1
|
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
|
2
|
Cai S, Li Q, Fan J, Zhong H, Cao L, Duan M. Therapeutic Hypothermia Combined with Hydrogen Sulfide Treatment Attenuated Early Blood-Brain Barrier Disruption and Brain Edema Induced by Cardiac Arrest and Resuscitation in Rat Model. Neurochem Res 2023; 48:967-979. [PMID: 36434369 PMCID: PMC9922226 DOI: 10.1007/s11064-022-03824-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/19/2022] [Accepted: 11/12/2022] [Indexed: 11/26/2022]
Abstract
Brain injury remains a major problem in patients suffering cardiac arrest (CA). Disruption of the blood-brain barrier (BBB) is an important factor leading to brain injury. Therapeutic hypothermia is widely accepted to limit neurological impairment. However, the efficacy is incomplete. Hydrogen sulfide (H2S), a signaling gas molecule, has protective effects after cerebral ischemia reperfusion injury. This study showed that combination of hypothermia and H2S after resuscitation was more beneficial for attenuated BBB disruption and brain edema than that of hypothermia or H2S treatment alone. CA was induced by ventricular fibrillation for 4 min. Hypothermia was performed by applying alcohol and ice bags to the body surface under anesthesia. We used sodium hydrosulphide (NaHS) as the H2S donor. We found that global brain ischemia induced by CA and cardiopulmonary resuscitation (CPR) resulted in brain edema and BBB disruption; Hypothermia or H2S treatment diminished brain edema, decreased the permeability and preserved the structure of BBB during the early period of CA and resuscitation, and more importantly, improved the neurologic function, increased the 7-day survival rate after resuscitation; the combination of hypothermia and H2S treatment was more beneficial than that of hypothermia or H2S treatment alone. The beneficial effects were associated with the inhibition of matrix metalloproteinase-9 expression, attenuated the degradation of the tight junction protein occludin, and subsequently protected the structure of BBB. These findings suggest that combined use of therapeutic hypothermia and hydrogen sulfide treatment during resuscitation of CA patients could be a potential strategy to improve clinical outcomes and survival rate.
Collapse
Affiliation(s)
- Shenquan Cai
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, No.305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Qian Li
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingjing Fan
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, No.305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Hao Zhong
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, No.305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Liangbin Cao
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, No.305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Manlin Duan
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, No.305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
3
|
Munteanu C, Rotariu M, Turnea M, Ionescu AM, Popescu C, Spinu A, Ionescu EV, Oprea C, Țucmeanu RE, Tătăranu LG, Silișteanu SC, Onose G. Main Cations and Cellular Biology of Traumatic Spinal Cord Injury. Cells 2022; 11:2503. [PMID: 36010579 PMCID: PMC9406880 DOI: 10.3390/cells11162503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023] Open
Abstract
Traumatic spinal cord injury is a life-changing condition with a significant socio-economic impact on patients, their relatives, their caregivers, and even the community. Despite considerable medical advances, there is still a lack of options for the effective treatment of these patients. The major complexity and significant disabling potential of the pathophysiology that spinal cord trauma triggers are the main factors that have led to incremental scientific research on this topic, including trying to describe the molecular and cellular mechanisms that regulate spinal cord repair and regeneration. Scientists have identified various practical approaches to promote cell growth and survival, remyelination, and neuroplasticity in this part of the central nervous system. This review focuses on specific detailed aspects of the involvement of cations in the cell biology of such pathology and on the possibility of repairing damaged spinal cord tissue. In this context, the cellular biology of sodium, potassium, lithium, calcium, and magnesium is essential for understanding the related pathophysiology and also the possibilities to counteract the harmful effects of traumatic events. Lithium, sodium, potassium-monovalent cations-and calcium and magnesium-bivalent cations-can influence many protein-protein interactions, gene transcription, ion channel functions, cellular energy processes-phosphorylation, oxidation-inflammation, etc. For data systematization and synthesis, we used the Preferred Reporting Items for Systematic Reviews and Meta-Analyzes (PRISMA) methodology, trying to make, as far as possible, some order in seeing the "big forest" instead of "trees". Although we would have expected a large number of articles to address the topic, we were still surprised to find only 51 unique articles after removing duplicates from the 207 articles initially identified. Our article integrates data on many biochemical processes influenced by cations at the molecular level to understand the real possibilities of therapeutic intervention-which must maintain a very narrow balance in cell ion concentrations. Multimolecular, multi-cellular: neuronal cells, glial cells, non-neuronal cells, but also multi-ionic interactions play an important role in the balance between neuro-degenerative pathophysiological processes and the development of effective neuroprotective strategies. This article emphasizes the need for studying cation dynamics as an important future direction.
Collapse
Affiliation(s)
- Constantin Munteanu
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
- Neuromuscular Rehabilitation Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Mariana Rotariu
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
| | - Marius Turnea
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
| | - Anca Mirela Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Cristina Popescu
- Neuromuscular Rehabilitation Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Aura Spinu
- Neuromuscular Rehabilitation Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Elena Valentina Ionescu
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
- Balneal and Rehabilitation Sanatorium of Techirghiol, 906100 Techirghiol, Romania
| | - Carmen Oprea
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
- Balneal and Rehabilitation Sanatorium of Techirghiol, 906100 Techirghiol, Romania
| | - Roxana Elena Țucmeanu
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
- Balneal and Rehabilitation Sanatorium of Techirghiol, 906100 Techirghiol, Romania
| | - Ligia Gabriela Tătăranu
- Neuromuscular Rehabilitation Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Sînziana Calina Silișteanu
- Faculty of Medicine and Biological Sciences, “Stefan cel Mare” University of Suceava, 720229 Suceava, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
4
|
Effect of Hypothermia Therapy on Children with Traumatic Brain Injury: A Meta-Analysis of Randomized Controlled Trials. Brain Sci 2022; 12:brainsci12081009. [PMID: 36009072 PMCID: PMC9406098 DOI: 10.3390/brainsci12081009] [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: 05/15/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Hypothermia therapy is a promising therapeutic strategy for traumatic brain injury (TBI); however, some trials have shown that hypothermia therapy has a negative effect on patients with TBI. The treatment of hypothermia in children with TBI remains controversial. We conducted a search of six online databases to validate the literature on comparing hypothermia with normal therapy for children with TBI. Eight randomized controlled trials (514 patients) were included. The meta-analysis indicated that hypothermia therapy may increase the Glasgow Outcome Scale (GOS) scores. However, in terms of improving the rate of complications, intracranial pressure (ICP), mortality, cerebral perfusion pressure (CPP), and length of stay both in hospital as well as pediatric ICU, the difference was not statistically significant. Hypothermia therapy may have clinical advantages in improving the GOS scores in children with TBI compared with normothermic therapy, but hypothermia therapy may have no benefit in improving the incidence of complications, ICP, mortality, CPP, and length of stay both in pediatric ICU as well as hospital. The decision to implement hypothermia therapy for children with TBI depends on the advantages and disadvantages from many aspects and these must be considered comprehensively.
Collapse
|
5
|
Lourbopoulos A, Mourouzis I, Xinaris C, Zerva N, Filippakis K, Pavlopoulos A, Pantos C. Translational Block in Stroke: A Constructive and "Out-of-the-Box" Reappraisal. Front Neurosci 2021; 15:652403. [PMID: 34054413 PMCID: PMC8160233 DOI: 10.3389/fnins.2021.652403] [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: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Why can we still not translate preclinical research to clinical treatments for acute strokes? Despite > 1000 successful preclinical studies, drugs, and concepts for acute stroke, only two have reached clinical translation. This is the translational block. Yet, we continue to routinely model strokes using almost the same concepts we have used for over 30 years. Methodological improvements and criteria from the last decade have shed some light but have not solved the problem. In this conceptual analysis, we review the current status and reappraise it by thinking "out-of-the-box" and over the edges. As such, we query why other scientific fields have also faced the same translational failures, to find common denominators. In parallel, we query how migraine, multiple sclerosis, and hypothermia in hypoxic encephalopathy have achieved significant translation successes. Should we view ischemic stroke as a "chronic, relapsing, vascular" disease, then secondary prevention strategies are also a successful translation. Finally, based on the lessons learned, we propose how stroke should be modeled, and how preclinical and clinical scientists, editors, grant reviewers, and industry should reconsider their routine way of conducting research. Translational success for stroke treatments may eventually require a bold change with solutions that are outside of the box.
Collapse
Affiliation(s)
- Athanasios Lourbopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurointensive Care Unit, Schoen Klinik Bad Aibling, Bad Aibling, Germany
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilian University, Munich, Germany
| | - Iordanis Mourouzis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christodoulos Xinaris
- IRCCS – Istituto di Ricerche Farmacologiche ‘Mario Negri’, Centro Anna Maria Astori, Bergamo, Italy
- University of Nicosia Medical School, Nicosia, Cyprus
| | - Nefeli Zerva
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Filippakis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Pavlopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Pantos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Wei H, Zhou W, Hu G, Shi C. Induction of mesenchymal stem cell‑like transformation in rat primary glial cells using hypoxia, mild hypothermia and growth factors. Mol Med Rep 2020; 23:121. [PMID: 33300053 PMCID: PMC7751450 DOI: 10.3892/mmr.2020.11760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
The transformation of rat primary glial cells into mesenchymal stem cells (MSCs) is intriguing as more seed cells can be harvested. The present study aimed to evaluate the effects of growth factors, hypoxia and mild hypothermia on the transformation of primary glial cells into MSCs. Rat primary glial cells were induced to differentiate by treatment with hypoxia, mild hypothermia and basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). Immunohistochemistry and western blotting were then used to determine the expression levels of glial fibrillary acidic protein (GFAP), nestin, musashi-1, neuron specific enolase (NSE) and neuronal nuclei (NeuN), in each treatment group. bFGF and EGF increased the proportion of CD44+ and CD105+ cells, while anaerobic mild hypothermia increased the proportion of CD90+ cells. The combination of bFGF and EGF, and anaerobic mild hypothermia increased the proportion of CD29+ cells and significantly decreased the proportions of GFAP+ cells and NSE+ cells. Treatment of primary glial cells with bFGF and EGF increased the expression levels of nestin, Musashi-1, NSE and NeuN. Anaerobic mild hypothermia increased the expression levels of Musashi-1 and decreased the expression levels of NSE and NeuN in glial cells. The results of the present study demonstrated that bFGF, EGF and anaerobic mild hypothermia treatments may promote the transformation of glial cells into MSC-like cells, and that the combination of these two treatments may have the optimal effect.
Collapse
Affiliation(s)
- Huiping Wei
- Department of Health Care for Cadres, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenyun Zhou
- Department of Prevention and Health Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guozhu Hu
- Institute of Clinical Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chunhua Shi
- Department of Rheumatology and Immunology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
7
|
Perez E, Viziano A, Al-Zaghal Z, Telischi FF, Sangaletti R, Jiang W, Dietrich WD, King C, Hoffer ME, Rajguru SM. Anatomical Correlates and Surgical Considerations for Localized Therapeutic Hypothermia Application in Cochlear Implantation Surgery. Otol Neurotol 2020; 40:1167-1177. [PMID: 31318786 PMCID: PMC6750193 DOI: 10.1097/mao.0000000000002373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Application of localized, mild therapeutic hypothermia during cochlear implantation (CI) surgery is feasible for residual hearing preservation.
Collapse
Affiliation(s)
| | - Andrea Viziano
- Department of Otolaryngology.,Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | - Weitao Jiang
- Department of Biomedical Engineering, University of Miami, Miami, Florida
| | - William Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, Florida
| | | | | | - Suhrud M Rajguru
- Department of Otolaryngology.,Department of Biomedical Engineering, University of Miami, Miami, Florida
| |
Collapse
|
8
|
Ávila-Gómez P, Vieites-Prado A, Dopico-López A, Bashir S, Fernández-Susavila H, Gubern C, Pérez-Mato M, Correa-Paz C, Iglesias-Rey R, Sobrino T, Bustamante A, Wellmann S, Montaner J, Serena J, Castillo J, Hervella P, Campos F. Cold stress protein RBM3 responds to hypothermia and is associated with good stroke outcome. Brain Commun 2020; 2:fcaa078. [PMID: 33585816 PMCID: PMC7869850 DOI: 10.1093/braincomms/fcaa078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
RNA-binding motif protein 3 is a molecular marker of hypothermia that has proved neuroprotective in neurodegenerative disease models. However, its relationship to the well-recognized therapeutic effect of hypothermia in ischaemic stroke had not been studied. In this work, the expression of RNA-binding motif protein 3 was investigated in ischaemic animal models subjected to systemic and focal brain hypothermia, specifically the effects of RNA-binding motif protein 3 silencing and overexpression on ischaemic lesions. Moreover, the association of RNA-binding motif protein 3 levels with body temperature and clinical outcome was evaluated in two independent cohorts of acute ischaemic stroke patients (n = 215); these levels were also determined in a third cohort of 31 patients derived from the phase III EuroHYP-1 trial of therapeutic cooling in ischaemic stroke. The preclinical data confirmed the increase of brain RNA-binding motif protein 3 levels in ischaemic animals subjected to systemic and focal hypothermia; this increase was selectively higher in the cooled hemisphere of animals undergoing focal brain hypothermia, thus confirming the direct effect of hypothermia on RNA-binding motif protein 3 expression, while RNA-binding motif protein 3 up-regulation in ischaemic brain regions led to functional recovery. Clinically, patients with body temperature <37.5°C in the first two cohorts had higher RNA-binding motif protein 3 values at 24 h and good outcome at 3 months post-ischaemic stroke, while RNA-binding motif protein 3 levels in the cooled third cohort tended to exceed those in placebo-treated patients. These results make RNA-binding motif protein 3 a molecular marker associated with the effect of hypothermia in ischaemic stroke and suggest its potential application as a promising protective target.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alba Vieites-Prado
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Saima Bashir
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - Héctor Fernández-Susavila
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Carme Gubern
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - María Pérez-Mato
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain.,Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Hospital La Paz Institute for Health Research (IdiPAZ), Autonomous University of Madrid, 28046, Madrid, Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Regensburg (KUNO), University of Regensburg, 93049 Regensburg, Germany
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Joaquín Serena
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| |
Collapse
|
9
|
Singh J, Barrett J, Sangaletti R, Dietrich WD, Rajguru SM. Additive Protective Effects of Delayed Mild Therapeutic Hypothermia and Antioxidants on PC12 Cells Exposed to Oxidative Stress. Ther Hypothermia Temp Manag 2020; 11:77-87. [PMID: 32302519 DOI: 10.1089/ther.2019.0034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mild therapeutic hypothermia is protective against several cellular stresses, but the mechanisms underlying this protection are not completely resolved. In the present study, we used an in vitro model to investigate whether therapeutic hypothermia at 33°C applied following a peroxide-induced oxidative stress would protect PC12 cells. A 1-hour exposure to tert-butyl peroxide increased cell death measured 24 hours later. This cell death was dose-dependent in the range of 100-1000 μM tert-butyl peroxide with ∼50% cell death observed at 24 hours from 500 μM peroxide exposure. Cell survival/death was measured with an alamarBlue viability assay, and propidium iodide/Hoechst imaging for counts of living and dead cells. Therapeutic hypothermia at 33°C applied for 2 hours postperoxide exposure significantly increased cell survival measured 24 hours postperoxide-induced stress. This protection was present even when delayed hypothermia, 15 minutes after the peroxide washout, was applied. Addition of any of the three FDA-approved antioxidants (Tempol, EUK134, Edaravone at 100 μM) in combination with hypothermia improved cell survival. With the therapeutic hypothermia treatment, a significant downregulation of caspases-3 and -8 and tumor necrosis factor-α was observed at 3 and 24 hours poststress. Consistent with this, a cell-permeable pan-caspase inhibitor Z-VAD-FMK applied in combination with hypothermia significantly increased cell survival. Overall, these results suggest that the antioxidants quenching of reactive oxygen species likely works with hypothermia to reduce mitochondrial damage and/or apoptotic mechanisms. Further studies are required to confirm and extend these results to other cell types, including neuronal cells, and other forms of oxidative stress as well as to optimize the critical parameters of hypothermia treatment such as target temperature and duration.
Collapse
Affiliation(s)
- Jayanti Singh
- Department of Otolaryngology, University of Miami, Miami, Florida, USA
| | - John Barrett
- Department of Physiology and Biophysics, University of Miami, Miami, Florida, USA
| | | | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, Florida, USA.,Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Suhrud M Rajguru
- Department of Otolaryngology, University of Miami, Miami, Florida, USA.,Department of Biomedical Engineering, University of Miami, Miami, Florida, USA
| |
Collapse
|
10
|
Scimone MT, Cramer HC, Hopkins P, Estrada JB, Franck C. Application of mild hypothermia successfully mitigates neural injury in a 3D in-vitro model of traumatic brain injury. PLoS One 2020; 15:e0229520. [PMID: 32236105 PMCID: PMC7112206 DOI: 10.1371/journal.pone.0229520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic hypothermia (TH) is an attractive target for mild traumatic brain injury (mTBI) treatment, yet significant gaps in our mechanistic understanding of TH, especially at the cellular level, remain and need to be addressed for significant forward progress to be made. Using a recently-established 3D in-vitro neural hydrogel model for mTBI we investigated the efficacy of TH after compressive impact injury and established critical treatment parameters including target cooling temperature, and time windows for application and maintenance of TH. Across four temperatures evaluated (31.5, 33, 35, and 37°C), 33°C was found to be most neuroprotective after 24 and 48 hours post-injury. Assessment of TH administration onset time and duration showed that TH should be administered within 4 hours post-injury and be maintained for at least 6 hours for achieving maximum viability. Cellular imaging showed TH reduced the percentage of cells positive for caspases 3/7 and increased the expression of calpastatin, an endogenous neuroprotectant. These findings provide significant new insight into the biological parameter space that renders TH effective in mitigating the deleterious effects of cellular mTBI and provides a quantitative foundation for the future development of animal and preclinical treatment protocols.
Collapse
Affiliation(s)
- Mark T. Scimone
- School of Engineering, Brown University, Providence, RI, United States of America
- Center for Biomedical Engineering, Brown University, Providence, RI, United States of America
| | - Harry C. Cramer
- School of Engineering, Brown University, Providence, RI, United States of America
- Center for Biomedical Engineering, Brown University, Providence, RI, United States of America
| | - Paul Hopkins
- School of Engineering, Brown University, Providence, RI, United States of America
- Center for Biomedical Engineering, Brown University, Providence, RI, United States of America
| | - Jonathan B. Estrada
- Department of Mechanical Engineering, University of Michigan—Ann Arbor, Ann Arbor, MI, United States of America
| | - Christian Franck
- Mechanical Engineering, University of Wisconsin–Madison, Madison, WI, United States of America
| |
Collapse
|
11
|
Ahn JH, Lee TK, Kim B, Lee JC, Tae HJ, Cho JH, Park Y, Shin MC, Ohk TG, Park CW, Cho JH, Hong S, Park JH, Choi SY, Won MH. Therapeutic Hypothermia Improves Hind Limb Motor Outcome and Attenuates Oxidative Stress and Neuronal Damage in the Lumbar Spinal Cord Following Cardiac Arrest. Antioxidants (Basel) 2020; 9:antiox9010038. [PMID: 31906329 PMCID: PMC7023071 DOI: 10.3390/antiox9010038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/30/2022] Open
Abstract
Hypothermia enhances outcomes of patients after resuscitation after cardiac arrest (CA). However, the underlying mechanism is not fully understood. In this study, we investigated effects of hypothermic therapy on neuronal damage/death, microglial activation, and changes of endogenous antioxidants in the anterior horn in the lumbar spinal cord in a rat model of asphyxial CA (ACA). A total of 77 adult male Sprague–Dawley rats were randomized into five groups: normal, sham ACA plus (+) normothermia, ACA + normothermia, sham ACA + hypothermia, and ACA + hypothermia. ACA was induced for 5 min by injecting vecuronium bromide. Therapeutic hypothermia was applied after return of spontaneous circulation (ROSC) via rapid cooling with isopropyl alcohol wipes, which was maintained at 33 ± 0.5 °C for 4 h. Normothermia groups were maintained at 37 ± 0.2 °C for 4 h. Neuronal protection, microgliosis, oxidative stress, and changes of endogenous antioxidants were evaluated at 12 h, 1 day, and 2 days after ROSC following ACA. ACA resulted in neuronal damage from 12 h after ROSC and evoked obvious degeneration/loss of spinal neurons in the ventral horn at 1 day after ACA, showing motor deficit of the hind limb. In addition, ACA resulted in a gradual increase in microgliosis with time after ACA. Therapeutic hypothermia significantly reduced neuronal loss and attenuated hind limb dysfunction, showing that hypothermia significantly attenuated microgliosis. Furthermore, hypothermia significantly suppressed ACA-induced increases of superoxide anion production and 8-hydroxyguanine expression, and significantly increased superoxide dismutase 1 (SOD1), SOD2, catalase, and glutathione peroxidase. Taken together, hypothermic therapy was found to have a substantial impact on changes in ACA-induced microglia activation, oxidative stress factors, and antioxidant enzymes in the ventral horn of the lumbar spinal cord, which closely correlate with neuronal protection and neurological performance after ACA.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (T.-K.L.); (B.K.); (J.-C.L.)
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (T.-K.L.); (B.K.); (J.-C.L.)
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (T.-K.L.); (B.K.); (J.-C.L.)
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Korea; (H.-J.T.); (J.H.C.)
| | - Jeong Hwi Cho
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Korea; (H.-J.T.); (J.H.C.)
| | - Yoonsoo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (Y.P.); (M.C.S.); (T.G.O.); (C.W.P.); (J.H.C.)
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (Y.P.); (M.C.S.); (T.G.O.); (C.W.P.); (J.H.C.)
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (Y.P.); (M.C.S.); (T.G.O.); (C.W.P.); (J.H.C.)
| | - Chan Woo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (Y.P.); (M.C.S.); (T.G.O.); (C.W.P.); (J.H.C.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (Y.P.); (M.C.S.); (T.G.O.); (C.W.P.); (J.H.C.)
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Korea;
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
- Correspondence: (S.Y.C.); (M.-H.W.); Tel.: +82-33-248-2112 (S.Y.C.); +82-33-250-8891 (M.-H.W.); Fax: +82-33-241-1463 (S.Y.C.); +82-33-256-1614 (M.-H.W.)
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (T.-K.L.); (B.K.); (J.-C.L.)
- Correspondence: (S.Y.C.); (M.-H.W.); Tel.: +82-33-248-2112 (S.Y.C.); +82-33-250-8891 (M.-H.W.); Fax: +82-33-241-1463 (S.Y.C.); +82-33-256-1614 (M.-H.W.)
| |
Collapse
|
12
|
Hiis HG, Cosson MV, Dahl CP, Fiane AE, Levy FO, Andersen GØ, Krobert KA. Hypothermia elongates the contraction-relaxation cycle in explanted human failing heart decreasing the time for ventricular filling during diastole. Am J Physiol Heart Circ Physiol 2018; 315:H1137-H1147. [DOI: 10.1152/ajpheart.00208.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Targeted temperature management is part of the standardized treatment for patients in cardiac arrest. Hypothermia decreases cerebral oxygen consumption and induces bradycardia; thus, increasing the heart rate may be considered to maintain cardiac output. We hypothesized that increasing heart rate during hypothermia would impair diastolic function. Human left ventricular trabeculae obtained from explanted hearts of patients with terminal heart failure were stimulated at 0.5 Hz, and contraction-relaxation cycles were recorded. Maximal developed force (Fmax), maximal rate of development of force [(dF/d t)max], time to peak force (TPF), time to 80% relaxation (TR80), and relaxation time (RT = TR80 − TPF) were measured at 37, 33, 31, and 29°C. At these temperatures, stimulation frequency was increased from 0.5 to 1.0 and to 1.5 Hz. At 1.5 Hz, concentration-response curves for the β-adrenergic receptor (β-AR) agonist isoproterenol were performed. Fmax, TPF, and RT increased when temperature was lowered, whereas (dF/d t)max decreased. At all temperatures, increasing stimulation frequency increased Fmax and (dF/d t)max, whereas TPF and RT decreased. At 31 and 29°C, resting tension increased at 1.5 Hz, which was ameliorated by β-AR stimulation. At all temperatures, maximal β-AR stimulation increased Fmax, (dF/d t)max, and maximal systolic force, whereas resting tension decreased progressively with lowering temperature. β-AR stimulation reduced TPF and RT to the same extent at all temperatures, despite the more elongated contraction-relaxation cycle at lower temperatures. Diastolic dysfunction during hypothermia results from an elongation of the contraction-relaxation cycle, which decreases the time for ventricular filling. Hypothermic bradycardia protects the heart from diastolic dysfunction and increasing the heart rate during hypothermia should be avoided. NEW & NOTEWORTHY Decreasing temperature increases the duration of the contraction-relaxation cycle in the human ventricular myocardium, significantly reducing the time for ventricular filling during diastole. During hypothermia, increasing heart rate further reduces the time for ventricular filling and in some situations increases resting tension further impairing diastolic function. Modest β-adrenergic receptor stimulation can ameliorate these potentially detrimental changes during diastole while improving contractile force generation during targeted temperature management.
Collapse
Affiliation(s)
- Halvard G. Hiis
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Marie V. Cosson
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Christen P. Dahl
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Cardiology-Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Arnt E. Fiane
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Finn Olav Levy
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Geir Ø. Andersen
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kurt A. Krobert
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Bi M, Wang J, Zhang Y, Li L, Wang L, Yao R, Duan S, Tong S, Li J. Bone mesenchymal stem cells transplantation combined with mild hypothermia improves the prognosis of cerebral ischemia in rats. PLoS One 2018; 13:e0197405. [PMID: 30067742 PMCID: PMC6070180 DOI: 10.1371/journal.pone.0197405] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/01/2018] [Indexed: 11/18/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are used as a great promising choice for the treatment of cerebral ischemia. Herein, we discuss the neuroprotective effects of the combination of BMSCs transplantation and mild hypothermia (MH) in an ischemia-reperfusion rat model. First, BMSCs were isolated using density gradient centrifugation and the adherent screening method, followed by culture, identification and labeling with DAPI. Second, adult male SD rats were divided into 5 groups: sham group (surgery without blockage of middle cerebral artery), model group (middle cerebral artery occlusion (MCAO) was established 2h prior to reperfusion), BMSCs group (injection of BMSCs via the lateral ventricle 24h after MCAO), MH group (mild hypothermia for 3h immediately after MCAO) and combination therapy group (combination of BMSCs and MH). Finally, the modified neurological severity score (mNSS) test was performed to assess behavioral function at different time points (before MCAO, before transplantation, at day 1, day 5 and day 10 after transplantation). After that, the brain was subjected to TTC staining, and the homing and angiogenesis were evaluated by immumofluorescence and immunohistochemistry. Immunofluorescence staining and Western Blot analysis were performed to calculate the percentage of the infarct area and explore glial fibrillary acidic protein (GFAP) and vascular endothelial growth factor (VEGF). Our results showed that the combination therapy significantly decreased mNSS scores (P<0.01) and reduced the percentage of the infarct area (P<0.01) than a single treatment. Moreover, the expression of GFAP and VEGF increased significantly in the combination therapy group (at day 5, day 10 after transplantation; at all time points after transplantation, respectively) compared to the single treatment groups. Taken together, it was suggested that the combination of BMSCs transplantation and MH can significantly reduce the percentage of the infarct area and improve functional recovery by promoting homing and angiogenesis, which may be a beneficial treatment for cerebral ischemia.
Collapse
Affiliation(s)
- Min Bi
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- The First Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Jiawei Wang
- Medical College of Xiamen University, Xiamen, Fujian, China
- Department of Neurology, The 184th Hospital of People’s Liberation Army of China, Yingtan, Jiangxi, China
| | - Yidan Zhang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Longzhu Li
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Linhui Wang
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Ran Yao
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Shijie Duan
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Suijun Tong
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- The First Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- * E-mail: (ST); (JL)
| | - Jianpeng Li
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- The First Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
- * E-mail: (ST); (JL)
| |
Collapse
|
14
|
Liu X, Wen S, Zhao S, Yan F, Zhao S, Wu D, Ji X. Mild Therapeutic Hypothermia Protects the Brain from Ischemia/Reperfusion Injury through Upregulation of iASPP. Aging Dis 2018; 9:401-411. [PMID: 29896428 PMCID: PMC5988595 DOI: 10.14336/ad.2017.0703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/03/2017] [Indexed: 11/16/2022] Open
Abstract
Mild therapeutic hypothermia, a robust neuroprotectant, reduces neuronal apoptosis, but the precise mechanism is not well understood. Our previous study showed that a novel inhibitor of an apoptosis-stimulating protein of p53 (iASPP) might be involved in neuronal death after stroke. The aim of this study was to confirm the role of iASPP after stroke treated with mild therapeutic hypothermia. To address this, we mimicked ischemia/reperfusion injury in vitro by using oxygen-glucose deprivation/reperfusion (OGD/R) in primary rat neurons. In our in vivo approach, we induced middle cerebral artery occlusion (MCAO) for 60 min in C57/B6 mice. From the beginning of ischemia, focal mild hypothermia was applied for two hours. To evaluate the role of iASPP, small interfering RNA (siRNA) was injected intracerebroventricularly. Our results showed that mild therapeutic hypothermia increased the expression of iASPP and decreased the expression of its targets, Puma and Bax, and an apoptosis marker, cleaved caspase-3, in primary neurons under OGD/R. Increased iASPP expression and decreased ASPP1/2 expression were observed under hypothermia treatment in MCAO mice. iASPP siRNA (iASPPi) or hypothermia plus iASPPi application increased infarct volume, apoptosis and aggravated the neurological deficits in MCAO mice. Furthermore, iASPPi downregulated iASPP expression, and upregulated the expression of proapoptotic effectors, Puma, Bax and cleaved caspase-3, in mice after stroke treated with mild therapeutic hypothermia. In conclusion, mild therapeutic hypothermia protects against ischemia/reperfusion brain injury in mice by upregulating iASPP and thus attenuating apoptosis. iASPP may be a potential target in the therapy of stroke.
Collapse
Affiliation(s)
- Xiangrong Liu
- 1China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,3Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shaohong Wen
- 1China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,3Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shunying Zhao
- 1China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Feng Yan
- 2 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,3Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shangfeng Zhao
- 4Department of Neurosurgery, Beijing Tongren Hospital, Capital University of Medical Sciences, Beijing, China
| | - Di Wu
- 1China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Xunming Ji
- 1China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,3Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.,5Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Liu X, Wu D, Wen S, Zhao S, Xia A, Li F, Ji X. Mild therapeutic hypothermia protects against cerebral ischemia/reperfusion injury by inhibiting miR-15b expression in rats. Brain Circ 2017; 3:219-226. [PMID: 30276328 PMCID: PMC6057705 DOI: 10.4103/bc.bc_15_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 07/10/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Mild hypothermia has a protective effect on ischemic stroke, but the mechanisms remain elusive. Here, we investigated microRNA (miRNA) profiles and the specific role of miRNAs in ischemic stroke treated with mild hypothermia. MATERIALS AND METHODS Male adult Sprague Dawley rats were subjected to focal transient cerebral ischemia. Mild hypothermia was induced by applying ice packs around the neck and head of the animals. miRNAs expression profiles were detected in ischemic stroke treated with mild therapeutic hypothermia through miRNA chips. Reverse transcription-polymerase chain reaction (RT-PCR) was used to verify the change of miRNA array. Western blot and adenosine triphosphate (ATP) assay kits were used to detect the changes of protein expression and ATP levels, respectively. miR-15b mimic and its control were injected into the right lateral ventricle 60 min before the induction of ischemia. RESULTS The results showed that mild hypothermia affected miRNAs profiles expression. We verified the expression of miR-15b and miR-598-3p by miRNA RT-PCR. miR-15b mimic inhibited the expression of its target, ADP ribosylation factor-like 2 (Arl2) protein, and decreased ATP levels in PC12 cells. Compared with the control, miR-15b mimic increased the infarct volume and aggravated the neurological function under normothermia or hypothermia treatment. Furthermore, the expression of Arl2 was decreased in the miR-15b mimic group under normothermia or hypothermia treatment. CONCLUSIONS Mild therapeutic hypothermia affected miRNA profiles and protected against cerebral ischemia/reperfusion by inhibiting miR-15b expression in rats. miR-15b may be a potential target for therapeutic intervention in stroke.
Collapse
Affiliation(s)
- Xiangrong Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Di Wu
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Shaohong Wen
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Shunying Zhao
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Ao Xia
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Fang Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Xunming Ji
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
| |
Collapse
|
16
|
Wowk S, Fagan KJ, Ma Y, Nichol H, Colbourne F. Examining potential side effects of therapeutic hypothermia in experimental intracerebral hemorrhage. J Cereb Blood Flow Metab 2017; 37:2975-2986. [PMID: 27899766 PMCID: PMC5536807 DOI: 10.1177/0271678x16681312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 11/16/2022]
Abstract
Studies treating intracerebral hemorrhage (ICH) with therapeutic hypothermia (TH) have shown inconsistent benefits. We hypothesized that TH's anti-inflammatory effects may be responsible as inflammatory cells are essential for removing degrading erythrocytes. Here, we subjected rats to a collagenase-induced striatal ICH followed by whole-body TH (∼33℃ for 11-72 h) or normothermia. We used X-ray fluorescence imaging to spatially quantify total and peri-hematoma iron three days post-injury. At three and seven days, we measured non-heme iron levels. Finally, hematoma volume was quantified on one, three, and seven days. In the injured hemisphere, total iron levels were elevated ( p < 0.001) with iron increasing in the peri-hematoma region ( p = 0.007). Non-heme iron increased from three to seven days (p < 0.001). TH had no effect on any measure of iron ( p ≥ 0.479). At one and three days, TH did not affect hematoma volume ( p ≥ 0.264); however, at seven days there was a four-fold increase in hematoma volume in 40% of treated animals ( p = 0.032). Thus, even when TH does not interfere with initial increases in total and non-heme iron or its containment, TH can cause re-bleeding post-treatment. This serious complication could partly account for the intermittent protection previously observed. This also raises serious concerns for clinical usage of TH for ICH.
Collapse
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kelly J Fagan
- Department of Biology, MacEwan University, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Helen Nichol
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Galgano M, Toshkezi G, Qiu X, Russell T, Chin L, Zhao LR. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant 2017; 26:1118-1130. [PMID: 28933211 PMCID: PMC5657730 DOI: 10.1177/0963689717714102] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) presents in various forms ranging from mild alterations of consciousness to an unrelenting comatose state and death. In the most severe form of TBI, the entirety of the brain is affected by a diffuse type of injury and swelling. Treatment modalities vary extensively based on the severity of the injury and range from daily cognitive therapy sessions to radical surgery such as bilateral decompressive craniectomies. Guidelines have been set forth regarding the optimal management of TBI, but they must be taken in context of the situation and cannot be used in every individual circumstance. In this review article, we have summarized the current status of treatment for TBI in both clinical practice and basic research. We have put forth a brief overview of the various subtypes of traumatic injuries, optimal medical management, and both the noninvasive and invasive monitoring modalities, in addition to the surgical interventions necessary in particular instances. We have overviewed the main achievements in searching for therapeutic strategies of TBI in basic science. We have also discussed the future direction for developing TBI treatment from an experimental perspective.
Collapse
Affiliation(s)
- Michael Galgano
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gentian Toshkezi
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| | - Thomas Russell
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Lawrence Chin
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| |
Collapse
|
18
|
Lee JH, Zhang J, Yu SP. Neuroprotective mechanisms and translational potential of therapeutic hypothermia in the treatment of ischemic stroke. Neural Regen Res 2017; 12:341-350. [PMID: 28469636 PMCID: PMC5399699 DOI: 10.4103/1673-5374.202915] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of disability and death, yet effective treatments for acute stroke has been very limited. Thus far, tissue plasminogen activator has been the only FDA-approved drug for thrombolytic treatment of ischemic stroke patients, yet its application is only applicable to less than 4–5% of stroke patients due to the narrow therapeutic window (< 4.5 hours after the onset of stroke) and the high risk of hemorrhagic transformation. Emerging evidence from basic and clinical studies has shown that therapeutic hypothermia, also known as targeted temperature management, can be a promising therapy for patients with different types of stroke. Moreover, the success in animal models using pharmacologically induced hypothermia (PIH) has gained increasing momentum for clinical translation of hypothermic therapy. This review provides an updated overview of the mechanisms and protective effects of therapeutic hypothermia, as well as the recent development and findings behind PIH treatment. It is expected that a safe and effective hypothermic therapy has a high translational potential for clinical treatment of patients with stroke and other CNS injuries.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - James Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| |
Collapse
|
19
|
Schieber AMP, Ayres JS. Thermoregulation as a disease tolerance defense strategy. Pathog Dis 2016; 74:ftw106. [PMID: 27815313 PMCID: PMC5975229 DOI: 10.1093/femspd/ftw106] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/03/2016] [Accepted: 11/02/2016] [Indexed: 12/28/2022] Open
Abstract
Physiological responses that occur during infection are most often thought of in terms of effectors of microbial destruction through the execution of resistance mechanisms, due to a direct action of the microbe, or are maladaptive consequences of host-pathogen interplay. However, an examination of the cellular and organ-level consequences of one such response, thermoregulation that leads to fever or hypothermia, reveals that these actions cannot be readily explained within the traditional paradigms of microbial killing or maladaptive consequences of host-pathogen interactions. In this review, the concept of disease tolerance is applied to thermoregulation during infection, inflammation and trauma, and we discuss the physiological consequences of thermoregulation during disease including tissue susceptibility to damage, inflammation, behavior and toxin neutralization.
Collapse
Affiliation(s)
- Alexandria M Palaferri Schieber
- The Salk Institute for Biological Studies, Immunobiology and Microbial Pathogenesis, 10010 North Torrey Pines Road, San DIego CA, USA
| | - Janelle S Ayres
- The Salk Institute for Biological Studies, Immunobiology and Microbial Pathogenesis, 10010 North Torrey Pines Road, San DIego CA, USA
| |
Collapse
|
20
|
Wang G, Zhang JN, Guo JK, Cai Y, Sun HS, Dong K, Wu CG. Neuroprotective effects of cold-inducible RNA-binding protein during mild hypothermia on traumatic brain injury. Neural Regen Res 2016; 11:771-8. [PMID: 27335561 PMCID: PMC4904468 DOI: 10.4103/1673-5374.182704] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cold-inducible RNA-binding protein (CIRP), a key regulatory protein, could be facilitated by mild hypothermia in the brain, heart and liver. This study observed the effects of mild hypothermia at 31 ± 0.5°C on traumatic brain injury in rats. Results demonstrated that mild hypothermia suppressed apoptosis in the cortex, hippocampus and hypothalamus, facilitated CIRP mRNA and protein expression in these regions, especially in the hypothalamus. The anti-apoptotic effect of mild hypothermia disappeared after CIRP silencing. There was no correlation between mitogen-activated extracellular signal-regulated kinase activation and CIRP silencing. CIRP silencing inhibited extracellular signal-regulated kinase-1/2 activation. These indicate that CIRP inhibits apoptosis by affecting extracellular signal-regulated kinase-1/2 activation, and exerts a neuroprotective effect during mild hypothermia for traumatic brain injury.
Collapse
Affiliation(s)
- Guan Wang
- Postgraduate Institution, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jian-Ning Zhang
- Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin, China
| | - Jia-Kui Guo
- Department of Neurosurgery, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying Cai
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Hong-Sheng Sun
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Kun Dong
- Department of Neurosurgery, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Cheng-Gang Wu
- Department of Neurosurgery, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
21
|
Tamames I, King C, Bas E, Dietrich WD, Telischi F, Rajguru SM. A cool approach to reducing electrode-induced trauma: Localized therapeutic hypothermia conserves residual hearing in cochlear implantation. Hear Res 2016; 339:32-9. [PMID: 27260269 DOI: 10.1016/j.heares.2016.05.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/19/2016] [Accepted: 05/26/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The trauma caused during cochlear implant insertion can lead to cell death and a loss of residual hair cells in the cochlea. Various therapeutic approaches have been studied to prevent cochlear implant-induced residual hearing loss with limited success. In the present study, we show the efficacy of mild to moderate therapeutic hypothermia of 4 to 6 °C applied to the cochlea in reducing residual hearing loss associated with the electrode insertion trauma. APPROACH Rats were randomly distributed in three groups: control contralateral cochleae, normothermic implanted cochleae and hypothermic implanted cochleae. Localized hypothermia was delivered to the middle turn of the cochlea for 20 min before and after implantation using a custom-designed probe perfused with cooled fluorocarbon. Auditory brainstem responses (ABRs) were recorded to assess the hearing function prior to and post-cochlear implantation at various time points up to 30 days. At the conclusion of the trials, inner ears were harvested for histology and cell count. The approach was extended to cadaver temporal bones to study the potential surgical approach and efficacy of our device. In this case, the hypothermia probe was placed next to the round window niche via the facial recess or a myringotomy. MAIN RESULTS A significant loss of residual hearing was observed in the normothermic implant group. Comparatively, the residual hearing in the cochleae receiving therapeutic hypothermia was significantly conserved. Histology confirmed a significant loss of outer hair cells in normothermic cochleae receiving the surgical trauma when compared to the hypothermia treated group. In human temporal bones, a controlled and effective cooling of the cochlea was achieved using our approach. SIGNIFICANCE Collectively, these results suggest that therapeutic hypothermia during cochlear implantation may reduce traumatic effects of electrode insertion and improve conservation of residual hearing.
Collapse
Affiliation(s)
- Ilmar Tamames
- Department of Biomedical Engineering, Seattle, WA, USA
| | | | - Esperanza Bas
- Department of Otolaryngology, University of Miami, Miami, FL, 33136, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, University of Miami, Miami, FL, 33136, USA
| | - Fred Telischi
- Department of Otolaryngology, University of Miami, Miami, FL, 33136, USA
| | - Suhrud M Rajguru
- Department of Biomedical Engineering, Seattle, WA, USA; Department of Otolaryngology, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
22
|
LaRocca TJ, Sosunov SA, Shakerley NL, Ten VS, Ratner AJ. Hyperglycemic Conditions Prime Cells for RIP1-dependent Necroptosis. J Biol Chem 2016; 291:13753-61. [PMID: 27129772 DOI: 10.1074/jbc.m116.716027] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Indexed: 12/27/2022] Open
Abstract
Necroptosis is a RIP1-dependent programmed cell death (PCD) pathway that is distinct from apoptosis. Downstream effector pathways of necroptosis include formation of advanced glycation end products (AGEs) and reactive oxygen species (ROS), both of which depend on glycolysis. This suggests that increased cellular glucose may prime necroptosis. Here we show that exposure to hyperglycemic levels of glucose enhances necroptosis in primary red blood cells (RBCs), Jurkat T cells, and U937 monocytes. Pharmacologic or siRNA inhibition of RIP1 prevented the enhanced death, confirming it as RIP1-dependent necroptosis. Hyperglycemic enhancement of necroptosis depends upon glycolysis with AGEs and ROS playing a role. Total levels of RIP1, RIP3, and mixed lineage kinase domain-like (MLKL) proteins were increased following treatment with high levels of glucose in Jurkat and U937 cells and was not due to transcriptional regulation. The observed increase in RIP1, RIP3, and MLKL protein levels suggests a potential positive feedback mechanism in nucleated cell types. Enhanced PCD due to hyperglycemia was specific to necroptosis as extrinsic apoptosis was inhibited by exposure to high levels of glucose. Hyperglycemia resulted in increased infarct size in a mouse model of brain hypoxia-ischemia injury. The increased infarct size was prevented by treatment with nec-1s, strongly suggesting that increased necroptosis accounts for exacerbation of this injury in conditions of hyperglycemia. This work reveals that hyperglycemia represents a condition in which cells are extraordinarily susceptible to necroptosis, that local glucose levels alter the balance of PCD pathways, and that clinically relevant outcomes may depend on glucose-mediated effects on PCD.
Collapse
Affiliation(s)
- Timothy J LaRocca
- From the Department of Basic and Social Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208
| | - Sergey A Sosunov
- the Department of Pediatrics, Columbia University, New York, New York 10032, and
| | - Nicole L Shakerley
- From the Department of Basic and Social Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208
| | - Vadim S Ten
- the Department of Pediatrics, Columbia University, New York, New York 10032, and
| | - Adam J Ratner
- the Department of Pediatrics, Columbia University, New York, New York 10032, and the Departments of Pediatrics and Microbiology, New York University, New York, New York 10016
| |
Collapse
|
23
|
Pramipexole-Induced Hypothermia Reduces Early Brain Injury via PI3K/AKT/GSK3β pathway in Subarachnoid Hemorrhage rats. Sci Rep 2016; 6:23817. [PMID: 27026509 PMCID: PMC4812308 DOI: 10.1038/srep23817] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/15/2016] [Indexed: 12/31/2022] Open
Abstract
Previous studies have shown neuroprotective effects of hypothermia. However, its effects on subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) remain unclear. In this study, a SAH rat model was employed to study the effects and mechanisms of pramipexole-induced hypothermia on EBI after SAH. Dose-response experiments were performed to select the appropriate pramipexole concentration and frequency of administration for induction of mild hypothermia (33–36 °C). Western blot, neurobehavioral evaluation, Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and Fluoro-Jade B (FJB) staining were used to detect the effects of pramipexole-induced hypothermia on SAH-induced EBI, as well as to study whether controlled rewarming could attenuate these effects. Inhibitors targeting the PI3K/AKT/GSK3β pathway were administered to determine whether the neuroprotective effect of pramipexole-induced hypothermia was mediated by PI3K/AKT/GSK3β signaling pathway. The results showed that intraperitoneal injection of pramipexole at 0.25 mg/kg body weight once per 8 hours was found to successfully and safely maintain rats at mild hypothermia. Pramipexole-induced hypothermia ameliorated SAH-induced brain cell death, blood-brain barrier damage and neurobehavioral deficits in a PI3K/AKT/GSK3β signaling-dependent manner. Therefore, we may conclude that pramipexole-induced hypothermia could effectively inhibit EBI after SAH in rats via PI3K/AKT/GSK3β signaling pathway.
Collapse
|
24
|
Sun HT, Zheng M, Wang Y, Diao Y, Zhao W, Wei Z. Monitoring intracranial pressure utilizing a novel pattern of brain multiparameters in the treatment of severe traumatic brain injury. Neuropsychiatr Dis Treat 2016; 12:1517-23. [PMID: 27382294 PMCID: PMC4922802 DOI: 10.2147/ndt.s106915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The aim of the study was to evaluate the clinical value of multiple brain parameters on monitoring intracranial pressure (ICP) procedures in the therapy of severe traumatic brain injury (sTBI) utilizing mild hypothermia treatment (MHT) alone or a combination strategy with other therapeutic techniques. A total of 62 patients with sTBI (Glasgow Coma Scale score <8) were treated using mild hypothermia alone or mild hypothermia combined with conventional ICP procedures such as dehydration using mannitol, hyperventilation, and decompressive craniectomy. The multiple brain parameters, which included ICP, cerebral perfusion pressure, transcranial Doppler, brain tissue partial pressure of oxygen, and jugular venous oxygen saturation, were detected and analyzed. All of these measures can control the ICP of sTBI patients to a certain extent, but multiparameters associated with brain environment and functions have to be critically monitored simultaneously because some procedures of reducing ICP can cause side effects for long-term recovery in sTBI patients. The result suggested that multimodality monitoring must be performed during the process of mild hypothermia combined with conventional ICP procedures in order to safely target different clinical methods to specific patients who may benefit from an individual therapy.
Collapse
Affiliation(s)
- Hong-Tao Sun
- Sixth Department of Neurosurgery, Affiliated Hospital of Logistics University of People's Armed Police Force, Tianjin
| | - Maohua Zheng
- Department of Neurosurgery, The First Hospital of Lanzhou University, Lanzhou, People's Republic of China
| | - Yanmin Wang
- Sixth Department of Neurosurgery, Affiliated Hospital of Logistics University of People's Armed Police Force, Tianjin
| | - Yunfeng Diao
- Sixth Department of Neurosurgery, Affiliated Hospital of Logistics University of People's Armed Police Force, Tianjin
| | - Wanyong Zhao
- Sixth Department of Neurosurgery, Affiliated Hospital of Logistics University of People's Armed Police Force, Tianjin
| | - Zhengjun Wei
- Sixth Department of Neurosurgery, Affiliated Hospital of Logistics University of People's Armed Police Force, Tianjin
| |
Collapse
|
25
|
Niquet J, Gezalian M, Baldwin R, Wasterlain CG. Neuroprotective effects of deep hypothermia in refractory status epilepticus. Ann Clin Transl Neurol 2015; 2:1105-15. [PMID: 26734661 PMCID: PMC4693587 DOI: 10.1002/acn3.262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/02/2015] [Indexed: 11/09/2022] Open
Abstract
Objective Pharmacoresistance develops quickly during repetitive seizures, and refractory status epilepticus (RSE) remains a therapeutic challenge. The outcome of RSE is poor, with high mortality and morbidity. New treatments are needed. Deep hypothermia (20°C) is used clinically during reconstructive cardiac surgery and neurosurgery, and has proved safe and effective in those indications. We tested the hypothesis that deep hypothermia reduces RSE and its long‐term consequences. Methods We used a model of SE induced by lithium and pilocarpine and refractory to midazolam. Several EEG measures were recorded in both hypothermic (n = 17) and normothermic (n = 20) animals. Neuronal injury (by Fluoro‐Jade B), cell‐mediated inflammation, and breakdown of the blood–brain barrier (BBB) (by immunohistochemistry) were studied 48 h following SE onset. Results Normothermic rats in RSE seized for 4.1 ± 1.1 h, and at 48 h they displayed extensive neuronal injury in many brain regions, including hippocampus, dentate gyrus, amygdala, entorhinal and pyriform cortices, thalamus, caudate/putamen, and the frontoparietal neocortex. Deep hypothermia (20°C) of 30 min duration terminated RSE within 12 min of initiation of hypothermia, reduced EEG power and seizure activity upon rewarming, and eliminated SE‐induced neuronal injury in most animals. Normothermic rats showed widespread breakdown of the BBB, and extensive macrophage infiltration in areas of neuronal injury, which were completely absent in animals treated with hypothermia. Interpretation These results suggest that deep hypothermia may open a new therapeutic avenue for the treatment of RSE and for the prevention of its long‐term consequences.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology David Geffen School of Medicine at UCLA Los Angeles California; Epilepsy Research Laboratory (151)Veterans Affairs Greater Los Angeles Healthcare System Los Angeles California
| | - Michael Gezalian
- Epilepsy Research Laboratory (151) Veterans Affairs Greater Los Angeles Healthcare System Los Angeles California
| | - Roger Baldwin
- Epilepsy Research Laboratory (151) Veterans Affairs Greater Los Angeles Healthcare System Los Angeles California
| | - Claude G Wasterlain
- Department of Neurology David Geffen School of Medicine at UCLA Los Angeles California; Epilepsy Research Laboratory (151) Veterans Affairs Greater Los Angeles Healthcare System Los Angeles California; Brain Research Institute David Geffen School of Medicine at UCLA Los Angeles California
| |
Collapse
|
26
|
Abstract
In a rat model of status epilepticus (SE) induced by lithium and pilocarpine and refractory to midazolam, deep hypothermia (20 °C for 30 min) reduced EEG power over 50-fold, stopped SE within 12 min, and reduced EEG spikes by 87%. Hypothermia deserves further investigation as a treatment of last resort for refractory SE. This article is part of a Special Issue entitled "Status Epilepticus".
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Roger Baldwin
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Michael Gezalian
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Claude G. Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA,Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
27
|
5'-adenosine monophosphate-induced hypothermia attenuates brain ischemia/reperfusion injury in a rat model by inhibiting the inflammatory response. Mediators Inflamm 2015; 2015:520745. [PMID: 25873763 PMCID: PMC4385688 DOI: 10.1155/2015/520745] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/03/2014] [Accepted: 10/22/2014] [Indexed: 12/24/2022] Open
Abstract
Hypothermia treatment is a promising therapeutic strategy for brain injury. We previously demonstrated that 5′-adenosine monophosphate (5′-AMP), a ribonucleic acid nucleotide, produces reversible deep hypothermia in rats when the ambient temperature is appropriately controlled. Thus, we hypothesized that 5′-AMP-induced hypothermia (AIH) may attenuate brain ischemia/reperfusion injury. Transient cerebral ischemia was induced by using the middle cerebral artery occlusion (MCAO) model in rats. Rats that underwent AIH treatment exhibited a significant reduction in neutrophil elastase infiltration into neuronal cells and matrix metalloproteinase 9 (MMP-9), interleukin-1 receptor (IL-1R), tumor necrosis factor receptor (TNFR), and Toll-like receptor (TLR) protein expression in the infarcted area compared to euthermic controls. AIH treatment also decreased the number of terminal deoxynucleotidyl transferase dUTP nick end labeling- (TUNEL-) positive neuronal cells. The overall infarct volume was significantly smaller in AIH-treated rats, and neurological function was improved. By contrast, rats with ischemic brain injury that were administered 5′-AMP without inducing hypothermia had ischemia/reperfusion injuries similar to those in euthermic controls. Thus, the neuroprotective effects of AIH were primarily related to hypothermia.
Collapse
|
28
|
Endocannabinoid Catabolic Enzymes Play Differential Roles in Thermal Homeostasis in Response to Environmental or Immune Challenge. J Neuroimmune Pharmacol 2015; 10:364-70. [PMID: 25715681 DOI: 10.1007/s11481-015-9593-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/11/2015] [Indexed: 01/12/2023]
Abstract
Cannabinoid receptor agonists, such as Δ(9)-THC, the primary active constituent of Cannabis sativa, have anti-pyrogenic effects in a variety of assays. Recently, attention has turned to the endogenous cannabinoid system and how endocannabinoids, including 2-arachidonoylglycerol (2-AG) and anandamide, regulate multiple homeostatic processes, including thermoregulation. Inhibiting endocannabinoid catabolic enzymes, monoacylglycerol lipase (MAGL) or fatty acid amide hydrolase (FAAH), elevates levels of 2-AG or anandamide in vivo, respectively. The purpose of this experiment was to test the hypothesis that endocannabinoid catabolic enzymes function to maintain thermal homeostasis in response to hypothermic challenge. In separate experiments, male C57BL/6J mice were administered a MAGL or FAAH inhibitor, and then challenged with the bacterial endotoxin lipopolysaccharide (LPS; 2 mg/kg ip) or a cold (4 °C) ambient environment. Systemic LPS administration caused a significant decrease in core body temperature after 6 h, and this hypothermia persisted for at least 12 h. Similarly, cold environment induced mild hypothermia that resolved within 30 min. JZL184 exacerbated hypothermia induced by either LPS or cold challenge, both of which effects were blocked by rimonabant, but not SR144528, indicating a CB1 cannabinoid receptor mechanism of action. In contrast, the FAAH inhibitor, PF-3845, had no effect on either LPS-induced or cold-induced hypothermia. These data indicate that unlike direct acting cannabinoid receptor agonists, which elicit profound hypothermic responses on their own, neither MAGL nor FAAH inhibitors affect normal body temperature. However, these endocannabinoid catabolic enzymes play distinct roles in thermoregulation following hypothermic challenges.
Collapse
|
29
|
Kowalik R, Szczerba E, Kołtowski Ł, Grabowski M, Chojnacka K, Golecki W, Hołubek A, Opolski G. Cardiac arrest survivors treated with or without mild therapeutic hypothermia: performance status and quality of life assessment. Scand J Trauma Resusc Emerg Med 2014; 22:76. [PMID: 25496708 PMCID: PMC4273459 DOI: 10.1186/s13049-014-0076-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 12/01/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy is the main determinant of clinical outcome after cardiac arrest. The study was designed to determine long-term neurological and psychological status in cardiac arrest survivors, as well as to compare neuropsychological outcomes between patients treated with mild therapeutic hypothermia (MTH) and patients who did not undergo hypothermia treatment. METHODS The article describes a single-center, retrospective, observational study on 28 post-cardiac arrest adult patients treated in the cardiac intensive care unit who qualified for MTH vs. 37 control group patients, hospitalized at the same center following cardiac arrest in the preceding years and fulfilling criteria for induced hypothermia, but who were not treated due to unavailability of the method at that time. Disability Rating Scale (DRS), Barthel Index and RAND-36 were used to assess performance status and quality of life in both study groups after hospital discharge. RESULTS There were no statistically significant differences in physical functioning found between groups either at the end of hospital treatment or at long-term follow-up (DRS: p = 0.11; Barthel Index: p = 0.83). In long-term follow-up, MTH patients showed higher vitality (p = 0.02) and reported fewer complaints on role limitations due to emotional problems (p = 0.04) compared to the control group. No significant differences were shown between study groups in terms of physical capacity and independent functioning. CONCLUSION To conclude, in long-term follow-up, MTH patients showed higher vitality and reported fewer complaints on role limitations due to emotional problems compared to the control group. This suggest that MTH helps to preserve global brain function in cardiac arrest survivors. However, the results can be biased by a small sample size and variable observation periods.
Collapse
|