1
|
Jiao C, Liu C, Yang Z, Jin C, Chen X, Xue J, Zhang G, Pan C, Jia J, Hou X. Brain Protection Effects of Mild Hypothermia Combined with Distant Ischemic Postconditioning and Thrombolysis in Patients with Acute Ischemic Stroke. Ther Hypothermia Temp Manag 2024; 14:172-178. [PMID: 37668993 DOI: 10.1089/ther.2023.0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
To assess the effectiveness and molecular mechanisms of mild hypothermia and remote ischemic postconditioning (RIPC) in patients with acute ischemic stroke (AIS) who have undergone thrombolysis therapy. A total of 58 AIS patients who received recombinant tissue plasmin activator (rt-PA) intravenous thrombolysis were included in this prospective study. Participants were randomly allocated to the experimental group (rt-PA intravenous thrombolysis plus mild hypothermic ice cap plus remote ischemic brain protection, n = 30) and the control group (rt-PA intravenous thrombolysis plus 0.9% saline, n = 28). The RIPC was performed for 14 consecutive days on both upper limb arteries spaced 2 minutes apart. Five cycles of ischemia-reperfusion were performed sequentially (2-2, 3-3, 4-4, 5-5, 5-0 minutes, respectively). The outcome measures of the National Institute of Health stroke scale (NIHSS) score, volume of cerebral infarction, serum levels of superoxide dismutase (SOD), malondialdehyde (MDA), interleukin-1β, tumor necrosis factor α, nuclear factors kappa B (NF-κB), and NOD-1ike receptor pyrin 3 (NLRP3) were evaluated at different time points after treatment. Similarly, the 90-day modified Rankin Scale (mRS) scores were compared between the two groups. After treatment, the NIHSS score, MDA, NF-κB, and NLRP3 levels in the experimental group were significantly lower than those in the control group (p < 0.05). While the SOD in the experimental group was significantly higher than in the control group (p < 0.05), the NIHSS scores decreased within groups (all p < 0.05) in both experimental and control groups. The 90-day mRS score (0-2 points) in the experimental group was significantly higher than that in the control group (73.33% vs. 53.57%, p < 0.05) and no significant differences were observed in the safety indices between the two groups (all p > 0.05). Our study shows that combining mild hypothermia and RIPC has a positive effect on brain protection and can significantly reduce the oxidative stress and associated outburst of inflammatory response. The Clinical Trial Registration number is ChiCTR2300073136.
Collapse
Affiliation(s)
- Changping Jiao
- Department of Neurosurgery, Heilongjiang Provincial Hospital, Harbin, China
| | - Cui Liu
- Department of Pediatrics, Qingdao Huangdao District Central Hospital, Qingdao, China
| | - Zhenhua Yang
- Department of Infectious Disease Prevention and Control, Qingdao West Coast New Area Disease Prevention and Control Center, Qingdao, China
| | - Chunfeng Jin
- Department of Neurology, The Second Hospital of Harbin, Harbin, China
| | - Xi Chen
- Clinical Laboratory, Heilongjiang Provincial Hospital, Harbin, China
| | - Jujun Xue
- Department of Geriatric Neurology, Heilongjiang Provincial Hospital, Harbin, China
| | - Ge Zhang
- Department of Geriatric Neurology, Heilongjiang Provincial Hospital, Harbin, China
| | - Chengli Pan
- Department of Geriatric Neurology, Heilongjiang Provincial Hospital, Harbin, China
| | - Jianrong Jia
- Department of Neurosurgery, Heilongjiang Provincial Hospital, Harbin, China
| | - Xiaojun Hou
- Department of Geriatric Neurology, Heilongjiang Provincial Hospital, Harbin, China
| |
Collapse
|
2
|
Li X, Deng J, Long Y, Ma Y, Wu Y, Hu Y, He X, Yu S, Li D, Li N, He F. Focus on brain-lung crosstalk: Preventing or treating the pathological vicious circle between the brain and the lung. Neurochem Int 2024; 178:105768. [PMID: 38768685 DOI: 10.1016/j.neuint.2024.105768] [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: 01/31/2024] [Revised: 05/05/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Recently, there has been increasing attention to bidirectional information exchange between the brain and lungs. Typical physiological data is communicated by channels like the circulation and sympathetic nervous system. However, communication between the brain and lungs can also occur in pathological conditions. Studies have shown that severe traumatic brain injury (TBI), cerebral hemorrhage, subarachnoid hemorrhage (SAH), and other brain diseases can lead to lung damage. Conversely, severe lung diseases such as acute respiratory distress syndrome (ARDS), pneumonia, and respiratory failure can exacerbate neuroinflammatory responses, aggravate brain damage, deteriorate neurological function, and result in poor prognosis. A brain or lung injury can have adverse effects on another organ through various pathways, including inflammation, immunity, oxidative stress, neurosecretory factors, microbiome and oxygen. Researchers have increasingly concentrated on possible links between the brain and lungs. However, there has been little attention given to how the interaction between the brain and lungs affects the development of brain or lung disorders, which can lead to clinical states that are susceptible to alterations and can directly affect treatment results. This review described the relationships between the brain and lung in both physiological and pathological conditions, detailing the various pathways of communication such as neurological, inflammatory, immunological, endocrine, and microbiological pathways. Meanwhile, this review provides a comprehensive summary of both pharmacological and non-pharmacological interventions for diseases related to the brain and lungs. It aims to support clinical endeavors in preventing and treating such ailments and serve as a reference for the development of relevant medications.
Collapse
Affiliation(s)
- Xiaoqiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jie Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yu Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yin Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yuanyuan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yue Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiaofang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shuang Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Fei He
- Department of Geratology, Yongchuan Hospital of Chongqing Medical University(the Fifth Clinical College of Chongqing Medical University), Chongqing, 402160, China.
| |
Collapse
|
3
|
Ionescu L, Morariu PC, Dascălu CG, Iov DE, Oancea AF, Chiriac CP, Sîrbu O, Timofte DV, Rezuş C, Șorodoc L, Şorodoc V, Baroi GL, Tanase DM, Floria M. Accidental hypothermia in the largest emergency hospital in North-Eastern Romania. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:295-306. [PMID: 38470364 DOI: 10.2478/rjim-2024-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Accidental hypothermia (AH) presents a significant mortality risk, even in individuals with good health. Early recognition of the parameters associated with negative prognosis could save more lives. METHODS This was a pilot, retrospective observational study, conducted in the largest Emergency Hospital in North Eastern Romania, which included all patients with AH (defined as body temperature below 35°C), hospitalized and treated in our hospital between 2019 and 2022. RESULTS A total of 104 patients with AH were included in our study, 90 of whom had data collected and statistically analyzed. The clinical, biological, and therapeutic parameters associated with negative outcomes were represented by a reduced GCS score (p=0.024), diminished systolic and diastolic blood pressure (p=0.007 respectively, 0.013), decreased bicarbonate (p=0.043) and hemoglobin levels (p=0.002), the presence of coagulation disorders (p=0.007), as well as the need for administration of inotropic or vasopressor medications (p=0.04). CONCLUSION In this pilot, retrospective, observational study, the negative outcomes observed in patients with AH hospitalized in the largest Emergency Hospital in North-Eastern Romania were associated with several clinical, biochemical, and therapeutic factors, which are easy to identify in clinical practice. Recognizing the significance of these associated factors empowers healthcare practitioners to intervene at an early stage to save more lives.
Collapse
Affiliation(s)
- Lidia Ionescu
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 2Surgery Clinic, "Sfântul Spiridon" Emergency Hospital, 700111 Iasi, Romania
| | - Paula Cristina Morariu
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Cristina Gena Dascălu
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 4Department of Medical Informatics and Biostatistics; University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
| | - Diana Elena Iov
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Alexandru Florinel Oancea
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 5Cardiology Clinic, "Sfântul Spiridon" Emergency Hospital, 700111 Iasi, Romania
| | - Cristina Petronela Chiriac
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Oana Sîrbu
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Daniel Vasile Timofte
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 2Surgery Clinic, "Sfântul Spiridon" Emergency Hospital, 700111 Iasi, Romania
| | - Ciprian Rezuş
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Laurenţiu Șorodoc
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Victoriţa Şorodoc
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Genoveva Livia Baroi
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 2Surgery Clinic, "Sfântul Spiridon" Emergency Hospital, 700111 Iasi, Romania
| | - Daniela Maria Tanase
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| | - Mariana Floria
- 1University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
- 3Medical Clinic, "Sfântul Spiridon" Emergency Hospital 700111 Iasi, Romania
| |
Collapse
|
4
|
Burrows DJ, McGown A, Abduljabbar O, Castelli LM, Shaw PJ, Hautbergue GM, Ramesh TM. RAN Translation of C9orf72-Related Dipeptide Repeat Proteins in Zebrafish Recapitulates Hallmarks of Amyotrophic Lateral Sclerosis and Identifies Hypothermia as a Therapeutic Strategy. Ann Neurol 2024. [PMID: 39215697 DOI: 10.1002/ana.27068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Hexanucleotide repeat expansions in the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). A large body of evidence implicates dipeptide repeats (DPRs) proteins as one of the main drivers of neuronal injury in cell and animal models. METHODS A pure repeat-associated non-AUG (RAN) translation zebrafish model of C9orf72-ALS/FTD was generated. Embryonic and adult transgenic zebrafish lysates were investigated for the presence of RAN-translated DPR species and adult-onset motor deficits. Using C9orf72 cell models as well as embryonic C9orf72-ALS/FTD zebrafish, hypothermic-therapeutic temperature management (TTM) was explored as a potential therapeutic option for C9orf72-ALS/FTD. RESULTS Here, we describe a pure RAN translation zebrafish model of C9orf72-ALS/FTD that exhibits significant RAN-translated DPR pathology and progressive motor decline. We further demonstrate that hypothermic-TTM results in a profound reduction in DPR species in C9orf72-ALS/FTD cell models as well as embryonic C9orf72-ALS/FTD zebrafish. INTERPRETATION The transgenic model detailed in this paper provides a medium throughput in vivo research tool to further investigate the role of RAN-translation in C9orf72-ALS/FTD and further understand the mechanisms that underpin neuroprotective strategies. Hypothermic-TTM presents a viable therapeutic avenue to explore in the context of C9orf72-ALS/FTD. ANN NEUROL 2024.
Collapse
Affiliation(s)
- David J Burrows
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexander McGown
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Olfat Abduljabbar
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, UK
| | - Tennore M Ramesh
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, UK
| |
Collapse
|
5
|
Xu Y, Duan Y, Xu S, He X, Guo J, Shi J, Zhang Y, Jia M, Li M, Wu C, Wu L, Jiang M, Chen X, Ji X, Wu D. Mild hypothermia therapy attenuates early BBB leakage in acute ischaemic stroke. J Cereb Blood Flow Metab 2024:271678X241275761. [PMID: 39157938 DOI: 10.1177/0271678x241275761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Reperfusion therapy inevitably leads to brain-blood barrier (BBB) disruption and promotes damage despite its benefits for acute ischaemic stroke (AIS). An effective brain cytoprotective treatment is still needed as an adjunct to reperfusion therapy. Here, we explore the potential benefits of therapeutic hypothermia (HT) in attenuating early BBB leakage and improving neurological outcomes. Mild HT was induced during the early and peri-recanalization stages in a mouse model of transient middle cerebral artery occlusion and reperfusion (tMCAO/R). The results showed that mild HT attenuated early BBB leakage in AIS, decreased the infarction volume, and improved functional outcomes. RNA sequencing data of the microvessels indicated that HT decreased the transcription of the actin polymerization-related pathway. We further discovered that HT attenuated the ROCK1/MLC pathway, leading to a decrease in the polymerization of G-actin to F-actin. Arachidonic acid (AA), a known structural ROCK agonist, partially counteracted the protective effects of HT in the tMCAO/R model. Our study highlights the importance of early vascular protection during reperfusion and provides a new strategy for attenuating early BBB leakage by HT treatment for ischaemic stroke.
Collapse
Affiliation(s)
- Yi Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Yunxia Duan
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaoduo He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Jingfei Shi
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Yang Zhang
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Milan Jia
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Ming Li
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Chuanjie Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Longfei Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Miaowen Jiang
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaonong Chen
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Munoz C, Acon-Chen C, Keith ZM, Shih TM. Hypothermia as potential therapeutic approach to attenuating soman-induced seizure, neuropathology, and mortality with an adenosine A 1 receptor agonist and body cooling. Neuropharmacology 2024; 253:109966. [PMID: 38677446 PMCID: PMC11197881 DOI: 10.1016/j.neuropharm.2024.109966] [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: 01/04/2024] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
Organophosphorus nerve agents, such as soman (GD), produce excitotoxic effects resulting in sustained status epilepticus (SSE) and brain damage. Previous work shows that neuronal inhibitory effects of A1 adenosine receptor (A1AR) agonists, such as N6- Bicyclo (2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (Cl-ENBA), suppresses GD-induced SSE and improves neuropathology. Some other physiologic effects of these agonists are hypothermia, hypotension, and sedation. Hypothermia may also shield the brain from injury by slowing down chemical insults, lessening inflammation, and contributing to improved neurological outcomes. Therefore, we attempted to isolate the hypothermic effect from ENBA by assessing the neuroprotective efficacy of direct surface body cooling in a rat GD-induced SSE model, and comparing the effects on seizure termination, neuropathology, and survival. Male rats implanted with a body temperature (Tb) transponder and electroencephalographic (EEG) electrodes were primed with asoxime (HI-6), exposed to GD 30 min later, and then treated with Cl-ENBA or had Tb lowered directly via body cooling at 30 min after the onset of seizure activity. Afterwards, they were either allowed to develop hypothermia as expected, or received thermal support to maintain normothermic Tb for a period of 6-h. Neuropathology was assessed at 24 h. Regardless of Cl-ENBA or surface cooling, all hypothermic GD-exposed groups had significantly improved 24-h survival compared to rats with normothermic Tb (81% vs. 39%, p < 0.001). Cl-ENBA offered neuroprotection independently of hypothermic Tb. While hypothermia enhanced the overall efficacy of Cl-ENBA by improving survival outcomes, body cooling didn't reduce seizure activity or neuropathology following GD-induced SSE.
Collapse
Affiliation(s)
- Crystal Munoz
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| | - Cindy Acon-Chen
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| | - Zora-Maya Keith
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| | - Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA.
| |
Collapse
|
7
|
King RM, Anagnostakou V, Shazeeb MS, Hornibrook S, Mitchell J, Epshtein M, Raskett C, Henninger N, Puri AS, Merrill TL, Gounis MJ. Selective brain cooling with a novel catheter reduces infarct growth after recanalization in a canine large vessel occlusion model. Interv Neuroradiol 2024:15910199241266010. [PMID: 39043215 DOI: 10.1177/15910199241266010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Therapeutic hypothermia has shown potential in cardiac intervention for years; however, its adoption into the neurovascular space has been limited. Studies have pointed to slow cooling and limited depth of hypothermia yielding negative outcomes. Here we present an insulated catheter that allows for consistent infusion of chilled saline directly to the brain. Direct delivery of cold saline allows a faster depth of hypothermia, which could have a benefit to the growth of ischemic lesions. METHODS Ten canines were randomized to either receive selective brain cooling or no additional therapy. Eight animals were successfully enrolled (n = 4 per group). Each animal underwent a temporary middle cerebral artery occlusion (MCAO) for a total of 45 min. Five minutes prior to flow restoration, chilled saline was injected through the ipsilateral internal carotid artery using an insulated catheter to ensure delivery temperature. The treatment continued for 20 min, after which the animal was transferred to an MRI scanner for imaging. RESULTS Of the 8 animals that were successfully enrolled in the study, 3 were able to survive to the 30-day endpoint with no differences between the cooled and control animals. There was no difference in the initial mean infarct size between the groups; however, animals that did not receive cooling had infarcts continuing to progress more rapidly after the MCAO was removed (13.8% vs 161.3%, p = 0.016, cooled vs control). CONCLUSIONS Selective hypothermia was able to reduce the post-MCAO infarct progression in a canine model of temporary MCAO.
Collapse
Affiliation(s)
- Robert M King
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vania Anagnostakou
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mohammed Salman Shazeeb
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Radiology, Image Processing and Analysis Core, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | - Mark Epshtein
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Christopher Raskett
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ajit S Puri
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Matthew J Gounis
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
8
|
Bardutzky J, Kollmar R, Al-Rawi F, Lambeck J, Fazel M, Taschner C, Niesen WD. COmbination of Targeted temperature management and Thrombectomy after acute Ischemic Stroke (COTTIS): a pilot study. Stroke Vasc Neurol 2024; 9:258-267. [PMID: 37612052 PMCID: PMC11221305 DOI: 10.1136/svn-2023-002420] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/05/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND To evaluate the feasibility and safety of a fast initiation of cooling to a target temperature of 35°C by means of transnasal cooling in patients with anterior circulation large vessel occlusion (LVO) undergoing endovascular thrombectomy (EVT). METHODS Patients with an LVO onset of <24 hour who had an indication for EVT were included in the study. Transnasal cooling (RhinoChill) was initiated immediately after the patient was intubated for EVT and continued until an oesophageal target temperature of 35°C was reached. Hypothermia was maintained with surface cooling for 6-hour postrecanalisation, followed by active rewarming (+0.2°C/hour). The primary outcome was defined as the time required to reach 35°C, while secondary outcomes comprised clinical, radiological and safety parameters. RESULTS Twenty-two patients (median age, 77 years) were included in the study (14 received additional thrombolysis, 4 additional stenting of the proximal internal carotid artery). The median time intervals were 309 min for last-seen-normal-to-groin, 58 min for door-to-cooling-initiation, 65 min for door-to-groin and 123 min for door-to-recanalisation. The target temperature of 35°C was reached within 30 min (range 13-78 min), corresponding to a cooling rate of 2.6 °C/hour. On recanalisation, 86% of the patients had a body temperature of ≤35°C. The median National Institutes of Health Stroke Scale at admission was 15 and improved to 2 by day 7, and 68% of patients had a good outcome (modified Rankin Scale 0-2) at 3 months. Postprocedure complications included asymptomatic bradycardia (32%), pneumonia (18%) and asymptomatic haemorrhagic transformation (18%). CONCLUSION The combined application of hypothermia and thrombectomy was found to be feasible in sedated and ventilated patents. Adverse events were comparable to those previously described for EVT in the absence of hypothermia. The effect of this procedure will next be evaluated in the randomised COmbination of Targeted temperature management and Thrombectomy after acute Ischemic Stroke-2 trial.
Collapse
Affiliation(s)
- Jürgen Bardutzky
- Department of Neurology and Neurophysiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rainer Kollmar
- Neurology and Neurointensive Care, Darmstadt Hospital, Darmstadt, Germany
| | - Forat Al-Rawi
- Department of Neurology and Neurophysiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johann Lambeck
- Department of Neurology and Neurophysiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Christian Taschner
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolf-Dirk Niesen
- Department of Neurology and Neurophysiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
auf dem Brinke K, Kück F, Jamous A, Ernst M, Kunze-Szikszay N, Psychogios MN, Maier IL. The effect of inadvertent systemic hypothermia after mechanical thrombectomy in patients with large-vessel occlusion stroke. Front Neurol 2024; 15:1381872. [PMID: 38903162 PMCID: PMC11188377 DOI: 10.3389/fneur.2024.1381872] [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: 02/04/2024] [Accepted: 05/08/2024] [Indexed: 06/22/2024] Open
Abstract
Background and aims Postinterventional hypothermia is a frequent complication in patients with large-vessel occlusion strokes (LVOS) after mechanical thrombectomy (MT). This inadvertent hypothermia might potentially have neuroprotective but also adverse effects on patients' outcomes. The aim of the study was to determine the rate of hypothermia in patients with LVOS receiving MT and its influence on functional outcome. Methods We performed a monocentric, retrospective study using a prospectively derived databank, including all LVOS patients receiving MT between 2015 and 2021. Predictive values of postinterventional body temperature and body temperature categories (hyperthermia (≥38°C), normothermia (35°C-37.9°C), and hypothermia (<35°C)) on functional outcome were analyzed using multivariable Bayesian logistic regression models. Favorable outcome was defined as modified Rankin Scale (mRS) ≤3. Results Of the 480 included LVOS patients with MT (46.0% men; mean ± SD age 73 ± 12.9 years), 5 (1.0%) were hyperthermic, 382 (79.6%) normothermic, and 93 (19.4%) hypothermic. Postinterventional hypothermia was significantly associated with unfavorable functional outcome (mRS > 3) after 90 days (OR 2.06, 95% CI 1.01-4.18, p = 0.045). For short-term functional outcome, patients with hypothermia had a higher discharge NIHSS (OR 1.38, 95% CI 1.06 to 1.79, p = 0.015) and a higher change of NIHSS from admission to discharge (OR 1.35, 95% CI 1.03 to 1.76, p = 0.029). Conclusion Approximately a fifth of LVOS patients in this cohort were hypothermic after MT. Hypothermia was an independent predictor of unfavorable functional outcomes. Our findings warrant a prospective trial investigating active warming during MT.
Collapse
Affiliation(s)
| | - Fabian Kück
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Ala Jamous
- Department of Neuroradiology, University Medical Center Göttingen, Göttingen, Germany
| | - Marielle Ernst
- Department of Neuroradiology, University Medical Center Göttingen, Göttingen, Germany
| | - Nils Kunze-Szikszay
- Department of Anesthesiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Ilko L. Maier
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
Bindal P, Kumar V, Kapil L, Singh C, Singh A. Therapeutic management of ischemic stroke. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2651-2679. [PMID: 37966570 DOI: 10.1007/s00210-023-02804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023]
Abstract
Stroke is the third leading cause of years lost due to disability and the second-largest cause of mortality worldwide. Most occurrences of stroke are brought on by the sudden occlusion of an artery (ischemic stroke), but sometimes they are brought on by bleeding into brain tissue after a blood vessel has ruptured (hemorrhagic stroke). Alteplase is the only therapy the American Food and Drug Administration has approved for ischemic stroke under the thrombolysis category. Current views as well as relevant clinical research on the diagnosis, assessment, and management of stroke are reviewed to suggest appropriate treatment strategies. We searched PubMed and Google Scholar for the available therapeutic regimes in the past, present, and future. With the advent of endovascular therapy in 2015 and intravenous thrombolysis in 1995, the therapeutic options for ischemic stroke have expanded significantly. A novel approach such as vagus nerve stimulation could be life-changing for many stroke patients. Therapeutic hypothermia, the process of cooling the body or brain to preserve organ integrity, is one of the most potent neuroprotectants in both clinical and preclinical contexts. The rapid intervention has been linked to more favorable clinical results. This study focuses on the pathogenesis of stroke, as well as its recent advancements, future prospects, and potential therapeutic targets in stroke therapy.
Collapse
Affiliation(s)
- Priya Bindal
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Lakshay Kapil
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt. Tehri Garhwal, Uttarakhand, 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India.
| |
Collapse
|
11
|
You Y, Gong Z, Zhang Y, Qiu L, Tang X. Observation of the effect of hypothermia therapy combined with optimized nursing on brain protection after cardiopulmonary resuscitation: A retrospective case-control study. Medicine (Baltimore) 2024; 103:e37776. [PMID: 38640316 PMCID: PMC11029950 DOI: 10.1097/md.0000000000037776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 04/21/2024] Open
Abstract
This study aimed to investigate the impact of optimized emergency nursing in conjunction with mild hypothermia nursing on neurological prognosis, hemodynamics, and complications in patients with cardiac arrest. A retrospective analysis was conducted on the medical records of 124 patients who received successful cardiopulmonary resuscitation (CPR) at Fujian Provincial Hospital South Branch. The patients were divided into control and observation groups, each consisting of 62 cases. The brain function of both groups was assessed using the Glasgow Coma Scale and the National Institutes of Health Stroke Scale. Additionally, serum neuron-specific enolase level was measured in both groups. The vital signs and hemodynamics of both groups were analyzed, and the complications and satisfaction experienced by the 2 groups were compared. The experimental group exhibited significantly improved neurological function than the control group (P < .05). Furthermore, the heart rate in the experimental group was significantly lower than the control group (P < .05). However, no significant differences were observed in blood oxygen saturation, mean arterial pressure, central venous pressure, and systolic blood pressure between the 2 groups (P > 0.05). Moreover, the implementation of optimized nursing practices significantly reduced complications and improved the quality of life and satisfaction of post-CPR patients (P < .05). The integration of optimized emergency nursing practices in conjunction with CPR improves neurological outcomes in patients with cardiac arrest.
Collapse
Affiliation(s)
- Yan You
- The Second Department of Intensive Care Unit, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Zheng Gong
- Department of Emergency Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, China
| | - Yaxu Zhang
- The Second Department of Intensive Care Unit, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Lirong Qiu
- The Second Department of Intensive Care Unit, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Xiahong Tang
- Department of Critical Care Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
12
|
Díaz-Peregrino R, Kentar M, Trenado C, Sánchez-Porras R, Albiña-Palmarola P, Ramírez-Cuapio FL, San-Juan D, Unterberg A, Woitzik J, Santos E. The neurophysiological effect of mild hypothermia in gyrencephalic brains submitted to ischemic stroke and spreading depolarizations. Front Neurosci 2024; 18:1302767. [PMID: 38567280 PMCID: PMC10986791 DOI: 10.3389/fnins.2024.1302767] [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: 09/27/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Characterize the neurophysiological effects of mild hypothermia on stroke and spreading depolarizations (SDs) in gyrencephalic brains. Methods Left middle cerebral arteries (MCAs) of six hypothermic and six normothermic pigs were permanently occluded (MCAo). Hypothermia began 1 h after MCAo and continued throughout the experiment. ECoG signals from both frontoparietal cortices were recorded. Five-minute ECoG epochs were collected 5 min before, at 5 min, 4, 8, 12, and 16 h after MCAo, and before, during, and after SDs. Power spectra were decomposed into fast (alpha, beta, and gamma) and slow (delta and theta) frequency bands. Results In the vascular insulted hemisphere under normothermia, electrodes near the ischemic core exhibited power decay across all frequency bands at 5 min and the 4th hour after MCAo. The same pattern was registered in the two furthest electrodes at the 12th and 16th hour. When mild hypothermia was applied in the vascular insulted hemispheres, the power decay was generalized and seen even in electrodes with uncompromised blood flow. During SD analysis, hypothermia maintained increased delta and beta power during the three phases of SDs in the furthest electrode from the ischemic core, followed by the second furthest and third electrode in the beta band during preSD and postSD segments. However, in hypothermic conditions, the third electrode showed lower delta, theta, and alpha power. Conclusion Mild hypothermia attenuates all frequency bands in the vascularly compromised hemisphere, irrespective of the cortical location. During SD formation, it preserves power spectra more significantly in electrodes further from the ischemic core.
Collapse
Affiliation(s)
- Roberto Díaz-Peregrino
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Modar Kentar
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
- Departement of Neurosurgery, Städtisches Klinikum Braunschweig gGmbH, Braunschweig, Germany
| | - Carlos Trenado
- Heinrich Heine University, Medical Faculty, Institute of Clinical Neuroscience and Medical Psychology, Düsseldorf, Germany
- Institute for the Future of Education Europe, Tecnológico de Monterrey, Cantabria, Spain
| | - Renán Sánchez-Porras
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Pablo Albiña-Palmarola
- Neuroradiologische Klinik, Klinikum Stuttgart, Stuttgart, Germany
- Medizinische Fakultät, Universität Duisburg-Essen, Essen, Germany
- Department of Anatomy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco L. Ramírez-Cuapio
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Daniel San-Juan
- Epilepsy Clinic, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Edgar Santos
- Department of Neurosurgery, University Hospital Heidelberg, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
13
|
Liddle LJ, Huang YG, Kung TFC, Mergenthaler P, Colbourne F, Buchan AM. An Assessment of Physical and N6-Cyclohexyladenosine-Induced Hypothermia in Rodent Distal Focal Ischemic Stroke. Ther Hypothermia Temp Manag 2024; 14:36-45. [PMID: 37339459 DOI: 10.1089/ther.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Abstract
Therapeutic hypothermia (TH) mitigates damage in ischemic stroke models. However, safer and easier TH methods (e.g., pharmacological) are needed to circumvent physical cooling complications. This study evaluated systemic and pharmacologically induced TH using the adenosine A1 receptor agonist, N6-cyclohexyladenosine (CHA), with control groups in male Sprague-Dawley rats. CHA was administered intraperitoneally 10 minutes following a 2-hour intraluminal middle cerebral artery occlusion. We used a 1.5 mg/kg induction dose, followed by three 1.0 mg/kg doses every 6 hours for a total of 4 doses, causing 20-24 hours of hypothermia. Animals assigned to physical hypothermia and CHA-hypothermia had similar induction rates and nadir temperatures, but forced cooling lasted ∼6 hours longer compared with CHA-treated animals. The divergence is likely attributable to individual differences in CHA metabolism, which led to varied durations at nadir, whereas physical hypothermia was better regulated. Physical hypothermia significantly reduced infarction (primary endpoint) on day 7 (mean reduction of 36.8 mm3 or 39% reduction; p = 0.021 vs. normothermic animals; Cohen's d = 0.75), whereas CHA-induced hypothermia did not (p = 0.33). Similarly, physical cooling improved neurological function (physical hypothermia median = 0, physical normothermia median = 2; p = 0.008) and CHA-induced cooling did not (p > 0.99). Our findings demonstrate that forced cooling was neuroprotective compared with controls, but prolonged CHA-induced cooling was not neuroprotective.
Collapse
Affiliation(s)
- Lane J Liddle
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Yi-Ge Huang
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tiffany F C Kung
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Philipp Mergenthaler
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, NeuroCure Clinical Research Center, Berlin, Germany
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Alastair M Buchan
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| |
Collapse
|
14
|
Zhang C, Ma Y, Zhao Y, Guo N, Han C, Wu Q, Mu C, Zhang Y, Tan S, Zhang J, Liu X. Systematic review of melatonin in cerebral ischemia-reperfusion injury: critical role and therapeutic opportunities. Front Pharmacol 2024; 15:1356112. [PMID: 38375039 PMCID: PMC10875093 DOI: 10.3389/fphar.2024.1356112] [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: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Cerebral ischemia-reperfusion (I/R) injury is the predominant causes for the poor prognosis of ischemic stroke patients after reperfusion therapy. Currently, potent therapeutic interventions for cerebral I/R injury are still very limited. Melatonin, an endogenous hormone, was found to be valid in preventing I/R injury in a variety of organs. However, a systematic review covering all neuroprotective effects of melatonin in cerebral I/R injury has not been reported yet. Thus, we perform a comprehensive overview of the influence of melatonin on cerebral I/R injury by collecting all available literature exploring the latent effect of melatonin on cerebral I/R injury as well as ischemic stroke. In this systematic review, we outline the extensive scientific studies and summarize the beneficial functions of melatonin, including reducing infarct volume, decreasing brain edema, improving neurological functions and attenuating blood-brain barrier breakdown, as well as its key protective mechanisms on almost every aspect of cerebral I/R injury, including inhibiting oxidative stress, neuroinflammation, apoptosis, excessive autophagy, glutamate excitotoxicity and mitochondrial dysfunction. Subsequently, we also review the predictive and therapeutic implications of melatonin on ischemic stroke reported in clinical studies. We hope that our systematic review can provide the most comprehensive introduction of current advancements on melatonin in cerebral I/R injury and new insights into personalized diagnosis and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chenguang Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yumei Ma
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yating Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shutong Tan
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Binda DD, Baker MB, Varghese S, Wang J, Badenes R, Bilotta F, Nozari A. Targeted Temperature Management for Patients with Acute Ischemic Stroke: A Literature Review. J Clin Med 2024; 13:586. [PMID: 38276093 PMCID: PMC10816923 DOI: 10.3390/jcm13020586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Despite significant advances in medical imaging, thrombolytic therapy, and mechanical thrombectomy, acute ischemic strokes (AIS) remain a major cause of mortality and morbidity globally. Targeted temperature management (TTM) has emerged as a potential therapeutic intervention, aiming to mitigate neuronal damage and improve outcomes. This literature review examines the efficacy and challenges of TTM in the context of an AIS. A comprehensive literature search was conducted using databases such as PubMed, Cochrane, Web of Science, and Google Scholar. Studies were selected based on relevance and quality. We identified key factors influencing the effectiveness of TTM such as its timing, depth and duration, and method of application. The review also highlighted challenges associated with TTM, including increased pneumonia rates. The target temperature range was typically between 32 and 36 °C, with the duration of cooling from 24 to 72 h. Early initiation of TTM was associated with better outcomes, with optimal results observed when TTM was started within the first 6 h post-stroke. Emerging evidence indicates that TTM shows considerable potential as an adjunctive treatment for AIS when implemented promptly and with precision, thereby potentially mitigating neuronal damage and enhancing overall patient outcomes. However, its application is complex and requires the careful consideration of various factors.
Collapse
Affiliation(s)
- Dhanesh D. Binda
- Department of Anesthesiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (D.D.B.); (M.B.B.); (S.V.); (J.W.); (A.N.)
| | - Maxwell B. Baker
- Department of Anesthesiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (D.D.B.); (M.B.B.); (S.V.); (J.W.); (A.N.)
| | - Shama Varghese
- Department of Anesthesiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (D.D.B.); (M.B.B.); (S.V.); (J.W.); (A.N.)
| | - Jennifer Wang
- Department of Anesthesiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (D.D.B.); (M.B.B.); (S.V.); (J.W.); (A.N.)
| | - Rafael Badenes
- Department Anesthesiology, Surgical-Trauma Intensive Care and Pain Clinic, Hospital Clínic Universitari, University of Valencia, 46010 Valencia, Spain
| | - Federico Bilotta
- Department of Anaesthesiology, Critical Care and Pain Medicine, Policlinico Umberto I Teaching Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Ala Nozari
- Department of Anesthesiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (D.D.B.); (M.B.B.); (S.V.); (J.W.); (A.N.)
| |
Collapse
|
16
|
Fang Y, Gao Q, Jin W, Li J, Yuan H, Lin Z, Pan G, Lin W. Clinical characteristics and prognostic analysis of acute necrotizing encephalopathy of childhood: a retrospective study at a single center in China over 3 years. Front Neurol 2023; 14:1308044. [PMID: 38178890 PMCID: PMC10766426 DOI: 10.3389/fneur.2023.1308044] [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: 10/05/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024] Open
Abstract
Objective Acute Necrotizing Encephalopathy of Childhood (ANEC) is a rare, fulminant neurological disease in children with unknown mechanisms and etiology. This study summarized the clinical characteristics, treatment, and prognosis of ANEC through a retrospective analysis, providing insights into the ANEC early diagnosis and prognosis assessment. Methods Clinical data of children diagnosed with ANEC at the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University from July 1, 2020, to June 30, 2023, were retrospectively analyzed. Results There were 25 cases, 14 males and 11 females, with a median age of 3 years. Hospital admissions were mainly in the winter (14/25, 56%) and spring (9/25, 36%). All patients presented with varying degrees of fever and altered consciousness, with 92% (23/25) experiencing high body temperatures (>39.1°C) and 88% (22/25) having a Glasgow coma scale (GCS) score of ≤8. Seizures were observed in 88% (22/25) of patients. Laboratory findings indicated 100% B lymphocyte activation (14/14), and 78% (14/18) of patients showed cytokine storm (interleukin (IL)-6, IL-8, IL-10, interferon (IFN)-α). Neuroimaging showed symmetrical thalamus involvement, commonly involving basal ganglia and brainstem regions. Viral infection (23/24, 96%) was the predominant etiological finding, with 42% (10/24) of cases due to SARS-CoV-2 infection and 42% (10/24) to influenza A virus infection. Multi-organ dysfunction occurred in 68% (17/25) of patients, and 52% (13/25) died. Correlation analysis revealed the death group exhibited higher proportion of male, lower GCS scores, higher IL-6 level and a greater likelihood of associated brainstem impairment (p < 0.05). Conclusion ANEC is more prevalent in the winter and spring, and its etiology may be associated with B lymphocyte activation and cytokine storm following viral infections. Clinical manifestations lack specific features, with fever, consciousness disturbances, and seizures being the main presentations, particularly in cases of high fever and hyperpyrexia. ANEC progresses rapidly and has a high mortality rate. The child's gender, GCS score, IL-6 levels, and the presence of brainstem involvement can serve as important risk factors for assessing the risk of mortality.
Collapse
Affiliation(s)
- Yu Fang
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiqi Gao
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenwen Jin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianshun Li
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao Yuan
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guoquan Pan
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Yenari MA. In cold blood: a new way to achieve therapeutic cooling? Sci Bull (Beijing) 2023; 68:2905-2906. [PMID: 37932199 DOI: 10.1016/j.scib.2023.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Affiliation(s)
- Midori A Yenari
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA.
| |
Collapse
|
18
|
Li SJ, Li F, Kong N, Liu JR, Zhu X. Near Infrared Emissive Lanthanide Luminescence Nanoparticle Used in Early Diagnosis and Brain Temperature Detection for Ischemic Stroke. Adv Healthc Mater 2023; 12:e2302276. [PMID: 37717206 DOI: 10.1002/adhm.202302276] [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: 07/18/2023] [Revised: 09/14/2023] [Indexed: 09/18/2023]
Abstract
Ischemic stroke (IS) is one of the most dangerous medical conditions resulting in high mortality and morbidity. The increased brain temperature after IS is closely related to prognosis, making it highly significant for the early diagnosis and the progression evaluation of IS. Herein, a temperature-responsive near infrared (NIR) emissive lanthanide luminescence nanoparticle is developed for the early diagnosis and brain temperature detection of IS. After intravenous injection, the nanoparticles can pass through the damaged blood-brain barrier of the ischemic region, allowing the extravasation and enrichment of nanoparticles into the ischemic brain tissue. The NIR luminescence signals of the nanoparticles are used not only to judge the location and severity of the cerebral ischemic injury but also to report the brain temperature variation in the ischemic area through a visualized way. The results show that the designed nanoparticles can be used for the early diagnosis of ischemic stroke and minimally invasive temperature detection of cerebral ischemic tissues in transient middle cerebral artery occlusion mice model, which is expected to make the clinical diagnosis of ischemic stroke more rapid and convenient, more accurately evaluate the state of brain injury in stroke patients and also guide stroke hypothermia treatment.
Collapse
Affiliation(s)
- Shen-Jie Li
- Department of Neurology, Stroke Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 ZhiZaoJu Road, Huangpu District, Shanghai, 200011, China
| | - Fang Li
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| | - Na Kong
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| | - Jian-Ren Liu
- Department of Neurology, Stroke Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 ZhiZaoJu Road, Huangpu District, Shanghai, 200011, China
| | - Xingjun Zhu
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| |
Collapse
|
19
|
Palà E, Penalba A, Bustamante A, García‐Berrocoso T, Lamana‐Vallverdú M, Meisel C, Meisel A, van der Worp HB, R Macleod M, Kallmünzer B, Schwab S, Montaner J. Blood biomarker changes following therapeutic hypothermia in ischemic stroke. Brain Behav 2023; 13:e3230. [PMID: 37721534 PMCID: PMC10636403 DOI: 10.1002/brb3.3230] [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: 03/25/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Therapeutic hypothermia is a promising candidate for stroke treatment although its efficacy has not yet been demonstrated in patients. Changes in blood molecules could act as surrogate markers to evaluate the efficacy and safety of therapeutic cooling. METHODS Blood samples from 54 patients included in the EuroHYP-1 study (27 treated with hypothermia, and 27 controls) were obtained at baseline, 24 ± 2 h, and 72 ± 4 h. The levels of a panel of 27 biomarkers, including matrix metalloproteinases and cardiac and inflammatory markers, were measured. RESULTS Metalloproteinase-3 (MMP-3), fatty-acid-binding protein (FABP), and interleukin-8 (IL-8) increased over time in relation to the hypothermia treatment. Statistically significant correlations between the minimum temperature achieved by each patient in the hypothermia group and the MMP-3 level measured at 72 h, FABP level measured at 24 h, and IL-8 levels measured at 24 and 72 h were found. No differential biomarker levels were observed in patients with poor or favorable outcomes according to modified Rankin Scale scores. CONCLUSION Although the exact roles of MMP3, FABP, and IL-8 in hypothermia-treated stroke patients are not known, further exploration is needed to confirm their roles in brain ischemia.
Collapse
Affiliation(s)
- Elena Palà
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
| | - Anna Penalba
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
| | - Alejandro Bustamante
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
- Stroke Unit, Hospital Universitari Germans Trias i PujolBadalonaSpain
| | - Teresa García‐Berrocoso
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
- CSIC/UAB Proteomics LaboratoryInstitute of Biomedical Research of BarcelonaSpanish National Research Council (IIBB‐CSIC/IDIBAPS)BarcelonaSpain
| | - Marcel Lamana‐Vallverdú
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
| | - Christian Meisel
- Institute for Medical ImmunologyCharité–Universitätsmedizin BerlinBerlinGermany
- Department of ImmunologyLabor Berlin–Charité VivantesBerlinGermany
| | - Andreas Meisel
- Department of Neurology and Center for Stroke Research BerlinCharité University Hospital BerlinBerlinGermany
| | - H. Bart van der Worp
- Department of Neurology and NeurosurgeryBrain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Malcolm R Macleod
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghScotlandUK
| | - Bernd Kallmünzer
- Department of NeurologyUniversitätsklinikum ErlangenErlangenGermany
| | - Stefan Schwab
- Department of NeurologyUniversitätsklinikum ErlangenErlangenGermany
| | - Joan Montaner
- Neurovascular Research LaboratoryVall d'Hebron Institute of Research (VHIR)–Universitat Autónoma de BarcelonaBarcelonaSpain
- Institute de Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville & Department of NeurologyHospital Universitario Virgen MacarenaSevilleSpain
| |
Collapse
|
20
|
Zhao J, Xia C, Tang Y, Wan H. Role of PERK-mediated pathway in the effect of mild hypothermia after cerebral ischaemia/reperfusion. Eur J Clin Invest 2023; 53:e14040. [PMID: 37337313 DOI: 10.1111/eci.14040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Hypothermia is an effective method of reducing brain injury caused by a variety of neurological insults. It is aimed to elucidate whether a change in the expression of PERK-mediated pathway proteins is an indicator of the neuroprotective effect of mild hypothermia after cerebral ischaemia/reperfusion. METHODS One hundred and ninety-two male C57BL/6 mice were randomly divided into three groups: a sham group, a cerebral normothermic ischaemia/reperfusion (I/R) group and a cerebral hypothermic I/R group. A cerebral ischaemia model was established by ligating the bilateral common carotid artery for 15 min. Mice in the hypothermia group stayed in a cage that was set at 33°C, sprayed with a spray of 70% ethanol, and blown with two high-speed fans. The state of neurons was assessed on micropreparations stained with haematoxylin-eosin and TUNEL. The expressions of GRP78, p-perk, p-eif2α, ATF4 and CHOP were measured by western blot analysis 6, 12, 24 and 72 h after reperfusion. RESULTS The number of surviving cells was significantly higher in the hypothermia group than in the group without hypothermia (p < .05). The GRP78 expression in the hypothermia group was statistically higher (p < .05) than in the ischaemia/reperfusion group. Optical densities of p-perk, p-eif2α and ATF4 in hippocampus CA1 neurons ischaemia were statistically significantly lower in the hypothermia group than in the ischaemia/reperfusion group (p < .05). The CHOP expression in the hypothermia group was statistically lower (p < .05) than in the ischaemia/reperfusion group. CONCLUSION Mild hypothermia for 6 h promoted moderate neuroprotection by mediating the expression of GRP78, p-PERK, p-eIF2α, ATF4 and CHOP.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Anesthesiology, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
| | - Chenzhong Xia
- Department of Anesthesiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yingying Tang
- Department of Anesthesiology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, China
| | - Haifang Wan
- Department of Anesthesiology, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
| |
Collapse
|
21
|
Wang R, Xiao L, Pan J, Bao G, Zhu Y, Zhu D, Wang J, Pei C, Ma Q, Fu X, Wang Z, Zhu M, Wang G, Gong L, Tong Q, Jiang M, Hu J, He M, Wang Y, Li T, Liang C, Li W, Xia C, Li Z, Ma DK, Tan M, Liu JY, Jiang W, Luo C, Yu B, Dang Y. Natural product P57 induces hypothermia through targeting pyridoxal kinase. Nat Commun 2023; 14:5984. [PMID: 37752106 PMCID: PMC10522591 DOI: 10.1038/s41467-023-41435-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
Induction of hypothermia during hibernation/torpor enables certain mammals to survive under extreme environmental conditions. However, pharmacological induction of hypothermia in most mammals remains a huge challenge. Here we show that a natural product P57 promptly induces hypothermia and decreases energy expenditure in mice. Mechanistically, P57 inhibits the kinase activity of pyridoxal kinase (PDXK), a key metabolic enzyme of vitamin B6 catalyzing phosphorylation of pyridoxal (PL), resulting in the accumulation of PL in hypothalamus to cause hypothermia. The hypothermia induced by P57 is significantly blunted in the mice with knockout of PDXK in the preoptic area (POA) of hypothalamus. We further found that P57 and PL have consistent effects on gene expression regulation in hypothalamus, and they may activate medial preoptic area (MPA) neurons in POA to induce hypothermia. Taken together, our findings demonstrate that P57 has a potential application in therapeutic hypothermia through regulation of vitamin B6 metabolism and PDXK serves as a previously unknown target of P57 in thermoregulation. In addition, P57 may serve as a chemical probe for exploring the neuron circuitry related to hypothermia state in mice.
Collapse
Affiliation(s)
- Ruina Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Xiao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Guangsen Bao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunmei Zhu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Di Zhu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chengfeng Pei
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Qinfeng Ma
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xian Fu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Ziruoyu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengdi Zhu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Guoxiang Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Gong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiuping Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Jiang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junchi Hu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiejun Li
- Department of Pharmacology, College of Pharmacy, Naval Medical University, Shanghai, China
| | - Chunmin Liang
- Lab of Tumor Immunology, Department of Human Anatomy, Histology and Embryology, Basic Medical School of Fudan University, Shanghai, China
| | - Wei Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zengxia Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dengke K Ma
- Department of Physiology, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan Liu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
22
|
Qin N, Wang J, Peng X, Wang L. Pathogenesis and Management of Acute Necrotizing Encephalopathy. Expert Rev Neurother 2023; 23:641-650. [PMID: 37309119 DOI: 10.1080/14737175.2023.2224503] [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: 02/02/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION During the COVID-19 pandemic, many cases of acute necrotizing encephalopathy (ANE) secondary to COVID-19 have been reported. ANE is characterized by a rapid onset, a fulminant course, and low morbidity and fatality rates. Therefore, clinicians need to be vigilant for such disorders, especially during the influenza virus and COVID-19 epidemics. AREAS COVERED The authors summarize the most recent studies on the clinical spectrum and treatment essentials of ANE to provide references for prompt diagnosis and improved treatment of this rare but fatal disease. EXPERT OPINION ANE is a type of necrotizing lesion of the brain parenchyma. There are two major types of reported cases. One is isolated and sporadic ANE, which is primarily caused by viral infections, particularly influenza and HHV-6 virus. The other type is familial recurrent ANE, which is caused by RANBP2 gene mutations. ANE patients have rapid progression and a very poor prognosis, with acute brain dysfunction occurring within days of viral infection and requiring admission to the intensive care unit. Clinicians still need to investigate and find solutions for the problems of early detection and treatment of ANE.
Collapse
Affiliation(s)
- Ningxiang Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Peng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
23
|
Huang J, Wang P, Wen H. The safety and efficacy of hypothermia combining mechanical thrombectomy or thrombolysis in the treatment of ischemic stroke: A systematic meta-analysis. Clinics (Sao Paulo) 2023; 78:100218. [PMID: 37269787 DOI: 10.1016/j.clinsp.2023.100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/26/2023] [Accepted: 04/24/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Stroke is a major global public health problem, affecting 13.7 million people worldwide. Previous studies have found a neuroprotective effect of hypothermia therapy and the efficacy and safety of combined hypothermia and mechanical thrombectomy or thrombolysis in the treatment of ischemic stroke have also attracted attention. OBJECTIVE In the present research, the authors conducted a meta-analysis to comprehensively assess the safety and efficacy of hypothermia combining mechanical thrombectomy or thrombolysis in the treatment of ischemic stroke. METHODS Articles published from January 2001 to May 2022 were searched from Google Scholar, Baidu Scholar and PubMed to evaluate the clinical significance of hypothermia treatment in ischemic stroke. Complications, short-term mortality, and the modified Rankin Scale (mRS) in the full text was extracted. RESULTS 89 publications were selected and 9 among them were included in this study with sample size of 643. All selected studies are in accordance with the inclusion criteria. Forest plot of clinical characteristics was as follows: complications (RR = 1.132, 95% CI 0.942‒1.361, p = 0.186, I2 = 37.2%), mortality within 3 months (RR = 1.076, 95% CI 0.694‒1.669, p = 0.744, I2 = 0.00%), mRS ≤ 1 at 3 months (RR = 1.138, 95% CI 0.829‒1.563, p = 0.423, I2 = 26.0%), mRS ≤ 2 at 3 months (RR = 1.672, 95% CI 1.236‒2.263, p = 0.001, I2=49.6%) and mRS ≤ 3 at 3 months (RR = 1.518, 95% CI 1.128‒2.043, p = 0.006, I2 = 0.00%). The funnel plot suggested that there was no significant publication bias in the meta-analysis on complications, mortality within 3 months, mRS ≤ 1 at 3 months and mRS ≤ 2 at 3 months. CONCLUSION In summary, the results showed that hypothermia treatment was correlated with mRS ≤ 2 at 3 months, but not linked with complications and mortality within 3 months.
Collapse
Affiliation(s)
- Jiankang Huang
- Department of Neurology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, Jiangshu 211200, China
| | - Peng Wang
- Department of Neurology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, Jiangshu 211200, China
| | - Hongbo Wen
- Department of Neurology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, Jiangshu 211200, China.
| |
Collapse
|
24
|
Kentar M, Ramirez-Cuapio FL, Gutiérrez-Herrera MA, Sanchez-Porras R, Díaz-Peregrino R, Holzwarth N, Maier-Hein L, Woitzik J, Santos E. Mild hypothermia reduces spreading depolarizations and infarct size in a swine model. J Cereb Blood Flow Metab 2023; 43:999-1009. [PMID: 36722153 PMCID: PMC10196741 DOI: 10.1177/0271678x231154604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 02/02/2023]
Abstract
Spreading depolarizations (SDs) have been linked to infarct volume expansion following ischemic stroke. Therapeutic hypothermia provides a neuroprotective effect after ischemic stroke. This study aimed to evaluate the effect of hypothermia on the propagation of SDs and infarct volume in an ischemic swine model. Through left orbital exenteration, middle cerebral arteries were surgically occluded (MCAo) in 16 swine. Extensive craniotomy and durotomy were performed. Six hypothermic and five normothermic animals were included in the analysis. An intracranial temperature probe was placed right frontal subdural. One hour after ischemic onset, mild hypothermia was induced and eighteen hours of electrocorticographic (ECoG) and intrinsic optical signal (IOS) recordings were acquired. Postmortem, 4 mm-thick slices were stained with 2,3,5-triphenyltetrazolium chloride to estimate the infarct volume. Compared to normothermia (36.4 ± 0.4°C), hypothermia (32.3 ± 0.2°C) significantly reduced the frequency and expansion of SDs (ECoG: 3.5 ± 2.1, 73.2 ± 5.2% vs. 1.0 ± 0.7, 41.9 ± 21.8%; IOS 3.9 ± 0.4, 87.6 ± 12.0% vs. 1.4 ± 0.7, 67.7 ± 8.3%, respectively). Further, infarct volume among hypothermic animals (23.2 ± 1.8% vs. 32.4 ± 2.5%) was significantly reduced. Therapeutic hypothermia reduces infarct volume and the frequency and expansion of SDs following cerebral ischemia.
Collapse
Affiliation(s)
- Modar Kentar
- Department of Neurosurgery,
University of Heidelberg, Heidelberg, Germany
| | | | | | - Renan Sanchez-Porras
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| | | | - Niklas Holzwarth
- Division of Intelligent Medical
Systems, German Cancer Research Center, Heidelberg, Germany
| | - Lena Maier-Hein
- Division of Intelligent Medical
Systems, German Cancer Research Center, Heidelberg, Germany
| | - Johannes Woitzik
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| | - Edgar Santos
- Department of Neurosurgery,
University of Heidelberg, Heidelberg, Germany
- Department of Neurosurgery,
Evangelisches Krankenhaus Oldenburg, Carl von Ossietzky University of Oldenburg,
Oldenburg, Germany
| |
Collapse
|
25
|
Lavinio A, Andrzejowski J, Antonopoulou I, Coles J, Geoghegan P, Gibson K, Gudibande S, Lopez-Soto C, Mullhi R, Nair P, Pauliah VP, Quinn A, Rasulo F, Ratcliffe A, Reddy U, Rhodes J, Robba C, Wiles M, Williams A. Targeted temperature management in patients with intracerebral haemorrhage, subarachnoid haemorrhage, or acute ischaemic stroke: updated consensus guideline recommendations by the Neuroprotective Therapy Consensus Review (NTCR) group. Br J Anaesth 2023:S0007-0912(23)00205-2. [PMID: 37225535 DOI: 10.1016/j.bja.2023.04.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND There is a lack of consistent, evidence-based guidelines for the management of patients with fever after brain injury. The aim was to update previously published consensus recommendations on targeted temperature management after intracerebral haemorrhage, aneurysmal subarachnoid haemorrhage, and acute ischaemic stroke in patients who require admission to critical care. METHODS A modified Delphi consensus, the Neuroprotective Therapy Consensus Review (NTCR), included 19 international neuro-intensive care experts with a subspecialty interest in the acute management of intracerebral haemorrhage, aneurysmal subarachnoid haemorrhage, and acute ischaemic stroke. An online, anonymised survey was completed ahead of the meeting before the group came together to consolidate consensus and finalise recommendations on targeted temperature management. A threshold of ≥80% for consensus was set for all statements. RESULTS Recommendations were formulated based on existing evidence, literature review, and consensus. After intracerebral haemorrhage, aneurysmal subarachnoid haemorrhage, and acute ischaemic stroke in patients who require critical care admission, core temperature should ideally be monitored continuously and maintained between 36.0°C and 37.5°C using automated feedback-controlled devices, where possible. Targeted temperature management should be commenced within 1 h of first fever identification with appropriate diagnosis and treatment of infection, maintained for as long as the brain remains at risk of secondary injury, and rewarming should be controlled. Shivering should be monitored and managed to limit risk of secondary injury. Following a single protocol for targeted temperature management across intracerebral haemorrhage, aneurysmal subarachnoid haemorrhage, and acute ischaemic stroke is desirable. CONCLUSIONS Based on a modified Delphi expert consensus process, these guidelines aim to improve the quality of targeted temperature management for patients after intracerebral haemorrhage, aneurysmal subarachnoid haemorrhage, and acute ischaemic stroke in critical care, highlighting the need for further research to improve clinical guidelines in this setting.
Collapse
Affiliation(s)
- Andrea Lavinio
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | | | | | - Jonathan Coles
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; University of Cambridge, Cambridge, UK
| | | | - Kyle Gibson
- National Hospital for Neurology and Neurosurgery, London, UK
| | | | | | - Randeep Mullhi
- University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Priya Nair
- The Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | - Aoife Quinn
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Frank Rasulo
- Spedali Civili University Hospital of Brescia, Brescia, Italy
| | | | - Ugan Reddy
- National Hospital for Neurology and Neurosurgery, London, UK
| | | | - Chiara Robba
- Ospedale Policlinicio San Martino, Genova, Italy
| | - Matthew Wiles
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | |
Collapse
|
26
|
Zhao Y, Liu J, Ding Z, Ge W, Wang S, Zhang J. ATP-induced hypothermia improves burn injury and relieves burn pain in mice. J Therm Biol 2023; 114:103563. [PMID: 37344025 DOI: 10.1016/j.jtherbio.2023.103563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/05/2023] [Accepted: 04/02/2023] [Indexed: 06/23/2023]
Abstract
Thermal burn injury is a severe and life-threatening form of trauma that presents a significant challenge to clinical therapy. Therapeutic hypothermia has been shown to be beneficial in various human pathologies. Adenosine triphosphate (ATP) induces a hypothermic state that resembles hibernation-like suspended animation in mammals. This study investigates the potential protective role of ATP-induced hypothermia in thermal burn injury. Male C57BL/6 mice underwent a sham procedure or third-degree burn, and ATP-induced hypothermia was applied immediately or 1 h after burn injury. Our results show that ATP-induced hypothermia significantly improved burn depth progression and reduced collagen degradation. Moreover, hypothermia induced by ATP alleviated burn-induced hyperinflammatory responses and oxidative stress. Metabolomic profiling revealed that ATP-induced hypothermia reversed the shifts of metabolic profiles of the skin in burn mice. In addition, ATP-induced hypothermia relieved nociceptive and inflammatory pain, as observed in the antinociceptive test. Our findings suggest that ATP-induced hypothermia attenuates burn injury and provides new insights into first-aid therapy after thermal burn injury.
Collapse
Affiliation(s)
- Yang Zhao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China
| | - Junhao Liu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China
| | - Zhao Ding
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China
| | - Wenhao Ge
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China
| | - Shiming Wang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, 210094, China.
| |
Collapse
|
27
|
Wei X, He Y, Wan H, Yin J, Lin B, Ding Z, Yang J, Zhou H. Integrated transcriptomics, proteomics and metabolomics to identify biomarkers of astragaloside IV against cerebral ischemic injury in rats. Food Funct 2023; 14:3588-3599. [PMID: 36946308 DOI: 10.1039/d2fo03030f] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The herb Astragali Radix is a food-medicine herb. A major component of Astragali Radix, astragaloside IV (AS-IV), has neuroprotective effects in IS, but its mechanisms are not well understood. Our research used a transient middle cerebral artery occlusion (MCAO) rat model for longitudinal multi-omics analyses of the side of the brain affected by ischemia. Based on transcriptomic and proteomic analysis, we found that 396 differential expression targets were up-regulated and 114 differential expression targets were down-regulated. A total of 117 differential metabolites were identified based on metabonomics. Finally, we found 8 hub genes corresponding to the compound-reaction-enzyme-gene network using the Metscape plug-in for Cytoscape 3.7.1. We found that the related key metabolites were 3,4-dihydroxy-L-phenylalanine, 2-aminomuconate semialdehyde, (R)-3-hydroxybutanoate, etc., and the affected pathways were tyrosine metabolism, tryptophan metabolism, butanoate metabolism, purine metabolism, etc. We further validated these targets using 4D-PRM proteomics and found that seven targets were significantly different, including Aprt, Atic, Gaa, Galk1, Glb1, Me2, and Hexa. We aimed to uncover the mechanism of AS-IV in the treatment of ischemic brain injury through a comprehensive strategy combining transcriptomics, proteomics, and metabolomics.
Collapse
Affiliation(s)
- Xiaoyu Wei
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Junjun Yin
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Bingying Lin
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, P. R. China.
| |
Collapse
|
28
|
Narayanamurthy R, Armstrong EA, Yang JLJ, Yager JY, Unsworth LD. Administration of selective brain hypothermia using a simple cooling device in neonatal rats. J Neurosci Methods 2023; 390:109838. [PMID: 36933705 DOI: 10.1016/j.jneumeth.2023.109838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The interruption of oxygen and blood supply to the newborn brain around the time of birth is a risk factor for hypoxic-ischemic encephalopathy and may lead to infant mortality or lifelong neurological impairments. Currently, therapeutic hypothermia, the cooling of the infant's head or entire body, is the only treatment to curb the extent of brain damage. NEW METHOD In this study, we designed a focal brain cooling device that circulates cooled water at a steady state temperature of 19 ± 1 °C through a coil of tubing fitted onto the neonatal rat's head. We tested its ability to selectively decrease brain temperature and offer neuroprotection in a neonatal rat model of hypoxic-ischemic brain injury. RESULTS Our method cooled the brain to 30-33 °C in conscious pups, while keeping the core body temperature approximately 3.2 °C warmer. Furthermore, the application of the cooling device to the neonatal rat model demonstrated a reduction in brain volume loss compared to pups maintained at normothermia and achieved a level of brain tissue protection the same as that of whole-body cooling. COMPARISON WITH EXISTING METHODS Prevailing methods of selective brain hypothermia are designed for adult animal models rather than for immature animals such as the rat as a conventional model of developmental brain pathology. Contrary to existing methods, our method of cooling does not require surgical manipulation or anaesthesia. CONCLUSION Our simple, economical, and effective method of selective brain cooling is a useful tool for rodent studies in neonatal brain injury and adaptive therapeutic interventions.
Collapse
Affiliation(s)
- Rukhmani Narayanamurthy
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Edward A Armstrong
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Jung-Lynn Jonathan Yang
- Department of Chemical and Materials Engineering, University of Alberta, 11487 89 Avenue, Edmonton, Alberta T6G 2M7, Canada
| | - Jerome Y Yager
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, 11487 89 Avenue, Edmonton, Alberta T6G 2M7, Canada.
| |
Collapse
|
29
|
Imataka G, Fujita Y, Kikuchi J, Wake K, Ono K, Yoshihara S. Brain Hypothermia Therapy and Targeted Temperature Management for Acute Encephalopathy in Children: Status and Prospects. J Clin Med 2023; 12:2095. [PMID: 36983098 PMCID: PMC10058746 DOI: 10.3390/jcm12062095] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/28/2023] [Accepted: 03/05/2023] [Indexed: 03/10/2023] Open
Abstract
In adult intensive care, brain hypothermia therapy (BHT) was reported to be effective in neuroprotection after resuscitation and cardiac arrest. By contrast, in neonatal intensive care, the pathophysiology of brain damage caused by hypoxic-ischemic encephalopathy (HIE) is attributed to circulatory disturbances resulting from ischemia/reperfusion, for which neonatal brain cryotherapy is used. The International Liaison Committee on Resuscitation, 2010, recommends cerebral cryotherapy for HIE associated with severe neonatal pseudoparenchyma death. The usefulness of BHT for neuroprotection in infants and children, especially in pediatric acute encephalopathy, is expected. Theoretically, BHT could be useful in basic medical science and animal experiments. However, there are limitations in clinical planning for treating pediatric acute encephalopathy. No international collaborative study has been conducted, and no clinical evidence exists for neuroprotection using BHT. In this review, we will discuss the pathogenesis of neuronal damage in hypoxic and hypoperfused brains; the history of BHT, its effects, and mechanisms of action; the success of BHT; cooling and monitoring methods of BHT; adverse reactions to BHT; literature on BHT. We will review the latest literature on targeted temperature management, which is used for maintaining and controlling body temperature in adults in intensive care. Finally, we will discuss the development of BHT and targeted temperature management as treatments for pediatric acute encephalopathy.
Collapse
Affiliation(s)
- George Imataka
- Department of Pediatrics, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| | - Yuji Fujita
- Department of Pediatrics, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| | - Jin Kikuchi
- Department of Emergency and Critical Care Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| | - Koji Wake
- Department of Emergency and Critical Care Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| | - Kazuyuki Ono
- Department of Emergency and Critical Care Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| | - Shigemi Yoshihara
- Department of Pediatrics, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan
| |
Collapse
|
30
|
Caffo M, Fusco R, Siracusa R, Caruso G, Barresi V, Di Paola R, Cuzzocrea S, Germanò AF, Cardali SM. Molecular Investigation of DKK3 in Cerebral Ischemic/Reperfusion Injury. Biomedicines 2023; 11:biomedicines11030815. [PMID: 36979794 PMCID: PMC10045463 DOI: 10.3390/biomedicines11030815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Dickkopf-3 (Dkk3) is an atypical member of the Dkk family of Wnt inhibitors, which has been implicated in the pathophysiology of neurodegenerative disorders. Its role in the mechanisms of cellular degeneration and protection is still unknown. The aim of our work is to investigate the endogenous activation of the DKK3 pathway in a model of transient occlusion of the middle cerebral artery in rats. In particular, the animals were subjected to 1 h of ischemia followed by different reperfusion times (1 h, 6 h, 12 h and 24 h) to evaluate the downstream pathway and the time course of its activation. Western blot analysis showed increased Dkk3 expression in animals with the highest time of reperfusion. The increased levels of Dkk3 were accompanied by reduced Wnt3a, Frz1 and PIWI1a expression in the cytosol while FOXM1 and β-catenin decreased in the nucleus. These molecular changes led to an increase in the apoptotic pathway, as showed by the increased expression of Caspase 3 and Bax and the reduced levels of Bcl-2, and to a decrease in neurogenesis, as shown by the decreased expression of Tbr2, Ngn2 and Pax6. In the second part of the study, we decided to employ curcumin, an activator of the Wnt/β-catenin signaling, to investigate its effect on Dkk3. In particular, curcumin was administered 1 and 6 h after ischemia, and animals were sacrificed 24 h later when the expression of Dkk3 was higher. Our data displayed that curcumin administration decreased Dkk3 expression, and increased Wnt3a, Frz1 and PIWI1a levels. Well in line with these data, curcumin administration increased nuclear β-catenin and FOXM1 expression. The down-regulation of Dkk3 by curcumin led to reduced apoptosis and increased neurogenesis. Summarizing, our results showed that Dkk3 acts as an inhibitor of Wnt/β-catenin signaling during cerebral ischemia. Additionally, its inhibition and the contextual activation of the Wnt/β-catenin pathway are protective against ischemic stroke.
Collapse
Affiliation(s)
- Maria Caffo
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Gerardo Caruso
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Piazzale Ludovico Antonio Scuro, 37124 Verona, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, 98166 Messina, Italy
| | - Antonino Francesco Germanò
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Salvatore Massimo Cardali
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
31
|
Zhou Z, Liu Z, Zhang C, Zhang W, Zhang C, Chen T, Wang Y. Mild hypothermia alleviates early brain injury after subarachnoid hemorrhage via suppressing pyroptosis through AMPK/NLRP3 inflammasome pathway in rats. Brain Res Bull 2023; 193:72-83. [PMID: 36535306 DOI: 10.1016/j.brainresbull.2022.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
As a subtype of stroke, subarachnoid hemorrhage (SAH) has a notoriously high rate of disability and mortality owing to the lack of effective intervention. Early brain injury (EBI) is the main factor responsible for the dismal prognosis of SAH patients. The current study intends to explore the molecular mechanism underlying the effect of MH on EBI after SAH from a novel perspective of pyroptosis, a highly specific inflammatory programmed cell death, in the SAH rat model. Sprague-Dawley (SD) rats were divided into different groups in accordance with various treatments. In the treatment group, the rats underwent mild hypothermia for 4 h after modeling; in the inhibitor group, Compound C (an inhibitor of AMPK) was administered intravenous injections (i.v.) 30 min before modeling. Neurological score, neuronal death, brain water content, inflammatory reaction, and expression levels of pyroptosis-related proteins were evaluated in the rats. Our results indicate that the MH therapy significantly increased the neurological score and assuaged brain edema, neuronal injury, and inflammatory reaction induced by SAH. Meanwhile, MH therapy upregulated the level of AMPK phosphorylation whereas downregulated the protein expressions of NLRP3, ASC, cleaved caspase-1, GSDMD, IL-1β, and IL-18. The reversed effect of MH therapy by Compound C concretely indicated that MH therapy inhibited pyroptosis through an AMPK-dependent pathway. Our study also found that MH therapy potently curbed the increasing trend of brain temperature (BT), rectal temperature (RT), and ICP after SAH. Taken together, our data indicate that the neuroprotective effects of MH therapy were manifested by inhibiting pyroptosis via the AMPK/NLRP3 inflammasome pathway, which may serve as a promising therapy for the intervention of SAH.
Collapse
Affiliation(s)
- Zhaopeng Zhou
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China
| | - Zhuanghua Liu
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China
| | - Chenxu Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China
| | - Wang Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China
| | - Chunlei Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China
| | - Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China.
| | - Yuhai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu, 214044, China.
| |
Collapse
|
32
|
Payal N, Sharma L, Sharma A, Hobanii YH, Hakami MA, Ali N, Rashid S, Sachdeva M, Gulati M, Yadav S, Chigurupati S, Singh A, Khan H, Behl T. Understanding the Therapeutic Approaches for Neuroprotection. Curr Pharm Des 2023; 29:3368-3384. [PMID: 38151849 DOI: 10.2174/0113816128275761231103102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/07/2023] [Indexed: 12/29/2023]
Abstract
The term "neurodegenerative disorders" refers to a group of illnesses in which deterioration of nerve structure and function is a prominent feature. Cognitive capacities such as memory and decision-making deteriorate as a result of neuronal damage. The primary difficulty that remains is safeguarding neurons since they do not proliferate or regenerate spontaneously and are therefore not substituted by the body after they have been damaged. Millions of individuals throughout the world suffer from neurodegenerative diseases. Various pathways lead to neurodegeneration, including endoplasmic reticulum stress, calcium ion overload, mitochondrial dysfunction, reactive oxygen species generation, and apoptosis. Although different treatments and therapies are available for neuroprotection after a brain injury or damage, the obstacles are inextricably connected. Several studies have revealed the pathogenic effects of hypothermia, different breathed gases, stem cell treatments, mitochondrial transplantation, multi-pharmacological therapy, and other therapies that have improved neurological recovery and survival outcomes after brain damage. The present review highlights the use of therapeutic approaches that can be targeted to develop and understand significant therapies for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Nazrana Payal
- Department of Pharmacy, School of Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Yahya Hosan Hobanii
- Department of Pharmacy, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | | | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Monika Sachdeva
- Department of Pharmacy, Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India
- ARCCIM, Faculty of Health, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 52571, Kingdom of Saudi Arabia
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Saveetha Nagar, Thandalam, Chennai 602105, India
| | - Abhiav Singh
- Department of Pharmacy, Indian Council of Medical Research, New Delhi, India
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Tapan Behl
- Department of Pharmacy, School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India
| |
Collapse
|
33
|
Kwon K, Song JH, Park H, Kwon OY, Kim SW. Regulation of Dihydropyrimidinase-like 3 Gene Expression by MicroRNAs in PC12 Cells with Induced Ischaemia and Hypothermia. Folia Biol (Praha) 2023; 69:69-73. [PMID: 38063003 DOI: 10.14712/fb2023069020069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Although hypothermic treatment has been reported to have some beneficial effects on ischaemia at the clinical level, the mechanism of ischaemia suppression by hypothermia remains unclear due to a lack of mechanism understanding and insufficient data. The aim of this study was to isolate and characterize microRNAs specifically expressed in ischaemia-hypothermia for the dihydropyrimidinase-like 3 (Dpysl3) gene. PC12 cells were induced with CoCl2 for chemical ischaemia and incubated at 32 ℃ for hypothermia. In ischaemia-hypothermia, four types of microRNAs (miR-106b-5p, miR-194-5p, miR-326-5p, and miR-497-5p) were highly related to the Dpysl3 gene based on exosomal microRNA analysis. Dpysl3 gene expression was up-regulated by miR-497-5p but down-regulated by miR-106b-5p, miR-194-5p and miR-326-5p. Our results suggest that these four microRNAs are involved in the regulation of Dpysl3 gene expression. These findings provide valuable clues that exosomal microRNAs could be used as therapeutic targets for effective treatment of ischaemia.
Collapse
Affiliation(s)
- Kisang Kwon
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan, South Korea
| | - Ji-Hye Song
- Institute of Bioscience and Integrative Medicine, College of Korean Medicine, Daejeon University, Daejeon, South Korea
| | - Hyewon Park
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - O-Yu Kwon
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon, South Korea.
| | - Seung-Whan Kim
- Department of Emergency Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
34
|
Zhou YH, Pang S, Miao GR, Zhao XY, Dong JZ. Combining the anion gap with the sequential organ failure assessment score to evaluate the short-term prognosis of patients in the cardiac intensive care unit. Int J Cardiol 2023; 370:381-387. [PMID: 36332753 DOI: 10.1016/j.ijcard.2022.10.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND We attempted to determine the predictive ability of the first-day Sequential Organ Failure Assessment (SOFA) score in the cardiac intensive care unit, as well as a new score combining the anion gap (AG) with the SOFA score (SOFA-AG). METHODS Information was obtained from the Medical Information Mart for Intensive Care III (MIMIC III 1.4) database. We plotted the relationship between the maximum first-day AG and 90-day mortality after admission to the care unit. Patients were divided into five groups based on the hazard ratio (HR) and assigned scores of 0, 1, 2, 3, or 4 points. We compared the area under the curve (AUC) for the receiver-operating characteristic curve of the SOFA and that of the SOFA-AG. RESULTS A total of 1316 patients were identified and divided into the following five groups: AG 8 to <16 mmol/L; AG 16 to <17 mmol/L; AG 17 to <19 mmol/L; AG 19 to <21 mmol/L; and AG ≥ 21 mmol/L. The SOFA-AG score had a greater AUC than the SOFA score at 7 days (0.770 vs. 0.711; P < 0.001), 14 days (0.751 vs. 0.692; P < 0.001), 28 days (0.741 vs. 0.684; P < 0.001), and 90 days (0.727 vs. 0.667; P < 0.001). CONCLUSIONS The SOFA score showed moderate predictive value only for 7-day mortality after admission to the cardiac intensive care unit, but the SOFA-AG score had improved predictive ability for up to 90 days after admission.
Collapse
Affiliation(s)
- Yuan-Hang Zhou
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuo Pang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guang-Rui Miao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiao-Yan Zhao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jian-Zeng Dong
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
35
|
The BE COOL Treatments (Batroxobin, oxygEn, Conditioning, and cOOLing): Emerging Adjunct Therapies for Ischemic Cerebrovascular Disease. J Clin Med 2022; 11:jcm11206193. [PMID: 36294518 PMCID: PMC9605177 DOI: 10.3390/jcm11206193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic cerebrovascular disease (ICD), the most common neurological disease worldwide, can be classified based on the onset time (acute/chronic) and the type of cerebral blood vessel involved (artery or venous sinus). Classifications include acute ischemic stroke (AIS)/transient ischemic attack (TIA), chronic cerebral circulation insufficiency (CCCI), acute cerebral venous sinus thrombosis (CVST), and chronic cerebrospinal venous insufficiency (CCSVI). The pathogenesis of cerebral arterial ischemia may be correlated with cerebral venous ischemia through decreased cerebral perfusion. The core treatment goals for both arterial and venous ICDs include perfusion recovery, reduction of cerebral ischemic injury, and preservation of the neuronal integrity of the involved region as soon as possible; however, therapy based on the current guidelines for either acute ischemic events or chronic cerebral ischemia is not ideal because the recurrence rate of AIS or CVST is still very high. Therefore, this review discusses the neuroprotective effects of four novel potential ICD treatments with high translation rates, known as the BE COOL treatments (Batroxobin, oxygEn, Conditioning, and cOOLing), and subsequently analyzes how BE COOL treatments are used in clinical settings. The combination of batroxobin, oxygen, conditioning, and cooling may be a promising intervention for preserving ischemic tissues.
Collapse
|
36
|
You JS, Kim JY, Yenari MA. Therapeutic hypothermia for stroke: Unique challenges at the bedside. Front Neurol 2022; 13:951586. [PMID: 36262833 PMCID: PMC9575992 DOI: 10.3389/fneur.2022.951586] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/08/2022] [Indexed: 12/24/2022] Open
Abstract
Therapeutic hypothermia has shown promise as a means to improving neurological outcomes at several neurological conditions. At the clinical level, it has been shown to improve outcomes in comatose survivors of cardiac arrest and in neonatal hypoxic ischemic encephalopathy, but has yet to be convincingly demonstrated in stroke. While numerous preclinical studies have shown benefit in stroke models, translating this to the clinical level has proven challenging. Major obstacles include cooling patients with typical stroke who are awake and breathing spontaneously but often have significant comorbidities. Solutions around these problems include selective brain cooling and cooling to lesser depths or avoiding hyperthermia. This review will cover the mechanisms of protection by therapeutic hypothermia, as well as recent progress made in selective brain cooling and the neuroprotective effects of only slightly lowering brain temperature. Therapeutic hypothermia for stroke has been shown to be feasible, but has yet to be definitively proven effective. There is clearly much work to be undertaken in this area.
Collapse
Affiliation(s)
- Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Midori A. Yenari
- Department of Neurology, The San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Midori A. Yenari
| |
Collapse
|
37
|
Zhang J, Lu Y, Yu P, Li Z, Liu Y, Zhang J, Tang X, Yu S. Therapeutic hypothermia alleviates myocardial ischaemia-reperfusion injury by inhibiting inflammation and fibrosis via the mediation of the SIRT3/NLRP3 signalling pathway. J Cell Mol Med 2022; 26:4995-5007. [PMID: 36036085 PMCID: PMC9549509 DOI: 10.1111/jcmm.17523] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/18/2022] [Accepted: 08/09/2022] [Indexed: 11/11/2022] Open
Abstract
Therapeutic hypothermia (TH) may attenuate myocardial ischaemia–reperfusion injury, thereby improving outcomes in acute myocardial infarction. However, the specific mechanism by which TH alleviates MIRI has not been elucidated so far. In this study, 120 healthy male Sprague‐Dawley rats were randomly divided into five groups. Haemodynamic parameters, myocardial infarction area, histological changes and the levels of cardiac enzymes, caspase‐1 and inflammatory cytokines were determined. In addition, the extent of myocardial fibrosis, the degree of cardiomyocyte apoptosis and the expression levels of SIRT3, GSDMD‐N, fibrosis‐related proteins and inflammation‐related proteins were estimated.TH reduced myocardial infarct area and cardiac enzyme levels, improved cardiomyopathic damage and haemodynamic indexes, and attenuated myocardial fibrosis, the protein expression levels of collagen I and III, myocardial apoptosis, the levels of inflammatory cytokines and inflammation‐related proteins. Notably, the immunofluorescence and protein expression levels of SIRT3 were upregulated in the 34H+DMSO group compared to the I/R group, but this protective effect was abolished by the SIRT3 inhibitor 3‐TYP. After administration of Mcc950, the reversal effects of 3‐TYP were significantly abolished, and TH could protect against MIRI in a rat isolated heart model by inhibiting inflammation and fibrosis. The SIRT3/NLRP3 signalling pathway is one of the most important signalling pathways in this regard.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yimei Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Yang Liu
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jun Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyi Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuchun Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
38
|
The effects of hypothermia on glutamate and γ-aminobutyric acid metabolism during ischemia in monkeys: a repeated-measures ANOVA study. Sci Rep 2022; 12:14470. [PMID: 36008544 PMCID: PMC9411555 DOI: 10.1038/s41598-022-18783-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
During an ischemic stroke, the brain releases various factors, including glutamate and γ-aminobutyric acid. Glutamate can cause neurotoxic effects through certain receptors and exacerbate neurological damage, while γ-aminobutyric acid as an inhibitory neurotransmitter can antagonize the excitotoxic effects of glutamate and enhance the tolerance of neurons to ischemia. Therefore, in this study, the content of amino acid neurotransmitters in brain tissue before ischemia, after 10 min of ischemia, hypothermic perfusion, and rewarming were analyzed by high-performance liquid chromatography-UV in an animal model of ischemic stroke generated by blocking the bilateral common carotid arteries of rhesus monkeys. The changes in amino acid neurotransmitters in the rhesus monkey brain during post-ischemia hypothermic perfusion and rewarming were investigated by statistical methods of repeated measures ANOVA, showing that the concentration change of glutamate had not only a temporal factor but also was influenced by temperature, and there was an interaction effect between the two. Time but not temperature affected the change in γ-aminobutyric acid concentration, and there was an interaction effect between the two. Accordingly, hypoperfusion exerts a protective effect during ischemia by inhibiting the release of excitatory amino acid neurotransmitters, while the antagonistic effect of GABA on Glu is not significant.
Collapse
|
39
|
Horn M, Diprose WK, Pichardo S, Demchuk A, Almekhlafi M. Non-invasive Brain Temperature Measurement in Acute Ischemic Stroke. Front Neurol 2022; 13:889214. [PMID: 35989905 PMCID: PMC9388770 DOI: 10.3389/fneur.2022.889214] [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: 03/03/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Selective therapeutic hypothermia in the setting of mechanical thrombectomy (MT) is promising to further improve the outcomes of large vessel occlusion stroke. A significant limitation in applying hypothermia in this setting is the lack of real-time non-invasive brain temperature monitoring mechanism. Non-invasive brain temperature monitoring would provide important information regarding the brain temperature changes during cooling, and the factors that might influence any fluctuations. This review aims to provide appraisal of brain temperature changes during stroke, and the currently available non-invasive modalities of brain temperature measurement that have been developed and tested over the past 20 years. We cover modalities including magnetic resonance spectroscopy imaging (MRSI), radiometric thermometry, and microwave radiometry, and the evidence for their accuracy from human and animal studies. We also evaluate the feasibility of using these modalities in the acute stroke setting and potential ways for incorporating brain temperature monitoring in the stroke workflow.
Collapse
Affiliation(s)
- MacKenzie Horn
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- *Correspondence: MacKenzie Horn
| | - William K Diprose
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Samuel Pichardo
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Andrew Demchuk
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Mohammed Almekhlafi
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Hypothermia Protects against Ischemic Stroke through Peroxisome-Proliferator-Activated-Receptor Gamma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6029445. [PMID: 35873794 PMCID: PMC9303492 DOI: 10.1155/2022/6029445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 11/17/2022]
Abstract
Ischemic stroke (IS) remains a global public health burden and requires novel strategies. Hypothermia plays a beneficial role in central nervous system diseases. However, the role of hypothermia in IS has not yet been elucidated. In this study, we determined the role of hypothermia in IS and explored its underlying mechanisms. The IS phenotype was detected based on infarct size, infarct volume, and brain edema in mice. Neuroinflammation was evaluated by the activation of microglial cells and the expression of inflammatory genes after ischemia/reperfusion (I/R) and oxygen-glucose deprivation/reperfusion (OGD/R). Neuronal cell apoptosis, cleaved caspase-3 and Bax/Bcl-2 expressions, cell viability, and lactate dehydrogenase (LDH) release were detected after I/R and OGD/R. Blood–brain barrier (BBB) permeability was calculated based on Evans blue extravasation, tight junction protein expression, cell viability, and LDH release after I/R and OGD/R. The expression of peroxisome proliferator-activated receptor gamma (PPARγ) was assessed after OGD/R. Our results suggested that hypothermia significantly reduced infarct size, brain edema, and neuroinflammation after I/R. Hypothermia increased PPARγ expression in microglial cells after OGD/R. Mechanistic studies revealed that hypothermia was a protectant against IS, including attenuated apoptosis of neuronal cells and BBB disruption after I/R and OGD/R, by upregulating PPARγ expression. The hypothermic effect was reversed by GW9662, a PPARγ inhibitor. Our data showed that hypothermia may reduce microglial cell-mediated neuroinflammation by upregulating PPARγ expression in microglial cells. Targeting hypothermia may be a feasible approach for IS treatment.
Collapse
|
41
|
Zhong C, Ye M, Hu L, Liu J. Association Between High Serum Anion Gap and All-Cause Mortality in Non-Traumatic Subarachnoid Hemorrhage: A Retrospective Analysis of the MIMIC-IV Database. Front Neurol 2022; 13:922099. [PMID: 35903118 PMCID: PMC9321397 DOI: 10.3389/fneur.2022.922099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022] Open
Abstract
Background: High serum anion gap (AG) on admission is often correlated with poor outcomes in critically ill patients; however, data in patients with non-traumatic subarachnoid hemorrhage (SAH) are lacking. Herein, we aimed to identify the association between serum AG and all-cause mortality in patients with non-traumatic SAH. Methods A retrospective analysis of data from the Medical Information Mart for Intensive Care (MIMIC-IV) database was performed on critically ill patients with non-traumatic SAH. Serum AG was collected on Intensive Care Unit (ICU) admission, and ICU and hospital all-cause mortality were analyzed. The multivariate Cox proportional hazard regression model and Kaplan-Meier survival curve analysis were used to analyze the correlation of serum AG with ICU and hospital all-cause mortality. Furthermore, interaction and subgroup analyses were evaluated for the consistency of these correlations. Results A total of 893 patients with non-traumatic SAH were included in this study. The all-cause mortality in ICU and hospital were 14.8% (132/893), and 18.9% (169/893), respectively. Multivariate analysis after adjusting for potential confounders indicated that high serum AG levels (≥16 mmol/L) were associated with increased risk of ICU and hospital all-cause mortality as compared to that with low serum AG levels (<16mmol/L), (hazards ratio (HR): 2.31 [95% CI: 1.58–3.38]) and HR: 1.91 [95% CI: 1.36–2.67)], respectively). Similarly, the Kaplan–Meier (K–M) survival curve also showed that patients with high serum AG levels presented with a lower survival rate. Stratified analyses further showed that depending on the variable testes, an association between higher serum AG levels and hospital all-cause mortality in different subgroups was observed. Conclusion Among patients with non-traumatic SAH, high serum AG level at ICU admission was associated with increased ICU and hospital all-cause mortality.
Collapse
Affiliation(s)
- Changli Zhong
- Department of Clinical Laboratory, The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Min Ye
- Department of Neurology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Liyi Hu
- Department of Clinical Laboratory, The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Jiuling Liu
- Department of Neurology, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Jiuling Liu
| |
Collapse
|
42
|
Zhang A, Rastogi R, Marsh KM, Yang B, Wu D, Kron IL, Yang Z. Topical Neck Cooling Without Systemic Hypothermia Attenuates Myocardial Ischemic Injury and Post-ischemic Reperfusion Injury. Front Cardiovasc Med 2022; 9:893837. [PMID: 35837603 PMCID: PMC9274088 DOI: 10.3389/fcvm.2022.893837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Following acute myocardial infarction (MI), irreversible damage to the myocardium can only be reduced by shortening the duration between symptom onset and revascularization. While systemic hypothermia has shown promising results in slowing pre-revascularization myocardial damage, it is resource intensive and not conducive to prehospital initiation. We hypothesized that topical neck cooling (NC), an easily implemented therapy for en route transfer to definitive therapy, could similarly attenuate myocardial ischemia-reperfusion injury (IRI). Methods Using an in vivo mouse model of myocardial IRI, moderate systemic hypothermia or NC was applied following left coronary artery (LCA) occlusion and subsequent reperfusion, at early, late, and post-reperfusion intervals. Vagotomy was performed after late NC in an additional group. Hearts were harvested to measure infarct size. Results Both hypothermia treatments equally attenuated myocardial infarct size by 60% compared to control. The infarct-sparing effect of NC was temperature-dependent and timing-dependent. Vagotomy at the gastroesophageal junction abolished the infarct-sparing effect of late NC. Cardiac perfusate isolated following ischemia had significantly reduced cardiac troponin T, HMGB1, cell-free DNA, and interferon α and β levels after NC. Conclusions Topical neck cooling attenuates myocardial IRI in a vagus nerve-dependent manner, with an effect comparable to that of systemic hypothermia. NC attenuated infarct size when applied during ischemia, with earlier initiation resulting in superior infarct sparing. This novel therapy exerts a cardioprotective effect without requiring significant change in core temperature and may be a promising practical strategy to attenuate myocardial damage while patients await definitive revascularization.
Collapse
|
43
|
Neuroprotective Effects of Pharmacological Hypothermia on Hyperglycolysis and Gluconeogenesis in Rats after Ischemic Stroke. Biomolecules 2022; 12:biom12060851. [PMID: 35740974 PMCID: PMC9220898 DOI: 10.3390/biom12060851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/26/2022] Open
Abstract
Stroke is a leading threat to human life. Metabolic dysfunction of glucose may play a key role in stroke pathophysiology. Pharmacological hypothermia (PH) is a potential neuroprotective strategy for stroke, in which the temperature is decreased safely. The present study determined whether neuroprotective PH with chlorpromazine and promethazine (C + P), plus dihydrocapsaicin (DHC) improved glucose metabolism in acute ischemic stroke. A total of 208 adult male Sprague Dawley rats were randomly divided into the following groups: sham, stroke, and stroke with various treatments including C + P, DHC, C + P + DHC, phloretin (glucose transporter (GLUT)-1 inhibitor), cytochalasin B (GLUT-3 inhibitor), TZD (thiazolidinedione, phosphoenolpyruvate carboxykinase (PCK) inhibitor), and apocynin (nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor). Stroke was induced by middle cerebral artery occlusion (MCAO) for 2 h followed by 6 or 24 h of reperfusion. Rectal temperature was monitored before, during, and after PH. Infarct volume and neurological deficits were measured to assess the neuroprotective effects. Reactive oxygen species (ROS), NOX activity, lactate, apoptotic cell death, glucose, and ATP levels were measured. Protein expression of GLUT-1, GLUT-3, phosphofructokinase (PFK), lactate dehydrogenase (LDH), PCK1, PCK2, and NOX subunit gp91 was measured with Western blotting. PH with a combination of C + P and DHC induced faster, longer, and deeper hypothermia, as compared to each alone. PH significantly improved every measured outcome as compared to stroke and monotherapy. PH reduced brain infarction, neurological deficits, protein levels of glycolytic enzymes (GLUT-1, GLUT-3, PFK and LDH), gluconeogenic enzymes (PCK1 and PCK2), NOX activity and its subunit gp91, ROS, apoptotic cell death, glucose, and lactate, while raising ATP levels. In conclusion, stroke impaired glucose metabolism by enhancing hyperglycolysis and gluconeogenesis, which led to ischemic injury, all of which were reversed by PH induced by a combination of C + P and DHC.
Collapse
|
44
|
Study on the Effects of Optimized Emergency Nursing Combined with Mild Hypothermia Nursing on Neurological Prognosis, Hemodynamics, and Cytokines in Patients with Cardiac Arrest. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1787312. [PMID: 35664942 PMCID: PMC9162833 DOI: 10.1155/2022/1787312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/05/2022] [Indexed: 11/17/2022]
Abstract
Purpose To study the effects of optimized emergency nursing combined with mild hypothermia nursing on neurological prognosis, hemodynamics, and cytokines in patients with cardiac arrest (CA). Methods The medical records of 147 patients who were successfully rescued by cardiopulmonary resuscitation (CPR) after CA in our hospital were retrospectively analyzed. The 56 patients admitted in 2020 who received optimized emergency nursing were recorded as the control group; and the 91 patients admitted in 2021 who received optimized emergency nursing combined with mild hypothermia nursing were recorded as the study group. The brain function of the two groups at 72 h after return of spontaneous circulation (ROSC) was analyzed: cerebral performance category (CPC) assessment method. The neurological function of the two groups before nursing and 7, 30, and 90 d after nursing was analyzed: National Institutes of Health Stroke Scale (NISHH) score. The vital signs of the two groups after 24 h of nursing were analyzed: heart rate, spontaneous breathing rate, and blood oxygen saturation. The hemodynamic indexes of the two groups at 24 hours after nursing were analyzed: mean arterial pressure (MAP), central venous pressure (CVP), systolic blood pressure (SBP), and diastolic blood pressure (DBP). The levels of cytokines of the two groups before nursing and 7 days after nursing were analyzed: tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8). The incidence of complications and the incidence of postresuscitation syndrome (PRS) during the nursing period were compared between the two groups. Results 72 h after ROSC, the CPC results in the study group were slightly better than those in the control group, but there was no significant difference in the number of cases of CPC Grade 1, CPC Grade 2, CPC Grade 3, CPC Grade 4, and CPC Grade 5 between the two groups (P > 0.05). Before nursing, there was no statistical difference in the NISHH total score between the two groups (P > 0.05). 7, 30, and 90 d after nursing, the NISHH total score between the two groups were lower than those before nursing, and the study group's score was lower than the control group's (P < 0.05). 24 h after nursing, the heart rate and spontaneous breathing rate of the study group were lower than those of the control group (P < 0.05), and there was no significant difference in blood oxygen saturation between the two groups (P > 0.05). 24 h after nursing, there was no significant difference in MAP, CVP, SBP, and DBP between the two groups (P > 0.05). Before nursing, there was no significant difference in the levels of TNF-α, IL-6, and IL-8 between the two groups (P > 0.05). 7 d after nursing, the levels of TNF-α, IL-6, and IL-8 between the two groups were lower than those before nursing, and the levels of the study group were lower than those of the control group (P < 0.05). During the nursing period, the total complication rates of the control group and the study group were 55.36% and 34.07%, respectively, with statistical difference (P < 0.05). During the nursing period, the incidences of PRS in the control group and the study group were 12.50% and 3.30%, respectively, with significant difference (P < 0.05). Conclusion The application of optimized emergency nursing combined with mild hypothermia nursing in CA can effectively improve the neurological prognosis and inflammatory levels of patients and reduce the incidence of body complications and PRS.
Collapse
|
45
|
Wang X, Wehbe A, Kaura S, Chaudhry N, Geng X, Ding Y. Updates on Selective Brain Hypothermia: Studies From Bench Work to Clinical Trials. Front Neurol 2022; 13:899547. [PMID: 35599727 PMCID: PMC9120368 DOI: 10.3389/fneur.2022.899547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/13/2022] [Indexed: 12/01/2022] Open
Abstract
Thrombectomy or thrombolysis are the current standards of care for acute ischemic stroke (AIS), however, due to time constraints regarding operations and a multitude of contraindications, AIS remains one of the leading causes of death and chronic disability worldwide. In recent years, therapeutic hypothermia has been explored as an adjuvant therapy for AIS treatment and has shown potential to improve outcomes in patients with AIS. In particular, selective therapeutic hypothermia has shown to markedly reduce infarct volumes and have neuroprotective effects, while also minimizing many systemic side effects seen with systemic therapeutic hypothermia. Both preclinical and clinical trials have demonstrated that selective therapeutic hypothermia is a safe and feasible therapy for patients who have suffered an AIS. In this review, we summarize the current update on selective hypothermia through major studies that have been conducted in rodents, large animals, and clinical trials, and briefly discuss the prospects of selective hypothermic research. We hope this review helps facilitate the exploration of other possible adjuvant treatment modalities in the neuroprotection of ischemic stroke, whether upon symptom onset or after vascular recanalization.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Alexandra Wehbe
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Shawn Kaura
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| | - Naveed Chaudhry
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| | - Xiaokun Geng
- Department of Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
- *Correspondence: Xiaokun Geng
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
- Yuchuan Ding
| |
Collapse
|
46
|
Brookes A, Ji L, Bradshaw TD, Stocks M, Gray D, Butler J, Gershkovich P. Is Oral Lipid-Based Delivery for Drug Targeting to the Brain Feasible? Eur J Pharm Biopharm 2022; 172:112-122. [PMID: 35149190 DOI: 10.1016/j.ejpb.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/21/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
This review outlines the feasibility of oral lipid-based targeted delivery of drugs to the brain, including permeation of the central nervous system's (CNS) protective blood-brain barrier (BBB). The structure of the BBB and disruption caused by varying disease states highlights the need for disease-specific approaches to alter permeation. Disruption during disease state, and the effects of certain molecules on the barrier, demonstrate the possibility of exploiting such BBB disruption for drug delivery. Many administration methods can be used to target the brain, but oral administration is considered ideal for chronic, long-term illnesses. Several lipids that have been shown to facilitate drug delivery into the brain after systemic administration, but could also be delivered orally are discussed, including oleic acid, triolein, alkylglycerol, and conjugates of linoleic and myristic acids. Current data reveal the potential for the use of such lipids as part of oral formulations for delivery to the brain by reaching sufficient plasma levels after administration to increase the permeability of the BBB. However, gaps in the literature remain regarding the concentrations and form of most lipids required to produce the desired effects. The use of lipids via oral delivery for brain targeting has not been investigated thoroughly enough to determine with certainty if similar permeability-enhancing effects would be observed as for parenteral administration. In conclusion, further research to fill research gaps is needed, but the limited evidence suggests that oral lipid-based drug delivery for brain targeting is potentially feasible.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Liuhang Ji
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Michael Stocks
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - David Gray
- Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK, LE12 5RD
| | - James Butler
- GlaxoSmithKline Research and Development, Park Road, Ware, Hertfordshire, UK, SG12 0DP
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD.
| |
Collapse
|
47
|
Wang L, Wu L, Duan Y, Xu S, Yang Y, Yin J, Lang Y, Gao Z, Wu C, Lv Z, Shi J, Wu D, Ji X. Phenotype Shifting in Astrocytes Account for Benefits of Intra-Arterial Selective Cooling Infusion in Hypertensive Rats of Ischemic Stroke. Neurotherapeutics 2022; 19:386-398. [PMID: 35044645 PMCID: PMC9130426 DOI: 10.1007/s13311-022-01186-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 01/03/2023] Open
Abstract
The translational failure of neuroprotective therapies in stroke may be influenced by the mismatch of existing comorbidities between animal models and patients. Previous studies found that single-target neuroprotective agents reduced infarction in Sprague-Dawley but not in spontaneously hypertensive rats. It is of great interest to explore whether multi-target neuroprotectants and stroke models with comorbidities should be used in further translational researches. Ischemic stroke was induced in normotensive or hypertensive rats by 90- or 120-min middle cerebral artery occlusion (MCAO) and reperfusion. Intra-Arterial Selective Cooling Infusion (IA-SCI) was started at the onset of reperfusion for 30 minutes. Acute neurological deficits, infarct volumes, gene expression and markers of A1-like and A2-like astrocytes were evaluated. In further analysis, TNFα and IL-1α were administrated intracerebroventricularly, phenotype shifting of astrocytes and infarct volumes were assessed. Normobaric oxygen treatment, as a negative control, was also assessed in hypertensive rats. IA-SCI led to similar benefits in normotensive rats with 120-min MCAO and hypertensive rats with both 90-min and 120-min MCAO, including mitigated functional deficit and reduced infarct volumes. IA-SCI shifted astrocyte phenotypes partly by downregulating A1-like astrocytes and upregulating A2-like astrocytes in both RNA and protein levels. Upregulated A1-type astrocyte markers levels, induced by intracerebroventricular injection of TNFα and IL-1α, were closely related to increased infarct volumes in hypertensive rats, despite receiving IA-SCI treatment. In addition, infarct volumes and A1/A2-like genes were not affected by normobaric oxygen treatment. IA-SCI reduced infarction in both normotensive and hypertensive rats. Our results demonstrated the neuroprotective effects of IA-SCI in hypertensive rats may be related with phenotype shifting of astrocytes.
Collapse
Affiliation(s)
- Luling Wang
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute of Brain Disorders, Beijing, China
- Department of Emergency, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Longfei Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yunxia Duan
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yuyao Yang
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jidong Yin
- Department of Emergency, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Ye Lang
- Department of Neurology, Shengli Oilfield Central Hospital, Shandong, China
| | - Zongen Gao
- Department of Neurology, Shengli Oilfield Central Hospital, Shandong, China
| | - Chuanjie Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Zaigang Lv
- Department of Neurology, Shengli Oilfield Central Hospital, Shandong, China
| | - Jingfei Shi
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.
- Center of Stroke, Beijing Institute of Brain Disorders, Beijing, China.
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.
- Center of Stroke, Beijing Institute of Brain Disorders, Beijing, China.
| |
Collapse
|
48
|
Lee H, Yun HJ, Ding Y. Timing is everything: Exercise therapy and remote ischemic conditioning for acute ischemic stroke patients. Brain Circ 2021; 7:178-186. [PMID: 34667901 PMCID: PMC8459690 DOI: 10.4103/bc.bc_35_21] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Physical exercise is a promising rehabilitative strategy for acute ischemic stroke. Preclinical trials suggest that exercise restores cerebral blood circulation and re-establishes the blood–brain barrier’s integrity with neurological function and motor skill improvement. Clinical trials demonstrated that exercise improves prognosis and decreases complications after ischemic events. Due to these encouraging findings, early exercise rehabilitation has been quickly adopted into stroke rehabilitation guidelines. Unfortunately, preclinical trials have failed to warn us of an adverse effect. Trials with very early exercise rehabilitation (within 24 h of ischemic attack) found an inferior prognosis at 3 months. It was not immediately clear as to why exercise was detrimental when performed very early while it was ameliorative just a few short days later. This review aimed to explore the potential mechanisms of harm seen in very early exercise administered to acute ischemic stroke patients. To begin, the mechanisms of exercise’s benefit were transposed onto the current understanding of acute ischemic stroke’s pathogenesis, specifically during the acute and subacute phases. Then, exercise rehabilitation’s mechanisms were compared to that of remote ischemic conditioning (RIC). This comparison may reveal how RIC may be providing clinical benefit during the acute phase of ischemic stroke when exercise proved to be harmful.
Collapse
Affiliation(s)
- Hangil Lee
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ho Jun Yun
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Research and Development Center, John D. Dingell VA Medical Center, Detroit, Michigan, USA
| |
Collapse
|
49
|
Omileke D, Pepperall D, Bothwell SW, Mackovski N, Azarpeykan S, Beard DJ, Coupland K, Patabendige A, Spratt NJ. Ultra-Short Duration Hypothermia Prevents Intracranial Pressure Elevation Following Ischaemic Stroke in Rats. Front Neurol 2021; 12:684353. [PMID: 34616350 PMCID: PMC8488292 DOI: 10.3389/fneur.2021.684353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
There is a transient increase in intracranial pressure (ICP) 18–24 h after ischaemic stroke in rats, which is prevented by short-duration hypothermia using rapid cooling methods. Clinical trials of long-duration hypothermia have been limited by feasibility and associated complications, which may be avoided by short-duration cooling. Animal studies have cooled faster than is achievable in patients. We aimed to determine whether gradual cooling at a rate of 2°C/h to 33°C or 1°C/h to 34.5°C, with a 30 min duration at target temperatures, prevented ICP elevation and reduced infarct volume in rats. Transient middle cerebral artery occlusion was performed, followed by gradual cooling to target temperature. Hypothermia to 33°C prevented significant ICP elevation (hypothermia ΔICP = 1.56 ± 2.26 mmHg vs normothermia ΔICP = 8.93 ± 4.82 mmHg; p = 0.02) and reduced infarct volume (hypothermia = 46.4 ± 12.3 mm3 vs normothermia = 85.0 ± 17.5 mm3; p = 0.01). Hypothermia to 34.5°C did not significantly prevent ICP elevation or reduce infarct volume. We showed that gradual cooling to 33°C, at cooling rates achievable in patients, had the same ICP preventative effect as traditional rapid cooling methods. This suggests that this paradigm could be translated to prevent delayed ICP rise in stroke patients.
Collapse
Affiliation(s)
- Daniel Omileke
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Debbie Pepperall
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Steven W Bothwell
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Nikolce Mackovski
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Sara Azarpeykan
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Daniel J Beard
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Kirsten Coupland
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Adjanie Patabendige
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Neil J Spratt
- The School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton, NSW, Australia.,Department of Neurology, John Hunter Hospital, Hunter New England Local Health District, New Lambton, NSW, Australia
| |
Collapse
|
50
|
Becker A, Helmuth M, Trzeczak D, Chindo BA. Methanol extract of Ficus platyphylla decreases cerebral ischemia induced injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114219. [PMID: 34058316 DOI: 10.1016/j.jep.2021.114219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Extracts of the stem bark of Ficus paltyphylla (FP) are used in the Nigerian traditional medicine to manage psychoses, depression, epilepsy, pain, and inflammation. Our previous studies revealed that the methanol extract of FP ameliorate body core temperature. AIM OF THE STUDY A number of pharmacological agents that utilize mechanisms that enhanced neuronal survival and/or neural regeneration have been developed for the treatment of stroke. Hypothermia protects the brain from damage caused by ischemia by attenuating destructive processes such as neuroinflammation, excitotoxicity, blood-brain barrier disruption, apoptosis, and free radical formation following cerebral ischemia. In the present study, we examined the neuroprotective potential of FP on permanent occlusion of the middle cerebral artery (MCAO)-induced ischemia in mice. MATERIAL AND METHODS C57Bl mice were subjected to MCAO. FP was administered 1 h prior to and immediately after surgery. The brains were collected 24 h later and infarct volumes were measured using immune-histochemical staining, DAPI, NeuN, synaptophysin, and NR2B were quantified. RESULTS Administration of FP prior to MCAO significantly reduced infarct volume, with no effect on infarct volume immediately after MCAO. Higher numbers of cells and neurons were observed in the peri-infarct area in both groups of mice. FP-induced hypothermia protected tissue in the peri-infarct region from synaptophysin reduction. NMDA receptor 2 (NR2B) immunoreactivity is enhanced by MCAO, with no difference observed in both sham-operated and FP-induced hypothermia groups of mice. CONCLUSIONS The data suggest that FP might be useful in the reduction of ischemia-induced infarct volume when administered prior to the initiation of ischemia with no effect observed after ischemia induction.
Collapse
Affiliation(s)
- Axel Becker
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - Martin Helmuth
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Doris Trzeczak
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ben A Chindo
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Kaduna State University, Kaduna, Nigeria; Department of Pharmacology and Toxicology, National Institute for Pharmaceutical Research & Development, Abuja, Nigeria
| |
Collapse
|