1
|
Zhu Y, Li X, Lei X, Tang L, Wen D, Zeng B, Zhang X, Huang Z, Guo Z. The potential mechanism and clinical application value of remote ischemic conditioning in stroke. Neural Regen Res 2025; 20:1613-1627. [PMID: 38845225 DOI: 10.4103/nrr.nrr-d-23-01800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/19/2024] [Indexed: 08/07/2024] Open
Abstract
Some studies have confirmed the neuroprotective effect of remote ischemic conditioning against stroke. Although numerous animal researches have shown that the neuroprotective effect of remote ischemic conditioning may be related to neuroinflammation, cellular immunity, apoptosis, and autophagy, the exact underlying molecular mechanisms are unclear. This review summarizes the current status of different types of remote ischemic conditioning methods in animal and clinical studies and analyzes their commonalities and differences in neuroprotective mechanisms and signaling pathways. Remote ischemic conditioning has emerged as a potential therapeutic approach for improving stroke-induced brain injury owing to its simplicity, non-invasiveness, safety, and patient tolerability. Different forms of remote ischemic conditioning exhibit distinct intervention patterns, timing, and application range. Mechanistically, remote ischemic conditioning can exert neuroprotective effects by activating the Notch1/phosphatidylinositol 3-kinase/Akt signaling pathway, improving cerebral perfusion, suppressing neuroinflammation, inhibiting cell apoptosis, activating autophagy, and promoting neural regeneration. While remote ischemic conditioning has shown potential in improving stroke outcomes, its full clinical translation has not yet been achieved.
Collapse
Affiliation(s)
- Yajun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Huang SF, Xu JL, Ren CH, Sim N, Han C, Han YQ, Zhao WB, Ding YC, Ji XM, Li SJ. Remote ischemic conditioning prevents ischemic cerebrovascular events in children with moyamoya disease: a randomized controlled trial. World J Pediatr 2024; 20:925-934. [PMID: 38951456 PMCID: PMC11422440 DOI: 10.1007/s12519-024-00824-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/29/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Moyamoya disease (MMD) is a significant cause of childhood stroke and transient ischemic attacks (TIAs). This study aimed to assess the safety and efficacy of remote ischemic conditioning (RIC) in children with MMD. METHODS In a single-center pilot study, 46 MMD patients aged 4 to 14 years, with no history of reconstructive surgery, were randomly assigned to receive either RIC or sham RIC treatment twice daily for a year. The primary outcome measured was the cumulative incidence of major adverse cerebrovascular events (MACEs). Secondary outcomes included ischemic stroke, recurrent TIA, hemorrhagic stroke, revascularization rates, and clinical improvement assessed using the patient global impression of change (PGIC) scale during follow-up. RIC-related adverse events were also recorded, and cerebral hemodynamics were evaluated using transcranial Doppler. RESULTS All 46 patients completed the final follow-up (23 each in the RIC and sham RIC groups). No severe adverse events associated with RIC were observed. Kaplan-Meier analysis indicated a significant reduction in MACEs frequency after RIC treatment [log-rank test (Mantel-Cox), P = 0.021]. At 3-year follow-up, two (4.35%) patients had an ischemic stroke, four (8.70%) experienced TIAs, and two (4.35%) underwent revascularization as the qualifying MACEs. The clinical improvement rate in the RIC group was higher than the sham RIC group on the PGIC scale (65.2% vs. 26.1%, P < 0.01). No statistical difference in cerebral hemodynamics post-treatment was observed. CONCLUSIONS RIC is a safe and effective adjunct therapy for asymptomatic children with MMD. This was largely due to the reduced incidence of ischemic cerebrovascular events.
Collapse
Affiliation(s)
- Shuang-Feng Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No 45, Changchun Street, Xicheng District, Beijing, 100053, China
| | - Jia-Li Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No 45, Changchun Street, Xicheng District, Beijing, 100053, China
- Department of Rehabilitation Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Chang-Hong Ren
- Beijing Institute of Brain Disorders, Capital Medical University, No.10, Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100053, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nathan Sim
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - Cong Han
- Department of Neurosurgery, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi-Qin Han
- Department of Neurosurgery, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Wen-Bo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No 45, Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yu-Chuan Ding
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - Xun-Ming Ji
- Beijing Institute of Brain Disorders, Capital Medical University, No.10, Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100053, China.
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Si-Jie Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No 45, Changchun Street, Xicheng District, Beijing, 100053, China.
- Beijing Institute of Brain Disorders, Capital Medical University, No.10, Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100053, China.
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Wang Q, Li WN, Otkur W, Cui Y, Chen HS. Neutrophil-to-Lymphocyte Ratio, Platelet-to-Lymphocyte Ratio, Systemic Immune Inflammation Index and Efficacy of Remote Ischemic Conditioning in Acute Ischemic Stroke: A Post Hoc Exploratory Analysis of the RICAMIS Study. J Inflamm Res 2024; 17:5543-5553. [PMID: 39185106 PMCID: PMC11344552 DOI: 10.2147/jir.s460928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Background We conducted a post-hoc analysis of the RICAMIS trial to investigate the effect of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune inflammation index (SII) on the efficacy of remote ischemic conditioning treatment. Methods In this post-hoc analysis, NLR, PLR, and SII were measured before randomization. Patients were divided into two groups based on their cut-off values: high vs low NLR, high vs low PLR, and high vs low SII groups. Each group was further subdivided into RIC and control groups. The primary endpoint was a poor outcome (mRS 2-6 at 90 days). Differences in the primary endpoint between the RIC and control subgroups were compared, and the interactions of treatment assignment with NLR, PLR, and SII were evaluated. Results A total of 1679 patients were included in the final analysis. Compared with the control group, RIC significantly improved functional outcomes regardless of the inflammation status. The improved probability of poor outcome in the RIC vs control group was numerically greater in the high vs low inflammation group (NLR, 7.8% vs 5.1%; PLR, 7% vs 6.5%; SII, 9% vs 5.3%). However, we did not find an interaction effect of an intervention (RIC or control) with different NLR, PLR, or SII on clinical outcomes (P > 0.05). In addition, the NLR and SII were independently associated with functional outcomes in all patients, regardless of whether they received RIC. Conclusion Inflammation may not affect the efficacy of RIC in patients with acute moderate ischemic stroke, although a lower probability of poor outcome at 90 days was identified in patients with a high vs low inflammatory status.
Collapse
Affiliation(s)
- Qi Wang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, 110016, People’s Republic of China
- Dalian Medical University, Dalian, People’s Republic of China
| | - Wen-Na Li
- Department of Neurology, Tangshan Central Hospital, Tangshan, People’s Republic of China
| | - Wuxiyar Otkur
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Yu Cui
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, 110016, People’s Republic of China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, 110016, People’s Republic of China
- Dalian Medical University, Dalian, People’s Republic of China
| |
Collapse
|
4
|
Zhao T, Li M, Yan Q, Gu J, Liu L. Effect of remote ischemic preconditioning intervention on serum levels of microRNA-582-5p/HMGB1 in patients with acute cerebral infarction. Clin Neurol Neurosurg 2024; 241:108291. [PMID: 38701547 DOI: 10.1016/j.clineuro.2024.108291] [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: 06/24/2023] [Revised: 08/29/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
OBJECTIVE Acute cerebral infarction (ACI) contributes to disability and death accross the globe. Remote ischemic preconditioning (RIPC) reduces cerebral infarct size and improves neurological function in ACI. We conducted this research to reveal the effects of RIPC intervention on serum levels of microRNA-582-5p (miR-582-5p)/high mobility group box-1 protein (HMGB1), inflammation, oxidative stress and neurological function in patients with ACI. METHODS In this study, 158 patients with ACI were prospectively selected and randomized into the control (administered symptomatic medication alone) and the RIPC (underwent RIPC of the limbs based on medication) groups, with their clinical baseline data documented. Serum levels of miR-582-5p, and HMGB1 and inflammatory factors [tumor necrosis factor alpha (TNF-α)/interleukin-1beta (IL-1β)/IL-10] were assessed by RT-qPCR/ELISA, followed by comparisons of oxidative stress indices [glutathione-peroxidase (GSH-Px)/catalase (CAT)/superoxide dismutase (SOD)] using a fully automatic biochemical analyzer. Correlations between serum miR-582-5p with serum HMGB1, and between their levels with TNF-α/IL-1β/IL-10 were analyzed by Pearson analysis. The NIHSS score/Barthel Index scale were used to assess neurological function/daily living ability. Intervention safety for ACI patients was evaluated. RESULTS RIPC intervention increased serum miR-582-5p levels and decreased serum HMGB1 levels in ACI patients. RIPC intervention significantly reduced inflammation (diminished TNF-α/IL-1β levels, increased IL-10 level) and oxidative stress (elevated GSH-Px/CAT/SOD levels) in ACI patients. Serum miR-582-5p was negatively correlated with TNF-α and IL-1β levels, while positively correlated with IL-10 level, while HMGB1 was positively correlated with TNF-α and IL-1β levels, while negatively correlated with IL-10 level. miR-582-5p was negatively correlated with HMGB1. RIPC intervention improved neurological function (reduced NIHSS, increased Barthel scores) in ACI patients to some extent. RIPC had certain effectiveness and safety in the treatment of ACI. CONCLUSION After RIPC intervention, serum miR-582-5p levels were increased, HMGB1 levels were decreased, and inflammation and oxidative stress were reduced in ACI patients, which mitigated neurological deficits, improved patients' ability to perform life activities, and exerted neuroprotective effects to some extent.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China.
| | - Meng Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Qiuyue Yan
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Juxian Gu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Lige Liu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| |
Collapse
|
5
|
Yan MY, Liu JM, Wu J, Chang Q. Impact of remote ischemic postconditioning on acute ischemic stroke in China: a systematic review and meta-analysis of randomized controlled trials. Syst Rev 2024; 13:141. [PMID: 38816852 PMCID: PMC11138007 DOI: 10.1186/s13643-024-02568-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Acute ischemic stroke (AIS) is a significant health burden in China, affecting a sizable portion of the population. Conventional pharmacological treatments frequently fall short of desirable outcomes. Therefore, exploring alternative therapies is crucial. Remote ischemic postconditioning (RIPostC) is a noninvasive and cost-effective adjunctive therapy. This study aimed to investigate the efficacy and safety of RIPostC as an adjunctive therapy for AIS to inform clinical practice. METHODS A comprehensive search was conducted across the PubMed, Embase, Web of Science, China National Knowledge Infrastructure (CNKI), WanFang, Weipu (VIP), and China Biology Medicine disc (CBM) databases up to October 2023. All included studies underwent bias risk assessment using the Cochrane risk-of-bias assessment tool. The primary outcome measure was the National Institute of Health Stroke Scale (NIHSS), with secondary outcomes including the Barthel index (BI), D-dimer, C-reactive protein (CRP), fibrinogen (FIB), brain-derived neurotrophic factor (BDNF), modified Rankin scale (mRS), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) levels. The data were analyzed using fixed-effects and random-effects models in Review Manager, with mean differences (MDs) and 95% confidence intervals (CIs) calculated for each outcome. The grading of recommendations, assessment, development, and evaluations (GRADE) approach was used to evaluate the level of evidence for each outcome measure. RESULTS This meta-analysis included 38 studies, encompassing 4334 patients. Compared with the control group, the RIPostC group had significantly lower NIHSS scores, serum CRP, D-dimer, IL-6, TNF-α, and FIB levels, and increased BDNF levels. Moreover, it improved the patient's BI and mRS scores. According to the GRADE approach, the quality of evidence for mRS was deemed "moderate," while the NIHSS, BI, and CRP were rated as "low" quality. IL-6, TNF-α, FIB, D-dimer, and BDNF received "very low" quality ratings. CONCLUSION The findings suggest that RIPostC activates endogenous protective mechanisms, providing benefits to patients with AIS.
Collapse
Affiliation(s)
- Ming-Yuan Yan
- Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Min Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Jing Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing Chang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
6
|
Kim ID, Ju H, Minkler J, Madkoor A, Park KW, Cho S. Obesity-induced Ly6C High and Ly6C Low monocyte subset changes abolish post-ischemic limb conditioning benefits in stroke recovery. J Cereb Blood Flow Metab 2024; 44:689-701. [PMID: 37974299 PMCID: PMC11197146 DOI: 10.1177/0271678x231215101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/28/2023] [Accepted: 10/15/2023] [Indexed: 11/19/2023]
Abstract
Remote limb conditioning (RLC), performed by intermittent interruption of blood flow to a limb, triggers endogenous tolerance mechanisms and improves stroke outcomes. The underlying mechanism for the protective effect involves a shift of circulating monocytes to a Ly6CHigh proinflammatory subset in normal metabolic conditions. The current study investigates the effect of RLC on stroke outcomes in subjects with obesity, a vascular comorbidity. Compared to lean mice, obese stroke mice displayed significantly higher circulating monocytes (monocytosis), increased CD45High monocytes/macrophages infiltration to the injured brain, worse acute outcomes, and delayed recovery. Unlike lean mice, obese mice with RLC at 2 hours post-stroke failed to shift circulating monocytes to pro-inflammatory status and nullified RLC-induced functional benefit. The absence of the monocyte shift was also observed in splenocytes incubated with RLC serum from obese mice, while the shift was observed in the cultures with RLC serum from lean mice. These results showed that the alteration of monocytosis and subsets underlies negating RLC benefits in obese mice and suggest careful considerations of comorbidities at the time of RLC application for stroke therapy.
Collapse
Affiliation(s)
- Il-doo Kim
- Burke Neurological Institute, White Plains, NY, USA
| | - Hyunwoo Ju
- Burke Neurological Institute, White Plains, NY, USA
| | | | | | | | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
7
|
Wyatt-Johnson SK, Afify R, Brutkiewicz RR. The immune system in neurological diseases: What innate-like T cells have to say. J Allergy Clin Immunol 2024; 153:913-923. [PMID: 38365015 PMCID: PMC10999338 DOI: 10.1016/j.jaci.2024.02.003] [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: 09/18/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
The immune system classically consists of 2 lines of defense, innate and adaptive, both of which interact with one another effectively to protect us against any pathogenic threats. Importantly, there is a diverse subset of cells known as innate-like T cells that act as a bridge between the innate and adaptive immune systems and are pivotal players in eliciting inflammatory immune responses. A growing body of evidence has demonstrated the regulatory impact of these innate-like T cells in central nervous system (CNS) diseases and that such immune cells can traffic into the brain in multiple pathological conditions, which can be typically attributed to the breakdown of the blood-brain barrier. However, until now, it has been poorly understood whether innate-like T cells have direct protective or causative properties, particularly in CNS diseases. Therefore, in this review, our attention is focused on discussing the critical roles of 3 unique subsets of unconventional T cells, namely, natural killer T cells, γδ T cells, and mucosal-associated invariant T cells, in the context of CNS diseases, disorders, and injuries and how the interplay of these immune cells modulates CNS pathology, in an attempt to gain a better understanding of their complex functions.
Collapse
Affiliation(s)
- Season K Wyatt-Johnson
- Department of Microbiology and Immunology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Ind
| | - Reham Afify
- Department of Microbiology and Immunology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Ind
| | - Randy R Brutkiewicz
- Department of Microbiology and Immunology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Ind.
| |
Collapse
|
8
|
Keevil H, Phillips BE, England TJ. Remote ischemic conditioning for stroke: A critical systematic review. Int J Stroke 2024; 19:271-279. [PMID: 37466245 PMCID: PMC10903142 DOI: 10.1177/17474930231191082] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
Remote ischemic conditioning (RIC) is the application of brief periods of ischemia to an organ or tissue with the aim of inducing protection from ischemia in a distant organ. It was first developed as a cardioprotective strategy but has been increasingly investigated as a neuroprotective intervention. The mechanisms by which RIC achieves neuroprotection are incompletely understood. Preclinical studies focus on the hypothesis that RIC can protect the brain from ischemia reperfusion (IR) injury following the restoration of blood flow after occlusion of a large cerebral artery. However, increasingly, a role of chronic RIC (CRIC) is being investigated as a means of promoting recovery following an ischemic insult to the brain. The recent publication of two large, randomized control trials has provided promise that RIC could improve functional outcomes after acute ischemic stroke, and that there may be a role for CRIC in the prevention of recurrent stroke. Although less developed, there is also proof-of-concept to suggest that RIC may be used to reduce vasospasm after subarachnoid hemorrhage or improve cognitive outcomes in vascular dementia. As a cheap, well-tolerated and almost universally applicable intervention, the motivation for investigating possible benefit of RIC in patients with cerebrovascular disease is great. In this review, we shall review the current evidence for RIC as applied to cerebrovascular disease.
Collapse
Affiliation(s)
- Harry Keevil
- Stroke Trials Unit, Division of Mental Health and Clinical Neuroscience, University of Nottingham, Nottingham, UK
- Medical Research Council Versus Arthritis Centre for Musculoskeletal Ageing Research, and NIHR Nottingham Biomedical Research Centre, Division of Injury, Recovery & Inflammation Sciences, University of Nottingham, Nottingham, UK
| | - Bethan E Phillips
- Medical Research Council Versus Arthritis Centre for Musculoskeletal Ageing Research, and NIHR Nottingham Biomedical Research Centre, Division of Injury, Recovery & Inflammation Sciences, University of Nottingham, Nottingham, UK
| | - Timothy J England
- Stroke Trials Unit, Division of Mental Health and Clinical Neuroscience, University of Nottingham, Nottingham, UK
- Department of Stroke, University Hospitals of Derby and Burton, Derby, UK
| |
Collapse
|
9
|
Zhang L, Zhou X, Zhao J, Wang X. Research hotspots and frontiers of preconditioning in cerebral ischemia: A bibliometric analysis. Heliyon 2024; 10:e24757. [PMID: 38317957 PMCID: PMC10839892 DOI: 10.1016/j.heliyon.2024.e24757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/13/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
Background Preconditioning is a promising strategy against ischemic brain injury, and numerous studies in vitro and in vivo have demonstrated its neuroprotective effects. However, at present there is no bibliometric analysis of preconditioning in cerebral ischemia. Therefore, a comprehensive overview of the current status, hot spots, and emerging trends in this research field is necessary. Materials and methods Studies on preconditioning in cerebral ischemia from January 1999-December 2022 were retrieved from the Web of Science Core Collection (WOSCC) database. CiteSpace was used for data mining and visual analysis. Results A total of 1738 papers on preconditioning in cerebral ischemia were included in the study. The annual publications showed an upwards and then downwards trend but currently remain high in terms of annual publications. The US was the leading country, followed by China, the most active country in recent years. Capital Medical University published the largest number of articles. Perez-Pinzon, Miguel A contributed the most publications, while KITAGAWA K was the most cited author. The focus of the study covered three areas: (1) relevant diseases and experimental models, (2) types of preconditioning and stimuli, and (3) mechanisms of ischemic tolerance. Remote ischemic preconditioning, preconditioning of mesenchymal stem cells (MSCs), and inflammation are the frontiers of research in this field. Conclusion Our study provides a visual and scientific overview of research on preconditioning in cerebral ischemia, providing valuable information and new directions for researchers.
Collapse
Affiliation(s)
- Long Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Traditional Chinese Medicine, Zibo TCM-Integrated Hospital, Zibo ,255026, China
| | - Xue Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jing Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xingchen Wang
- Division of Neurology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| |
Collapse
|
10
|
Ma Y, Sun W, Bai J, Gao F, Ma H, Liu H, Hu J, Xu C, Zhang X, Liu Z, Yuan T, Sun C, Huang Y, Wang R. Targeting blood brain barrier-Remote ischemic conditioning alleviates cognitive impairment in female APP/PS1 rats. CNS Neurosci Ther 2024; 30:e14613. [PMID: 38379185 PMCID: PMC10879645 DOI: 10.1111/cns.14613] [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: 06/16/2023] [Revised: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a significant global health concern, and it is crucial that we find effective methods to prevent or slow down AD progression. Recent studies have highlighted the essential role of blood vessels in clearing Aβ, a protein that contributes to AD. Scientists are exploring blood biomarkers as a potential tool for future AD diagnosis. One promising method that may help prevent AD is remote ischemic conditioning (RIC). RIC involves using sub-lethal ischemic-reperfusion cycles on limbs. However, a comprehensive understanding of how RIC can prevent AD and its long-term effectiveness is still lacking. Further research is essential to fully comprehend the potential benefits of RIC in preventing AD. METHODS Female wild-type (WT) and APP/PS1 transgenic rats, aged 12 months, underwent ovariectomy and were subsequently assigned to WT, APP/PS1, and APP/PS1 + RIC groups. RIC was conducted five times a week for 4 weeks. The rats' depressive and cognitive behaviors were evaluated using force swimming, open-field tests, novel objective recognition, elevated plus maze, and Barnes maze tests. Evaluation of the neurovascular unit (NVU), synapses, vasculature, astrocytes, and microglia was conducted using immunofluorescence staining (IF), Western blot (WB), and transmission electron microscopy (TEM). Additionally, the cerebro-vasculature was examined using micro-CT, and cerebral blood flow (CBF) was measured using Speckle Doppler. Blood-brain barrier (BBB) permeability was determined by measuring the Evans blue leakage. Finally, Aβ levels in the rat frontal cortex were measured using WB, ELISA, or IF staining. RESULTS RIC enhanced memory-related protein expression and rescued depressive-like behavior and cognitive decline in APP/PS1 transgenic rats. Additionally, the intervention protected NVU in the rat frontal cortex, as evidenced by (1) increased expression of TJ (tight junction) proteins, pericyte marker PDGFRβ, and glucose transporter 1 (GLUT1), as well as decreased VCAM1; (2) mitigation of ultrastructure impairment in neuron, cerebral vascular, and astrocyte; (3) upregulation of A2 astrocyte phenotype markers and downregulation of A1 phenotype markers, indicating a shift toward a healthier phenotype. Correspondingly, RIC intervention alleviated neuroinflammation, as evidenced by the decreased Iba1 level, a microglia marker. Meanwhile, RIC intervention elevated CBF in frontal cortex of the rats. Notably, RIC intervention effectively suppressed Aβ toxicity, as demonstrated by the enhancement of α-secretase and attenuation of β-secretase (BACE1) and γ- secretase and Aβ1-42 and Aβ1-40 levels as well. CONCLUSION Chronic RIC intervention exerts vascular and neuroprotective roles, suggesting that RIC could be a promising therapeutic strategy targeting the BBB and NVU during AD development.
Collapse
Affiliation(s)
- Yuxuan Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Wuxiang Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Jing Bai
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Fujia Gao
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Haoran Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Huiyu Liu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Jiewei Hu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Chao Xu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Xin Zhang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Zixuan Liu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Tao Yuan
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Chenxu Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Yuanyuan Huang
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Ruimin Wang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| |
Collapse
|
11
|
Liang H, Ye R, Zhang X, Ye H, Ouyang W, Cai S, Wei L. Autonomic function may mediate the neuroprotection of remote ischemic postconditioning in stroke: A randomized controlled trial. J Stroke Cerebrovasc Dis 2023; 32:107198. [PMID: 37329785 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107198] [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: 01/26/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/19/2023] Open
Abstract
OBJECTIVES To evaluate the effect of remote ischemic postconditioning (RIPostC) on the prognosis of acute ischemic stroke(AIS) patients and investigate the mediating role of autonomic function in the neuroprotection of RIPostC. MATERIALS AND METHODS 132 AIS patients were randomized into two groups. Patients received four cycles of 5-min inflation to a pressure of 200 mmHg(i.e., RIPostC) or patients' diastolic BP(i.e., shame), followed by 5 min of deflation on healthy upper limbs once a day for 30 days. The main outcome was neurological outcome including the National Institutes of Health Stroke Scale (NIHSS), modified Rankin Scale (mRS), and Barthel index(BI). The second outcome measure was autonomic function measured by heart rate variability(HRV). RESULTS Compared with the baseline, the post-intervention NIHSS score was significantly reduced in both groups (P<0.001). NIHSS score was significantly lower in the control group than intervention group at day 7.[RIPostC:3(1,5) versus shame:2(1,4); P=0.030]. mRS scored lower in the intervention group compared with the control group at day 90 follow-up(RIPostC:0.5±2.0 versus shame:1.0±2.0;P=0.016). The goodness-of-fit test revealed a significant difference between the generalized estimating equation model of mRS and BI scores of uncontrolled-HRV and controlled-HRV(P<0.05, both). The results of bootstrap revealed a complete mediation effect of HRV between group on mRS[indirect effect: -0.267 (LLCI = -0.549, ULCI = -0.048), the direct effect: -0.443 (LLCI = -0.831, ULCI = 0.118)]. CONCLUSION This is the first human-based study providing evidence for a mediation role of autonomic function between RIpostC and prognosis in AIS patients. It indicated that RIPostC could improve the neurological outcome of AIS patients. Autonomic function may play a mediating role in this association. TRIAL REGISTRATION The clinical trials registration number for this study is NCT02777099 (ClinicalTrials.gov Identifier).
Collapse
Affiliation(s)
- Hao Liang
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Richun Ye
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaopei Zhang
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huanwen Ye
- Department of Cardiac Function, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenwei Ouyang
- Key Unit of Methodology in Clinical Research, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shuang Cai
- Tongde Hospital of Zhejiang Province, Zhejiang, China
| | - Lin Wei
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Yang H, Hu Z, Gao X, Su J, Jiang H, Yang S, Zhang Q, Ni W, Gu Y. Safety and efficacy of remote ischemic conditioning in adult moyamoya disease patients undergoing revascularization surgery: a pilot study. Front Neurol 2023; 14:1200534. [PMID: 37576009 PMCID: PMC10419176 DOI: 10.3389/fneur.2023.1200534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Background and purpose Revascularization surgery for patients with moyamoya disease (MMD) is very complicated and has a high rate of postoperative complications. This pilot study aimed to prove the safety and efficacy of remote ischemic conditioning (RIC) in adult MMD patients undergoing revascularization surgery. Methods A total of 44 patients with MMD were enrolled in this single-center, open-label, prospective, parallel randomized study, including 22 patients assigned to the sham group and 22 patients assigned to the RIC group. The primary outcome was the incidence of major neurologic complications during the perioperative period. Secondary outcomes were the modified Rankin Scale (mRS) score at discharge, at 90 days post-operation, and at 1 year after the operation. The outcome of safety was the incidence of adverse events associated with RIC. Blood samples were obtained to monitor the serum concentrations of cytokines (VEGF, IL-6). Results No subjects experienced adverse events during RIC intervention, and all patients could tolerate the RIC intervention in the perioperative period. The incidence of major neurologic complications was significantly lower in the RIC group compared with the control group (18.2% vs. 54.5%, P = 0.027). The mRS score at discharge in the RIC group was also lower than the control group (0.86 ± 0.99 vs. 1.18 ± 1.22, P = 0.035). In addition, the serum IL-6 level increased significantly at 7 days after bypass surgery in the control group and the serum level of VEGF at 7 days post-operation in the RIC group. Conclusion In conclusion, our study demonstrated the neuroprotective effect of RIC by reducing perioperative complications and improving cerebral blood flow in adult MMD patients undergoing revascularization surgery. Thus, RIC seems to be a potential treatment method for MMD. Clinical trial registration ClinicalTrials.gov, identifier: NCT05860946.
Collapse
Affiliation(s)
- Heng Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| | - Zhenzhen Hu
- Department of Nursing, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xinjie Gao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| | - Jiabin Su
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hanqiang Jiang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaoxuan Yang
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| | - Qing Zhang
- Department of Nursing, Huashan Hospital North, Fudan University, Shanghai, China
| | - Wei Ni
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| | - Yuxiang Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Xu Y, Wang Y, Ji X. Immune and inflammatory mechanism of remote ischemic conditioning: A narrative review. Brain Circ 2023; 9:77-87. [PMID: 37576576 PMCID: PMC10419737 DOI: 10.4103/bc.bc_57_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 08/15/2023] Open
Abstract
The benefits of remote ischemic conditioning (RIC) on multiple organs have been extensively investigated. According to existing research, suppressing the immune inflammatory response is an essential mechanism of RIC. Based on the extensive effects of RIC on cardiovascular and cerebrovascular diseases, this article reviews the immune and inflammatory mechanisms of RIC and summarizes the effects of RIC on immunity and inflammation from three perspectives: (1) the mechanisms of the impact of RIC on inflammation and immunity; (2) evidence of the effects of RIC on immune and inflammatory processes in ischaemic stroke; and (3) possible future applications of this effect, especially in systemic infectious diseases such as sepsis and sepsis-associated encephalopathy. This review explores the possibility of using RIC as a treatment in more inflammation-related diseases, which will provide new ideas for the treatment of this kind of disease.
Collapse
Affiliation(s)
- Yi Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Li X, Ren C, Li S, Zhao W, Wang P, Ji X. The antihypertensive effect of remote ischemic conditioning in spontaneously hypertensive rats. Front Immunol 2023; 13:1093262. [PMID: 36713422 PMCID: PMC9878686 DOI: 10.3389/fimmu.2022.1093262] [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: 11/09/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Purpose Limb remote ischemic conditioning (LRIC) may be an effective method to control hypertension. This study investigated whether LRIC decreases blood pressure by regulating the hypertensive inflammatory response in spontaneously hypertensive rats (SHR). Method The SHR and aged-matched Wistar rats with different ages were randomly assigned to the SHR group, SHR+LRIC group, Wistar group, and Wistar + LRIC group. LRIC was conducted by tightening a tourniquet around the upper thigh and releasing it for three cycles daily (10 mins x3 cycles). Blood pressure, the percentage of monocytes and T lymphocytes, and the concentration of pro-inflammatory cytokines in the blood were analyzed. Results The blood pressure of SHR was significantly higher than that of age-matched Wistar rats. LRIC decreased blood pressure in SHR at different ages (4, 8, and 16 weeks old), but had no effect on the blood pressure in Wistar rats. Flow cytometry analysis showed that blood monocytes and CD8 T cells of SHR were higher than those of Wistar rats. LRIC significantly decreased the percentage of monocytes and CD8 T cells in SHR. Consistent with the changes of immune cells, the levels of plasma IL-6 and TNF-α in SHR were also higher. And LRIC attenuated the plasma IL-6 and TNF-α levels in SHR. Conclusion LRIC may decreased the blood pressure via modulation of the inflammatory response in SHR.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Neurology, Aerospace center Hospital, Beijing, China
- Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Peifu Wang
- Department of Neurology, Aerospace center Hospital, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Rizzo PA, Bellavia S, Scala I, Colò F, Broccolini A, Antonica R, Vitali F, Angeloni BM, Brunetti V, Di Iorio R, Monforte M, Della Marca G, Calabresi P, Luigetti M, Frisullo G. COVID-19 Vaccination Is Associated with a Better Outcome in Acute Ischemic Stroke Patients: A Retrospective Observational Study. J Clin Med 2022; 11:jcm11236878. [PMID: 36498464 PMCID: PMC9737827 DOI: 10.3390/jcm11236878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Background: It is unclear whether and how COVID-19 vaccination may affect the outcome of patients with acute ischemic stroke (AIS). We investigated this potential association in a retrospective study by comparing previously vaccinated (VAX) versus unvaccinated (NoVAX) stroke patients. Methods: We collected clinical reports for all consecutive AIS patients admitted to our hospital and evaluated the outcome predictors in VAX and NoVAX groups. Adjustments were made for possible confounders in multivariable logistic regression analysis, and adjusted hazard ratios were calculated. Results: A total of 466 AIS patients (287 VAX and 179 NoVAX) were included in this study. The NIHSS score at discharge and mRS score at a 3-month follow-up visit were significantly lower in VAX patients compared to NoVAX patients (p < 0.001). Good outcomes (mRS 0−2) were significantly associated with COVID-19 vaccination before AIS (adjusted hazard ratio, 0.400 [95% CI = 0.216−0.741]). Conclusions: The observation that COVID-19 vaccination can influence the outcome of AIS provides support for further studies investigating the role of immunity in ischemic brain damage.
Collapse
Affiliation(s)
- Pier Andrea Rizzo
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Bellavia
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Irene Scala
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Francesca Colò
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Aldobrando Broccolini
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Riccardo Antonica
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Francesca Vitali
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | | | - Valerio Brunetti
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Riccardo Di Iorio
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Mauro Monforte
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Giacomo Della Marca
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Paolo Calabresi
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Marco Luigetti
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-3015-4435
| | - Giovanni Frisullo
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
16
|
Cerebral Ischemia/Reperfusion Injury and Pharmacologic Preconditioning as a Means to Reduce Stroke-induced Inflammation and Damage. Neurochem Res 2022; 47:3598-3614. [DOI: 10.1007/s11064-022-03789-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
|
17
|
Li Y, Xin G, Li S, Dong Y, Zhu Y, Yu X, Wan C, Li F, Wei Z, Wang Y, Zhang K, Chen Q, Niu H, Huang W. PD-L1 Regulates Platelet Activation and Thrombosis via Caspase-3/GSDME Pathway. Front Pharmacol 2022; 13:921414. [PMID: 35784685 PMCID: PMC9240427 DOI: 10.3389/fphar.2022.921414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets play a central role in hemostasis and thrombosis, regulating the occurrence and development of thrombotic diseases, including ischemic stroke. Programmed death ligand 1 (PD-L1) has recently been detected in platelet, while the function of PD-L1 in platelets remain elusive. Our data reveal a novel mechanism for the role of PD-L1 on platelet activation and arterial thrombosis. PD-L1 knockout does not affect platelet morphology, count, and mean volume under homeostasis and without risk of bleeding, which inhibits platelet activation by suppressing outside-in-activation of integrin by downregulating the Caspase-3/GSDME pathway. Platelet adoptive transfer experiments demonstrate that PD-L1 knockout inhibits thrombosis. And the absence of PD-L1 improves ischemic stroke severity and increases mice survival. Immunohistochemical staining of the internal structure of the thrombus proves that PD-L1 enhances the seriousness of the thrombus by inhibiting platelet activation. This work reveals a regulatory role of PD-L1 on platelet activation and thrombosis while providing novel platelet intervention strategies to prevent thrombosis.
Collapse
|
18
|
Mollet I, Martins C, Ângelo-Dias M, Carvalho AS, Aloria K, Matthiesen R, Baptista MV, Borrego LM, Vieira HL. Pilot study in human healthy volunteers on the mechanisms underlying remote ischemic conditioning (RIC) – Targeting circulating immune cells and immune-related proteins. J Neuroimmunol 2022; 367:577847. [DOI: 10.1016/j.jneuroim.2022.577847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022]
|
19
|
Zhang B, Zhao W, Ma H, Zhang Y, Che R, Bian T, Yan H, Xu J, Wang L, Yu W, Liu J, Song H, Duan J, Chang H, Ma Q, Zhang Q, Ji X. Remote Ischemic Conditioning in the Prevention for Stroke-Associated Pneumonia: A Pilot Randomized Controlled Trial. Front Neurol 2022; 12:723342. [PMID: 35185744 PMCID: PMC8850400 DOI: 10.3389/fneur.2021.723342] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/21/2021] [Indexed: 01/02/2023] Open
Abstract
BackgroundDespite the continuing effort in investigating the preventive therapies for stroke-associated pneumonia (SAP), which is closely associated with unfavorable outcomes, conclusively effective therapy for the prevention of SAP is still lacking. Remote ischemic conditioning (RIC) has been proven to improve the survival in the sepsis model and inflammatory responses have been indicated as important mechanisms involved in the multi-organ protection effect of RIC. This study aimed to assess the safety and the preliminary efficacy of RIC in the prevention of SAP in patients with acute ischemic stroke.MethodsWe performed a proof-of-concept, pilot open-label randomized controlled trial. Eligible patients (age > 18 years) within 48 h after stroke onset between March 2019 and October 2019 with acute ischemic stroke were randomly allocated (1:1) to the RIC group and the control group. All participants received standard medical therapy. Patients in the RIC group underwent RIC twice daily for 6 consecutive days. The safety outcome included any adverse events associated with RIC procedures. The efficacy outcome included the incidence of SAP, changes of immunological profiles including mHLA-DR, TLR-2, and TLR-4 as well as other plasma parameters from routine blood tests.ResultsIn total, 46 patients aged 63.1 ± 12.5 years, were recruited (23 in each group). Overall, 19 patients in the RIC group and 22 patients in the control group completed this study. No severe adverse event was attributed to RIC procedures. The incidence of SAP was lower in the remote ischemic conditioning group (2 patients [10.5%]) than that in the control group (6 patients [27.3%]), but no significant difference was detected in both univariate and multivariate analysis (p = 0.249 and adjusted p = 0.666). No significance has been found in this pilot trial in the level of immunological profiles HLA-DR, TLR4 and TLR2 expressed on monocytes as well as blood parameters tested through routine blood tests between the two groups (p > 0.05). The IL-6 and IL-1β levels at day 5 after admission in the RIC group were lower than those in the control group (p < 0.05).InterpretationThis proof-of-concept pilot randomized controlled trial was to investigate RIC as a prevention method for SAP. Remote ischemic conditioning is safe in the prevention of SAP in patients with acute ischemic stroke. The preventive effect of RIC on SAP should be further validated in future studies.
Collapse
Affiliation(s)
- Bowei Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongrui Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yunzhou Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ruiwen Che
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tingting Bian
- Department of Neurology, Beijing Fengtai You'anmen Hospital, Beijing, China
| | - Heli Yan
- Department of Neurology, Beijing Fengtai You'anmen Hospital, Beijing, China
| | - Jiali Xu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lin Wang
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wantong Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jia Liu
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jiangang Duan
- Department of Emergency, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hong Chang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qingfeng Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qian Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xunming Ji
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- *Correspondence: Xunming Ji
| |
Collapse
|
20
|
Synergetic protective effect of remote ischemic preconditioning and prolyl 4‑hydroxylase inhibition in ischemic cardiac injury. Mol Med Rep 2022; 25:80. [PMID: 35029283 PMCID: PMC8778658 DOI: 10.3892/mmr.2022.12596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
It has been reported that hypoxia-inducible factor 1α (HIF-1α) serves a key role in the protective effect of remote ischemic preconditioning (RIP) in ischemia/reperfusion (I/R)-induced cardiac injury. Moreover, inhibition of prolyl 4-hydroxylase (PHD), an enzyme responsible for HIF-1α degradation, prevents I/R-induced cardiac injury. However, whether their protective effects are synergetic remains to be elucidated. The present study aimed to investigate the protective effect of RIP, PHD inhibition using dimethyloxalylglycine (DMOG) and their combination on I/R-induced cardiac injury. Rabbits were randomly divided into seven groups: i) Sham; ii) I/R; iii) lung RIP + I/R; iv) thigh RIP + I/R; v) DMOG + I/R; vi) DMOG + lung RIP + I/R; and vii) DMOG + thigh RIP + I/R. I/R models were established via 30 min left coronary artery occlusion and 3 h reperfusion. For lung/thigh RIP, rabbits received left pulmonary artery (or left limb) ischemia for 25 min and followed by release for 5 min. Some rabbits were administered 20 mg/kg DMOG. The results demonstrated that both lung/thigh RIP and DMOG significantly decreased myocardial infarct size, creatine kinase activity and myocardial apoptosis in I/R rabbits. Furthermore, the combination of RIP and PHD inhibition exerted synergetic protective effects on these aforementioned changes. The mechanistic study indicated that both treatments increased mRNA and protein expression levels of HIF-1α and its downstream regulators, including vascular endothelial growth factor (VEGF), AKT and endothelial nitric oxide synthase (eNOS). In conclusion, the present study demonstrated that RIP and PHD inhibition exerted synergetic protective effects on cardiac injury via activation of HIF-1α and the downstream VEGF/AKT-eNOS signaling pathway.
Collapse
|
21
|
Abstract
Cerebral ischemic injury may lead to a series of serious brain diseases, death or different degrees of disability. Hypoxia-inducible factor-1α (HIF-1α) is an oxygen-sensitive transcription factor, which mediates the adaptive metabolic response to hypoxia and serves a key role in cerebral ischemia. HIF-1α is the main molecule that responds to hypoxia. HIF-1α serves an important role in the development of cerebral ischemia by participating in numerous processes, including metabolism, proliferation and angiogenesis. The present review focuses on the endogenous protective mechanism of cerebral ischemia and elaborates on the role of HIF-1α in cerebral ischemia. In addition, it focuses on cerebral ischemia interventions that act on the HIF-1α target, including biological factors, non-coding RNA, hypoxic-ischemic preconditioning and drugs, and expands upon the measures to strengthen the endogenous compensatory response to support HIF-1α as a therapeutic target, thus providing novel suggestions for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Qingna Li
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Hua Han
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
22
|
Abbasi-Habashi S, Jickling GC, Winship IR. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front Neurol 2021; 12:746486. [PMID: 34956045 PMCID: PMC8695500 DOI: 10.3389/fneur.2021.746486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic conditioning (RIC), which involves a series of short cycles of ischemia in an organ remote to the brain (typically the limbs), has been shown to protect the ischemic penumbra after stroke and reduce ischemia/reperfusion (IR) injury. Although the exact mechanism by which this protective signal is transferred from the remote site to the brain remains unclear, preclinical studies suggest that the mechanisms of RIC involve a combination of circulating humoral factors and neuronal signals. An improved understanding of these mechanisms will facilitate translation to more effective treatment strategies in clinical settings. In this review, we will discuss potential protective mechanisms in the brain and cerebral vasculature associated with RIC. We will discuss a putative role of the immune system and circulating mediators of inflammation in these protective processes, including the expression of pro-and anti-inflammatory genes in peripheral immune cells that may influence the outcome. We will also review the potential role of extracellular vesicles (EVs), biological vectors capable of delivering cell-specific cargo such as proteins and miRNAs to cells, in modulating the protective effects of RIC in the brain and vasculature.
Collapse
Affiliation(s)
- Sima Abbasi-Habashi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
23
|
Torres-Querol C, Quintana-Luque M, Arque G, Purroy F. Preclinical evidence of remote ischemic conditioning in ischemic stroke, a metanalysis update. Sci Rep 2021; 11:23706. [PMID: 34887465 PMCID: PMC8660795 DOI: 10.1038/s41598-021-03003-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/12/2021] [Indexed: 01/13/2023] Open
Abstract
Remote ischemic conditioning (RIC) is a promising therapeutic approach for ischemic stroke patients. It has been proven that RIC reduces infarct size and improves functional outcomes. RIC can be applied either before ischemia (pre-conditioning; RIPreC), during ischemia (per-conditioning; RIPerC) or after ischemia (post-conditioning; RIPostC). Our aim was to systematically determine the efficacy of RIC in reducing infarct volumes and define the cellular pathways involved in preclinical animal models of ischemic stroke. A systematic search in three databases yielded 50 peer-review articles. Data were analyzed using random effects models and results expressed as percentage of reduction in infarct size (95% CI). A meta-regression was also performed to evaluate the effects of covariates on the pooled effect-size. 95.3% of analyzed experiments were carried out in rodents. Thirty-nine out of the 64 experiments studied RIPostC (61%), sixteen examined RIPreC (25%) and nine tested RIPerC (14%). In all studies, RIC was shown to reduce infarct volume (- 38.36%; CI - 42.09 to - 34.62%) when compared to controls. There was a significant interaction caused by species. Short cycles in mice significantly reduces infarct volume while in rats the opposite occurs. RIPreC was shown to be the most effective strategy in mice. The present meta-analysis suggests that RIC is more efficient in transient ischemia, using a smaller number of RIC cycles, applying larger length of limb occlusion, and employing barbiturates anesthetics. There is a preclinical evidence for RIC, it is safe and effective. However, the exact cellular pathways and underlying mechanisms are still not fully determined, and its definition will be crucial for the understanding of RIC mechanism of action.
Collapse
Affiliation(s)
- Coral Torres-Querol
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Manuel Quintana-Luque
- Epilepsy Unit, Neurology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gloria Arque
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
- Experimental Medicine Department, Universitat de Lleida, Lleida, Spain
| | - Francisco Purroy
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain.
- Medicine Department, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain.
- Stroke Unit, Department of Neurology, Universitat de Lleida, Hospital Universitari Arnau de Vilanova, Clinical Neurosciences Group IRBLleida, Avda Rovira Roure 80, 25198, Lleida, Spain.
| |
Collapse
|
24
|
Wang X, Ji X. Interactions between remote ischemic conditioning and post-stroke sleep regulation. Front Med 2021; 15:867-876. [PMID: 34811643 DOI: 10.1007/s11684-021-0887-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/31/2021] [Indexed: 12/31/2022]
Abstract
Sleep disturbances are common in patients with stroke, and sleep quality has a critical role in the onset and outcome of stroke. Poor sleep exacerbates neurological injury, impedes nerve regeneration, and elicits serious complications. Thus, exploring a therapy suitable for patients with stroke and sleep disturbances is imperative. As a multi-targeted nonpharmacological intervention, remote ischemic conditioning can reduce the ischemic size of the brain, improve the functional outcome of stroke, and increase sleep duration. Preclinical/clinical evidence showed that this method can inhibit the inflammatory response, mediate the signal transductions of adenosine, activate the efferents of the vagal nerve, and reset the circadian clocks, all of which are involved in sleep regulation. In particular, cytokines tumor necrosis factor α (TNFα) and adenosine are sleep factors, and electrical vagal nerve stimulation can improve insomnia. On the basis of the common mechanisms of remote ischemic conditioning and sleep regulation, a causal relationship was proposed between remote ischemic conditioning and post-stroke sleep quality.
Collapse
Affiliation(s)
- Xian Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, 100069, China.
| |
Collapse
|
25
|
Mollet I, Marto JP, Mendonça M, Baptista MV, Vieira HLA. Remote but not Distant: a Review on Experimental Models and Clinical Trials in Remote Ischemic Conditioning as Potential Therapy in Ischemic Stroke. Mol Neurobiol 2021; 59:294-325. [PMID: 34686988 PMCID: PMC8533672 DOI: 10.1007/s12035-021-02585-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Stroke is one of the main causes of neurological disability worldwide and the second cause of death in people over 65 years old, resulting in great economic and social burden. Ischemic stroke accounts for 85% of total cases, and the approved therapies are based on re-establishment of blood flow, and do not directly target brain parenchyma. Thus, novel therapies are urgently needed. In this review, limb remote ischemic conditioning (RIC) is revised and discussed as a potential therapy against ischemic stroke. The review targets both (i) fundamental research based on experimental models and (ii) clinical research based on clinical trials and human interventional studies with healthy volunteers. Moreover, it also presents two approaches concerning RIC mechanisms in stroke: (i) description of the underlying cerebral cellular and molecular mechanisms triggered by limb RIC that promote neuroprotection against stroke induced damage and (ii) the identification of signaling factors involved in inter-organ communication following RIC procedure. Limb to brain remote signaling can occur via circulating biochemical factors, immune cells, and/or stimulation of autonomic nervous system. In this review, these three hypotheses are explored in both humans and experimental models. Finally, the challenges involved in translating experimentally generated scientific knowledge to a clinical setting are also discussed.
Collapse
Affiliation(s)
- Inês Mollet
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Campus de Caparica, 2829-526, Caparica, Portugal.,CEDOC, Faculdade de Ciências Médicas/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João Pedro Marto
- CEDOC, Faculdade de Ciências Médicas/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Marcelo Mendonça
- CEDOC, Faculdade de Ciências Médicas/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Champalimaud Research, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Miguel Viana Baptista
- CEDOC, Faculdade de Ciências Médicas/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Helena L A Vieira
- UCIBIO, Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Campus de Caparica, 2829-526, Caparica, Portugal. .,CEDOC, Faculdade de Ciências Médicas/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal. .,Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal.
| |
Collapse
|
26
|
Yan Y, Gu T, Christensen SDK, Su J, Lassen TR, Hjortbak MV, Lo IJ, Venø ST, Tóth AE, Song P, Nielsen MS, Bøtker HE, Blagoev B, Drasbek KR, Kjems J. Cyclic Hypoxia Conditioning Alters the Content of Myoblast-Derived Extracellular Vesicles and Enhances Their Cell-Protective Functions. Biomedicines 2021; 9:biomedicines9091211. [PMID: 34572398 PMCID: PMC8471008 DOI: 10.3390/biomedicines9091211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Remote ischemic conditioning (RIC) is a procedure that can attenuate ischemic-reperfusion injury by conducting brief cycles of ischemia and reperfusion in the arm or leg. Extracellular vesicles (EVs) circulating in the bloodstream can release their content into recipient cells to confer protective function on ischemia-reperfusion injured (IRI) organs. Skeletal muscle cells are potential candidates to release EVs as a protective signal during RIC. In this study, we used C2C12 cells as a model system and performed cyclic hypoxia-reoxygenation (HR) to mimic RIC. EVs were collected and subjected to small RNA profiling and proteomics. HR induced a distinct shift in the miRNA profile and protein content in EVs. HR EV treatment restored cell viability, dampened inflammation, and enhanced tube formation in in vitro assays. In vivo, HR EVs showed increased accumulation in the ischemic brain compared to EVs secreted from normoxic culture (N EVs) in a mouse undergoing transient middle cerebral artery occlusion (tMCAO). We conclude that HR conditioning changes the miRNA and protein profile in EVs released by C2C12 cells and enhances the protective signal in the EVs to recipient cells in vitro.
Collapse
Affiliation(s)
- Yan Yan
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Omiics ApS, 8200 Aarhus, Denmark;
| | - Tingting Gu
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Stine Duelund Kaas Christensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Junyi Su
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | - Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - IJu Lo
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Andrea Erzsebet Tóth
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.E.T.); (M.S.N.)
| | - Ping Song
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Kim Ryun Drasbek
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Correspondence: ; Tel.: +45-289-920-86
| |
Collapse
|
27
|
Quantitative Proteomic Analysis of Plasma after Remote Ischemic Conditioning in a Rhesus Monkey Ischemic Stroke Model. Biomolecules 2021; 11:biom11081164. [PMID: 34439830 PMCID: PMC8393806 DOI: 10.3390/biom11081164] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Animal and clinical studies have shown that remote ischemic conditioning (RIC) has protective effects for cerebral vascular diseases, with induced humoral factor changes in the peripheral blood. However, many findings are heterogeneous, perhaps due to differences in the RIC intervention schemes, enrolled populations, and sample times. This study aimed to examine the RIC-induced changes in the plasma proteome using rhesus monkey models of strokes. Methods: Two adult rhesus monkeys with autologous blood clot-induced middle cerebral artery (MCA) occlusion underwent RIC interventions twice a week for five consecutive weeks. Each RIC treatment included five cycles of five minutes of ischemia alternating with five minutes of reperfusion of the forearm. The blood samples were taken from the median cubital vein of the monkeys at baseline and immediately after each week’s RIC stimulus. The plasma samples were isolated for a proteomic analysis using mass spectrometry (MS). Results: Several proteins related to lipid metabolism (Apolipoprotein A-II and Apolipoprotein C-II), coagulation (Fibrinogen alpha chain and serpin), immunoinflammatory responses (complement C3 and C1), and endovascular hemostasis (basement membrane-specific heparan sulfate proteoglycan) were significantly modulated after the RIC intervention. Many of these induced changes, such as in the lipid metabolism regulation and anticoagulation responses, starting as early as two weeks following the RIC intervention. The complementary activation and protection of the endovascular cells occurred more than three weeks postintervention. Conclusions: Multiple protective effects were induced by RIC and involved lipid metabolism regulation (anti-atherogenesis), anticoagulation (antithrombosis), complement activation, and endovascular homeostasis (anti-inflammation). In conclusion, this study indicates that RIC results in significant modulations of the plasma proteome. It also provides ideas for future research and screening targets.
Collapse
|
28
|
Liu J, Gu Y, Guo M, Ji X. Neuroprotective effects and mechanisms of ischemic/hypoxic preconditioning on neurological diseases. CNS Neurosci Ther 2021; 27:869-882. [PMID: 34237192 PMCID: PMC8265941 DOI: 10.1111/cns.13642] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
As the organ with the highest demand for oxygen, the brain has a poor tolerance to ischemia and hypoxia. Despite severe ischemia/hypoxia induces the occurrence and development of various central nervous system (CNS) diseases, sublethal insult may induce strong protection against subsequent fatal injuries by improving tolerance. Searching for potential measures to improve brain ischemic/hypoxic is of great significance for treatment of ischemia/hypoxia related CNS diseases. Ischemic/hypoxic preconditioning (I/HPC) refers to the approach to give the body a short period of mild ischemic/hypoxic stimulus which can significantly improve the body's tolerance to subsequent more severe ischemia/hypoxia event. It has been extensively studied and been considered as an effective therapeutic strategy in CNS diseases. Its protective mechanisms involved multiple processes, such as activation of hypoxia signaling pathways, anti-inflammation, antioxidant stress, and autophagy induction, etc. As a strategy to induce endogenous neuroprotection, I/HPC has attracted extensive attention and become one of the research frontiers and hotspots in the field of neurotherapy. In this review, we discuss the basic and clinical research progress of I/HPC on CNS diseases, and summarize its mechanisms. Furthermore, we highlight the limitations and challenges of their translation from basic research to clinical application.
Collapse
Affiliation(s)
- Jia Liu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Ran Y, Ye L, Ding Z, Gao F, Yang S, Fang B, Liu Z, Xi J. Melatonin Protects Against Ischemic Brain Injury by Modulating PI3K/AKT Signaling Pathway via Suppression of PTEN Activity. ASN Neuro 2021; 13:17590914211022888. [PMID: 34120482 PMCID: PMC8207287 DOI: 10.1177/17590914211022888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide with limited therapeutic options. Melatonin can attenuate ischemic brain damage with improved functional outcomes. However, the cellular mechanisms of melatonin-driven neuroprotection against post-stroke neuronal death remain unknown. Here, distal middle cerebral artery occlusion (dMCAO) was performed in C57BL/6j mice to develop an ischemic stroke in vivo model. Melatonin was injected intraperitoneally immediately after ischemia, and 24 and 48 hours later. Melatonin treatment, with 5 to 20 mg/kg, elicited a dose-dependent decrease in infarct volume and concomitant increase in sensorimotor function. At the molecular level, phosphorylation of PTEN and Akt were increased, whereas PTEN activity was decreased in melatonin treated animals 72 hours after dMCAO. At the cellular level, oxygenglucose deprivation (OGD) challenge of neuronal cell line Neuro-2a (N2a) and primary neurons supported melatonin’s direct protection against neuronal cell death. Melatonin treatment reduced LDH release and neuronal apoptosis at various time points, markedly increased Akt phosphorylation in neuronal membrane, but significantly suppressed it in the cytoplasm of post-OGD neurons. Mechanistically, melatonin-induced Akt phosphorylation and neuronal survival was blocked by Wortmannin, a potent PIP3 inhibitor, exposing increased PI3K/Akt activation as a central player in melatonin-driven neuroprotection. Finally, PTEN knock-down through siRNA significantly inhibited PI3K/Akt activation and cell survival following melatonin treatment, suggesting that melatonin protection against ischemic brain damage, is at least partially, dependent on modulation of the PTEN/PI3K/Akt signaling axis.
Collapse
Affiliation(s)
- Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Haidian, China
| | - Zitong Ding
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Fuhai Gao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Shuiqing Yang
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Boyan Fang
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| | - Jianing Xi
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Shijingshan, China
| |
Collapse
|
30
|
Wang L, Ren C, Li Y, Gao C, Li N, Li H, Wu D, He X, Xia C, Ji X. Remote ischemic conditioning enhances oxygen supply to ischemic brain tissue in a mouse model of stroke: Role of elevated 2,3-biphosphoglycerate in erythrocytes. J Cereb Blood Flow Metab 2021; 41:1277-1290. [PMID: 32933360 PMCID: PMC8142126 DOI: 10.1177/0271678x20952264] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxygen supply for ischemic brain tissue during stroke is critical to neuroprotection. Remote ischemic conditioning (RIC) treatment is effective for stroke. However, it is not known whether RIC can improve brain tissue oxygen supply. In current study, we employed a mouse model of stroke created by middle cerebral artery occlusion (MCAO) to investigate the effect of RIC on oxygen supply to the ischemic brain tissue using a hypoxyprobe system. Erythrocyte oxygen-carrying capacity and tissue oxygen exchange were assessed by measuring oxygenated hemoglobin and oxygen dissociation curve. We found that RIC significantly mitigated hypoxic signals and decreased neural cell death, thereby preserving neurological functions. The tissue oxygen exchange was markedly enhanced, along with the elevated hemoglobin P50 and right-shifted oxygen dissociation curve. Intriguingly, RIC markedly elevated 2,3-biphosphoglycerate (2,3-BPG) levels in erythrocyte, and the erythrocyte 2,3-BPG levels were highly negatively correlated with the hypoxia in the ischemic brain tissue. Further, adoptive transfusion of 2,3-BPG-rich erythrocytes prepared from RIC-treated mice significantly enhanced the oxygen supply to the ischemic tissue in MCAO mouse model. Collectively, RIC protects against ischemic stroke through improving oxygen supply to the ischemic brain tissue where the enhanced tissue oxygen delivery and exchange by RIC-induced 2,3-BPG-rich erythrocytes may play a role.
Collapse
Affiliation(s)
- Lin Wang
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Yang Li
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ning Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoduo He
- Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changqing Xia
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Geriatric Medical Research Center, Beijing, China.,Deparment of Neurology, China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| |
Collapse
|
31
|
O'Brien L, Jacobs I. Methodological Variations Contributing to Heterogenous Ergogenic Responses to Ischemic Preconditioning. Front Physiol 2021; 12:656980. [PMID: 33995123 PMCID: PMC8117357 DOI: 10.3389/fphys.2021.656980] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
Ischemic preconditioning (IPC) has been repeatedly reported to augment maximal exercise performance over a range of exercise durations and modalities. However, an examination of the relevant literature indicates that the reproducibility and robustness of ergogenic responses to this technique are variable, confounding expectations about the magnitude of its effects. Considerable variability among study methodologies may contribute to the equivocal responses to IPC. This review focuses on the wide range of methodologies used in IPC research, and how such variability likely confounds interpretation of the interactions of IPC and exercise. Several avenues are recommended to improve IPC methodological consistency, which should facilitate a future consensus about optimizing the IPC protocol, including due consideration of factors such as: location of the stimulus, the time between treatment and exercise, individualized tourniquet pressures and standardized tourniquet physical characteristics, and the incorporation of proper placebo treatments into future study designs.
Collapse
Affiliation(s)
- Liam O'Brien
- Human Physiology Laboratory, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Ira Jacobs
- Human Physiology Laboratory, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Pearce L, Davidson SM, Yellon DM. Does remote ischaemic conditioning reduce inflammation? A focus on innate immunity and cytokine response. Basic Res Cardiol 2021; 116:12. [PMID: 33629195 PMCID: PMC7904035 DOI: 10.1007/s00395-021-00852-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
The benefits of remote ischaemic conditioning (RIC) have been difficult to translate to humans, when considering traditional outcome measures, such as mortality and heart failure. This paper reviews the recent literature of the anti-inflammatory effects of RIC, with a particular focus on the innate immune response and cytokine inhibition. Given the current COVID-19 pandemic, the inflammatory hypothesis of cardiac protection is an attractive target on which to re-purpose such novel therapies. A PubMed/MEDLINE™ search was performed on July 13th 2020, for the key terms RIC, cytokines, the innate immune system and inflammation. Data suggest that RIC attenuates inflammation in animals by immune conditioning, cytokine inhibition, cell survival and the release of anti-inflammatory exosomes. It is proposed that RIC inhibits cytokine release via a reduction in nuclear factor kappa beta (NF-κB)-mediated NLRP3 inflammasome production. In vivo, RIC attenuates pro-inflammatory cytokine release in myocardial/cerebral infarction and LPS models of endotoxaemia. In the latter group, cytokine inhibition is associated with a profound survival benefit. Further clinical trials should establish whether the benefits of RIC in inflammation can be observed in humans. Moreover, we must consider whether uncomplicated MI and elective surgery are the most suitable clinical conditions in which to test this hypothesis.
Collapse
Affiliation(s)
- Lucie Pearce
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
33
|
Kim E, Cho S. CNS and peripheral immunity in cerebral ischemia: partition and interaction. Exp Neurol 2021; 335:113508. [PMID: 33065078 PMCID: PMC7750306 DOI: 10.1016/j.expneurol.2020.113508] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/28/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Stroke elicits excessive immune activation in the injured brain tissue. This well-recognized neural inflammation in the brain is not just an intrinsic organ response but also a result of additional intricate interactions between infiltrating peripheral immune cells and the resident immune cells in the affected areas. Given that there is a finite number of immune cells in the organism at the time of stroke, the partitioned immune systems of the central nervous system (CNS) and periphery must appropriately distribute the limited pool of immune cells between the two domains, mounting a necessary post-stroke inflammatory response by supplying a sufficient number of immune cells into the brain while maintaining peripheral immunity. Stroke pathophysiology has mainly been neurocentric in focus, but understanding the distinct roles of the CNS and peripheral immunity in their concerted action against ischemic insults is crucial. This review will discuss stroke-induced influences of the peripheral immune system on CNS injury/repair and of neural inflammation on peripheral immunity, and how comorbidity influences each.
Collapse
Affiliation(s)
- Eunhee Kim
- Vivian L. Smith Department of Neurosurgery at University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, United States of America; Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, United States of America.
| |
Collapse
|
34
|
Liu J, Sun X, Jin H, Yan XL, Huang S, Guo ZN, Yang Y. Remote ischemic conditioning: A potential therapeutic strategy of type 2 diabetes. Med Hypotheses 2020; 146:110409. [PMID: 33277103 DOI: 10.1016/j.mehy.2020.110409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/12/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is one of the major public diseases which is characterized by peripheral insulin resistance (IR) and progressive pancreatic β-cell failure. While in the past few years, some new factors, such as inflammation, oxidative stress, immune responses and other potential pathways, have been identified to play critical roles in T2D, and thereby provide novel promising targets for the treatment of T2D. Remote ischemic conditioning (RIC) is a non-invasive and convenient operation performed by transient, repeated ischemia in distant place. Nowadays, RIC has been established as a potentially powerful therapeutic tool for many diseases, especially in I/R injuries. Through activating a series of neural, humoral and immune pathways, it can release multiple protective signals, which then regulating inflammation, oxidative stress, immune response and so on. Interestingly, several recent studies have discovered that the beneficial effects of RIC on I/R injuries might be abolished by T2D, wherein the higher basal levels of inflammation and oxidative stress, dysregulation of immune system and some potential pathways secondary to hyperglycemia may play critical roles. In contrast, a higher intensity of conditioning could restore the protective effects. Based on the overlapped mechanisms RIC and T2D performs, we provide a hypothesis that RIC may also play a protective role in T2D via targeting these signaling pathways.
Collapse
Affiliation(s)
- Jie Liu
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China
| | - Xin Sun
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Hang Jin
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Xiu-Li Yan
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
| | - Shuo Huang
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China
| | - Zhen-Ni Guo
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China.
| | - Yi Yang
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, the First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China; China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China.
| |
Collapse
|
35
|
Huang L, Wan Y, Dang Z, Yang P, Yang Q, Wu S. Hypoxic preconditioning ameliorated neuronal injury after middle cerebral artery occlusion by promoting neurogenesis. Brain Behav 2020; 10:e01804. [PMID: 32841552 PMCID: PMC7559635 DOI: 10.1002/brb3.1804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/27/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Sequelae of stroke were mainly caused by neuronal injury. Oxygen is a key factor affecting the microenvironment of neural stem cells (NSCs), and oxygen levels are used to promote NSC neurogenesis. In this study, effects of intermittent hypoxic preconditioning (HPC) on neurogenesis were investigated in a rat model of middle cerebral artery occlusion (MCAO). METHODS SD rats were used to establish the MCAO model. Nissl staining and Golgi staining were used to confirm the neuronal injury status in the MCAO model. Immunofluorescence, transmission electron microscopy, Western blot, and qPCR were used to observe the effects of HPC on neurogenesis. At the same time, the hypothesis that HPC could affect proliferation, apoptosis, differentiation, and migration of NSC was verified in vitro. RESULTS Hypoxic preconditioning significantly ameliorated the neuronal injury induced by MCAO. Compared with MCAO group, the dendrites, Edu+ /SOX2+ , Edu+ /DCX+ , Edu+ /NeuN+ , Edu+ /GFAP+ , and Edu+ /Tubulin+ positive cells in the HPC + MCAO group exhibited significantly difference. Similarly, axonal and other neuronal injuries in the HPC + MCAO group were also ameliorated. In the in vitro experiments, mild HPC significantly enhanced the viability of NSCs, promoted the migration of differentiated cells, and reduced apoptosis. CONCLUSIONS Our results showed that HPC significantly promotes neurogenesis after MCAO and ameliorates neuronal injury.
Collapse
Affiliation(s)
- Lu Huang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Xining, China.,Qinghai Provincial People's Hospital, Xining, China
| | - Yaqi Wan
- Qinghai Provincial People's Hospital, Xining, China
| | - Zhancui Dang
- Qinghai University Medical College, Xining, China
| | - Peng Yang
- Qinghai Provincial People's Hospital, Xining, China
| | - Quanyu Yang
- Qinghai University Medical College, Xining, China
| | - Shizheng Wu
- Qinghai Provincial People's Hospital, Xining, China
| |
Collapse
|
36
|
Hao Y, Xin M, Feng L, Wang X, Wang X, Ma D, Feng J. Review Cerebral Ischemic Tolerance and Preconditioning: Methods, Mechanisms, Clinical Applications, and Challenges. Front Neurol 2020; 11:812. [PMID: 33071923 PMCID: PMC7530891 DOI: 10.3389/fneur.2020.00812] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide, and it is increasing in prevalence. The limited therapeutic window and potential severe side effects prevent the widespread clinical application of the venous injection of thrombolytic tissue plasminogen activator and thrombectomy, which are regarded as the only approved treatments for acute ischemic stroke. Triggered by various types of mild stressors or stimuli, ischemic preconditioning (IPreC) induces adaptive endogenous tolerance to ischemia/reperfusion (I/R) injury by activating a multitude cascade of biomolecules, for example, proteins, enzymes, receptors, transcription factors, and others, which eventually lead to transcriptional regulation and epigenetic and genomic reprogramming. During the past 30 years, IPreC has been widely studied to confirm its neuroprotection against subsequent I/R injury, mainly including local ischemic preconditioning (LIPreC), remote ischemic preconditioning (RIPreC), and cross preconditioning. Although LIPreC has a strong neuroprotective effect, the clinical application of IPreC for subsequent cerebral ischemia is difficult. There are two main reasons for the above result: Cerebral ischemia is unpredictable, and LIPreC is also capable of inducing unexpected injury with only minor differences to durations or intensity. RIPreC and pharmacological preconditioning, an easy-to-use and non-invasive therapy, can be performed in a variety of clinical settings and appear to be more suitable for the clinical management of ischemic stroke. Hoping to advance our understanding of IPreC, this review mainly focuses on recent advances in IPreC in stroke management, its challenges, and the potential study directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Wei L, Liang H, Mo M, Liu Z, Ye R, Ye H, Ouyang W, Yu W, Zhao W, Zhang X. The effect of remote ischemic postconditioning on autonomic function in patients with acute ischemic stroke: A Randomized Controlled Trail. Complement Ther Med 2020; 54:102541. [PMID: 33183660 DOI: 10.1016/j.ctim.2020.102541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/22/2020] [Accepted: 08/13/2020] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE The evidence for the effect of remote ischemic postconditioning(RIpostC) on autonomic function in patients with acute ischemic stroke(AIS) is lacking and the neural mechanism underlying the protection of RIpostC remains speculative. This trial was aimed to evaluated the efficiency of RIpostC on autonomic function in AIS patients. DESIGN One hundred and six AIS patients were included in this prospective, randomized, placebo-controlled trial. Patients in intervention group (n = 57) received 4 cycles of alternating inflation (cuff inflation to 200 mmHg) and deflation for 5 min on healthy upper arm once a day for 30 days. The control group underwent a sham inflation and deflation cycles. Autonomic function was evaluated by heart rate variability (HRV). RESULTS All HRV parameters except for the ratio of low frequency to high frequency (P = 0.101) increased significantly with time (P < 0.001) in the two groups. The value of standard deviation of all normal R-R intervals(SDNN) and high frequency at day7 and day30 and the value of the percent of difference between adjacent normal R-R intervals (pNN50) at day 30 in RIpostC group was significantly higher than that of the sham-RIpostC group(P < 0.05). A significant time-by-group interaction was observed in SDNN、pNN50、and high frequency over time between two groups (P < 0.05). CONCLUSIONS 30-day RIpostC could improve autonomic function in AIS patients through the enhancement of the total autonomic nerve activity and vagus nerve activity. The mechanism of RIpostC mediating autonomic function needs to be further investigated.
Collapse
Affiliation(s)
- Lin Wei
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Hao Liang
- School of Nursing, Guangzhou University of Chinese Medicine, Airport Road 12, Baiyun District, Guangzhou 510405, Guangdong, China
| | - Miaomiao Mo
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Zhuyun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Richun Ye
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Huanwen Ye
- Department of Cardiac Function, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Wenwei Ouyang
- Key Unit of Methodology in Clinical Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Wenqi Yu
- Geriatrics dept(neurology), The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Inner Ring West Road 55, Panyu District, Guangzhou 510006, Guangdong, China
| | - Wenbo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Tianhe Road 600, Tianhe District, Guangzhou 510632, Guangdong, China.
| | - Xiaopei Zhang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
38
|
Sharma D, Maslov LN, Singh N, Jaggi AS. Remote ischemic preconditioning-induced neuroprotection in cerebral ischemia-reperfusion injury: Preclinical evidence and mechanisms. Eur J Pharmacol 2020; 883:173380. [PMID: 32693098 DOI: 10.1016/j.ejphar.2020.173380] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 12/29/2022]
Abstract
Remote ischemic preconditioning (RIPC) is an intrinsic protective phenomenon in which 3 to 4 interspersed cycles of non-fatal regional ischemia followed by reperfusion to the remote tissues protect the vital organs including brain, heart and kidney against sustained ischemia-reperfusion-induced injury. There is growing preclinical evidence supporting the usefulness of RIPC in eliciting neuroprotection against focal and global cerebral ischemia-reperfusion injury. Scientists have explored the involvement of HIF-1α, oxidative stress, apoptotic pathway, Lcn-2, platelets-derived microparticles, splenic response, adenosine A1 receptors, adenosine monophosphate activated protein kinase and neurogenic pathway in mediating RIPC-induced neuroprotection. The present review discusses the early and late phases of neuroprotection induced by RIPC against cerebral ischemic injury in animals along with the various possible mechanisms.
Collapse
Affiliation(s)
- Diwakar Sharma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India
| | - Leonid N Maslov
- Labortary of Experimental Cardiology, Institute of Cardiology, Kyevskaya 111, 634012 Tomsk, Russia
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India.
| |
Collapse
|
39
|
Cui J, Liu N, Chang Z, Gao Y, Bao M, Xie Y, Xu W, Liu X, Jiang S, Liu Y, Shi R, Xie W, Jia X, Shi J, Ren C, Gong K, Zhang C, Bade R, Shao G, Ji X. Exosomal MicroRNA-126 from RIPC Serum Is Involved in Hypoxia Tolerance in SH-SY5Y Cells by Downregulating DNMT3B. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:649-660. [PMID: 32380415 PMCID: PMC7210387 DOI: 10.1016/j.omtn.2020.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/18/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Abstract
Ischemic tolerance in the brain can be induced by transient limb ischemia, and this phenomenon is termed remote ischemic preconditioning (RIPC). It still remains elusive how this transfer of tolerance occurs. Exosomes can cross the blood-brain barrier, and some molecules may transfer neuroprotective signals from the periphery to the brain. Serum miRNA-126 is associated with ischemic stroke, and exosomal miRNA-126 has shown protective effects against acute myocardial infarction. Therefore, this study aims to explore whether exosomal miRNA-126 from RIPC serum can play a similar neuroprotective role. Exosomes were isolated from the venous serum of four healthy young male subjects, both before and after RIPC. Exosomal miRNA-126 was measured by real-time PCR. The miRNA-126 target sequence was predicted by bioinformatics software. SH-SY5Y neuronal cells were incubated with exosomes, and the cell cycle was analyzed by flow cytometry. The expression and activity of DNA methyltransferase (DNMT) 3B, a potential target gene of miRNA-126, were examined in SH-SY5Y cells. The cell viability of SH-SY5Y cells exposed to oxygen-glucose deprivation (OGD) was also investigated. To confirm the association between miRNA-126 and DNMT3B, we overexpressed miRNA-126 in SH-SY5Y cells using lentiviral transfection. miRNA-126 expression was upregulated in RIPC exosomes, and bioinformatics prediction showed that miRNA-126 could bind with DNMT3B. DNMT levels and DNMT3B activity were downregulated in SH-SY5Y cells incubated with RIPC exosomes. After overexpression of miRNA-126 in SH-SY5Y cells, global methylation levels and DNMT3B gene expression were downregulated in these cells, consistent with the bioinformatics predictions. RIPC exosomes can affect the cell cycle and increase OGD tolerance in SH-SY5Y cells. RIPC seems to have neuroprotective effects by downregulating the expression of DNMTs in neural cells through the upregulation of serum exosomal miRNA-126.
Collapse
Affiliation(s)
- Junhe Cui
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Na Liu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Zhehan Chang
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Yongsheng Gao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Mulan Bao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Yabin Xie
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Wenqiang Xu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Xiaolei Liu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Shuyuan Jiang
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - You Liu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Rui Shi
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Wei Xie
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Xiaoe Jia
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Jinghua Shi
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China
| | - Rengui Bade
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC.
| | - Guo Shao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, PRC; Biomedicine Research Center, Basic Medical College and Baotou Medical College of the Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC; Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China.
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC.
| |
Collapse
|
40
|
RIPC provides neuroprotection against ischemic stroke by suppressing apoptosis via the mitochondrial pathway. Sci Rep 2020; 10:5361. [PMID: 32210331 PMCID: PMC7093414 DOI: 10.1038/s41598-020-62336-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Ischemic stroke is a common disease with high morbidity and mortality. Remote ischemic preconditioning (RIPC) can stimulate endogenous protection mechanisms by inducing ischemic tolerance to reduce subsequent damage caused by severe or fatal ischemia to non-ischemic organs. This study was designed to assess the therapeutic properties of RIPC in ischemic stroke and to elucidate their underlying mechanisms. Neurobehavioral function was evaluated with the modified neurological severity score (mNSS) test and gait analysis. PET/CT was used to detect the ischemic volume and level of glucose metabolism. The protein levels of cytochrome c oxidase-IV (COX-IV) and heat shock protein 60 (HSP60) were tested by Western blotting. TUNEL and immunofluorescence staining were used to analyze apoptosis and to observe the nuclear translocation and colocalization of apoptosis-inducing factor (AIF) and endonuclease G (EndoG) in apoptotic cells. Transmission electron microscopy (TEM) was used to detect mitochondrial-derived vesicle (MDV) production and to assess mitochondrial ultrastructure. The experimental results showed that RIPC exerted significant neuroprotective effects, as indicated by improvements in neurological dysfunction, reductions in ischemic volume, increases in glucose metabolism, inhibition of apoptosis, decreased nuclear translocation of AIF and EndoG from mitochondria and improved MDV formation. In conclusion, RIPC alleviates ischemia/reperfusion injury after ischemic stroke by inhibiting apoptosis via the endogenous mitochondrial pathway.
Collapse
|
41
|
Du X, Yang J, Liu C, Wang S, Zhang C, Zhao H, Du H, Geng X. Hypoxia-Inducible Factor 1α and 2α Have Beneficial Effects in Remote Ischemic Preconditioning Against Stroke by Modulating Inflammatory Responses in Aged Rats. Front Aging Neurosci 2020; 12:54. [PMID: 32210788 PMCID: PMC7076079 DOI: 10.3389/fnagi.2020.00054] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/18/2020] [Indexed: 11/29/2022] Open
Abstract
Limb remote ischemic preconditioning (RIPC) has been proven to alleviate stroke injury in young rats, but its protective effect and its mechanism in aged rats are still unclear. Hypoxia-inducible factor (HIF) is one of the important markers of stroke, and its high expression plays an important role in the pathogenesis of stroke. In this study, we tested the hypothesis that RIPC could regulate the expression of HIF, leading to reduced inflammatory responses in aged rats. Stroke was induced by transient middle cerebral artery occlusion (MCAo) in aged rats, and RIPC was conducted in both hind limbs. The HIF-1α and HIF-2α mRNA and protein were examined by real-time RT-PCR and western blotting (WB). Inflammatory cytokines in the peripheral blood and brain were measured using AimPlex multiplex immunoassays. The protein levels of p-Akt, Akt, p-ERK, and ERK were examined by WB. We investigated that RIPC reduced the infarct size, improved neurological functions, and decreased the expression of HIF-1α and HIF-2α in the ischemic brain. RIPC reduced the levels of IL-1β, IL-6 and IFN-γ in the peripheral blood and the levels of IL-1β and IFN-γ in the ischemic brain 48 h post-stroke. Moreover, intraperitoneal injection of the HIF inhibitor, acriflavine hydrochloride (ACF), abolished the protection of RIPC with respect to infarct size and neurological functions and neutralized the downregulation of pro-inflammatory IL-1β, IL-6 and IFN-γ. ACF also reversed the activation of the Akt signaling pathway induced by RIPC following stroke. HIF may play a key role in RIPC, which was likely mediated by the Akt signaling pathway and systemic modulation of the inflammatory response in aged rats.
Collapse
Affiliation(s)
- Xiangnan Du
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jian Yang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Cuiying Liu
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Sainan Wang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chencheng Zhang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Heng Zhao
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Huishan Du
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
42
|
Qin C, Yan X, Jin H, Zhang R, He Y, Sun X, Zhang Y, Guo ZN, Yang Y. Effects of Remote Ischemic Conditioning on Cerebral Hemodynamics in Ischemic Stroke. Neuropsychiatr Dis Treat 2020; 16:283-299. [PMID: 32021218 PMCID: PMC6988382 DOI: 10.2147/ndt.s231944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is one of the most common cerebrovascular diseases and is the leading cause of disability all over the world. It is well known that cerebral blood flow (CBF) is disturbed or even disrupted when ischemic stroke happens. The imbalance between demand and shortage of blood supply makes ischemic stroke take place or worsen. The search for treatments that can preserve CBF, especially during the acute phase of ischemic stroke, has become a research hotspot. Animal and clinical experiments have proven that remote ischemic conditioning (RIC) is a beneficial therapeutic strategy for the treatment of ischemic stroke. However, the mechanism by which RIC affects CBF has not been fully understood. This review aims to discuss several possible mechanisms of RIC on the cerebral hemodynamics in ischemic stroke, such as the improvement of cardiac function and collateral circulation of cerebral vessels, the protection of neurovascular units, the formation of gas molecules, the effect on the function of vascular endothelial cells and the nervous system. RIC has the potential to become a therapeutic treatment to improve CBF in ischemic stroke. Future studies are needed to highlight our understanding of RIC as well as accelerate its clinical translation.
Collapse
Affiliation(s)
- Chen Qin
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Xiuli Yan
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Hang Jin
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Ruyi Zhang
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Yaode He
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Xin Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Yihe Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Zhen-Ni Guo
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
43
|
Qin Y, Zhang Q, Liu Y. Analysis of knowledge bases and research focuses of cerebral ischemia-reperfusion from the perspective of mapping knowledge domain. Brain Res Bull 2019; 156:15-24. [PMID: 31843561 DOI: 10.1016/j.brainresbull.2019.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia-reperfusion (IR) has attracted wide attention as a serious clinical problem. So far, the field has accumulated a large amount of scientific research literature. To clarify the temporal and spatial distribution characteristics of research resources, knowledge bases and research focuses, a visual analysis was performed on 5814 articles cited in the WoS databases from 2004 to 2019. This analysis was based on bibliometrics and mapping knowledge domain (MKD) analysis with VOSviewer, and CiteSpace 5.4.R4. The results can be elaborated from four aspects. First, the volume of publications in this area is on the rise. Second, the United States and China are the active regions. The USA is the central region of cerebral ischemia-reperfusion research. Third, the knowledge bases of IR have focused on five major areas of "Suitable small-animal models", "A framework with further study", "Molecular signaling targets by oxidative stress", "Finding new potential targets for therapy" and "Protective effect of multiple transient ischemia". Fourth, the research focuses consist of three representative areas: "Oxidative stress closelyd with cerebral ischemia-reperfusion", "Neuronal apoptosis and neuronal protection", and "Neuroprotective effect of the blood-brain barrier".
Collapse
Affiliation(s)
- Yi Qin
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qing Zhang
- No.4 Hospital Beijing University of Chinese Medicine, Zaozhuang, Shandong 277000
| | - Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
44
|
Zhou D, Ding J, Ya J, Pan L, Wang Y, Ji X, Meng R. Remote ischemic conditioning: a promising therapeutic intervention for multi-organ protection. Aging (Albany NY) 2019; 10:1825-1855. [PMID: 30115811 PMCID: PMC6128414 DOI: 10.18632/aging.101527] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022]
Abstract
Despite decades of formidable exploration, multi-organ ischemia-reperfusion injury (IRI) encountered, particularly amongst elderly patients with clinical scenarios, such as age-related arteriosclerotic vascular disease, heart surgery and organ transplantation, is still an unsettled conundrum that besets clinicians. Remote ischemic conditioning (RIC), delivered via transient, repetitive noninvasive IR interventions to distant organs or tissues, is regarded as an innovative approach against IRI. Based on the available evidence, RIC holds the potential of affording protection to multiple organs or tissues, which include not only the heart and brain, but also others that are likely susceptible to IRI, such as the kidney, lung, liver and skin. Neuronal and humoral signaling pathways appear to play requisite roles in the mechanisms of RIC-related beneficial effects, and these pathways also display inseparable interactions with each other. So far, several hurdles lying ahead of clinical translation that remain to be settled, such as establishment of biomarkers, modification of RIC regimen, and deep understanding of underlying minutiae through which RIC exerts its powerful function. As this approach has garnered an increasing interest, herein, we aim to encapsulate an overview of the basic concept and postulated protective mechanisms of RIC, highlight the main findings from proof-of-concept clinical studies in various clinical scenarios, and also to discuss potential obstacles that remain to be conquered. More well designed and comprehensive experimental work or clinical trials are warranted in future research to confirm whether RIC could be utilized as a non-invasive, inexpensive and efficient adjunct therapeutic intervention method for multi-organ protection.
Collapse
Affiliation(s)
- Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Jiayue Ding
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Jingyuan Ya
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
45
|
Liu C, Zhang C, Du H, Geng X, Zhao H. Remote ischemic preconditioning protects against ischemic stroke in streptozotocin-induced diabetic mice via anti-inflammatory response and anti-apoptosis. Brain Res 2019; 1724:146429. [PMID: 31476295 DOI: 10.1016/j.brainres.2019.146429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/15/2019] [Accepted: 08/29/2019] [Indexed: 01/03/2023]
Abstract
OBJECTIVE It has been shown that remote ischemic preconditioning (RIPreC) attenuates ischemic injury after stroke in healthy rats or mice. The present study aims to examine whether RIPreC offers neuroprotection against ischemic stroke in streptozotocin-induced diabetic mice. METHODS Streptozotocin (STZ, 120 mg/kg) was intraperitoneally injected into the mice to induce type 1 diabetic model. The immune and inflammatory changes were analyzed 2 days after reperfusion by flow cytometry and multiplex cytokine assay analysis, respectively. RESULTS We found that RIPreC reduced infarct sizes and alleviated neurological impairment in diabetic mice. RIPreC decreased CD8 T cells infiltrated into the brain, and attenuated the decreases of CD8 T cells in the blood, CD4 T cells and CD8 T cells in the spleen. Results from multiplex cytokine assay showed that RIPreC treatment decreased IL-6, IL-1 beta and TNF alpha levels in the cortex, while it inhibited IL-6 level in the hippocampus and striatum, and TNF alpha level in the hippocampus. RIPreC treatment also downregulated IL-6 and IFN gamma level in the blood, which increased after cerebral ischemic injury. In addition, RIPreC reduced pro-apoptotic protein BAX expression in the ischemic brain. CONCLUSIONS Our results indicate that RIPreC attenuates cerebral injuries in streptozotocin-induced diabetic mice via anti-inflammatory response and anti-apoptosis in the ischemic brain.
Collapse
Affiliation(s)
- Cuiying Liu
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Chencheng Zhang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Huishan Du
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
46
|
Remote Postischemic Conditioning Promotes Stroke Recovery by Shifting Circulating Monocytes to CCR2 + Proinflammatory Subset. J Neurosci 2019; 39:7778-7789. [PMID: 31427395 DOI: 10.1523/jneurosci.2699-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/17/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
Brain injury from stroke is typically considered an event exclusive to the CNS, but injury progression and repair processes are profoundly influenced by peripheral immunity. Stroke stimulates an acute inflammatory response that results in a massive infiltration of peripheral immune cells into the ischemic area. While these cells contribute to the development of brain injury, their recruitment has been considered as a key step for tissue repair. The paradoxical role of inflammatory monocytes in stroke raises the possibility that the manipulation of peripheral immune cells before infiltration into the brain could influence stroke outcome. One such manipulation is remote ischemic limb conditioning (RLC), which triggers an endogenous tolerance mechanism. We observed that mice subjected to poststroke RLC shifted circulating monocytes to a CCR2+ proinflammatory monocyte subset and had reduced acute brain injury, swelling, and improved motor/gait function in chronic stroke. The RLC benefits were observed regardless of injury severity, with a greater shift to a CCR2+ subset in severe stroke. Adoptive transfer of CCR2-deficient monocytes abolished RLC-mediated protection. The study demonstrates the importance of RLC-induced shift of monocytes to a CCR2+ proinflammatory subset in attenuating acute injury and promoting functional recovery in chronic stroke. The defined immune-mediated mechanism underlying RLC benefits allows for an evidence-based framework for the development of immune-based therapeutic strategies for stroke patients.SIGNIFICANCE STATEMENT Stroke is the leading cause of physical disability worldwide but has few treatment options for patients. Because remote ischemic limb conditioning (RLC) elicits endogenous tolerance in neither an organ- nor a tissue-specific manner, the immune system has been considered a mediator for an RLC-related benefit. Application of RLC after stroke increased a proinflammatory CCR2+ monocyte subset in the blood and the brain. RLC reduced acute stroke injury and promoted motor/gait function during the recovery phase. The RLC benefits were absent in mice that received CCR2-deficient monocytes. This preclinical study shows the importance of CCR2+ proinflammatory monocytes in RLC benefits in stroke and provides a therapeutic RLC platform as a novel immune strategy to improve outcomes in stroke patients.
Collapse
|
47
|
Malone K, Amu S, Moore AC, Waeber C. Immunomodulatory Therapeutic Strategies in Stroke. Front Pharmacol 2019; 10:630. [PMID: 31281252 PMCID: PMC6595144 DOI: 10.3389/fphar.2019.00630] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
The role of immunity in all stages of stroke is increasingly being recognized, from the pathogenesis of risk factors to tissue repair, leading to the investigation of a range of immunomodulatory therapies. In the acute phase of stroke, proposed therapies include drugs targeting pro-inflammatory cytokines, matrix metalloproteinases, and leukocyte infiltration, with a key objective to reduce initial brain cell toxicity. Systemically, the early stages of stroke are also characterized by stroke-induced immunosuppression, where downregulation of host defences predisposes patients to infection. Therefore, strategies to modulate innate immunity post-stroke have garnered greater attention. A complementary objective is to reduce longer-term sequelae by focusing on adaptive immunity. Following stroke onset, the integrity of the blood–brain barrier is compromised, exposing central nervous system (CNS) antigens to systemic adaptive immune recognition, potentially inducing autoimmunity. Some pre-clinical efforts have been made to tolerize the immune system to CNS antigens pre-stroke. Separately, immune cell populations that exhibit a regulatory phenotype (T- and B- regulatory cells) have been shown to ameliorate post-stroke inflammation and contribute to tissue repair. Cell-based therapies, established in oncology and transplantation, could become a strategy to treat the acute and chronic stages of stroke. Furthermore, a role for the gut microbiota in ischaemic injury has received attention. Finally, the immune system may play a role in remote ischaemic preconditioning-mediated neuroprotection against stroke. The development of stroke therapies involving organs distant to the infarct site, therefore, should not be overlooked. This review will discuss the immune mechanisms of various therapeutic strategies, surveying published data and discussing more theoretical mechanisms of action that have yet to be exploited.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| | - Sylvie Amu
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
48
|
Early Immunological Effects of Ischemia-Reperfusion Injury: No Modulation by Ischemic Preconditioning in a Randomised Crossover Trial in Healthy Humans. Int J Mol Sci 2019; 20:ijms20122877. [PMID: 31200465 PMCID: PMC6628232 DOI: 10.3390/ijms20122877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/06/2019] [Accepted: 06/08/2019] [Indexed: 12/14/2022] Open
Abstract
Ischemic preconditioning (IPC) has been protective against ischemia-reperfusion injury (IRI), but the underlying mechanism is poorly understood. We examined whether IPC modulates the early inflammatory response after IRI. Nineteen healthy males participated in a randomised crossover trial with and without IPC before IRI. IPC and IRI were performed by cuff inflation on the forearm. IPC consisted of four cycles of five minutes followed by five minutes of reperfusion. IRI consisted of twenty minutes followed by 15 min of reperfusion. Blood was collected at baseline, 0 min, 85 min and 24 h after IRI. Circulating monocytes, T-cells subsets and dendritic cells together with intracellular activation markers were quantified by flow cytometry. Luminex measured a panel of inflammation-related cytokines in plasma. IRI resulted in dynamic regulations of the measured immune cells and their intracellular activation markers, however IPC did not significantly alter these patterns. Neither IRI nor the IPC protocol significantly affected the levels of inflammatory-related cytokines. In healthy volunteers, it was not possible to detect an effect of the investigated IPC-protocol on early IRI-induced inflammatory responses. This study indicates that protective effects of IPC on IRI is not explained by direct modulation of early inflammatory events.
Collapse
|
49
|
Effects of Combined Remote Ischemic Pre-and Post-Conditioning on Neurologic Complications in Moyamoya Disease Patients Undergoing Superficial Temporal Artery-Middle Cerebral Artery Anastomosis. J Clin Med 2019; 8:jcm8050638. [PMID: 31075871 PMCID: PMC6572043 DOI: 10.3390/jcm8050638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/07/2019] [Accepted: 05/07/2019] [Indexed: 01/01/2023] Open
Abstract
Superficial temporal artery-middle cerebral artery (STA-MCA) anastomosis is the most commonly used treatment for Moyamoya disease. During the perioperative period, however, these patients are vulnerable to ischemic injury or hyperperfusion syndrome. This study investigated the ability of combined remote ischemic pre-conditioning (RIPC) and remote ischemic post-conditioning (RIPostC) to reduce the occurrence of major neurologic complications in Moyamoya patients undergoing STA-MCA anastomosis. The 108 patients were randomly assigned to a RIPC with RIPostC group (n = 54) or a control group (n = 54). Patients in the RIPC with RIPostC group were treated with four cycles of 5-min ischemia and 5-min reperfusion before craniotomy and after STA-MCA anastomosis (RIPostC). The incidence of postoperative neurologic complications and the duration of hospital stay were determined. The overall incidence of neurologic complication was significantly higher in the control group than in the RIPC with RIPostC group (13 vs. 3, p = 0.013). The duration of hospital stay was significantly longer in the control group than in the RIPC with RIPostC group (17.8 (11.3) vs. 13.8 (5.9) days, p = 0.023). Combined remote ischemic pre- and post-conditioning can be effective in reducing neurologic complications and the duration of hospitalization in Moyamoya patients undergoing STA-MCA anastomosis.
Collapse
|
50
|
Limb Remote Ischemic Preconditioning Reduces Repeated Ketamine Exposure-Induced Adverse Effects in the Developing Brain of Rats. J Mol Neurosci 2019; 68:58-65. [DOI: 10.1007/s12031-019-01282-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 02/18/2019] [Indexed: 11/26/2022]
|