1
|
Kofke WA, Miano TA. Failed Neuroprotection Trials: An Evaluation of Complexity and Clinical Trial Design. Anesthesiology 2025:00000542-990000000-00624. [PMID: 39813404 DOI: 10.1097/aln.0000000000005244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Affiliation(s)
- W Andrew Kofke
- Department of Anesthesiology and Critical Care, Neuroanesthesia and Critical Care Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd A Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Suerte ACC, Liddle LJ, Abrahart A, Khiabani E, Colbourne F. A Systematic Review and Meta-Analysis of Therapeutic Hypothermia and Pharmacological Cotherapies in Animal Models of Ischemic Stroke. Ther Hypothermia Temp Manag 2024; 14:229-242. [PMID: 38946643 PMCID: PMC11685787 DOI: 10.1089/ther.2024.0012] [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: 07/02/2024] Open
Abstract
Therapeutic hypothermia (TH) lessens ischemic brain injury. Cytoprotective agents can augment protection, although it is unclear which combinations are most effective. The objective of this study is to identify which cytoprotective drug works best with delayed TH. Following PRISMA guidelines, a systematic review (PubMed, Web of Science, MEDLINE, Scopus) identified controlled experiments that used an in vivo focal ischemic stroke model and evaluated the efficacy of TH (delay of ≥1 hour) coupled with cytoprotective agents. This combination was our main intervention compared with single treatments with TH, drug, or no treatment. Endpoints were brain injury and neurological impairment. The CAMARADES checklist for study quality and the SYRCLE's risk of bias tool gauged study quality. Twenty-five studies were included. Most used young, healthy male rats, with only one using spontaneously hypertensive rats. Two studies used mice models, and six used adult animals. Study quality was moderate (median score = 6), and risk of bias was high. Pharmacological agents provided an additive effect on TH for all outcomes measured. Magnesium coupled with TH had the greatest impact compared with other agent-TH combinations on all outcomes. Longer TH durations improved both behavioral and histological outcomes and had greater cytoprotective efficacy than shorter durations. Anti-inflammatories were the most effective in reducing infarction (standardized mean difference [SMD]: -1.64, confidence interval [CI]: [-2.13, -1.15]), sulfonylureas reduced edema the most (SMD: -2.32, CI: [-3.09, -1.54]), and antiapoptotic agents improved behavioral outcomes the most (normalized mean difference: 52.38, CI: [45.29, 59.46]). Statistically significant heterogeneity was observed (I2 = 82 - 98%, all p < 0.001), indicating that studies wildly differ in their effect size estimates. Our results support the superiority of adding cytoprotective therapies with TH (vs. individual or no therapy). Additional exploratory and confirmatory studies are required to identify and thoroughly assess combination therapies owing to limited work and inconsistent translational quality.
Collapse
Affiliation(s)
| | - Lane J. Liddle
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, Canada
| | - Ashley Abrahart
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, Canada
| | - Elmira Khiabani
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
3
|
Pawluk H, Tafelska-Kaczmarek A, Sopońska M, Porzych M, Modrzejewska M, Pawluk M, Kurhaluk N, Tkaczenko H, Kołodziejska R. The Influence of Oxidative Stress Markers in Patients with Ischemic Stroke. Biomolecules 2024; 14:1130. [PMID: 39334896 PMCID: PMC11430825 DOI: 10.3390/biom14091130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke is the second leading cause of death worldwide, and its incidence is rising rapidly. Acute ischemic stroke is a subtype of stroke that accounts for the majority of stroke cases and has a high mortality rate. An effective treatment for stroke is to minimize damage to the brain's neural tissue by restoring blood flow to decreased perfusion areas of the brain. Many reports have concluded that both oxidative stress and excitotoxicity are the main pathological processes associated with ischemic stroke. Current measures to protect the brain against serious damage caused by stroke are insufficient. For this reason, it is important to investigate oxidative and antioxidant strategies to reduce oxidative damage. This review focuses on studies assessing the concentration of oxidative stress biomarkers and the level of antioxidants (enzymatic and non-enzymatic) and their impact on the clinical prognosis of patients after stroke. Mechanisms related to the production of ROS/RNS and the role of oxidative stress in the pathogenesis of ischemic stroke are presented, as well as new therapeutic strategies aimed at reducing the effects of ischemia and reperfusion.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Agnieszka Tafelska-Kaczmarek
- Department of Organic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100 Torun, Poland
| | - Małgorzata Sopońska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Marta Porzych
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
4
|
Gusev EI, Martynov MY. [Stroke: current state of the problem]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:7-18. [PMID: 39690546 DOI: 10.17116/jnevro20241241117] [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: 12/19/2024]
Abstract
We review the current and emerging topics in ischemic stroke (IS) and intracerebral hemorrhage (ICH). We discuss the association of genetic predisposition and healthy lifestyle, ambient particulate air pollution, weather parameters, variations in the anatomy of cerebral blood vessels, psychological stress, depression, insonmia with the development of IS or ICH. Also, the role of oral anticoagulants (AC) as a new risk factor for ICH is presented. The issues of pathophysiology of IS and ICH are considered, in particular, the discrepancy between blood flow and metabolism and penumbra stability in IS and changes in perfusion and cerebral blood flow (CBF) in stroke, autoimmune mechanisms of brain damage. Widening of therapeutic window, introduction of new generation of thrombolytic medications, tele-thrombolysis and thrombolysis in mobile stroke units are discussed. In patients taking AC, the tactics of prescribing IIa and Xa factor blockers during telethrombolysis and thrombolysis are discussed. The issues of urgent correction of hemostasis in the development of ICH while taking AC, as well as the resumption of their use in the future, are considered. The issues of neuroprotection, including the combination of neuroprotection and telethrombolysis and thrombolysis, and the translation of experimental studies into clinical practice, as well as the implementation of immunomodulatory therapy are discussed.
Collapse
Affiliation(s)
- E I Gusev
- Pirogov Russian National Research Medical University (Pirogov University), Moscow, Russia
| | - M Yu Martynov
- Pirogov Russian National Research Medical University (Pirogov University), Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
5
|
Martynov MY, Zhuravleva MV, Vasyukova NS, Kuznetsova EV, Kameneva TR. [Oxidative stress in the pathogenesis of stroke and its correction]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:16-27. [PMID: 36719115 DOI: 10.17116/jnevro202312301116] [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: 02/01/2023]
Abstract
We reviewed the role of oxidative stress (OS) in the pathogenesis of ischemic (IS) and hemorrhagic stroke (HS). OS plays a major role in programmed cell death, increased permeability of the blood-brain barrier, astroglial and microglial activation, and local inflammatory response. We also reviewed the current state of neuro- and cytoprotection studies and their translation in clinical practice. With respect to experimental and clinical data the efficacy of long term administration of multimodal cytoprotective drug with antioxidant effect - ethylmethylhydroxypyridine succinate (Mexidol) is discussed during the acute and early recovery period after stroke.
Collapse
Affiliation(s)
- M Yu Martynov
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - M V Zhuravleva
- Research Center for Examination of Medical Devices, Moscow, Russia.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - N S Vasyukova
- Skriabin and Kovalenko Institute of Experimental Veterinary Medicine, Moscow, Russia
| | - E V Kuznetsova
- Research Institute for Healthcare and Medical Management, Moscow, Russia
| | - T R Kameneva
- Konchalovsky City Clinical Hospital, Moscow, Russia
| |
Collapse
|
6
|
Li Z, Bi R, Sun S, Chen S, Chen J, Hu B, Jin H. The Role of Oxidative Stress in Acute Ischemic Stroke-Related Thrombosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8418820. [PMID: 36439687 PMCID: PMC9683973 DOI: 10.1155/2022/8418820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/13/2022] [Accepted: 11/02/2022] [Indexed: 09/22/2023]
Abstract
Acute ischemic stroke is a serious life-threatening disease that affects almost 600 million people each year throughout the world with a mortality of more than 10%, while two-thirds of survivors remain disabled. However, the available treatments for ischemic stroke are still limited to thrombolysis and/or mechanical thrombectomy, and there is an urgent need for developing new therapeutic target. Recently, intravascular oxidative stress, derived from endothelial cells, platelets, and leukocytes, has been found to be tightly associated with stroke-related thrombosis. It not only promotes primary thrombus formation by damaging endothelial cells and platelets but also affects thrombus maturation and stability by modifying fibrin components. Thus, oxidative stress is expected to be a novel target for the prevention and treatment of ischemic stroke. In this review, we first discuss the mechanisms by which oxidative stress promotes stroke-related thrombosis, then summarize the oxidative stress biomarkers of stroke-related thrombosis, and finally put forward an antithrombotic therapy targeting oxidative stress in ischemic stroke.
Collapse
Affiliation(s)
- Zhifang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Jurcau A, Ardelean AI. Oxidative Stress in Ischemia/Reperfusion Injuries following Acute Ischemic Stroke. Biomedicines 2022; 10:biomedicines10030574. [PMID: 35327376 PMCID: PMC8945353 DOI: 10.3390/biomedicines10030574] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Recanalization therapy is increasingly used in the treatment of acute ischemic stroke. However, in about one third of these patients, recanalization is followed by ischemia/reperfusion injuries, and clinically to worsening of the neurological status. Much research has focused on unraveling the involved mechanisms in order to prevent or efficiently treat these injuries. What we know so far is that oxidative stress and mitochondrial dysfunction are significantly involved in the pathogenesis of ischemia/reperfusion injury. However, despite promising results obtained in experimental research, clinical studies trying to interfere with the oxidative pathways have mostly failed. The current article discusses the main mechanisms leading to ischemia/reperfusion injuries, such as mitochondrial dysfunction, excitotoxicity, and oxidative stress, and reviews the clinical trials with antioxidant molecules highlighting recent developments and future strategies.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Neurology, Clinical Municipal Hospital Oradea, Louis Pasteur Street nr 26, 410054 Oradea, Romania
- Correspondence: ; Tel.: +40-744-600-833
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Universitatii Street nr 1, 410087 Oradea, Romania;
- Department of Cardiology, Clinical Emergency County Hospital Oradea, Gh. Doja Street nr 65, 410169 Oradea, Romania
| |
Collapse
|
8
|
Oxidative Stress in the Brain: Basic Concepts and Treatment Strategies in Stroke. Antioxidants (Basel) 2021; 10:antiox10121886. [PMID: 34942989 PMCID: PMC8698986 DOI: 10.3390/antiox10121886] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022] Open
Abstract
The production of free radicals is inevitably associated with metabolism and other enzymatic processes. Under physiological conditions, however, free radicals are effectively eliminated by numerous antioxidant mechanisms. Oxidative stress occurs due to an imbalance between the production and elimination of free radicals under pathological conditions. Oxidative stress is also associated with ageing. The brain is prone to oxidative damage because of its high metabolic activity and high vulnerability to ischemic damage. Oxidative stress, thus, plays a major role in the pathophysiology of both acute and chronic pathologies in the brain, such as stroke, traumatic brain injury or neurodegenerative diseases. The goal of this article is to summarize the basic concepts of oxidative stress and its significance in brain pathologies, as well as to discuss treatment strategies for dealing with oxidative stress in stroke.
Collapse
|
9
|
Wang Z, He C, Shi JS. Natural Products for the Treatment of Neurodegenerative Diseases. Curr Med Chem 2020; 27:5790-5828. [PMID: 31131744 DOI: 10.2174/0929867326666190527120614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/06/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders characterized by the progressive degeneration of the structure and function of the central nervous system or peripheral nervous system. Alzheimer's Disease (AD), Parkinson's Disease (PD) and Spinal Cord Injury (SCI) are the common neurodegenerative diseases, which typically occur in people over the age of 60. With the rapid development of an aged society, over 60 million people worldwide are suffering from these uncurable diseases. Therefore, the search for new drugs and therapeutic methods has become an increasingly important research topic. Natural products especially those from the Traditional Chinese Medicines (TCMs), are the most important sources of drugs, and have received extensive interest among pharmacist. In this review, in order to facilitate further chemical modification of those useful natural products by pharmacists, we will bring together recent studies in single natural compound from TCMs with neuroprotective effect.
Collapse
Affiliation(s)
- Ze Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China.,Generic Drug Research Center of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Chunyang He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China.,Generic Drug Research Center of Guizhou Province, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Guizhou 563003, China
| |
Collapse
|
10
|
Zhang Y, Li H, Guo H, Li B, Zhao Z, Wang P, Wu H, Liu J, Chen Y, Zhang X, Wu P, Wang Z, Wang J. Genome Analysis Reveals a Synergistic Mechanism of Ursodeoxycholic Acid and Jasminoidin in Mice Brain Repair After Ischemia/Reperfusion: Crosstalk Among Muti-Pathways. Front Pharmacol 2019; 10:1383. [PMID: 31920636 PMCID: PMC6920098 DOI: 10.3389/fphar.2019.01383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 10/30/2019] [Indexed: 01/19/2023] Open
Abstract
Studies have shown that combination drug therapy which corresponding treatment involves multiple genes and targets is more effective against cerebral ischemia. To identify the synergistic mechanism of ursodeoxycholic acid and jasminoidin based on differential pathway network, which protect against brain ischemia-reperfusion injury. Totally 115 mice with focal cerebral ischemia-reperfusion injury were allocated into five groups: sham, vehicle, ursodeoxycholic acid (UA), jasminoidin (JA), and JA and UA combination group (JU). The differentially expressed genes identified by microarray which consisted of 11,644 complementary DNAs were loaded to the GeneGo MetaCore™ software to analyze the enriched pathways and processes among different groups. Of the top 10 pathways and process networks, 5, 6, and 3 overlapping pathways as well as 5, 3, and 4 overlapping process networks were observed between UA and JA, UA and JU, and JA and JU, respectively. Of these, three pathways and three process networks overlapped across the three groups. Interestingly, four representative pathways and six process networks were only noted in the JU group. Gene Ontology process analysis showed 2 processes were shared by all three treatment groups in the top 10 processes. The UA and JA combination resulted in synergistic effects through affecting multi-signal transduction pathways, different locations in the same pathway, and the new signaling pathway emerged in drug combination group, those together may enhance the treatment of cerebral ischemia-reperfusion injury through promoting neural cell apoptosis, decreasing calcium levels, inhibiting inflammation, and protecting neurons.
Collapse
Affiliation(s)
- Yingying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haixia Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huan Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bing Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zide Zhao
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongli Wu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinying Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxu Zhang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Li F, Liu WC, Wang Q, Sun Y, Wang H, Jin X. NG2-glia cell proliferation and differentiation by glial growth factor 2 (GGF2), a strategy to promote functional recovery after ischemic stroke. Biochem Pharmacol 2019; 171:113720. [PMID: 31751533 DOI: 10.1016/j.bcp.2019.113720] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022]
Abstract
Stroke is the leading cause of adult disability. Spontaneous functional recovery occurs after ischemic stroke, but it is very limited. Therefore, it is urgent to find a strategy to promote functional recovery after stroke in clinical setting. Gray matter damage has received extensive attention owing to the important roles of the gray matter in synaptic plasticity, cognitive, and motor function. However, stroke also causes white matter damage, which accounts for half of the infarct volume and can be aggravated by blood brain barrier damage. Disruption of white matter integrity, which is characterized by death of oligodendrocytes (OLs), loss of myelin, and axonal injury, greatly contributes to impaired neurological function. Impaired proliferation and differentiation of OL precursor cell (OPC, NG2-glia cells) play an important role in limited functional recovery after ischemic stroke and inhibitor of differentiation 2 (ID2) is a key factor controlling NG2-glia cells differentiation. It has been reported that the number of NG2-glia cells in the peri-infarction area significantly increases after ischemic stroke and glial growth factor (GGF2) administration promotes the proliferation and differentiation of NG2-glia cells as well as functional recovery after spinal cord injury. On the basis of the important roles of GGF2 in functional recovery and those of ID2 in NG2-glia cell proliferation and differentiation, we propose that after binding with the ErBb receptor on the surface of NG2-glia cells, GGF2 promotes NG2-glia cell proliferation and differentiation, thereby repairing BBB and white matter integrity and promoting neural functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Fei Li
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Wen-Cao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanyun Sun
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| | - Xinchun Jin
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| |
Collapse
|
12
|
Ke Z, Hu S, Cui W, Sun J, Zhang S, Mak S, Wang J, Tang J, Pang Y, Han Y, Tong K. Bis(propyl)-cognitin potentiates rehabilitation of treadmill exercise after a transient focal cerebral ischemia, possibly via inhibiting NMDA receptor and regulating VEGF expression. Neurochem Int 2019; 128:143-153. [DOI: 10.1016/j.neuint.2019.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/18/2022]
|
13
|
Lalu MM, Fergusson DA, Cheng W, Avey MT, Corbett D, Dowlatshahi D, Macleod MR, Sena ES, Moher D, Shorr R, McCann SK, Gray LJ, Hill MD, O'Connor A, Thayer K, Haggar F, Dobriyal A, Chung HS, Welton NJ, Hutton B. Identifying stroke therapeutics from preclinical models: A protocol for a novel application of network meta-analysis. F1000Res 2019; 8:11. [PMID: 30906535 PMCID: PMC6426098 DOI: 10.12688/f1000research.15869.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction: Globally, stroke is the second leading cause of death. Despite the burden of illness and death, few acute interventions are available to patients with ischemic stroke. Over 1,000 potential neuroprotective therapeutics have been evaluated in preclinical models. It is important to use robust evidence synthesis methods to appropriately assess which therapies should be translated to the clinical setting for evaluation in human studies. This protocol details planned methods to conduct a systematic review to identify and appraise eligible studies and to use a network meta-analysis to synthesize available evidence to answer the following questions: in preclinical in vivo models of focal ischemic stroke, what are the relative benefits of competing therapies tested in combination with the gold standard treatment alteplase in (i) reducing cerebral infarction size, and (ii) improving neurobehavioural outcomes? Methods: We will search Ovid Medline and Embase for articles on the effects of combination therapies with alteplase. Controlled comparison studies of preclinical in vivo models of experimentally induced focal ischemia testing the efficacy of therapies with alteplase versus alteplase alone will be identified. Outcomes to be extracted include infarct size (primary outcome) and neurobehavioural measures. Risk of bias and construct validity will be assessed using tools appropriate for preclinical studies. Here we describe steps undertaken to perform preclinical network meta-analysis to synthesise all evidence for each outcome and obtain a comprehensive ranking of all treatments. This will be a novel use of this evidence synthesis approach in stroke medicine to assess pre-clinical therapeutics. Combining all evidence to simultaneously compare mutliple therapuetics tested preclinically may provide a rationale for the clinical translation of therapeutics for patients with ischemic stroke. Dissemination: Review findings will be submitted to a peer-reviewed journal and presented at relevant scientific meetings to promote knowledge transfer. Registration: PROSPERO number to be submitted following peer review.
Collapse
Affiliation(s)
- Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Canada.,Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Regenerative Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Wei Cheng
- Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Marc T Avey
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dale Corbett
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, Canada
| | - Dar Dowlatshahi
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, Canada.,Department of Medicine. Division of Neurology, The Ottawa Hospital, Ottawa, Canada.,Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Emily S Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - David Moher
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, Canada
| | - Sarah K McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Laura J Gray
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael D Hill
- Cumming School of Medicine, Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Annette O'Connor
- College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Kristina Thayer
- National Institutes of Environmental Health Sciences, Durham, North Carolina, USA
| | - Fatima Haggar
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Aditi Dobriyal
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Hee Sahng Chung
- Clinical Epidemiology Program, Blueprint Translational Research Group, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Nicky J Welton
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Brian Hutton
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada.,Knowledge Synthesis Group, Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
14
|
Lapchak PA, Boitano PD, Bombien R, Cook DJ, Doyan S, Lara JM, Schubert DR. CNB-001, a pleiotropic drug is efficacious in embolized agyrencephalic New Zealand white rabbits and ischemic gyrencephalic cynomolgus monkeys. Exp Neurol 2018; 313:98-108. [PMID: 30521790 DOI: 10.1016/j.expneurol.2018.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/16/2018] [Accepted: 11/30/2018] [Indexed: 01/10/2023]
Abstract
Ischemic stroke is an acute neurodegenerative disease that is extremely devastating to patients, their families and society. Stroke is inadequately treated even with endovascular procedures and reperfusion therapy. Using an extensive translational screening process, we have developed a pleiotropic cytoprotective agent with the potential to positively impact a large population of brain ischemia patients and revolutionize the process used for the development of new drugs to treat complex brain disorders. In this unique translational study article, we document that the novel curcumin-based compound, CNB-001, when administered as a single intravenous dose, has significant efficacy to attenuate clinically relevant behavioral deficits following ischemic events in agyrencephalic rabbits when administered 1 h post-embolization and reduces infarct growth in gyrencephalic non-human primates, when administered 5 min after initiation of middle cerebral artery occlusion. CNB-001 is safe and does not increase morbidity or mortality in either research species. Mechanistically, CNB-001 inhibits human 5- and 15-lipoxygenase in vitro, and can attenuate ischemia-induced inflammatory markers, and oxidative stress markers, while potentially promoting synaptic plasticity mediated by enhanced brain-derived neurotrophic factor (BDNF).
Collapse
Affiliation(s)
- Paul A Lapchak
- Neurocore LLC, Western University of Health Sciences, Pomona, CA 91766, USA.
| | | | | | - Douglas J Cook
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | | | | | - David R Schubert
- Cellular Neurobiology Laboratories, The Salk Institute, La Jolla, CA, USA
| |
Collapse
|
15
|
Hietamies TM, Ostrowski C, Pei Z, Feng L, McCabe C, Work LM, Quinn TJ. Variability of functional outcome measures used in animal models of stroke and vascular cognitive impairment - a review of contemporary studies. J Cereb Blood Flow Metab 2018; 38:1872-1884. [PMID: 30203705 PMCID: PMC6259321 DOI: 10.1177/0271678x18799858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite promising preclinical data, few novel stroke therapies have shown efficacy in man. Efforts to improve standards in conduct and reporting of preclinical research are ongoing. In clinical trials, inconsistency in outcome measures led to regulatory agencies and funders mandating use of a core set of functional outcomes. Our aim was to describe functional outcome measures in preclinical stroke and vascular cognitive impairment (VCI) studies. From 14 high impact journals (January 2005-December 2015 inclusive), 91,956 papers were screened with 1302 full texts analyzed for stroke (ischemic and hemorrhagic) and 56 for VCI studies. In total, 636 (49%) stroke and 37 (66%) VCI papers reported functional outcome measures. There were 74 different functional assessments reported in stroke and 20 in VCI studies. Neurological deficit scores (74%) and Morris water maze (60%) were most commonly used in stroke and VCI, respectively. However, inconsistencies in methods used to assess and score recovery were noted. Neurological and behavioural functional outcome measures are increasingly used in preclinical stroke or VCI studies; however, there is substantial variation in methods. A strict standardized outcome set may not be suitable for translational work, but greater consistency in choice, application and reporting of outcomes may improve the science.
Collapse
Affiliation(s)
- Tuuli M Hietamies
- 1 Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences; University of Glasgow, Glasgow, UK
| | - Caroline Ostrowski
- 1 Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences; University of Glasgow, Glasgow, UK
| | - Zhong Pei
- 2 Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou Shi, Guangdong Sheng, China
| | - Luyang Feng
- 2 Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou Shi, Guangdong Sheng, China
| | - Christopher McCabe
- 3 Institute of Neuroscience & Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Lorraine M Work
- 1 Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences; University of Glasgow, Glasgow, UK
| | - Terence J Quinn
- 1 Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences; University of Glasgow, Glasgow, UK
| |
Collapse
|
16
|
Suvanish Kumar VS, Pretorius E, Rajanikant GK. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol Neurobiol 2018; 38:1383-1397. [PMID: 30062636 DOI: 10.1007/s10571-018-0605-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023]
Abstract
Ischemic stroke is a debilitating multi-factorial cerebrovascular disorder, representing an area of tremendous unmet medical need. Combination treatment has been proposed as a promising therapeutic approach towards combating ischemic stroke. The present study employs in vitro oxygen glucose deprivation (OGD) model to evaluate the post-ischemic neuroprotective efficacy of Everolimus and Paroxetine, alone and in combination. Post-OGD treatment with Everolimus and Paroxetine, alone or in combination, significantly improved the cell survival (~ 80%) when compared to the cells subjected to ischemic injury alone. The individual neuroprotective doses of Everolimus and Paroxetine were found to be at 6.25 and 25 nM, respectively. Whereas, the synergistic neuroprotective dose for Everolimus:Paroxetine was 2:10 nM, calculated using the Chou-Talalay combination index and other four mathematical models. The synergistic combination dose downregulated neuroinflammatory genes (Tnf-α, Il1b, Nf-κB, and iNos) and upregulated the neuroprotective genes (Bcl-2, Bcl-xl, Hif-1, and Epo). The mitochondrial functioning and ROS neutralizing ability increased with combination treatment. Further, the active role of nitric oxide synthase and calmodulin were revealed while exploring the bio-activity of Everolimus and Paroxetine through network pharmacology. The present study for the first time demonstrates the synergistic post-ischemic neuroprotective efficacy of combination treatment with Everolimus and Paroxetine in vitro. Taken together, these findings clearly suggest that Everolimus in combination with Paroxetine may represent a promising therapeutic strategy for the treatment of ischemic stroke, further supporting the combination treatment strategy for this debilitating disorder.
Collapse
Affiliation(s)
- V S Suvanish Kumar
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1, Matieland, 7602, South Africa
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
17
|
Intravenous xenogeneic human cardiosphere-derived cell extracellular vesicles (exosomes) improves behavioral function in small-clot embolized rabbits. Exp Neurol 2018; 307:109-117. [DOI: 10.1016/j.expneurol.2018.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022]
|
18
|
Kofke WA, Ren Y, Augoustides JG, Li H, Nathanson K, Siman R, Meng QC, Bu W, Yandrawatthana S, Kositratna G, Kim C, Bavaria JE. Reframing the Biological Basis of Neuroprotection Using Functional Genomics: Differentially Weighted, Time-Dependent Multifactor Pathogenesis of Human Ischemic Brain Damage. Front Neurol 2018; 9:497. [PMID: 29997569 PMCID: PMC6028620 DOI: 10.3389/fneur.2018.00497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Neuroprotection studies are generally unable to demonstrate efficacy in humans. Our specific hypothesis is that multiple pathophysiologic pathways, of variable importance, contribute to ischemic brain damage. As a corollary to this, we discuss the broad hypothesis that a multifaceted approach will improve the probability of efficacious neuroprotection. But to properly test this hypothesis the nature and importance of the multiple contributing pathways needs elucidation. Our aim is to demonstrate, using functional genomics, in human cardiac surgery procedures associated with cerebral ischemia, that the pathogenesis of perioperative human ischemic brain damage involves the function of multiple variably weighted proteins involving several pathways. We then use these data and literature to develop a proposal for rational design of human neuroprotection protocols. Methods: Ninety-four patients undergoing deep hypothermic circulatory arrest (DHCA) and/or aortic valve replacement surgery had brain damage biomarkers, S100β and neurofilament H (NFH), assessed at baseline, 1 and 24 h post-cardiopulmonary bypass (CPB) with analysis for association with 92 single nucleotide polymorphisms (SNPs) (selected by co-author WAK) related to important proteins involved in pathogenesis of cerebral ischemia. Results: At the nominal significance level of 0.05, changes in S100β and in NFH at 1 and 24 h post-CPB were associated with multiple SNPs involving several prospectively determined pathophysiologic pathways, but were not individually significant after multiple comparison adjustments. Variable weights for the several evaluated SNPs are apparent on regression analysis and, notably, are dissimilar related to the two biomarkers and over time post CPB. Based on our step-wise regression model, at 1 h post-CPB, SOD2, SUMO4, and GP6 are related to relative change of NFH while TNF, CAPN10, NPPB, and SERPINE1 are related to the relative change of S100B. At 24 h post-CPB, ADRA2A, SELE, and BAX are related to the relative change of NFH while SLC4A7, HSPA1B, and FGA are related to S100B. Conclusions: In support of the proposed hypothesis, association SNP data suggest function of specific disparate proteins, as reflected by genetic variation, may be more important than others with variation at different post-insult times after human brain ischemia. Such information may support rational design of post-insult time-sensitive multifaceted neuroprotective therapies.
Collapse
Affiliation(s)
- William A Kofke
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Yue Ren
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, United States
| | - John G Augoustides
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongzhe Li
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Katherine Nathanson
- Department of Medicine, Division of Translational Medicine and Human Genetics Abramson Cancer Center Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Robert Siman
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Qing Cheng Meng
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Weiming Bu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Sukanya Yandrawatthana
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Guy Kositratna
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, United States
| | - Cecilia Kim
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Joseph E Bavaria
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
19
|
Malá H, Rasmussen CP. The effect of combined therapies on recovery after acquired brain injury: Systematic review of preclinical studies combining enriched environment, exercise, or task-specific training with other therapies. Restor Neurol Neurosci 2018; 35:25-64. [PMID: 27858724 DOI: 10.3233/rnn-160682] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Acquired brain injuries (ABI) have devastating effects for the affected individual as well as society. Many studies have investigated the effect of different monotherapies. However, functional recovery is typically only partial. One possible strategy to promote a greater degree of recovery is to apply monotherapies in combination with one or more treatments. OBJECTIVE The objective of this systematic review is to investigate if approaches combining enriched environment (EE), exercise, or task-specific training with other monotherapies, further enhance the degree of recovery after ABI. METHOD Scopus, PsychINFO, and PubMed databases were searched in March 2016 with the following search strings: exercise (or) enriched environment (or) environmental enrichment (or) rehabilitation (and) traumatic brain injury (or) ischemia (or) stroke (and) rat (or) rodent. Studies were included if they (1) were in English, (2) used adult animals subjected to brain injury, (3) included EE, and/or exercise, and/or task-specific training as post-injury treatment strategies, (4) included at least one group receiving another monotherapy. Out of 2.168 hits, 29 studies fulfilled the inclusion criteria. RESULTS Despite several trends for enhanced recovery after combined therapies, this systematic review of 29 studies does not indicate that combined therapies confer consistent combined effects on motor, cognitive, or cerebral recovery according to present criteria for combined effect. CONCLUSION Combined treatments continue to provide hope for enhanced recovery after ABI, however, the research area is in its infancy. This systematic review does not provide conclusive evidence. This is likely due to sparse knowledge regarding optimal treatment parameters. Combined treatments, however, hold the best promise regarding treatment of the complex changes induced by ABI.
Collapse
|
20
|
Co-administration of liposomal fasudil and tissue plasminogen activator ameliorated ischemic brain damage in occlusion model rats prepared by photochemically induced thrombosis. Biochem Biophys Res Commun 2017; 495:873-877. [PMID: 29162447 DOI: 10.1016/j.bbrc.2017.11.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Delivery of neuroprotectants with liposomes has been shown to be effective for the treatment of ischemic stroke. We have recently revealed that intravenous administration of liposomal fasudil (Fasudil-Lip), a Rho-kinase inhibitor, prior to thrombolysis with tissue plasminogen activator (t-PA) can extend the narrow therapeutic time window (TTW) of t-PA. In the present study, we examined the influence of t-PA treatment on liposomal accumulation into the ischemic region and cerebroprotective effect of combined treatment with Fasudil-Lip and t-PA performed at the same timing after the onset of ischemia in middle cerebral artery occlusion (MCAO) prepared by photochemically induced thrombosis. The t-PA administration into MCAO rats 3 h after occlusion brought about significantly higher accumulation of intravenously injected PEGylated liposomes in wide area of ischemic region. Confocal images showed that extravasation of the liposomes from cerebral vessels into brain parenchyma was markedly facilitated by the t-PA treatment which increased blood flow in cerebral vessels. Importantly, co-administration of Fasudil-Lip and t-PA after 3 h occlusion, beyond the TTW of t-PA in MCAO rats, significantly suppressed brain cell damage compared with t-PA treatment alone. These findings suggest that co-administration of Fasudil-Lip and t-PA should lead to prolong t-PA's TTW and become a useful therapeutic option for ischemic stroke.
Collapse
|
21
|
Anesthetic Neuroprotection in Experimental Stroke in Rodents: A Systematic Review and Meta-analysis. Anesthesiology 2017; 126:653-665. [PMID: 28182585 DOI: 10.1097/aln.0000000000001534] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Patients undergoing endovascular therapy for acute ischemic stroke may require general anesthesia to undergo the procedure. At present, there is little clinical evidence to guide the choice of anesthetic in this acute setting. The clinical implications of experimental studies demonstrating anesthetic neuroprotection are poorly understood. Here, the authors evaluated the impact of anesthetic treatment on neurologic outcome in experimental stroke. METHODS Controlled studies of anesthetics in stroke using the filament occlusion model were identified in electronic databases up to December 15, 2015. The primary outcome measures, infarct volume, and neurologic deficit score were used to calculate the normalized mean difference for each comparison. Meta-analysis of normalized mean difference values provided estimates of neuroprotection and contributions of predefined factors: study quality, the timing of treatment, and the duration of ischemia. RESULTS In 80 retrieved publications anesthetic treatment reduced neurologic injury by 28% (95% CI, 24 to 32%; P < 0.0001). Internal validity was high: publication bias enhanced the effect size by 4% or less, effect size increased with study quality (P = 0.0004), and approximately 70% of studies were adequately powered. Apart from study quality, no predefined factor influenced neuroprotection. Neuroprotection failed in animals with comorbidities. Neuroprotection by anesthetics was associated with prosurvival mechanisms. CONCLUSIONS Anesthetic neuroprotection is a robust finding in studies using the filament occlusion model of ischemic stroke and should be assumed to influence outcomes in studies using this model. Neuroprotection failed in female animals and animals with comorbidities, suggesting that the results in young male animals may not reflect human stroke.
Collapse
|
22
|
Abstract
The presence of a salvageable penumbra, a region of ischemic brain tissue with sufficient energy for short-term survival, has been widely agreed as the premise for thrombolytic therapy with tissue plasminogen activator (tPA), which remains the only United States Food and Drug Administration (FDA) approved treatment for acute ischemia stroke. However, the use of tPA has been profoundly constrained due to its narrow therapeutic time window and the increased risk of potentially deadly hemorrhagic transformation (HT). Blood brain barrier (BBB) damage within the thrombolytic time window is an indicator for tPA-induced HT and both normobaric hyperoxia (NBO) and hypothermia have been shown to protect the BBB from ischemia/reperfusion injury. Therefore, providing the O2 as soon as possible (NBO treatment), freezing the brain (hypothermia treatment) to slow down ischemia-induced BBB damage or their combined use may extend the time window for the treatment of tPA. In this review, we summarize the protective effects of NBO, hypothermia or their use combined with tPA on ischemia stroke, based on which, the combination of NBO and hypothermia may be an ideal early stroke treatment to preserve the ischemic penumbra. Given this, there is an urge for large randomized controlled trials to address the effect.
Collapse
Affiliation(s)
- Wen-Cao Liu
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Xin-Chun Jin
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
23
|
Fukuta T, Asai T, Yanagida Y, Namba M, Koide H, Shimizu K, Oku N. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke. FASEB J 2017; 31:1879-1890. [PMID: 28082354 DOI: 10.1096/fj.201601209r] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022]
Abstract
For ischemic stroke treatment, extension of the therapeutic time window (TTW) of thrombolytic therapy with tissue plasminogen activator (tPA) and amelioration of secondary ischemia/reperfusion (I/R) injury are most desirable. Our previous studies have indicated that liposomal delivery of neuroprotectants into an ischemic region is effective for stroke treatment. In the present study, for solving the above problems in the clinical setting, the usefulness of combination therapy with tPA and liposomal fasudil (fasudil-Lip) was investigated in ischemic stroke model rats with photochemically induced thrombosis, with clots that were dissolved by tPA. Treatment with tPA 3 h after occlusion markedly increased blood-brain barrier permeability and activated matrix metalloproteinase (MMP)-2 and -9, which are involved in cerebral hemorrhage. However, an intravenous administration of fasudil-Lip before tPA markedly suppressed the increase in permeability and the MMP activation stemming from tPA. The combination treatment showed significantly larger neuroprotective effects, even in the case of delayed tPA administration compared with each treatment alone or the tPA/fasudil-treated group. These findings suggest that treatment with fasudil-Lip before tPA could decrease the risk of tPA-derived cerebral hemorrhage and extend the TTW of tPA and that the combination therapy could be a useful therapeutic option for ischemic stroke.-Fukuta, T., Asai, T., Yanagida, Y., Namba, M., Koide, H., Shimizu, K., Oku, N. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Tomohiro Asai
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Yosuke Yanagida
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Mio Namba
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Hiroyuki Koide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan; and
| |
Collapse
|
24
|
Abstract
Involvement of the Anesthesiologist in the early stages of care for acute ischemic stroke patient undergoing endovascular treatment is essential. Anesthetic management includes the anesthetic technique (general anesthesia vs sedation), a matter of much debate and an area in need of well-designed prospective studies. The large numbers of confounding factors make the design of such studies a difficult process. A universally agreed point in the endovascular management of acute ischemic stroke is the importance of decreasing the time to revascularization. Hemodynamic and ventilatory management and implementation of neuroprotective modalities and treatment of acute procedural complications are important components of the anesthetic plan.
Collapse
Affiliation(s)
- Rafi Avitsian
- Department of General Anesthesiology, Cleveland Clinic, 9500 Euclid Avenue E-31, Cleveland, OH 44195, USA.
| | - Sandra B Machado
- Department of General Anesthesiology, Cleveland Clinic, 9500 Euclid Avenue E-31, Cleveland, OH 44195, USA
| |
Collapse
|
25
|
Normobaric Hyperoxia Extends Neuro- and Vaso-Protection of N-Acetylcysteine in Transient Focal Ischemia. Mol Neurobiol 2016; 54:3418-3427. [PMID: 27177548 DOI: 10.1007/s12035-016-9932-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/04/2016] [Indexed: 01/04/2023]
Abstract
N-acetylcysteine (NAC), a precursor of glutathione that reduces reperfusion-induced injury, has been shown protection when it was administered pre-ischemia. However, less is known about the effect when it was given post-ischemia and there is no positive result associated with anti-oxidant in clinical trials. This study investigated the neuro- and vaso-protection of post-ischemia NAC administration as well as combining NAC with normobaric hyperoxia (NBO). Male Sprague-Dawley rats were exposed to NBO or normoxia during 2-h occlusion of the middle cerebral artery, followed by 48-h reperfusion. NAC or vehicle was intraperitoneally administered to rats immediately before reperfusion onset. NAC and NBO treatments produced 1.2 and 30 % reduction of infarction volume, respectively, and combination treatment showed greater reduction (59.8 %) as well as more decrease of hemispheric swelling volume. Of note, combination therapy showed improved neurological assessment and motor function which were sustained for 7 days after reperfusion. We also determined that the combination therapy showed greater inhibitory effects on tight junction protein degradation accompanied by Evan's blue extravasation, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) induction, and poly ADP-ribose polymerase (PARP)-1 activation in ischemic brain tissue. Our results showed that although post-ischemia NAC administration had limited protection, combination treatment of NAC plus NBO effectively prevented blood-brain barrier (BBB) damage and significantly improved the outcome of brain injury, providing new evidence to support the concept that "cocktail" treatment targeting different stages provides better neuro- and vaso-protection than current individual treatment that has all failed in their clinical trials.
Collapse
|
26
|
Subirós N, Pérez-Saad HM, Berlanga JA, Aldana L, García-Illera G, Gibson CL, García-del-Barco D. Assessment of dose–effect and therapeutic time window in preclinical studies of rhEGF and GHRP-6 coadministration for stroke therapy. Neurol Res 2016; 38:187-95. [DOI: 10.1179/1743132815y.0000000089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
27
|
Xie CL, Li JH, Wang WW, Zheng GQ, Wang LX. Neuroprotective effect of ginsenoside-Rg1 on cerebral ischemia/reperfusion injury in rats by downregulating protease-activated receptor-1 expression. Life Sci 2015; 121:145-51. [DOI: 10.1016/j.lfs.2014.12.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 11/30/2022]
|
28
|
Lu H, Hu H, He Z, Han X, Chen J, Tu R. Therapeutic imaging window of cerebral infarction revealed by multisequence magnetic resonance imaging: An animal and clinical study. Neural Regen Res 2014; 7:2446-55. [PMID: 25337095 PMCID: PMC4200719 DOI: 10.3969/j.issn.1673-5374.2012.31.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/09/2012] [Indexed: 12/17/2022] Open
Abstract
In this study, we established a Wistar rat model of right middle cerebral artery occlusion and observed pathological imaging changes (T2-weighted imaging [T2WI], T2FLAIR, and diffusion-weighted imaging [DWI]) following cerebral infarction. The pathological changes were divided into three phases: early cerebral infarction, middle cerebral infarction, and late cerebral infarction. In the early cerebral infarction phase (less than 2 hours post-infarction), there was evidence of intracellular edema, which improved after reperfusion. This improvement was defined as the ischemic penumbra. In this phase, a high DWI signal and a low apparent diffusion coefficient were observed in the right basal ganglia region. By contrast, there were no abnormal T2WI and T2FLAIR signals. For the middle cerebral infarction phase (2–4 hours post-infarction), a mixed edema was observed. After reperfusion, there was a mild improvement in cell edema, while the angioedema became more serious. A high DWI signal and a low apparent diffusion coefficient signal were observed, and some rats showed high T2WI and T2FLAIR signals. For the late cerebral infarction phase (4–6 hours post-infarction), significant angioedema was visible in the infarction site. After reperfusion, there was a significant increase in angioedema, while there was evidence of hemorrhage and necrosis. A mixed signal was observed on DWI, while a high apparent diffusion coefficient signal, a high T2WI signal, and a high T2FLAIR signal were also observed. All 86 cerebral infarction patients were subjected to T2WI, T2FLAIR, and DWI. MRI results of clinic data similar to the early infarction phase of animal experiments were found in 51 patients, for which 10 patients (10/51) had an onset time greater than 6 hours. A total of 35 patients had MRI results similar to the middle and late infarction phase of animal experiments, of which eight patients (8/35) had an onset time less than 6 hours. These data suggest that defining the “therapeutic time window” as the time 6 hours after infarction may not be suitable for all patients. Integrated application of MRI sequences including T2WI, T2FLAIR, DW-MRI, and apparent diffusion coefficient mapping should be used to examine the ischemic penumbra, which may provide valuable information for identifying the “therapeutic time window”.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Hui Hu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Zhanping He
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Xiangjun Han
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Jing Chen
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Rong Tu
- Department of Radiology, Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan Province, China
| |
Collapse
|
29
|
|
30
|
Systems pharmacology dissection of multi-scale mechanisms of action for herbal medicines in stroke treatment and prevention. PLoS One 2014; 9:e102506. [PMID: 25093322 PMCID: PMC4122378 DOI: 10.1371/journal.pone.0102506] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/18/2014] [Indexed: 11/19/2022] Open
Abstract
Annually, tens of millions of first-ever strokes occur in the world; however, currently there is lack of effective and widely applicable pharmacological treatments for stroke patients. Herbal medicines, characterized as multi-constituent, multi-target and multi-effect, have been acknowledged with conspicuous effects in treating stroke, and attract extensive interest of researchers although the mechanism of action is yet unclear. In this work, we introduce an innovative systems-pharmacology method that combines pharmacokinetic prescreening, target fishing and network analysis to decipher the mechanisms of action of 10 herbal medicines like Salvia miltiorrhizae, Ginkgo biloba and Ephedrae herba which are efficient in stroke treatment and prevention. Our systematic analysis results display that, in these anti-stroke herbal medicines, 168 out of 1285 constituents with the favorable pharmacokinetic profiles might be implicated in stroke therapy, and the systematic use of these compounds probably acts through multiple mechanisms to synergistically benefit patients with stroke, which can roughly be classified as preventing ischemic inflammatory response, scavenging free radicals and inhibiting neuronal apoptosis against ischemic cerebral damage, as well as exhibiting lipid-lowering, anti-diabetic, anti-thrombotic and antiplatelet effects to decrease recurrent strokes. Relying on systems biology-based analysis, we speculate that herbal medicines, being characterized as the classical combination therapies, might be not only engaged in multiple mechanisms of action to synergistically improve the stroke outcomes, but also might be participated in reducing the risk factors for recurrent strokes.
Collapse
|
31
|
Oxidative Stress and the Use of Antioxidants in Stroke. Antioxidants (Basel) 2014; 3:472-501. [PMID: 26785066 PMCID: PMC4665418 DOI: 10.3390/antiox3030472] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/08/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022] Open
Abstract
Transient or permanent interruption of cerebral blood flow by occlusion of a cerebral artery gives rise to an ischaemic stroke leading to irreversible damage or dysfunction to the cells within the affected tissue along with permanent or reversible neurological deficit. Extensive research has identified excitotoxicity, oxidative stress, inflammation and cell death as key contributory pathways underlying lesion progression. The cornerstone of treatment for acute ischaemic stroke remains reperfusion therapy with recombinant tissue plasminogen activator (rt-PA). The downstream sequelae of events resulting from spontaneous or pharmacological reperfusion lead to an imbalance in the production of harmful reactive oxygen species (ROS) over endogenous anti-oxidant protection strategies. As such, anti-oxidant therapy has long been investigated as a means to reduce the extent of injury resulting from ischaemic stroke with varying degrees of success. Here we discuss the production and source of these ROS and the various strategies employed to modulate levels. These strategies broadly attempt to inhibit ROS production or increase scavenging or degradation of ROS. While early clinical studies have failed to translate success from bench to bedside, the combination of anti-oxidants with existing thrombolytics or novel neuroprotectants may represent an avenue worthy of clinical investigation. Clearly, there is a pressing need to identify new therapeutic alternatives for the vast majority of patients who are not eligible to receive rt-PA for this debilitating and devastating disease.
Collapse
|
32
|
Antonic A, Dottori M, Leung J, Sidon K, Batchelor PE, Wilson W, Macleod MR, Howells DW. Hypothermia protects human neurons. Int J Stroke 2014; 9:544-52. [PMID: 24393199 PMCID: PMC4235397 DOI: 10.1111/ijs.12224] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/14/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Hypothermia provides neuroprotection after cardiac arrest, hypoxic-ischemic encephalopathy, and in animal models of ischemic stroke. However, as drug development for stroke has been beset by translational failure, we sought additional evidence that hypothermia protects human neurons against ischemic injury. METHODS Human embryonic stem cells were cultured and differentiated to provide a source of neurons expressing β III tubulin, microtubule-associated protein 2, and the Neuronal Nuclei antigen. Oxygen deprivation, oxygen-glucose deprivation, and H2 O2 -induced oxidative stress were used to induce relevant injury. RESULTS Hypothermia to 33°C protected these human neurons against H2 O2 -induced oxidative stress reducing lactate dehydrogenase release and Terminal deoxynucleotidyl transferase dUTP nick end labeling-staining by 53% (P ≤ 0·0001; 95% confidence interval 34·8-71·04) and 42% (P ≤ 0·0001; 95% confidence interval 27·5-56·6), respectively, after 24 h in culture. Hypothermia provided similar protection against oxygen-glucose deprivation (42%, P ≤ 0·001, 95% confidence interval 18·3-71·3 and 26%, P ≤ 0·001; 95% confidence interval 12·4-52·2, respectively) but provided no protection against oxygen deprivation alone. Protection (21%) persisted against H2 O2 -induced oxidative stress even when hypothermia was initiated six-hours after onset of injury (P ≤ 0·05; 95% confidence interval 0·57-43·1). CONCLUSION We conclude that hypothermia protects stem cell-derived human neurons against insults relevant to stroke over a clinically relevant time frame. Protection against H2 O2 -induced injury and combined oxygen and glucose deprivation but not against oxygen deprivation alone suggests an interaction in which protection benefits from reduction in available glucose under some but not all circumstances.
Collapse
Affiliation(s)
- Ana Antonic
- Florey Institute of Neuroscience and Mental HealthHeidelberg, Vic, Australia
- Department of Medicine, University of MelbourneHeidelberg, Vic, Australia
| | - Mirella Dottori
- Centre for Neuroscience Research, Department of Anatomy and Neuroscience, University of MelbourneMelbourne, Vic, Australia
| | - Jessie Leung
- Centre for Neuroscience Research, Department of Anatomy and Neuroscience, University of MelbourneMelbourne, Vic, Australia
| | - Kate Sidon
- Florey Institute of Neuroscience and Mental HealthHeidelberg, Vic, Australia
- Department of Medicine, University of MelbourneHeidelberg, Vic, Australia
| | - Peter E Batchelor
- Department of Medicine, University of MelbourneHeidelberg, Vic, Australia
| | - William Wilson
- CSIRO Mathematics, Informatics and Statistics, Riverside Life Sciences PrecinctNorth Ryde, NSW, Australia
| | - Malcolm R Macleod
- Department of Clinical Neurosciences, Western General Hospital, University of EdinburghEdinburgh, UK
| | - David W Howells
- Florey Institute of Neuroscience and Mental HealthHeidelberg, Vic, Australia
- Department of Medicine, University of MelbourneHeidelberg, Vic, Australia
| |
Collapse
|
33
|
Pösel C, Scheibe J, Kranz A, Bothe V, Quente E, Fröhlich W, Lange F, Schäbitz WR, Minnerup J, Boltze J, Wagner DC. Bone marrow cell transplantation time-dependently abolishes efficacy of granulocyte colony-stimulating factor after stroke in hypertensive rats. Stroke 2014; 45:2431-7. [PMID: 24984745 DOI: 10.1161/strokeaha.113.004460] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE We aimed to determine a possible synergistic effect of granulocyte colony-stimulating factor (G-CSF) and bone marrow-derived mononuclear cells (BM MNC) after stroke in spontaneously hypertensive rats. METHODS Male spontaneously hypertensive rats were subjected to middle cerebral artery occlusion and randomly assigned to daily injection of 50 μg/kg G-CSF for 5 days starting 1 hour after stroke (groups 1, 2, and 3) with additional intravenous transplantation of 1.5×10E7 BM MNC per kilogram at 6 hours (group 2) or 48 hours (group 3) after stroke, or control treatment (group 4). Circulating leukocyte counts and functional deficits, infarct volume, and brain edema were repeatedly assessed in the first week and first month. RESULTS G-CSF treatment led to a significant neutrophilia, to a reversal of postischemic depression of circulating leukocytes, and to a significantly improved functional recovery without affecting the infarct volume or brain edema. BM MNC cotransplantation was neutral after 6 hours, but reversed the functional effect of G-CSF after 48 hours. Short-term investigation of combined G-CSF and BM MNC treatment at 48 hours indicated splenic accumulation of granulocytes and transplanted cells, accompanied by a significant rise of granulocytes in the circulation and the ischemic brain. CONCLUSIONS G-CSF improved functional recovery in spontaneously hypertensive rats, but this effect was abolished by cotransplantation of BM MNC after 48 hours. In the spleen, transplanted cells may hinder the clearance of granulocytes that were massively increased by G-CSF. Increased circulation and infiltration of granulocytes into the ischemic brain may be detrimental for stroke outcome.
Collapse
Affiliation(s)
- Claudia Pösel
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.).
| | - Johanna Scheibe
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Alexander Kranz
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Viktoria Bothe
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Elfi Quente
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Wenke Fröhlich
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Franziska Lange
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Wolf-Rüdiger Schäbitz
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Jens Minnerup
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Johannes Boltze
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Daniel-Christoph Wagner
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| |
Collapse
|
34
|
Wu S, Sena E, Egan K, Macleod M, Mead G. Edaravone Improves Functional and Structural Outcomes in Animal Models of Focal Cerebral Ischemia: A Systematic Review. Int J Stroke 2013; 9:101-6. [PMID: 24148907 DOI: 10.1111/ijs.12163] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Edaravone has been used in patients with acute ischemic stroke in Japan for over 10 years but does not have marketing authorization in Europe or America. Either patients in Europe and America are not receiving an effective treatment, or those in Asia are being given a treatment which is not effective. Finding out which of these is true will require further clinical trials, and a better understanding of its efficacy in animal models may help inform the design of those trials so that it might be tested under conditions where there is the greatest prospect of success. We systematically reviewed the efficacy of edaravone in animal models of focal ischemia and summarized data using weighted mean difference DerSimonian and Laird random-effects modeling. We used stratified meta-analysis and metaregression to assess the influence of study design and methodological quality. We identified 49 experiments describing outcome in 814 animals; 30 experiments (519 animals) reported functional and 35 experiments (503 animals) reported structural outcome. Edaravone improved functional and structural outcome by 30.3% (95% confidence interval 23.4–37.2%) and 25.5% (95% confidence interval, 21.1–29.9%), respectively. For functional outcome, there was an inverse relationship between study quality and effect size ( P < 0.0017). Effect sizes were larger in studies where randomization or blinded assessment was not reported. There was no evidence of publication bias. Edaravone is a promising treatment for stroke. However, because of the methodological weakness in current animal studies, no sufficient preclinical evidence is available to optimize the study design of clinical trials. Higher quality animal studies are expected to inform further clinical study.
Collapse
Affiliation(s)
- Simiao Wu
- University of Edinburgh, Centre for Clinical Brain Sciences, Edinburgh, UK
- Sichuan University, West China Hospital, Department of Neurology, Chengdu, Sichuan, China
| | - Emily Sena
- University of Edinburgh, Centre for Clinical Brain Sciences, Edinburgh, UK
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, 245 Burgundy St., Heidelberg Vic 3084, Australia
| | - Kieren Egan
- University of Edinburgh, Centre for Clinical Brain Sciences, Edinburgh, UK
| | - Malcolm Macleod
- University of Edinburgh, Centre for Clinical Brain Sciences, Edinburgh, UK
| | - Gillian Mead
- University of Edinburgh, Centre for Clinical Brain Sciences, Edinburgh, UK
| |
Collapse
|
35
|
Pitkänen A, Nehlig A, Brooks-Kayal AR, Dudek FE, Friedman D, Galanopoulou AS, Jensen FE, Kaminski RM, Kapur J, Klitgaard H, Löscher W, Mody I, Schmidt D. Issues related to development of antiepileptogenic therapies. Epilepsia 2013; 54 Suppl 4:35-43. [PMID: 23909852 DOI: 10.1111/epi.12297] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Several preclinical proof-of-concept studies have provided evidence for positive treatment effects on epileptogenesis. However, none of these hypothetical treatments has advanced to the clinic. The experience in other fields of neurology such as stroke, Alzheimer's disease, or amyotrophic lateral sclerosis has indicated several problems in the design of preclinical studies, which likely contribute to failures in translating the positive preclinical data to the clinic. The Working Group on "Issues related to development of antiepileptogenic therapies" of the International League Against Epilepsy (ILAE) and the American Epilepsy Society (AES) has considered the possible problems that arise when moving from proof-of-concept antiepileptogenesis (AEG) studies to preclinical AEG trials, and eventually to clinical AEG trials. This article summarizes the discussions and provides recommendations on how to design a preclinical AEG monotherapy trial in adult animals. We specifically address study design, animal and model selection, number of studies needed, issues related to administration of the treatment, outcome measures, statistics, and reporting. In addition, we give recommendations for future actions to advance the preclinical AEG testing.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
O'Collins VE, Donnan GA, Macleod MR, Howells DW. Hypertension and experimental stroke therapies. J Cereb Blood Flow Metab 2013; 33:1141-7. [PMID: 23736641 PMCID: PMC3734784 DOI: 10.1038/jcbfm.2013.88] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/26/2013] [Accepted: 04/30/2013] [Indexed: 01/20/2023]
Abstract
UNLABELLED Hypertension is an established target for long-term stroke prevention but procedures for management of hypertension in acute stroke are less certain. Here, we analyze basic science data to examine the impact of hypertension on candidate stroke therapies and of anti-hypertensive treatments on stroke outcome. METHODS Data were pooled from 3,288 acute ischemic stroke experiments (47,899 animals) testing the effect of therapies on infarct size (published 1978-2010). Data were combined using meta-analysis and meta-regression, partitioned on the basis of hypertension, stroke model, and therapy. RESULTS Hypertensive animals were used in 10% of experiments testing 502 therapies. Hypertension was associated with lower treatment efficacy, especially in larger infarcts. Overall, anti-hypertensives did not provide greater benefit than other drugs, although benefits were evident in hypertensive animals even when given after stroke onset. Fifty-eight therapies were tested in both normotensive and hypertensive animals: some demonstrated superior efficacy in hypertensive animals (hypothermia) while others worked better in normotensive animals (tissue plasminogen activator, anesthetic agents). DISCUSSION Hypertension has a significant effect on the efficacy of candidate stroke drugs: standard basic science testing may overestimate the efficacy which could be reasonably expected from certain therapies and for hypertensive patients with large or temporary occlusions.
Collapse
Affiliation(s)
- Victoria E O'Collins
- Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia.
| | | | | | | |
Collapse
|
37
|
Combined antiapoptotic and antioxidant approach to acute neuroprotection for stroke in hypertensive rats. J Cereb Blood Flow Metab 2013; 33:1215-24. [PMID: 23632970 PMCID: PMC3734772 DOI: 10.1038/jcbfm.2013.70] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 04/03/2013] [Accepted: 04/05/2013] [Indexed: 12/17/2022]
Abstract
We hypothesized that targeting key points in the ischemic cascade with combined neuroglobin (Ngb) overexpression and c-jun N-terminal kinase (JNK) inhibition (SP600125) would offer greater neuroprotection than single treatment after in vitro hypoxia/reoxygenation and in a randomized, blinded in vivo experimental stroke study using a clinically relevant rat strain. Male spontaneously hypertensive stroke-prone rats underwent transient middle cerebral artery occlusion (tMCAO) and were divided into the following groups: tMCAO; tMCAO+control GFP-expressing canine adenovirus-2, CAVGFP; tMCAO+Ngb-expressing CAV-2, CAVNgb; tMCAO+SP600125; tMCAO+CAVNgb+SP600125; or sham procedure. Rats were assessed till day 14 for neurologic outcome before infarct determination. In vitro, combined lentivirus-mediated Ngb overexpression+SP600125 significantly reduced oxidative stress and apoptosis compared with single treatment(s) after hypoxia/reoxygenation in B50 cells. In vivo, infarct volume was significantly reduced by CAVNgb, SP600125, and further by CAVNgb+SP600125. The number of Ngb-positive cells in the peri-infarct cortex and striatum was significantly increased 14 days after tMCAO in animals receiving CAVNgb. Neurologic outcome, measured using a 32-point neurologic score, significantly improved with CAVNgb+SP600125 compared with single treatments at 14 days after tMCAO. Combined Ngb overexpression with JNK inhibition reduced hypoxia/reoxygenation-induced oxidative stress and apoptosis in cultured neurons and reduced infarct and improved neurologic outcome more than single therapy after in vivo experimental stroke in hypertensive rats.
Collapse
|
38
|
Abstract
Stroke is one of the leading causes of death worldwide and the biggest reason for long-term disability. Basic research has formed the modern understanding of stroke pathophysiology, and has revealed important molecular, cellular and systemic mechanisms. However, despite decades of research, most translational stroke trials that aim to introduce basic research findings into clinical treatment strategies - most notably in the field of neuroprotection - have failed. Among other obstacles, poor methodological and statistical standards, negative publication bias, and incomplete preclinical testing have been proposed as 'translational roadblocks'. In this article, we introduce the models commonly used in preclinical stroke research, discuss some of the causes of failed translational success and review potential remedies. We further introduce the concept of modeling 'care' of stroke patients, because current preclinical research models the disorder but does not model care or state-of-the-art clinical testing. Stringent statistical methods and controlled preclinical trials have been suggested to counteract weaknesses in preclinical research. We conclude that preclinical stroke research requires (1) appropriate modeling of the disorder, (2) appropriate modeling of the care of stroke patients and (3) an approach to preclinical testing that is similar to clinical testing, including Phase 3 randomized controlled preclinical trials as necessary additional steps before new therapies enter clinical testing.
Collapse
Affiliation(s)
- Philipp Mergenthaler
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10098 Berlin, Germany.
| | | |
Collapse
|
39
|
Campbell K, Knuckey NW, Brookes LM, Meloni BP. Efficacy of mild hypothermia (35°C) and moderate hypothermia (33°C) with and without magnesium when administered 30min post-reperfusion after 90min of middle cerebral artery occlusion in Spontaneously Hypertensive rats. Brain Res 2013; 1502:47-54. [PMID: 23370002 DOI: 10.1016/j.brainres.2013.01.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/20/2012] [Accepted: 01/22/2013] [Indexed: 11/30/2022]
Abstract
In this study we compared the efficacy of mild (35°C) and moderate (33°C) hypothermia alone and when combined with magnesium in a transient focal cerebral ischaemia rat model. Spontaneously Hypertensive rats were subjected to 90min of transient intraluminal thread middle cerebral artery occlusion (MCAO). Thirty minutes after reperfusion animals were treated with mild (35°C/24h) or moderate (33°C/24h) hypothermia combined with either magnesium (intravenous MgSO4 infusion: 360μmol/kg, then 120μmol/kg/h for 24h) or a similar volume of saline. Control animals were maintained normothermic (37°C/24h) and received vehicle infusion (saline for 24h). Infarct volumes and functional assessment (bi-symmetrical adhesive tape removal) were measured 48h after MCAO induction. After transient MCAO, only moderate hypothermia and mild hypothermia combined with magnesium treatment significantly reduced infarct volumes by 32.9% (P=0.01) and by 24.8% (P=0.046), respectively. Mild hypothermia alone reduced infarct volume by 23.8%, but did not reach statistical significance (P=0.054), while moderate hypothermia combined with magnesium reduced infarct volume by 17.3% (P=0.17). No treatment improved adhesive tape removal time. In summary, moderate hypothermia and mild hypothermia with or without magnesium can reduce infarct volume, however magnesium may reduce the efficacy of moderate hypothermia. Given the potential advantages of mild hypothermia over moderate hypothermia in terms of side-effects and induction, and the potential beneficial effects of magnesium, these findings have important implications for the use of hypothermia for stroke.
Collapse
Affiliation(s)
- Kym Campbell
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia
| | | | | | | |
Collapse
|
40
|
Gibson CL, Murphy AN, Murphy SP. Stroke outcome in the ketogenic state--a systematic review of the animal data. J Neurochem 2012; 123 Suppl 2:52-7. [PMID: 23050642 DOI: 10.1111/j.1471-4159.2012.07943.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
As a predictor of potential clinical outcome, we performed a systematic review of controlled studies that assessed experimental stroke outcome in rodents maintained on special diets (calorie restriction and ketogenic diet) or following the direct administration of ketone bodies. Pre-clinical studies were identified by searching web databases and the reference lists of relevant original articles and reviews. Sixteen published studies (a total of 733 experimental animals) met specific criteria and were analyzed using Cochrane Review Manager software. This resulted in objective evidence to suggest beneficial effects of the ketogenic pathway on pathological and functional outcomes following experimental stroke.
Collapse
Affiliation(s)
- Claire L Gibson
- School of Psychology, University of Leicester, Leicester, UK
| | | | | |
Collapse
|
41
|
Jin X, Liu J, Liu KJ, Rosenberg GA, Yang Y, Liu W. Normobaric hyperoxia combined with minocycline provides greater neuroprotection than either alone in transient focal cerebral ischemia. Exp Neurol 2012. [PMID: 23195595 DOI: 10.1016/j.expneurol.2012.11.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Normobaric hyperoxia (NBO), which maintains penumbral oxygenation, reduces brain injury during cerebral ischemia, and minocycline, a tetracycline derivative, reduces reperfusion injury, including inflammation, apoptosis and matrix metalloproteinases (MMPs) activation. Since they have different mechanisms of action, we hypothesized that combining them would provide greater neuroprotection. To test the hypothesis, we evaluated the neuroprotective effects of the combination of NBO with minocycline. Male Sprague-Dawley rats were exposed to NBO (95% O(2)) or normoxia (21% O(2)) during 90-min filament occlusion of the middle cerebral artery, followed by 48 h of reperfusion. Minocycline (3 mg/kg) or vehicle was intravenously administered to rats 15 min after reperfusion onset. Treatment with NBO and minocycline alone resulted in 36% and 30% reductions in infarction volume, respectively. When the two treatments were combined, there was a 68% reduction in infarction volume. The combination therapy also significantly reduced hemispheric swelling, which was absent with monotherapy. In agreement with its greater neuro- and vasoprotection, the combination therapy showed greater inhibitory effects on MMP-2/9 induction, occludin degradation, caspase-3 and -9 activation and apoptosis inducing factor (AIF) induction in ischemic brain tissue. Our results show that NBO plus minocycline effectively reduces brain injury in transient focal cerebral ischemia with protection due to inhibition on MMP-2/9-mediated occludin degradation and attenuation of caspase-dependent and independent apoptotic pathways.
Collapse
Affiliation(s)
- Xinchun Jin
- College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | | | |
Collapse
|
42
|
Colbourne F. Comparing hypothermia and magnesium after global ischemia-editorial commentary on li et Al., 2011. Ther Hypothermia Temp Manag 2012; 2:10. [PMID: 24717132 DOI: 10.1089/ther.2012.1503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|