9.5 min CA followed by cardiopulmonary resuscitation (CPR) or sham surgery. CORM-3 (30 mg/kg) or vehicle (normal saline) were administered at 1 h after return of spontaneous circulation or sham surgery. Survival, neurologic deficits, alterations in the permeability of the brain-blood barrier and cerebral blood flow, changes of oxidative stress level, level of neuroinflammation and neuronal degeneration, and the activation of Nrf2/HO-1 signaling pathway were measured. Results In CORM-3 treated mice that underwent CA/CPR, significantly improved survival (75.00% vs. 58.33%, P = 0.0146 (24 h) and 66.67% vs. 16.67%, P < 0.0001 (72 h)) and neurological function were observed at 24 h and 72 h after ROSC (P < 0.05 for each). Additionally, increased cerebral blood flow, expression of tight junctions, and reduced reactive oxygen species generation at 24 h after ROSC were observed (P < 0.05 for each). CORM-3 treated mice had less neuron death and alleviated neuroinflammation at 72 h after ROSC (P < 0.05 for each). Notably, the Nrf2/HO-1 signaling pathway was significantly activated in mice subjected to CA/CPR with CORM-3 treatment. Conclusions CORM-3 could improve survival and exert neuroprotection after CA/CPR in mice. CORM-3 may be a novel and promising pharmacological therapy for PCABI.
Collapse
Affiliation(s)
- Yuanrui Zhao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhun Yao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liping Lu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianfei Sun
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Zhu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanping Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Magliocca A, Perego C, Motta F, Merigo G, Micotti E, Olivari D, Fumagalli F, Lucchetti J, Gobbi M, Mandelli A, Furlan R, Skrifvars MB, Latini R, Bellani G, Ichinose F, Ristagno G. Indoleamine 2,3-Dioxygenase Deletion to Modulate Kynurenine Pathway and to Prevent Brain Injury after Cardiac Arrest in Mice. Anesthesiology 2023; 139:628-645. [PMID: 37487175 PMCID: PMC10566599 DOI: 10.1097/aln.0000000000004713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND The catabolism of the essential amino acid tryptophan to kynurenine is emerging as a potential key pathway involved in post-cardiac arrest brain injury. The aim of this study was to evaluate the effects of the modulation of kynurenine pathway on cardiac arrest outcome through genetic deletion of the rate-limiting enzyme of the pathway, indoleamine 2,3-dioxygenase. METHODS Wild-type and indoleamine 2,3-dioxygenase-deleted (IDO-/-) mice were subjected to 8-min cardiac arrest. Survival, neurologic outcome, and locomotor activity were evaluated after resuscitation. Brain magnetic resonance imaging with diffusion tensor and diffusion-weighted imaging sequences was performed, together with microglia and macrophage activation and neurofilament light chain measurements. RESULTS IDO-/- mice showed higher survival compared to wild-type mice (IDO-/- 11 of 16, wild-type 6 of 16, log-rank P = 0.036). Neurologic function was higher in IDO-/- mice than in wild-type mice after cardiac arrest (IDO-/- 9 ± 1, wild-type 7 ± 1, P = 0.012, n = 16). Indoleamine 2,3-dioxygenase deletion preserved locomotor function while maintaining physiologic circadian rhythm after cardiac arrest. Brain magnetic resonance imaging with diffusion tensor imaging showed an increase in mean fractional anisotropy in the corpus callosum (IDO-/- 0.68 ± 0.01, wild-type 0.65 ± 0.01, P = 0.010, n = 4 to 5) and in the external capsule (IDO-/- 0.47 ± 0.01, wild-type 0.45 ± 0.01, P = 0.006, n = 4 to 5) in IDO-/- mice compared with wild-type ones. Increased release of neurofilament light chain was observed in wild-type mice compared to IDO-/- (median concentrations [interquartile range], pg/mL: wild-type 1,138 [678 to 1,384]; IDO-/- 267 [157 to 550]; P < 0.001, n = 3 to 4). Brain magnetic resonance imaging with diffusion-weighted imaging revealed restriction of water diffusivity 24 h after cardiac arrest in wild-type mice; indoleamine 2,3-dioxygenase deletion prevented water diffusion abnormalities, which was reverted in IDO-/- mice receiving l-kynurenine (apparent diffusion coefficient, μm2/ms: wild-type, 0.48 ± 0.07; IDO-/-, 0.59 ± 0.02; IDO-/- and l-kynurenine, 0.47 ± 0.08; P = 0.007, n = 6). CONCLUSIONS The kynurenine pathway represents a novel target to prevent post-cardiac arrest brain injury. The neuroprotective effects of indoleamine 2,3-dioxygenase deletion were associated with preservation of brain white matter microintegrity and with reduction of cerebral cytotoxic edema. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Aurora Magliocca
- Department of Pathophysiology and Transplants, University of Milan, Milan, Italy; and Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carlo Perego
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Motta
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giulia Merigo
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Davide Olivari
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Fumagalli
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessandra Mandelli
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology–INSpe, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology–INSpe, San Raffaele Scientific Institute, Milan, Italy
| | - Markus B. Skrifvars
- Department of Emergency Care and Services, Helsinki University Hospital and University of Helsinki, Finland
| | - Roberto Latini
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giacomo Bellani
- Centre for Medical Sciences−CISMed, University of Trento, Italy; and Department of Anesthesia and Intensive Care, Santa Chiara Hospital, Trento, Italy
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts; and Harvard Medical School, Boston, Massachusetts
| | - Giuseppe Ristagno
- Department of Pathophysiology and Transplants, University of Milan, Milan, Italy; and Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda−Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
6
|
Li R, Duan W, Zhang D, Hoffmann U, Yao J, Yang W, Sheng H. Mouse Cardiac Arrest Model for Brain Imaging and Brain Physiology Monitoring During Ischemia and Resuscitation. J Vis Exp 2023:10.3791/65340. [PMID: 37125804 PMCID: PMC10910853 DOI: 10.3791/65340] [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] [Indexed: 05/02/2023] Open
Abstract
Most cardiac arrest (CA) survivors experience varying degrees of neurologic deficits. To understand the mechanisms that underpin CA-induced brain injury and, subsequently, develop effective treatments, experimental CA research is essential. To this end, a few mouse CA models have been established. In most of these models, the mice are placed in the supine position in order to perform chest compression for cardiopulmonary resuscitation (CPR). However, this resuscitation procedure makes the real-time imaging/monitoring of brain physiology during CA and resuscitation challenging. To obtain such critical knowledge, the present protocol presents a mouse asphyxia CA model that does not require the chest compression CPR step. This model allows for the study of dynamic changes in blood flow, vascular structure, electrical potentials, and brain tissue oxygen from the pre-CA baseline to early post-CA reperfusion. Importantly, this model applies to aged mice. Thus, this mouse CA model is expected to be a critical tool for deciphering the impact of CA on brain physiology.
Collapse
Affiliation(s)
- Ran Li
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University Medical Center
| | - Weina Duan
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University Medical Center
| | - Dong Zhang
- Department of Biomedical Engineering, Duke University
| | - Ulrike Hoffmann
- Department of Anesthesiology and Pain Management, UT Southwestern University Medical Center
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University
| | - Wei Yang
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University Medical Center;
| | - Huaxin Sheng
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University Medical Center;
| |
Collapse
|
7
|
Xu S, Guo L, Shao W, Liang L, Shu T, Zhang Y, Huang H, Guo G, Zhang Q, Sun P. Vagus nerve stimulation protects against cerebral injury after cardiopulmonary resuscitation by inhibiting inflammation through the TLR4/NF-κB and α7nAChR/JAK2 signaling pathways. World J Emerg Med 2023; 14:462-470. [PMID: 37969224 PMCID: PMC10632751 DOI: 10.5847/wjem.j.1920-8642.2023.102] [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: 03/09/2023] [Accepted: 06/20/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Our previous research proved that vagus nerve stimulation (VNS) improved the neurological outcome after cardiopulmonary resuscitation (CPR) by activating α7 nicotinic acetylcholine receptor (α7nAChR) in a rat model, but the underlying mechanism of VNS in neuroprotection after CPR remains unclear. METHODS In vivo, we established a mouse model of cardiac arrest (CA)/CPR to observe the survival rate, and the changes in inflammatory factors and brain tissue after VNS treatment. In vitro, we examined the effects of α7nAChR agonist on ischemia/reperfusion (I/R)-induced inflammation in BV2 cells under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions. We observed the changes in cell survival rate, the levels of inflammatory factors, and the expressions of α7nAChR/Janus kinase 2 (JAK2) and toll-like receptor 4 (TLR4) /nuclear factor-κB (NF-κB). RESULTS In vivo, VNS preconditioning enhanced functional recovery, improved the survival rate, and reduced hippocampal CA1 cell damage, and the levels of inflammatory mediators after CA/CPR. The application of α7nAChR agonists provided similar effects against cerebral injury after the return of spontaneous circulation (ROSC), while α7nAChR antagonists reversed these neuroprotective impacts. The in vitro results mostly matched the findings in vivo. OGD/R increased the expression of tumor necrosis factor-alpha (TNF-α), TLR4 and NF-κB p65. When nicotine was added to the OGD/R model, the expression of TLR4, NF-κB p65, and TNF-α decreased, while the phosphorylation of JAK2 increased, which was prevented by preconditioning with α7nAChR or JAK2 antagonists. CONCLUSION The neuroprotective effect of VNS correlated with the activation of α7nAChR. VNS may alleviate cerebral IR injury by inhibiting TLR4/NF-κB and activating the α7nAChR/JAK2 signaling pathway.
Collapse
Affiliation(s)
- Shuang Xu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lang Guo
- Department of Urology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Weijing Shao
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Licai Liang
- Department of Emergency Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tingting Shu
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, Wuhan 430000, China
| | - Yuhan Zhang
- Department of Intensive Care Unit, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
| | - He Huang
- Department of Emergency, General Hospital of Central Theatre Command of the Chinese People’s Liberation Army, Wuhan 430070, China
| | - Guangqi Guo
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qing Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Sun
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
8
|
Yuan Z, Lu L, Lian Y, Zhao Y, Tang T, Xu S, Yao Z, Yu Z. AA147 ameliorates post-cardiac arrest cerebral ischemia/reperfusion injury through the co-regulation of the ATF6 and Nrf2 signaling pathways. Front Pharmacol 2022; 13:1028002. [PMID: 36506549 PMCID: PMC9727236 DOI: 10.3389/fphar.2022.1028002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Ischemia/reperfusion caused by cardiac arrest (CA) disturbs endoplasmic reticulum (ER) homeostasis and redox balance in neurons. AA147, originally developed as a pharmacologic activator of the activating transcription factor 6 (ATF6), can protect multiple tissues from ischemia/reperfusion injury (IRI) by decreasing reactive oxygen species (ROS) and restoring ER function. However, it is unclear whether pharmacologic treatment of AA147 could ameliorate post-CA cerebral IRI and whether it is associated with proteostasis regulation and anti-oxidative stress mechanism. In the present study, mice were subjected to 9 min-CA surgery followed by cardiopulmonary resuscitation (CPR). AA147 or vehicle was administered 1 day before the operation and 15 min after the return of spontaneous circulation. We found that AA147 restored neurological function and reduced dead neurons in mice suffering from CA. Moreover, AA147 inhibited CA/CPR-caused neuronal apoptosis and ER stress, indicated by reduced TUNEL-positive neurons, surged expression of Bcl-2/Bax, and down expression of cleaved caspase-3, caspase-12, C/EBP homologous protein (CHOP). The expression of ATF6 and its regulated gene glucose-regulated protein 78 (GRP78) increased significantly after the administration of AA147, suggesting the activation of the ATF6 pathway. In addition, AA147 also alleviated the upsurge of the ROS generation and MDA levels as well as increased SOD activity, accompanied by enhancement of the nuclear factor E2-related factor 2 (Nrf2) and its modulated heme-oxygenase-1 (HO-1) expressions. Cotreatment of AA147 with inhibitors of the ATF6 or Nrf2 significantly suppressed AA147-dependent reductions in ROS scavenging and neuronal death after CA/CPR. The results suggested that AA147 could confer neuroprotection against post-CA cerebral IRI through inhibition of oxidative stress along with ER stress-associated apoptosis, which is attributed to the coregulation of both ATF6 and Nrf2 signaling pathways activity. Our findings support the potential for AA147 as a therapeutic approach to improve post-CA brain injury.
Collapse
Affiliation(s)
- Zhu Yuan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liping Lu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingtao Lian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanrui Zhao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tingting Tang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhun Yao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China,*Correspondence: Zhui Yu,
| |
Collapse
|
9
|
Manganese Porphyrin Promotes Post Cardiac Arrest Recovery in Mice and Rats. BIOLOGY 2022; 11:biology11070957. [PMID: 36101338 PMCID: PMC9312251 DOI: 10.3390/biology11070957] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/11/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022]
Abstract
Introduction Cardiac arrest (CA) and resuscitation induces global cerebral ischemia and reperfusion, causing neurologic deficits or death. Manganese porphyrins, superoxide dismutase mimics, are reportedly able to effectively reduce ischemic injury in brain, kidney, and other tissues. This study evaluates the efficacy of a third generation lipophilic Mn porphyrin, MnTnBuOE-2-PyP5+, Mn(III) ortho meso-tetrakis (N-n-butoxyethylpyridinium-2-yl)porphyrin (MnBuOE, BMX-001), in both mouse and rat models of CA. Methods Forty-eight animals were subjected to 8 min of CA and resuscitated subsequently by chest compression and epinephrine infusion. Vehicle or MnBuOE was given immediately after resuscitation followed by daily subcutaneous injections. Body weight, spontaneous activity, neurologic deficits, rotarod performance, and neuronal death were assessed. Kidney tubular injury was assessed in CA mice. Data were collected by the investigators who were blinded to the treatment groups. Results Vehicle mice had a mortality of 20%, which was reduced by 50% by MnBuOE. All CA mice had body weight loss, spontaneous activity decline, neurologic deficits, and decreased rotarod performance that were significantly improved at three days post MnBuOE daily treatment. MnBuOE treatment reduced cortical neuronal death and kidney tubular injury in mice (p < 0.05) but not hippocampus neuronal death (23% MnBuOE vs. 34% vehicle group, p = 0.49). In rats, they had a better body-weight recovery and increased rotarod latency after MnBuOE treatment when compared to vehicle group (p < 0.01 vs. vehicle). MnBuOE-treated rats had a low percentage of hippocampus neuronal death (39% MnBuOE vs. 49% vehicle group, p = 0.21) and less tubular injury (p < 0.05) relative to vehicle group. Conclusions We demonstrated the ability of MnBuOE to improve post-CA survival, as well as functional outcomes in both mice and rats, which jointly account for the improvement not only of brain function but also of the overall wellbeing of the animals. While MnBuOE bears therapeutic potential for treating CA patients, the females and the animals with comorbidities must be further evaluated before advancing toward clinical trials.
Collapse
|
10
|
Duan W, Sun Q, Wu X, Xia Z, Warner DS, Ulloa L, Yang W, Sheng H. Cervical Vagus Nerve Stimulation Improves Neurologic Outcome After Cardiac Arrest in Mice by Attenuating Oxidative Stress and Excessive Autophagy. Neuromodulation 2022; 25:414-423. [PMID: 35131154 DOI: 10.1016/j.neurom.2021.12.014] [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: 08/24/2021] [Revised: 12/11/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cerebral ischemia and reperfusion (I/R) induces oxidative stress and activates autophagy, leading to brain injury and neurologic deficits. Cervical vagus nerve stimulation (VNS) increases cerebral blood flow (CBF). In this study, we investigate the effect of VNS-induced CBF increase on neurologic outcomes after cardiac arrest (CA). MATERIALS AND METHODS A total of 40 male C57Bl/6 mice were subjected to ten minutes of asphyxia CA and randomized to vagus nerve isolation (VNI) or VNS treatment group. Eight mice received sham surgery and VNI. Immediately after resuscitation, 20 minutes of electrical stimulation (1 mA, 1 ms, and 10 Hz) was started in the VNS group. Electrocardiogram, blood pressure, and CBF were monitored. Neurologic and histologic outcomes were evaluated at 72 hours. Oxidative stress and autophagy were assessed at 3 hours and 24 hours after CA. RESULTS Baseline characteristics were not different among groups. VNS mice had better behavioral performance (ie, open field, rotarod, and neurologic score) and less neuronal death (p < 0.05, vs VNI) in the hippocampus. CBF was significantly increased in VNS-treated mice at 20 minutes after return of spontaneous circulation (ROSC) (p < 0.05). Furthermore, levels of 8-hydroxy-2'-deoxyguanosine in the blood and autophagy-related proteins (ie, LC-3Ⅱ/Ⅰ, Beclin-1, and p62) in the brain were significantly decreased in VNS mice. Aconitase activity was also reduced, and the p-mTOR/mTOR ratio was increased in VNS mice. CONCLUSIONS Oxidative stress induced by global brain I/R following CA/ROSC leads to early excessive autophagy and impaired autophagic flux. VNS promoted CBF recovery, ameliorating these changes. Neurologic and histologic outcomes were also improved.
Collapse
Affiliation(s)
- Weina Duan
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - David S Warner
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- 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.
| |
Collapse
|
11
|
Choi ES, Park GH, Kim DS, Shin HS, Park SY, Kim M, Hong JM. A novel global ischemia-reperfusion rat model with asymmetric brain damage simulating post-cardiac arrest brain injury. J Neurosci Methods 2022; 372:109554. [DOI: 10.1016/j.jneumeth.2022.109554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
|
12
|
Jing W, Tuxiu X, Xiaobing L, Guijun J, Lulu K, Jie J, Lu Y, Liying Z, Xiaoxing X, Jingjun L. LncRNA GAS5/miR-137 Is a Hypoxia-Responsive Axis Involved in Cardiac Arrest and Cardiopulmonary Cerebral Resuscitation. Front Immunol 2022; 12:790750. [PMID: 35087519 PMCID: PMC8787067 DOI: 10.3389/fimmu.2021.790750] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) represents one of the devastating medical emergencies and is associated with high mortality and neuro-disability. Post-cardiac arrest syndrome (PCAS) is mechanistically ascribed to acute systemic ischemia/reperfusion(I/R) injury. The lncRNA/microRNA/mRNA networks have been found to play crucial roles in the pathogenesis of the hypoxia-responsive diseases. Nonetheless, the precise molecular mechanisms by which lncRNA/miRNA/mRNA axes are involved in the astrocyte-microglia crosstalk in CA/CPR have not been fully elucidated. Methods We collected and purified the exosomes from the blood of CA/CPR patients and supernatant of OGD/R-stimulated astrocytes. On the basis of microarray analysis, bioinformatic study, and luciferase activity determination, we speculated that lncRNA GAS5/miR-137 is implicated in the astrocyte-microglia crosstalk under the insult of systemic I/R injury. The regulation of lncRNA GAS5/miR-137 on INPP4B was examined by cellular transfection in OGD/R cell culture and by lateral ventricle injection with miR-137 agomir in CA/CPR mice model. Flow cytometry and immunofluorescence staining were performed to detect the microglial apoptosis, M1/M2 phenotype transformation, and neuroinflammation. Neurological scoring and behavior tests were conducted in CA/CPR group, with miR-137 agomir lateral-ventricle infusion and in their controls. Results In all the micRNAs, miR-137 was among the top 10 micRNAs that experienced greatest changes, in both the blood of CA/CPR patients and supernatant of OGD/R-stimulated astrocytes. Bioinformatic analysis revealed that miR-137 was sponged by lncRNA GAS5, targeting INPP4B, and the result was confirmed by Luciferase activity assay. qRT-PCR and Western blotting showed that lncRNA GAS5 and INPP4B were over-expressed whereas miR-137 was downregulated in the blood of CA/CPR patients, OGD/R-stimulated astrocytes, and brain tissue of CA/CPR mice. Silencing lncRNA GAS5 suppressed INPP4B expression, but over-expression of miR-137 negatively modulated its expression. Western blotting exhibited that PI3K and Akt phosphorylation was increased when lncRNA GAS5 was silenced or miR-137 was over-expressed. However, PI3K and Akt phosphorylation was notably suppressed in the absence of miR-137, almost reversing their phosphorylation in the silencing lncRNA GAS5 group. Then we found that GAS5 siRNA or miR-137 mimic significantly increased cell viability and alleviated apoptosis after OGD/R injury. Furthermore, over-expression of miR-137 attenuated microglial apoptosis and neuroinflammation in CA/CPR mice model, exhibiting significantly better behavioral tests after CA/CPR. Conclusion LncRNA GAS5/miR-137 may be involved in the astrocyte-microglia communication that inhibits PI3K/Akt signaling activation via regulation of INPP4B during CA/CPR.
Collapse
Affiliation(s)
- Wang Jing
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xie Tuxiu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, China
| | - Long Xiaobing
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiang Guijun
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kang Lulu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiang Jie
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ye Lu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhan Liying
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiong Xiaoxing
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lyu Jingjun
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Li R, Shen Y, Li X, Lu L, Wang Z, Sheng H, Hoffmann U, Yang W. Activation of the XBP1s/O-GlcNAcylation Pathway Improves Functional Outcome After Cardiac Arrest and Resuscitation in Young and Aged Mice. Shock 2021; 56:755-761. [PMID: 34652341 PMCID: PMC9059164 DOI: 10.1097/shk.0000000000001732] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
ABSTRACT After cardiac arrest (CA) and resuscitation, the unfolded protein response (UPR) is activated in various organs including the brain. However, the role of the UPR in CA outcome remains largely unknown. One UPR branch involves spliced X-box-binding protein-1 (XBP1s). Notably, XBP1s, a transcriptional factor, can upregulate expression of specific enzymes related to glucose metabolism, and subsequently boost O-linked β-N-acetylglucosamine modification (O-GlcNAcylation). The current study is focused on effects of the XBP1 UPR branch and its downstream O-GlcNAcylation on CA outcome. Using both loss-of-function and gain-of-function mouse genetic tools, we provide the first evidence that activation of the XBP1 UPR branch in the post-CA brain is neuroprotective. Specifically, neuron-specific Xbp1 knockout mice had worse CA outcome, while mice with neuron-specific expression of Xbp1s in the brain had better CA outcome. Since it has been shown that the protective role of the XBP1s signaling pathway under ischemic conditions is mediated by increasing O-GlcNAcylation, we then treated young mice with glucosamine, and found that functional deficits were mitigated on day 3 post CA. Finally, after confirming that glucosamine can boost O-GlcNAcylation in the aged brain, we subjected aged mice to 8 min CA, and then treated them with glucosamine. We found that glucosamine-treated aged mice performed significantly better in behavioral tests. Together, our data indicate that the XBP1s/O-GlcNAc pathway is a promising target for CA therapy.
Collapse
Affiliation(s)
- Ran Li
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Shen Y, Li R, Yu S, Zhao Q, Wang Z, Sheng H, Yang W. Activation of the ATF6 (Activating Transcription Factor 6) Signaling Pathway in Neurons Improves Outcome After Cardiac Arrest in Mice. J Am Heart Assoc 2021; 10:e020216. [PMID: 34111943 PMCID: PMC8477867 DOI: 10.1161/jaha.120.020216] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/21/2021] [Indexed: 11/16/2022]
Abstract
Background Ischemia/reperfusion injury impairs proteostasis, and triggers adaptive cellular responses, such as the unfolded protein response (UPR), which functions to restore endoplasmic reticulum homeostasis. After cardiac arrest (CA) and resuscitation, the UPR is activated in various organs including the brain. However, the role of the UPR in CA has remained largely unknown. Here we aimed to investigate effects of activation of the ATF6 (activating transcription factor 6) UPR branch in CA. Methods and Results Conditional and inducible sATF6-KI (short-form ATF6 knock-in) mice and a selective ATF6 pathway activator 147 were used. CA was induced in mice by KCl injection, followed by cardiopulmonary resuscitation. We first found that neurologic function was significantly improved, and neuronal damage was mitigated after the ATF6 pathway was activated in neurons of sATF6-KI mice subjected to CA/cardiopulmonary resuscitation. Further RNA sequencing analysis indicated that such beneficial effects were likely attributable to increased expression of pro-proteostatic genes regulated by ATF6. Especially, key components of the endoplasmic reticulum-associated degradation process, which clears potentially toxic unfolded/misfolded proteins in the endoplasmic reticulum, were upregulated in the sATF6-KI brain. Accordingly, the CA-induced increase in K48-linked polyubiquitin in the brain was higher in sATF6-KI mice relative to control mice. Finally, CA outcome, including the survival rate, was significantly improved in mice treated with compound 147. Conclusions This is the first experimental study to determine the role of the ATF6 UPR branch in CA outcome. Our data indicate that the ATF6 UPR branch is a prosurvival pathway and may be considered as a therapeutic target for CA.
Collapse
Affiliation(s)
- Yuntian Shen
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Ran Li
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Shu Yu
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Qiang Zhao
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Zhuoran Wang
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Huaxin Sheng
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| | - Wei Yang
- Department of AnesthesiologyCenter for Perioperative Organ ProtectionDuke University Medical CenterDurhamNC
| |
Collapse
|
15
|
Wang W, Li R, Miao W, Evans C, Lu L, Lyu J, Li X, Warner DS, Zhong X, Hoffmann U, Sheng H, Yang W. Development and Evaluation of a Novel Mouse Model of Asphyxial Cardiac Arrest Revealed Severely Impaired Lymphopoiesis After Resuscitation. J Am Heart Assoc 2021; 10:e019142. [PMID: 34013738 PMCID: PMC8483518 DOI: 10.1161/jaha.120.019142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Animal disease models represent the cornerstone in basic cardiac arrest (CA) research. However, current experimental models of CA and resuscitation in mice are limited. In this study, we aimed to develop a mouse model of asphyxial CA followed by cardiopulmonary resuscitation (CPR), and to characterize the immune response after asphyxial CA/CPR. Methods and Results CA was induced in mice by switching from an O2/N2 mixture to 100% N2 gas for mechanical ventilation under anesthesia. Real-time measurements of blood pressure, brain tissue oxygen, cerebral blood flow, and ECG confirmed asphyxia and ensuing CA. After a defined CA period, mice were resuscitated with intravenous epinephrine administration and chest compression. We subjected young adult and aged mice to this model, and found that after CA/CPR, mice from both groups exhibited significant neurologic deficits compared with sham mice. Analysis of post-CA brain confirmed neuroinflammation. Detailed characterization of the post-CA immune response in the peripheral organs of both young adult and aged mice revealed that at the subacute phase following asphyxial CA/CPR, the immune system was markedly suppressed as manifested by drastic atrophy of the spleen and thymus, and profound lymphopenia. Finally, our data showed that post-CA systemic lymphopenia was accompanied with impaired T and B lymphopoiesis in the thymus and bone marrow, respectively. Conclusions In this study, we established a novel validated asphyxial CA model in mice. Using this new model, we further demonstrated that asphyxial CA/CPR markedly affects both the nervous and immune systems, and notably impairs lymphopoiesis of T and B cells.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Ran Li
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Wanying Miao
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Cody Evans
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Liping Lu
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Jingjun Lyu
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Xuan Li
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - David S Warner
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Xiaoping Zhong
- Department of Pediatrics Duke University Medical Center Durham NC
| | - Ulrike Hoffmann
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Huaxin Sheng
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| | - Wei Yang
- Department of Anesthesiology Center for Perioperative Organ Protection Duke University Medical Center Durham NC
| |
Collapse
|
16
|
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
|
17
|
Rutledge CA, Chiba T, Redding K, Dezfulian C, Sims-Lucas S, Kaufman BA. A novel ultrasound-guided mouse model of sudden cardiac arrest. PLoS One 2020; 15:e0237292. [PMID: 33275630 PMCID: PMC7717537 DOI: 10.1371/journal.pone.0237292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
AIM Mouse models of sudden cardiac arrest are limited by challenges with surgical technique and obtaining reliable venous access. To overcome this limitation, we sought to develop a simplified method in the mouse that uses ultrasound-guided injection of potassium chloride directly into the heart. METHODS Potassium chloride was delivered directly into the left ventricular cavity under ultrasound guidance in intubated mice, resulting in immediate asystole. Mice were resuscitated with injection of epinephrine and manual chest compressions and evaluated for survival, body temperature, cardiac function, kidney damage, and diffuse tissue injury. RESULTS The direct injection sudden cardiac arrest model causes rapid asystole with high surgical survival rates and short surgical duration. Sudden cardiac arrest mice with 8-min of asystole have significant cardiac dysfunction at 24 hours and high lethality within the first seven days, where after cardiac function begins to improve. Sudden cardiac arrest mice have secondary organ damage, including significant kidney injury but no significant change to neurologic function. CONCLUSIONS Ultrasound-guided direct injection of potassium chloride allows for rapid and reliable cardiac arrest in the mouse that mirrors human pathology without the need for intravenous access. This technique will improve investigators' ability to study the mechanisms underlying post-arrest changes in a mouse model.
Collapse
Affiliation(s)
- Cody A. Rutledge
- Division of Cardiology, Cardiovascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Takuto Chiba
- Rangos Research Center, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States of America
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Kevin Redding
- Division of Cardiology, Cardiovascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Cameron Dezfulian
- Safar Center for Resuscitation Research and Critical Care Medicine Department, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sunder Sims-Lucas
- Rangos Research Center, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States of America
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Brett A. Kaufman
- Division of Cardiology, Cardiovascular Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
18
|
Shen Y, Yan B, Zhao Q, Wang Z, Wu J, Ren J, Wang W, Yu S, Sheng H, Crowley SD, Ding F, Paschen W, Yang W. Aging Is Associated With Impaired Activation of Protein Homeostasis-Related Pathways After Cardiac Arrest in Mice. J Am Heart Assoc 2018; 7:e009634. [PMID: 30371162 PMCID: PMC6201440 DOI: 10.1161/jaha.118.009634] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/02/2018] [Indexed: 01/08/2023]
Abstract
Background The mechanisms underlying worse outcome at advanced age after cardiac arrest ( CA ) and resuscitation are not well understood. Because protein homeostasis (proteostasis) is essential for cellular and organismal health, but is impaired after CA , we investigated the effects of age on proteostasis-related prosurvival pathways activated after CA . Methods and Results Young (2-3 months old) and aged (21-22 months old) male C57Bl/6 mice were subjected to CA and cardiopulmonary resuscitation ( CPR ). Functional outcome and organ damage were evaluated by assessing neurologic deficits, histological features, and creatinine level. CA / CPR -related changes in small ubiquitin-like modifier conjugation, ubiquitination, and the unfolded protein response were analyzed by measuring mRNA and protein levels in the brain, kidney, and spinal cord. Thiamet-G was used to increase O-linked β-N-acetylglucosamine modification. After CA / CPR , aged mice had trended lower survival rates, more severe tissue damage in the brain and kidney, and poorer recovery of neurologic function compared with young mice. Furthermore, small ubiquitin-like modifier conjugation, ubiquitination, unfolded protein response, and O-linked β-N-acetylglucosamine modification were activated after CA / CPR in young mice, but their activation was impaired in aged mice. Finally, pharmacologically increasing O-linked β-N-acetylglucosamine modification after CA improved outcome. Conclusions Results suggest that impaired activation of prosurvival pathways contributes to worse outcome after CA / CPR in aged mice because restoration of proteostasis is critical to the survival of cells stressed by ischemia. Therefore, a pharmacologic intervention that targets aging-related impairment of proteostasis-related pathways after CA / CPR may represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yuntian Shen
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Baihui Yan
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
- Department of AnesthesiologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Qiang Zhao
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
- Department of NeurologyTianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Zhuoran Wang
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
| | - Jiangbo Wu
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
| | - Jiafa Ren
- Division of NephrologyDepartment of MedicineDuke University and Durham VA Medical CentersDurhamNC
| | - Wei Wang
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
- Department of AnesthesiologySouthern Medical University Nanfang HospitalGuangzhouChina
| | - Shu Yu
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Huaxin Sheng
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
| | - Steven D. Crowley
- Division of NephrologyDepartment of MedicineDuke University and Durham VA Medical CentersDurhamNC
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationCo‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Wulf Paschen
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
| | - Wei Yang
- Center for Perioperative Organ ProtectionDepartment of AnesthesiologyDuke University Medical CenterDurhamNC
| |
Collapse
|
19
|
Impact of Different Serum Potassium Levels on Postresuscitation Heart Function and Hemodynamics in Patients with Nontraumatic Out-of-Hospital Cardiac Arrest. Bioinorg Chem Appl 2018; 2018:5825929. [PMID: 29849540 PMCID: PMC5907484 DOI: 10.1155/2018/5825929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/25/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Background Sustained return of spontaneous circulation (ROSC) can be initially established in patients with out-of-hospital cardiac arrest (OHCA); however, the early postresuscitation hemodynamics can still be impaired by high levels of serum potassium (hyperkalemia). The impact of different potassium levels on early postresuscitation heart function has remained unclear. We aim to analyze the relationship between different levels of serum potassium and postresuscitation heart function during the early postresuscitation period (the first hour after achieving sustained ROSC). Methods Information on 479 nontraumatic OHCA patients with sustained ROSC was retrospectively obtained. Measures of early postresuscitation heart function (rate, blood pressure, and rhythm), hemodynamics (urine output and blood pH), and the duration of survival were analyzed in the case of different serum potassium levels (low: <3.5; normal: 3.5–5; high: >5 mmol/L). Results Most patients (59.9%, n = 287) had previously presented with high levels of potassium. Bradycardia, nonsinus rhythm, urine output <1 ml/kg/hr, and acidosis (pH < 7.35) were more common in patients with high levels of potassium (all p < 0.05). Compared with hyperkalemia, a normal potassium level was more likely to be associated with a normal heart rate (OR: 2.97, 95% CI: 1.74–5.08) and sinus rhythm (OR: 2.28, 95% CI: 1.45–3.58). A low level of potassium was more likely to be associated with tachycardia (OR: 3.54, 95% CI: 1.32–9.51), urine output >1 ml/kg/hr (OR: 5.35, 95% CI: 2.58–11.10), and nonacidosis (blood pH >7.35, OR: 7.74, 95% CI: 3.78–15.58). The duration of survival was shorter in patients with hyperkalemia than that in patients whose potassium levels were low or normal (p < 0.05). Conclusion Early postresuscitation heart function and hemodynamics were associated with the serum potassium level. A high potassium level was more likely to be associated with bradycardia, nonsinus rhythm, urine output <1 ml/kg/hr, and acidosis. More importantly, a high potassium level decreased the duration of survival.
Collapse
|