101
|
Wu L, Wu D, Yang T, Xu J, Chen J, Wang L, Xu S, Zhao W, Wu C, Ji X. Hypothermic neuroprotection against acute ischemic stroke: The 2019 update. J Cereb Blood Flow Metab 2020; 40:461-481. [PMID: 31856639 PMCID: PMC7026854 DOI: 10.1177/0271678x19894869] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023]
Abstract
Acute ischemic stroke is a leading cause of death and disability worldwide. Therapeutic hypothermia has long been considered as one of the most robust neuroprotective strategies. Although the neuroprotective effects of hypothermia have only been confirmed in patients with global cerebral ischemia after cardiac arrest and in neonatal hypoxic ischemic encephalopathy, establishing standardized protocols and strictly controlling the key parameters may extend its application in other brain injuries, such as acute ischemic stroke. In this review, we discuss the potential neuroprotective effects of hypothermia, its drawbacks evidenced in previous studies, and its potential clinical application for acute ischemic stroke especially in the era of reperfusion. Based on the different conditions between bench and bedside settings, we demonstrate the importance of vascular recanalization for neuroprotection of hypothermia by analyzing numerous literatures regarding hypothermia in focal cerebral ischemia. Then, we make a thorough analysis of key parameters of hypothermia and introduce novel hypothermic therapies. We advocate in favor of the process of clinical translation of intra-arterial selective cooling infusion in the era of reperfusion and provide insights into the prospects of hypothermia in acute ischemic stroke.
Collapse
Affiliation(s)
- Longfei Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jin Xu
- Department of Library, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jian Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Luling Wang
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chuanjie Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
102
|
Williams EI, Betterton RD, Davis TP, Ronaldson PT. Transporter-Mediated Delivery of Small Molecule Drugs to the Brain: A Critical Mechanism That Can Advance Therapeutic Development for Ischemic Stroke. Pharmaceutics 2020; 12:pharmaceutics12020154. [PMID: 32075088 PMCID: PMC7076465 DOI: 10.3390/pharmaceutics12020154] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke is the 5th leading cause of death in the United States. Despite significant improvements in reperfusion therapies, stroke patients still suffer from debilitating neurocognitive deficits. This indicates an essential need to develop novel stroke treatment paradigms. Endogenous uptake transporters expressed at the blood-brain barrier (BBB) provide an excellent opportunity to advance stroke therapy via optimization of small molecule neuroprotective drug delivery to the brain. Examples of such uptake transporters include organic anion transporting polypeptides (OATPs in humans; Oatps in rodents) and organic cation transporters (OCTs in humans; Octs in rodents). Of particular note, small molecule drugs that have neuroprotective properties are known substrates for these transporters and include 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (i.e., statins) for OATPs/Oatps and 1-amino-3,5-dimethyladamantane (i.e., memantine) for OCTs/Octs. Here, we review current knowledge on specific BBB transporters that can be targeted for improvement of ischemic stroke treatment and provide state-of-the-art perspectives on the rationale for considering BBB transport properties during discovery/development of stroke therapeutics.
Collapse
|
103
|
Wang R, Pu H, Ye Q, Jiang M, Chen J, Zhao J, Li S, Liu Y, Hu X, Rocha M, Jadhav AP, Chen J, Shi Y. Transforming Growth Factor Beta-Activated Kinase 1-Dependent Microglial and Macrophage Responses Aggravate Long-Term Outcomes After Ischemic Stroke. Stroke 2020; 51:975-985. [PMID: 32078472 DOI: 10.1161/strokeaha.119.028398] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background and Purpose- Microglia/macrophages (Mi/MΦ) can profoundly influence stroke outcomes by acquiring functionally dominant phenotypes (proinflammatory or anti-inflammatory; deleterious or salutary). Identification of the molecular mechanisms that dictate the functional status of Mi/MΦ after brain ischemia/reperfusion may reveal novel therapeutic targets for stroke. We hypothesized that activation of TAK1 (transforming growth factor beta-activated kinase 1), a key MAP3K upstream of multiple inflammation-regulating pathways, drives Mi/MΦ toward a proinflammatory phenotype and potentiates ischemia/reperfusion brain injury. Methods- Young adult mice were subjected to 1 hour of middle cerebral artery occlusion (MCAO) followed by reperfusion. TAK1 was targeted by tamoxifen-induced Mi/MΦ-specific knockout or administration of a selective inhibitor 5Z-7-Oxozeaenol after MCAO. Neurobehavioral deficits and long-term gray matter and white matter injury were assessed up to 35 days after MCAO. Mi/MΦ functional status and brain inflammatory profiles were assessed 3 days after MCAO by RNA-seq, flow cytometry, and immunohistochemistry. Results- TAK1 Mi/MΦ-specific knockout markedly ameliorated neurological deficits in the rotarod and cylinder tests for at least 35 days after MCAO. Mechanistically, RNA-seq of purified brain Mi/MΦ demonstrated that proinflammatory genes and their predicted biological functions were downregulated or inhibited in microglia and macrophages from TAK1 Mi/MΦ-specific knockout mice versus WT mice 3 days after MCAO. Consistent with the anti-inflammatory phenotype of Mi/MΦ-specific knockout, oxozeaenol treatment mitigated neuroinflammation 3 days after MCAO, manifested by less Iba1+/CD16+ proinflammatory Mi/MΦ and suppressed brain invasion of various peripheral immune cells. Oxozeaenol treatment beginning 2 hours after MCAO improved long-term sensorimotor and cognitive functions in the foot fault, rotarod, and water maze tests. Furthermore, Oxozeaenol promoted both gray matter and white matter integrity 35 days after MCAO. Conclusions- TAK1 promotes ischemia/reperfusion-induced inflammation, brain injury, and maladaptive behavior by enhancing proinflammatory and deleterious Mi/MΦ responses. Therefore, TAK1 inhibition is a promising therapy to improve long-term stroke outcomes.
Collapse
Affiliation(s)
- Rongrong Wang
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Hongjian Pu
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Qing Ye
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.).,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (Q.Y., X.H., Jun Chen, Y.S.)
| | - Ming Jiang
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Jie Chen
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Jingyan Zhao
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Sicheng Li
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Yaan Liu
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.)
| | - Xiaoming Hu
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.).,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (Q.Y., X.H., Jun Chen, Y.S.)
| | - Marcelo Rocha
- Department of Neurology, UPMC Stroke Institute, University of Pittsburgh, PA (M.R., A.P.J.)
| | - Ashutosh P Jadhav
- Department of Neurology, UPMC Stroke Institute, University of Pittsburgh, PA (M.R., A.P.J.)
| | - Jun Chen
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.).,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (Q.Y., X.H., Jun Chen, Y.S.)
| | - Yejie Shi
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA (R.W., H.P., Q.Y., M.J., Jie Chen, J.Z., S.L., Y.L., X.H., Jun Chen, Y.S.).,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (Q.Y., X.H., Jun Chen, Y.S.)
| |
Collapse
|
104
|
Affiliation(s)
- K-O Lövblad
- Service de Neuroradiologie Diagnostique et Interventionnelle, Département Diagnostique Hôpitaux Universitaires de Genève Genève, Switzerland
| |
Collapse
|
105
|
Duan Y, Wu D, Huber M, Shi J, An H, Wei W, He X, Ding Y, Ji X. New Endovascular Approach for Hypothermia With Intrajugular Cooling and Neuroprotective Effect in Ischemic Stroke. Stroke 2020; 51:628-636. [PMID: 31884905 DOI: 10.1161/strokeaha.119.026523] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background and Purpose—
Induction of hypothermia as a stroke therapy has been limited by logistical challenges. This study was designed to determine the hypothermic and neuroprotective efficacy of infusing cold saline directly into the internal jugular (IJ) vein and compare the effects of IJ hypothermia to those achieved by intracarotid artery hypothermia in an ischemic stroke model.
Methods—
The right middle cerebral artery was occluded in rats using an intraluminal filament. Immediately following reperfusion, hypothermia was achieved by infusing isotonic saline through microcatheter into the right IJ or right intracarotid over 30 minutes. Infarct sizes, neurological deficits, blood-brain barrier damage, edema volume, blood-brain barrier associated molecules (MMP-9 [matrix metallopeptidase 9] and AQP-4 [aquaporin 4]), and apoptosis-associated proteins (Bcl-2 and cleaved Caspase-3) were measured.
Results—
We found that both IJ- and intracarotid-based infusion cooled the brain robustly with a minimal effect on rectal temperatures. This brain cooling led to significantly reduced infarct volumes at 24 hours after reperfusion, as well as decreased expression of the proapoptotic protein cleaved Caspase-3 and increased expression of the antiapoptotic protein Bcl-2. Intracarotid and IJ cooling also aided in blood-brain barrier maintenance, as shown by decreased edema volumes, reduced Evans Blue leakage, and decreased expression of edema-facilitating proteins (MMP-9 and AQP-4). Both cooling methods then translated to preserved neurological function as determined by multiple functional tests over a 28-day observation period. Most importantly, the cooling and neuroprotective efficacy of IJ cooling was comparable to intracarotid cooling by almost every metric evaluated.
Conclusions—
Compared with intracarotid infusion, IJ infusion conferred a similar degree of hypothermia and neuroprotection following ischemic stroke. Given the ease of establishing vascular access via the internal jugular vein and the powerful neuroprotection that hypothermia provides, IJ brain cooling could be used as a promising hypothermia-induction modality going forward.
Collapse
Affiliation(s)
- Yunxia Duan
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (Y. Duan, D.W., X.J.), Xuanwu Hospital, Capital Medical University, China
- Center of Stroke, Beijing Institute for Brain Disorders, China (Y. Duan., D.W., X.J.)
| | - Di Wu
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (Y. Duan, D.W., X.J.), Xuanwu Hospital, Capital Medical University, China
- Center of Stroke, Beijing Institute for Brain Disorders, China (Y. Duan., D.W., X.J.)
| | - Mitchell Huber
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI (M.H., Y. Ding.)
| | - Jingfei Shi
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
| | - Hong An
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
| | - Wenjing Wei
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
| | - Xiaoduo He
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
| | - Yuchuan Ding
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI (M.H., Y. Ding.)
| | - Xunming Ji
- From the Department of Neurology, China-America Institute of Neuroscience (Y. Duan, D.W., J.S., H.A., W.W., X.H., Y. Ding, X.J.), Xuanwu Hospital, Capital Medical University, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (Y. Duan, D.W., X.J.), Xuanwu Hospital, Capital Medical University, China
- Center of Stroke, Beijing Institute for Brain Disorders, China (Y. Duan., D.W., X.J.)
| |
Collapse
|
106
|
Ting SM, Zhao X, Sun G, Obertas L, Ricote M, Aronowski J. Brain Cleanup as a Potential Target for Poststroke Recovery: The Role of RXR (Retinoic X Receptor) in Phagocytes. Stroke 2020; 51:958-966. [PMID: 31914884 DOI: 10.1161/strokeaha.119.027315] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Phagocytic cells, such as microglia and blood-derived macrophages, are a key biological modality responsible for phagocytosis-mediated clearance of damaged, dead, or displaced cells that are compromised during senescence or pathological processes, including after stroke. This process of clearance is essential to eliminate the source of inflammation and to allow for optimal brain repair and functional recovery. Transcription factor, RXR (retinoic-X-receptor) is strongly implicated in phagocytic functions regulation, and as such could represent a novel target for brain recovery after stroke. Methods- Primary cultured microglia and bone marrow macrophages were used for phagocytic study. Mice with deleted RXR-α in myeloid phagocytes (Mac-RXR-α-/-) were subjected to transient middle cerebral artery occlusion to mimic ischemic stroke and then treated with RXR agonist bexarotene. RNA-sequencing and long-term recovery were evaluated. Results- Using cultured microglia, we demonstrated that the RXR-α promotes the phagocytic functions of microglia toward apoptotic neurons. Using mice with deleted RXR-α in myeloid phagocytes (Mac-RXR-α-/-), we have shown that despite behaving similarly to the control at early time points (up to 3 days, damage established histologically and behaviorally), these Mac-RXR-α-/- mice demonstrated worsened late functional recovery and developed brain atrophy that was larger in size than that seen in control mice. The RXR-α deficiency was associated with reduced expression of genes known to be under control of the prominent transcriptional RXR partner, PPAR (peroxisome proliferator-activated receptor)-γ, as well as genes encoding for scavenger receptors and genes that signify microglia/macrophages polarization to a reparative phenotype. Finally, we demonstrated that the RXR agonist, bexarotene, administered as late as 1 day after middle cerebral artery occlusion, improved neurological recovery, and reduced the atrophy volume as assessed 28 days after stroke. Bexarotene did not improve outcome in Mac-RXR-α-/- mice. Conclusions- Altogether, these data suggest that phagocytic cells control poststroke recovery and that RXR in these cells represents an attractive target with exceptionally long therapeutic window.
Collapse
Affiliation(s)
- Shun-Ming Ting
- From the Department of Neurology, University of Texas HSC, McGovern Medical School, Houston (S.-M.T., X.Z., G.S., L.O., J.A.)
| | - Xiurong Zhao
- From the Department of Neurology, University of Texas HSC, McGovern Medical School, Houston (S.-M.T., X.Z., G.S., L.O., J.A.)
| | - Guanghua Sun
- From the Department of Neurology, University of Texas HSC, McGovern Medical School, Houston (S.-M.T., X.Z., G.S., L.O., J.A.)
| | - Lidiya Obertas
- From the Department of Neurology, University of Texas HSC, McGovern Medical School, Houston (S.-M.T., X.Z., G.S., L.O., J.A.)
| | - Mercedes Ricote
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.R.)
| | - Jaroslaw Aronowski
- From the Department of Neurology, University of Texas HSC, McGovern Medical School, Houston (S.-M.T., X.Z., G.S., L.O., J.A.)
| |
Collapse
|
107
|
Cao W, Zhang C, Chen R, Wu Q, Xu R, Zhang L, Zhang X. A Novel Cerebroprotein Hydrolysate, CH1, Ameliorates Chronic Focal Cerebral Ischemia Injury by Promoting White Matter Integrity via the Shh/Ptch-1/Gli-1 Signaling Pathway. Neuropsychiatr Dis Treat 2020; 16:3209-3224. [PMID: 33380798 PMCID: PMC7767750 DOI: 10.2147/ndt.s289990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Strokes are devastating as there are no current therapies to prevent long-term neurological deficits. Previous studies reported that cerebroprotein hydrolysate (CH) plays a role in neuronal protection in acute phase after ischemic stroke, while the long-term effects of CH upon brain plasticity and neurological outcomes after stroke are still uncertain. To address these gaps, we assessed the effect of a new cerebroprotein hydrolysate, CH1, on long-term gray and white matter integrity as well as axonal plasticity in the late phase after ischemic stroke and the potential mechanisms. METHODS Adult male mice were subjected to permanent distal middle cerebral artery occlusion (dMCAO), followed by daily intraperitoneal injection of CH1 for 14 days. Motor function was measured weekly through behavioral neurological evaluations. Gray matter intensity and white matter intensity were examined by immunofluorescence staining. The sonic hedgehog (Shh) inhibitor cyclopamine (CYC) was injected to determine the involvement of the Shh pathway in the therapeutic effects of CH1. RESULTS We found that intraperitoneal delivery of CH1, compared to vehicle administration, significantly improved long-term neurological outcomes at various times and promoted neuronal viability at 14 days but not at 28 days after stroke. Importantly, CH1 mitigated stroke-induced white matter injury and facilitated axonal plasticity in the late stage after stroke. CONCLUSION These results unveil a previously unappreciated role for CH in the repair of white matter and brain plasticity after stroke.
Collapse
Affiliation(s)
- Wen Cao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China.,Hebei Vascular Homeostasis Key Laboratory for Neurology, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Qianqian Wu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China.,Hebei Vascular Homeostasis Key Laboratory for Neurology, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China.,Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China.,Hebei Vascular Homeostasis Key Laboratory for Neurology, Shijiazhuang, Hebei 050000, People's Republic of China
| |
Collapse
|
108
|
Abstract
Novel therapeutic intervention that aims to enhance the endogenous recovery potential of the brain during the subacute phase of stroke has produced promising results. The paradigm shift in treatment approaches presents new challenges to preclinical and clinical researchers alike, especially in the functional endpoints domain. Shortcomings of the "neuroprotection" era of stroke research are yet to be fully addressed. Proportional recovery observed in clinics, and potentially in animal models, requires a thorough reevaluation of the methods used to assess recovery. To this end, this review aims to give a detailed evaluation of functional outcome measures used in clinics and preclinical studies. Impairments observed in clinics and animal models will be discussed from a functional testing perspective. Approaches needed to bridge the gap between clinical and preclinical research, along with potential means to measure the moving target recovery, will be discussed. Concepts such as true recovery of function and compensation and methods that are suitable for distinguishing the two are examined. Often-neglected outcomes of stroke, such as emotional disturbances, are discussed to draw attention to the need for further research in this area.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Burke Neurological Research Institute, White Plains, NY, USA
| | - Sunghee Cho
- Burke Neurological Research Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Neurological Research Institute, White Plains, NY, USA
| |
Collapse
|
109
|
Abstract
The recently completed EXTEND trial tested the idea that tissue plasminogen activator thrombolysis can be safely extended up to 9 h after stroke onset if automated perfusion imaging indicates the presence of a salvageable penumbra. This important trial contributes to an ongoing paradigm shift for stroke therapy. Combined with the introduction of endovascular therapy, image-guided patient selection is expanding the toolbox of the stroke practitioner. At the same time, pushing the limits of reperfusion has raised important questions about mechanisms to pursue for combination therapy as well as potential approaches to mitigate side effects and optimize treatments for patients with various co-morbidities.
Collapse
Affiliation(s)
- Klaus van Leyen
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiaoying Wang
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Magdy Selim
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
110
|
Broocks G, Kemmling A, Aberle J, Kniep H, Bechstein M, Flottmann F, Leischner H, Faizy TD, Nawabi J, Schön G, Sporns P, Thomalla G, Fiehler J, Hanning U. Elevated blood glucose is associated with aggravated brain edema in acute stroke. J Neurol 2019; 267:440-448. [PMID: 31667625 DOI: 10.1007/s00415-019-09601-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND PURPOSE Clinical outcome after endovascular thrombectomy in patients with acute ischemic stroke still varies significantly. Higher blood glucose levels (BGL) have been associated with worse clinical outcome, but the pathophysiological causes are not yet understood. We hypothesized that higher levels of BGL are associated with more pronounced ischemic brain edema and worse clinical outcome mediated by cerebral collateral circulation. METHODS 178 acute ischemic stroke patients who underwent mechanical thrombectomy were included. Early ischemic brain edema was determined using quantitative lesion water uptake on initial computed tomography (CT) and collateral status was assessed with an established 5-point scoring system in CT-angiography. Good clinical outcome was defined as functional independence (modified Rankin Scale [mRS] score 0-2). Multivariable logistic regression analysis was performed to predict functional independence and linear regression analyses to investigate the impact of BGL and collateral status on water uptake. RESULTS The mean BGL at admission was significantly lower in patients with good outcome at 90 days (116.5 versus 138.5 mg/dl; p < 0.001) and early water uptake was lower (6.3% versus 9.6%; p < 0.001). The likelihood for good outcome declined with increasing BGL (odds ratio [OR] per 100 mg/dl BGL increase: 0.15; 95% CI 0.02-0.86; p = 0.039). Worse collaterals (1% water uptake per point, 95% CI 0.4-1.7%) and higher BGL (0.6% per 10 mg/dl BGL, 95% CI 0.3-0.8%) were significantly associated with increased water uptake. CONCLUSION Elevated admission BGL were associated with increased early brain edema and poor clinical outcome mediated by collateral status. Patients with higher BGL might be targeted by adjuvant anti-edematous treatment.
Collapse
Affiliation(s)
- Gabriel Broocks
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Andre Kemmling
- Department of Neuroradiology, Westpfalz-Klinikum, Kaiserslautern, Germany.,Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jens Aberle
- Department of Endocrinology and Diabetology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helge Kniep
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Matthias Bechstein
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Fabian Flottmann
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Hannes Leischner
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Tobias D Faizy
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jawed Nawabi
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Department of Radiology, Charité University Medical Center, Berlin, Germany
| | - Gerhard Schön
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Sporns
- Department of Neuroradiology, Westpfalz-Klinikum, Kaiserslautern, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Fiehler
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Uta Hanning
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
111
|
Almekhlafi MA, Poli S, Goyal M, Demchuk AM. Therapeutic hypothermia in stroke: Quo Vadis? Brain Circ 2019; 5:157-159. [PMID: 31950090 PMCID: PMC6950514 DOI: 10.4103/bc.bc_62_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/24/2022] Open
Affiliation(s)
- Mohammed A. Almekhlafi
- Department of Clinical Neurosciences and Radiology, Calgary Stroke Program, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sven Poli
- Department of Neurology and Stroke, Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Mayank Goyal
- Department of Clinical Neurosciences and Radiology, Calgary Stroke Program, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Andrew M. Demchuk
- Department of Clinical Neurosciences and Radiology, Calgary Stroke Program, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
112
|
Chiu CC, Lin JM, Wu LY, Hsu TC, Tzang BS. The Beneficial Effects of Raffinee in Permanent Occulted Stroke Mice. J Med Food 2019; 22:1226-1234. [PMID: 31545135 DOI: 10.1089/jmf.2019.4438] [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: 11/12/2022] Open
Abstract
Ischemic stroke is a major cause of disability and mortality globally. Although thrombolytic therapy is routinely adopted in cases of ischemic stroke, various alternative natural neuroprotectants are also used as effective adjuvant therapies to recover neurofunction following ischemic stroke. Raffinee, a natural fermented product with strong antioxidant and neuroprotective activities, has antiatherogenic effects in animals and has exhibited neuroprotective effects in a clinical trial by recovering motor and sensory function following spinal cord lesion. This study reveals the advantageous effects of Raffinee on PC12 cells by decreasing hypoxia-induced apoptosis in mice with permanent middle cerebral artery occlusion (pMCAO) by increasing the levels of neurotrophic factors such as S100β, reducing serum inflammatory factors such as matrix metalloproteinases (MMP)-9/MMP-2 ratio, tumor necrosis factor-α, and interleukin (IL)-6 level, and increasing IL-10 levels. Significantly reduced brain infarct volume along with a favorable survival ratio was observed for pMCAO mice that received Raffinee, suggesting a neuroprotective potential of Raffinee in cases of acute ischemic stroke by suppressing apoptosis.
Collapse
Affiliation(s)
- Chun-Ching Chiu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Intensive Care Unit, Changhua Christian Hospital, Changhua, Taiwan
| | - Jer-Min Lin
- Ziel Enterprise Co., Ltd., Kaohsiung, Taiwan
| | - Li-Yi Wu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Tsai-Ching Hsu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.,Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan
| | - Bor-Show Tzang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.,Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan.,Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
113
|
Bahr Hosseini M, Hou J, Bikson M, Iacoboni M, Gornbein J, Saver JL. Central Nervous System Electrical Stimulation for Neuroprotection in Acute Cerebral Ischemia: Meta-Analysis of Preclinical Studies. Stroke 2019; 50:2892-2901. [PMID: 31480966 DOI: 10.1161/strokeaha.119.025364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background and Purpose- Brain electrical stimulation, widely studied to facilitate recovery from stroke, has also been reported to confer direct neuroprotection in preclinical models of acute cerebral ischemia. Systematic review of controlled preclinical acute cerebral ischemia studies would aid in planning for initial human clinical trials. Methods- A systematic Medline search identified controlled, preclinical studies of central nervous system electrical stimulation in acute cerebral ischemia. Studies were categorized among 6 stimulation strategies. Three strategies applied different stimulation types to tissues within the ischemic zone (cathodal hemispheric stimulation [CHS], anodal hemispheric stimulation, and pulsed hemispheric stimulation), and 3 strategies applied deep brain stimulation to different neuronal targets remote from the ischemic zone (fastigial nucleus stimulation, subthalamic vasodilator area stimulation, and dorsal periaqueductal gray stimulation). Random-effects meta-analysis assessed electrical stimulation modification of final infarct volume. Study-level risk of bias and intervention-level readiness-for-translation were assessed using formal rating scales. Results- Systematic search identified 28 experiments in 21 studies, including a total of 350 animals, of electrical stimulation in preclinical acute cerebral ischemia. Overall, in animals undergoing electrical stimulation, final infarct volumes were reduced by 37% (95% CI, 34%-40%; P<0.001), compared with control. There was evidence of heterogeneity of efficacy among stimulation strategies (I2=93.1%, Pheterogeneity<0.001). Among the within-ischemic zone stimulation strategies, only CHS significantly reduced the infarct volume (27 %; 95% CI, 22%-33%; P<0.001); among the remote-from ischemic zone approaches, all (fastigial nucleus stimulation, subthalamic vasodilator area stimulation, and dorsal periaqueductal gray stimulation) reduced infarct volumes by approximately half. On formal rating scales, CHS studies had the lowest risk of bias, and CHS had the highest overall quality of intervention-level evidence supporting readiness to proceed to clinical testing. Conclusions- Electrical stimulation reduces final infarct volume across preclinical studies. CHS shows the most robust evidence and is potentially appropriate for progression to early-stage human clinical trial testing as a promising neuroprotective intervention.
Collapse
Affiliation(s)
- Mersedeh Bahr Hosseini
- From the Department of Neurology and Comprehensive Stroke Center (M.B.H., J.H., J.L.S.), David Geffen School of Medicine at UCLA
| | - Jesse Hou
- From the Department of Neurology and Comprehensive Stroke Center (M.B.H., J.H., J.L.S.), David Geffen School of Medicine at UCLA
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York (CCNY) (M.B.)
| | - Marco Iacoboni
- Department of Psychiatry and Biobehavioral Sciences (M.I.), David Geffen School of Medicine at UCLA
| | - Jeffrey Gornbein
- Department of Biomedical Engineering, The City College of New York (CCNY) (M.B.)
| | - Jeffrey L Saver
- From the Department of Neurology and Comprehensive Stroke Center (M.B.H., J.H., J.L.S.), David Geffen School of Medicine at UCLA
| |
Collapse
|
114
|
Li C, Zhang L, Wang C, Teng H, Fan B, Chopp M, Zhang ZG. N-Acetyl-Seryl-Aspartyl-Lysyl-Proline Augments Thrombolysis of tPA (Tissue-Type Plasminogen Activator) in Aged Rats After Stroke. Stroke 2019; 50:2547-2554. [PMID: 31387512 PMCID: PMC6710137 DOI: 10.1161/strokeaha.119.026212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background and Purpose- Stroke is a leading cause of disability worldwide, mainly affecting the elderly. However, preclinical studies in aged ischemic animals are limited. N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) is a naturally occurring tetrapeptide with vascular-protective properties. The present study investigated the effect of AcSDKP on tPA (tissue-type plasminogen activator)-induced thrombolysis in aged rats after ischemic stroke. Methods- Aged male rats (18 months) were subjected to embolic middle cerebral artery occlusion. Rats subjected to 4 hours of middle cerebral artery occlusion were randomized into the following groups: (1) AcSDKP; (2) tPA; (3) AcSDKP in combination with tPA; and (4) saline. Neurological deficits, cerebral microvascular patency and integrity, and infarction were examined at 1 day and 7 days after middle cerebral artery occlusion. In vitro experiments were performed to examine the effect of AcSDKP on aged cerebral endothelial cell permeability. Results- Compared with saline, AcSDKP, or tPA as monotherapy did not have any therapeutic effects, whereas AcSDKP in combination with tPA significantly reduced cerebral tissue infarction and improved neurological outcome without increasing cerebral hemorrhage. Concurrently, the combination treatment significantly augmented microvascular perfusion and reduced thrombosis and blood-brain barrier leakage. In vitro, compared with cerebral endothelial cells from ischemic adult rats, the endothelial cells from ischemic aged rats exhibited significantly increased leakage. AcSDKP suppressed tPA-induced aged endothelial cell leakage and reduced expression of ICAM-1 (intercellular adhesion molecule 1) and NF (nuclear factor)-κB. Conclusions- The present study provides evidence for the therapeutic efficacy of AcSDKP in combination tPA for the treatment of embolic stroke in aged rats at 4 hours after stroke onset. AcSDKP likely acts on cerebral endothelial cells to enhance the benefits of tPA by increasing tissue perfusion and augmenting the integrity of the blood-brain barrier. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| | - Chunyang Wang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| | - Hua Teng
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| | - Baoyan Fan
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
- Department of Physics, Oakland University, Rochester, Michigan, 48309
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202
| |
Collapse
|
115
|
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]
|
116
|
|
117
|
Han QY, Zhang H, Zhang X, He DS, Wang SW, Cao X, Dai YT, Xu Y, Han LJ. dl-3-n-butylphthalide preserves white matter integrity and alleviates cognitive impairment in mice with chronic cerebral hypoperfusion. CNS Neurosci Ther 2019; 25:1042-1053. [PMID: 31334611 PMCID: PMC6698981 DOI: 10.1111/cns.13189] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 01/01/2023] Open
Abstract
Aims Effects of dl‐3‐n‐butylphthalide (NBP) on white matter damage and cognitive impairment in vascular cognitive impairment (VCI) have not been well studied. This study aimed to investigate the effects of NBP treatment on chronic cerebral hypoperfusion‐induced white matter lesions and cognitive dysfunction in mice. Methods Mice were subjected to bilateral common carotid artery stenosis (BCAS) for over 30 days. The cerebral blood flow was detected using a laser Doppler flowmetry. Cognitive functions were assessed by several behavioral tests. We also evaluated the effects of NBP on the blood‐brain barrier (BBB) disruption and reactive astrogliosis, using Evans Blue extravasation, Western blot, CBA, and immunofluorescence in BCAS mice and cultured astrocytes. Results The results indicated that NBP treatment attenuated spatial memory dysfunction while promoted cerebral perfusion and white matter integrity in BCAS mice. Moreover, NBP treatment prevented BBB leakage and damage of endothelial cells, as well as disruption of endothelial tight junctions. Furthermore, NBP administration effectively decreased the number of activated astrocytes and pro‐inflammatory cytokines, as well as the production of MMPs, in BCAS‐induced mice and LPS‐stimulated astrocytes. Conclusion Our results indicated that NBP represents a promising therapy for chronic cerebral hypoperfusion‐induced white matter damage and cognitive impairment.
Collapse
Affiliation(s)
- Qin-Yu Han
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - He Zhang
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xi Zhang
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Dong-Sheng He
- Department of Neurology, The Northern Area of Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Sun-Wei Wang
- Department of Neurology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yu-Tian Dai
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Li-Juan Han
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
118
|
Mu HF, Gao XG, Li SC, Wei PJ, Zhao YF, Zhang WT, Wang Y, Gao YQ. Distinctive functional deficiencies in axonal conduction associated with two forms of cerebral white matter injury. CNS Neurosci Ther 2019; 25:1018-1029. [PMID: 31140740 PMCID: PMC6698976 DOI: 10.1111/cns.13155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/09/2019] [Accepted: 05/11/2019] [Indexed: 01/19/2023] Open
Abstract
Aims This study determines whether assessment with compound action potentials (CAPs) can distinguish two different forms of cerebral white matter injury at the functional levels. Methods A pure demyelination model was induced in C57/BL6 adult mice by dietary supplementation of cuprizone (0.2%) for 6 weeks. Callosal L‐N5‐(1‐Iminoethyl) ornithine (L‐NIO) hydrochloride (27 mg/mL) was injected into the corpus callosum (CC) to induce a focal white matter stroke (WMS), resulting in both demyelination and axonal injury. White matter integrity was assessed by performing CAP recording, electron microscopy, and immunohistological and luxol fast blue (LFB) staining. Results Immunohistological and electron microscopic analyses confirmed the induction of robust demyelination in CC with cuprizone, and mixed demyelination and axonal damage with L‐NIO. Electrophysiologically, cuprizone‐induced demyelination significantly reduced the amplitude of negative peak 1 (N1), but increased the amplitude of negative peak 2 (N2), of the CAPs compared to the sham controls. However, cuprizone did not affect the axonal conduction velocity. In contrast, the amplitude and area of both N1 and N2 along with N1 axonal conduction velocity were dramatically decreased in L‐NIO‐induced WMS. Conclusions Concertedly, parameters of the CAPs offer a novel functional assessment strategy for cerebral white matter injury in rodent models.
Collapse
Affiliation(s)
- Hong-Feng Mu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu-Guang Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Si-Cheng Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng-Ju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong-Fang Zhao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Ting Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan-Qin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
119
|
Xie YZ, Zhang XJ, Zhang C, Yang Y, He JN, Chen YX. Protective effects of leonurine against ischemic stroke in mice by activating nuclear factor erythroid 2-related factor 2 pathway. CNS Neurosci Ther 2019; 25:1006-1017. [PMID: 31087454 PMCID: PMC6698971 DOI: 10.1111/cns.13146] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Aims Leonurine has been shown to trigger antioxidant responses during ischemic stroke, and nuclear factor erythroid 2‐related factor 2 (Nrf‐2) imparts protective effects against oxidative injury. The present study has determined that leonurine prevents ischemic injury of brain tissues via Nrf‐2 pathway activation. Methods Male ICR mice and Nrf‐2−/− mice were subjected to permanent middle cerebral artery occlusion (pMCAO) and received leonurine treatment at 2 hours after pMCAO by intraperitoneal injection. Neurological deficit scores as well as infarct volume were assessed to determine the neuroprotective role of leonurine. Nrf‐2 was investigated using Western blotting and real‐time polymerase chain reaction (RT‐PCR) analysis to elucidate the neuroprotective mechanism of leonurine. Commercial kits were employed to determine reactive oxygen species (ROS), superoxide (SOD), catalase (CAT), glutathione peroxidase (GSH‐Px), malonaldehyde (MDA), and glutathione (GSH). Vascular endothelial growth factor (VEGF) was evaluated by Western blotting and RT‐PCR analysis, and VEGF was localized using immunofluorescence. Results The application of leonurine on ICR mice resulted in an improvement in neurological deficit scores and a reduction in infarct volume. Leonurine upregulated nuclear Nrf‐2 protein and increased total Nrf‐2 protein expression and mRNA levels. Leonurine regulated SOD, MDA, CAT, GSH, and GSH‐Px, and it significantly inhibited ROS production in ICR mice. Leonurine improved VEGF expression and increased VEGF expression in neurons, astrocytes, and endothelial cells. However, leonurine had no obvious beneficial effects on Nrf‐2−/− mice. Conclusions Leonurine exerted neuroprotective effects, promoted antioxidant responses, and upregulated VEGF expression by activating the Nrf‐2 pathway.
Collapse
Affiliation(s)
- Yan-Zhao Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Jian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yang Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Jun-Na He
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yan-Xia Chen
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
120
|
Protease-independent action of tissue plasminogen activator in brain plasticity and neurological recovery after ischemic stroke. Proc Natl Acad Sci U S A 2019; 116:9115-9124. [PMID: 30996120 DOI: 10.1073/pnas.1821979116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wild-type mice, suggesting that endogenously expressed tPA promotes long-term neurological recovery after stroke. In tPA knockout mice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective as wild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke.
Collapse
|
121
|
Wang Z, Yang W. Impaired capacity to restore proteostasis in the aged brain after ischemia: Implications for translational brain ischemia research. Neurochem Int 2018; 127:87-93. [PMID: 30599146 DOI: 10.1016/j.neuint.2018.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022]
Abstract
Brain ischemia induced by cardiac arrest or ischemic stroke is a severe form of metabolic stress that substantially disrupts cellular homeostasis, especially protein homeostasis (proteostasis). As proteostasis is fundamental for cellular and organismal health, cells have developed a complex network to restore proteostasis impaired by stress. Many components of this network - including ubiquitination, small ubiquitin-like modifier (SUMO) conjugation, autophagy, and the unfolded protein response (UPR) - are activated in the post-ischemic brain, and play a crucial role in cell survival and recovery of neurologic function. Importantly, recent studies have shown that ischemia-induced activation of these proteostasis-related pathways in the aged brain is impaired, indicating an aging-related decline in the self-healing capacity of the brain. This impaired capacity is a significant factor for consideration in the field of brain ischemia because the vast majority of cardiac arrest and stroke patients are elderly. In this review, we focus on the effects of aging on these critical proteostasis-related pathways in the brain, and discuss their implications in translational brain ischemia research.
Collapse
Affiliation(s)
- Zhuoran Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
122
|
Lo EH, Moskowitz MA. Introduction to the special issue honoring Richard Traystman. J Cereb Blood Flow Metab 2018; 38:2055-2056. [PMID: 30497333 PMCID: PMC6282219 DOI: 10.1177/0271678x18803702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Eng H Lo
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Michael A Moskowitz
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
123
|
Post-stroke administration of omega-3 polyunsaturated fatty acids promotes neurovascular restoration after ischemic stroke in mice: Efficacy declines with aging. Neurobiol Dis 2018; 126:62-75. [PMID: 30218758 DOI: 10.1016/j.nbd.2018.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023] Open
Abstract
Post-stroke treatment with omega-3 polyunsaturated fatty acids (n-3 PUFAs) may be a promising therapy in young animals but this has not been tested in aged subjects, a population at most risk of ischemic stroke. Herein we examined the therapeutic efficacy of n-3 PUFAs after distal middle cerebral artery occlusion (dMCAO) in young (10-12 weeks old) and aged (18 months old) mice. Post-ischemic mice were randomly assigned to 4 groups that received: 1) regular food with low content of n-3 PUFAs, 2) intraperitoneal docosahexaenoic acid (DHA, a major component of n-3 PUFAs) injections, 3) Fish oil (FO, containing high concentration of n-3 PUFAs) dietary supplement, or 4) combined treatment with DHA and FO dietary supplement. Long-term neurorestoration induced by n-3 PUFA post-stroke administration and its underlying mechanism(s) were analyzed up to 35 days after dMCAO. Aged mice showed more severe neurological deficits than young mice after dMCAO with histological lesions extended to the striatum. Notably, post-stroke treatment with combined DHA injections and FO dietary supplementation was more effective in reducing brain injury and improving sensorimotor function in aged mice than either treatment alone, albeit to a lesser extent than in the young mice. Unlike the improvement in spatial cognitive function observed in young mice, the combined treatment regimen failed to improve cognitive function in aged mice. The reduction in stroke-induced neurological deficits with n-3 PUFA post-treatment was associated with enhanced angiogenesis, oligodendrogenesis, neuron survival and white matter restoration. Together, these results indicate that the neurological benefits of n-3 PUFA administration after stroke extend to older animals and are associated with improved neuronal survival and brain remodeling, therefore suggesting that post-stroke administration of n-3 PUFAs is a viable clinically relevant treatment option against stroke.
Collapse
|