1
|
Lin Y, Pang Q, Shi Y, Chen X, Tu F. Long noncoding RNA MALAT1 promotes angiogenesis through the caveolin-1/VEGF pathway after cerebral ischemic injury. Neuroreport 2025; 36:350-363. [PMID: 40203233 DOI: 10.1097/wnr.0000000000002157] [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: 04/11/2025]
Abstract
The long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) might protect against cerebral ischemic injury. This study explored MALAT1's function in ischemic stroke and whether it acts through the caveolin-1/vascular endothelial growth factor (VEGF) pathway. A mouse model of middle cerebral artery occlusion/reperfusion (MCAO/R) and a human brain microvascular endothelial cell (HBMEC) model of oxygen-glucose deprivation/reoxygenation (OGD/R) were established. Lentiviral vectors for MALAT1 knockdown, caveolin-1 knockdown, and MALAT1 overexpression were used for gene regulation studies. Neurological deficits, endothelial cell proliferation, cell apoptosis, cell viability, in vitro angiogenesis, cell migration, and the expression of related gene and protein were evaluated using the Zea Longa five-point scale, VEGF receptor 2/CD34 double immunofluorescence, TdT-mediated dUTP nick end labeling staining, cell counting kit-8 assay, tube formation assay, transwell assay, quantitative real time PCR, and western blot. In mouse MCAO/R model and HBMEC OGD/R model, the expression levels of MALAT1, caveolin-1, and VEGF were significantly upregulated compared to the control group. In vivo, downregulation of MALAT1 expression exacerbated cerebral ischemic injury as manifested by severe neurological deficits, larger infarct volume, increased apoptosis, decreased numbers of VEGF receptor 2+/CD34+ endothelial progenitor cells, increased cell apoptosis, and the downregulation of caveolin-1 and VEGF. Conversely, overexpression of MALAT1 partially reversed the inhibition of cell migration and tubule formation by caveolin-1 gene downregulation, and restored in the expression of caveolin-1 and VEGF. MALAT1 promotes angiogenesis after cerebral ischemic injury, likely in part via the caveolin-1/VEGF pathway. Thus, MALAT1 may serve as a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yao Lin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
2
|
Zhou MY, Zhang YJ, Ding HM, Wu WF, Cai WW, Wang YQ, Geng DQ. Diprotin A TFA Exerts Neurovascular Protection in Ischemic Cerebral Stroke. Front Neurosci 2022; 16:861059. [PMID: 35615279 PMCID: PMC9125038 DOI: 10.3389/fnins.2022.861059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIt has been established that the dipeptidyl peptidase-4 (DPP-4) inhibitor Diprotin A TFA can reduce vascular endothelial (VE)-cadherin disruption by inhibiting the increase in cleaved β-catenin in response to hypoxia, thereby protecting the vascular barrier of human umbilical vein endothelial cells. In this study, we sought to investigate the possible effect of Diprotin A TFA on the VE barrier after cerebral ischemic stroke in mice.MethodsC57BL/6J mice were divided into five groups, namely, (1) sham, (2) stroke, (3) stroke + dimethyl sulfoxide (DMSO), (4) stroke + Diprotin A TFA, and (5) stroke + Diprotin A TFA + XAV-939. First, the cerebral ischemia model was established by photothrombotic ischemia, followed by intraperitoneal injection with Diprotin A TFA and XAV-939 at doses of 70 μg/kg and 40 mg/kg 30 min once in the morning and once in the evening for 3 days. Immunofluorescence staining and Western blot methods were used to analyze the expression of vascular and blood-brain barrier (BBB)-associated molecular markers in the peri-infarct area.ResultsCompared with the vehicle control group, we found that mice injected with Diprotin A TFA exhibited reduced cerebral infarction volume, increased vascular area and length around the brain injury, increased pericyte and basement membrane coverage, upregulated expression of BBB tight junction proteins, and improved their BBB permeability, whereas the group injected with both drug and inhibitor exhibited significantly aggravated vascular injury and BBB permeability.ConclusionDiprotin A TFA can reduce VE-cadherin disruption by inhibiting ischemia-hypoxia-induced β-catenin cleavage to protect blood vessels.
Collapse
Affiliation(s)
- Ming-Yue Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ya-Jie Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hong-Mei Ding
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Nanjing Medical University, Nanjing, China
| | - Wei-Feng Wu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei-Wei Cai
- Department of Neurology, The Third Hospital of Huai'an, Huai'an, China
| | - Yan-Qiang Wang
- Department of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Yan-Qiang Wang
| | - De-Qin Geng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Nanjing Medical University, Nanjing, China
- De-Qin Geng
| |
Collapse
|
3
|
Scott LA, Dickie BR, Rawson SD, Coutts G, Burnett TL, Allan SM, Parker GJ, Parkes LM. Characterisation of microvessel blood velocity and segment length in the brain using multi-diffusion-time diffusion-weighted MRI. J Cereb Blood Flow Metab 2021; 41:1939-1953. [PMID: 33325766 PMCID: PMC8323340 DOI: 10.1177/0271678x20978523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multi-diffusion-time diffusion-weighted MRI can probe tissue microstructure, but the method has not been widely applied to the microvasculature. At long diffusion-times, blood flow in capillaries is in the diffusive regime, and signal attenuation is dependent on blood velocity (v) and capillary segment length (l). It is described by the pseudo-diffusion coefficient (D*=vl/6) of intravoxel incoherent motion (IVIM). At shorter diffusion-times, blood flow is in the ballistic regime, and signal attenuation depends on v, and not l. In theory, l could be estimated using D* and v. In this study, we compare the accuracy and repeatability of three approaches to estimating v, and therefore l: the IVIM ballistic model, the velocity autocorrelation model, and the ballistic approximation to the velocity autocorrelation model. Twenty-nine rat datasets from two strains were acquired at 7 T, with b-values between 0 and 1000 smm-2 and diffusion times between 11.6 and 50 ms. Five rats were scanned twice to assess scan-rescan repeatability. Measurements of l were validated using corrosion casting and micro-CT imaging. The ballistic approximation of the velocity autocorrelation model had lowest bias relative to corrosion cast estimates of l, and had highest repeatability.
Collapse
Affiliation(s)
- Lauren A Scott
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Ben R Dickie
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Shelley D Rawson
- The Henry Royce Institute, Department of Materials, The University of Manchester, Manchester, UK
| | - Graham Coutts
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Timothy L Burnett
- The Henry Royce Institute, Department of Materials, The University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Geoff Jm Parker
- The Henry Royce Institute, Department of Materials, The University of Manchester, Manchester, UK.,Bioxydyn Limited, Manchester, UK
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Zheng J, Wei Z, Yang K, Lu Y, Lu P, Zhao J, Du Y, Zhang H, Li R, Lei S, Lv H, Chen X, Liu Y, Chen YM, Zhang Q, Zhang P. Neural Stem Cell-Laden Self-Healing Polysaccharide Hydrogel Transplantation Promotes Neurogenesis and Functional Recovery after Cerebral Ischemia in Rats. ACS APPLIED BIO MATERIALS 2021; 4:3046-3054. [PMID: 35014393 DOI: 10.1021/acsabm.0c00934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Exploring a strategy to effectively repair cerebral ischemic injury is a critical requirement for neuroregeneration. Herein, we transplanted a neural stem cell (NSC)-laden self-healing and injectable hydrogel into the brains of ischemic rats and evaluated its therapeutic effects. We observed an improvement in neurological functions in rats transplanted with the NSC-laden hydrogel. This strategy is sufficiently efficient to support neuroregeneration evidenced by NSC proliferation, differentiation, and athletic movement recovery of rats. This therapeutic effect relates to the inhibition of the astrocyte reaction and the increased expression of vascular endothelial growth factor. This work provides a novel approach to repair cerebral ischemic injury.
Collapse
Affiliation(s)
- Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Zhao Wei
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Kuan Yang
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Jingyi Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Yin Du
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University, Xi'an 710016, P. R. China
| | - Yong Mei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Qiqing Zhang
- Institute of Biomedical Engineering, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen 518020, P. R. China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P. R. China
| |
Collapse
|
5
|
Prakash R, Izraely S, Thareja NS, Lee RH, Rappaport M, Kawaguchi R, Sagi-Assif O, Ben-Menachem S, Meshel T, Machnicki M, Ohe S, Hoon DS, Coppola G, Witz IP, Carmichael ST. Regeneration Enhances Metastasis: A Novel Role for Neurovascular Signaling in Promoting Melanoma Brain Metastasis. Front Neurosci 2019; 13:297. [PMID: 31024232 PMCID: PMC6465799 DOI: 10.3389/fnins.2019.00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
Neural repair after stroke involves initiation of a cellular proliferative program in the form of angiogenesis, neurogenesis, and molecular growth signals in the surrounding tissue elements. This cellular environment constitutes a niche in which regeneration of new blood vessels and new neurons leads to partial tissue repair after stroke. Cancer metastasis has similar proliferative cellular events in the brain and other organs. Do cancer and CNS tissue repair share similar cellular processes? In this study, we identify a novel role of the regenerative neurovascular niche induced by stroke in promoting brain melanoma metastasis through enhancing cellular interactions with surrounding niche components. Repair-mediated neurovascular signaling induces metastatic cells to express genes crucial to metastasis. Mimicking stroke-like conditions in vitro displays an enhancement of metastatic migration potential and allows for the determination of cell-specific signals produced by the regenerative neurovascular niche. Comparative analysis of both in vitro and in vivo expression profiles reveals a major contribution of endothelial cells in mediating melanoma metastasis. These results point to a previously undiscovered role of the regenerative neurovascular niche in shaping the tumor microenvironment and brain metastatic landscape.
Collapse
Affiliation(s)
- Roshini Prakash
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sivan Izraely
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rex H Lee
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maya Rappaport
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shlomit Ben-Menachem
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Machnicki
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shuichi Ohe
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Dave S Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isaac P Witz
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
6
|
Baker EW, Kinder HA, West FD. Neural stem cell therapy for stroke: A multimechanistic approach to restoring neurological function. Brain Behav 2019; 9:e01214. [PMID: 30747485 PMCID: PMC6422715 DOI: 10.1002/brb3.1214] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/02/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Neural stem cells (NSCs) have demonstrated multimodal therapeutic function for stroke, which is the leading cause of long-term disability and the second leading cause of death worldwide. In preclinical stroke models, NSCs have been shown to modulate inflammation, foster neuroplasticity and neural reorganization, promote angiogenesis, and act as a cellular replacement by differentiating into mature neural cell types. However, there are several key technical questions to address before NSC therapy can be applied to the clinical setting on a large scale. PURPOSE OF REVIEW In this review, we will discuss the various sources of NSCs, their therapeutic modes of action to enhance stroke recovery, and considerations for the clinical translation of NSC therapies. Understanding the key factors involved in NSC-mediated tissue recovery and addressing the current translational barriers may lead to clinical success of NSC therapy and a first-in-class restorative therapy for stroke patients.
Collapse
Affiliation(s)
- Emily W Baker
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Holly A Kinder
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Franklin D West
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| |
Collapse
|
7
|
Chuang BTC, Liu X, Lundberg AJ, Toung TJK, Ulatowski JA, Koehler RC. Refinement of embolic stroke model in rats: Effect of post-embolization anesthesia duration on arterial blood pressure, cerebral edema and mortality. J Neurosci Methods 2018; 307:8-13. [PMID: 29935198 DOI: 10.1016/j.jneumeth.2018.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Injection of a clot into the internal carotid artery is an experimental model of ischemic stroke that is considered to closely mimic embolic stroke in humans. In this model, the common carotid artery typically remains temporarily occluded to permit time for stabilization of the clot in the middle cerebral artery. However, the associated lengthening of the anesthesia duration could affect arterial blood pressure and stroke outcome. NEW METHOD We refined the model by examining how increasing isoflurane anesthesia duration from 30 to 60 min after clot embolization affects mortality, infarct volume, edema, blood-brain barrier permeability, and the 8-h post-ischemic time course of blood pressure, which has not been reported previously in this model. RESULTS We found that arterial pressure increased after discontinuing anesthesia in both embolized groups and that the increase was greater than in the corresponding non-embolized sham-operated rats. At 24 h, the group with 60-min post-ischemia anesthesia exhibited greater brain water content and a greater ipsilateral-to-contralateral ratio of extravasated Evans blue dye. Mortality was greater in the 60-min group, but infarct volume among survivors was not different from that in the 30-min anesthesia group. COMPARISON WITH EXISTING METHODS This study refines the embolic stroke model by demonstrating the importance of minimizing the duration of anesthesia after embolization. CONCLUSIONS These data indicate that early discontinuation of isoflurane anesthesia after clot embolization permits an earlier hypertensive response that limits edema formation and mortality without significantly affecting infarct volume in survivors, thereby decreasing the required number of animals.
Collapse
Affiliation(s)
- Bryan T C Chuang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Anesthesiology, Saint Mary's Hospital Luodong, Taiwan
| | - Xiaoguang Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alexander J Lundberg
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Tommy J K Toung
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - John A Ulatowski
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Delayed histochemical alterations within the neurovascular unit due to transient focal cerebral ischemia and experimental treatment with neurotrophic factors. PLoS One 2017; 12:e0174996. [PMID: 28445478 PMCID: PMC5405989 DOI: 10.1371/journal.pone.0174996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 03/17/2017] [Indexed: 02/03/2023] Open
Abstract
Current stroke therapy is focused on recanalizing strategies, but neuroprotective co-treatments are still lacking. Modern concepts of the ischemia-affected neurovascular unit (NVU) and surrounding penumbra emphasize the complexity during the transition from initial damaging to regenerative processes. While early treatment with neurotrophic factors was shown to result in lesion size reduction and blood-brain barrier (BBB) stabilization, cellular consequences from these treatments are poorly understood. This study explored delayed cellular responses not only to ischemic stroke, but also to an early treatment with neurotrophic factors. Rats underwent 60 minutes of focal cerebral ischemia. Fluorescence labeling was applied to sections from brains perfused 7 days after ischemia. Analyses focused on NVU constituents including the vasculature, astrocytes and microglia in the ischemic striatum, the border zone and the contralateral hemisphere. In addition to histochemical signs of BBB breakdown, a strong up-regulation of collagen IV and microglia activation occurred within the ischemic core with simultaneous degradation of astrocytes and their endfeet. Activated astroglia were mainly depicted at the border zone in terms of a glial scar formation. Early treatment with pigment epithelium-derived factor (PEDF) resulted in an attenuation of the usually up-regulated collagen IV-immunoreactivity. However, glial activation was not influenced by treatment with PEDF or the epidermal growth factor (EGF). In conclusion, these data on ischemia-induced cellular reactions within the NVU might help to develop treatments addressing the transition from injury towards regeneration. Thereby, the integrity of the vasculature in close relation to neighboring structures like astrocytes appears as a promising target.
Collapse
|
9
|
Lake EM, Mester J, Thomason LAM, Adams C, Bazzigaluppi P, Koletar M, Janik R, Carlen P, McLaurin J, Stanisz GJ, Stefanovic B. Modulation of the peri-infarct neurogliovascular function by delayed COX-1 inhibition. J Magn Reson Imaging 2016; 46:505-517. [DOI: 10.1002/jmri.25541] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/20/2016] [Indexed: 11/06/2022] Open
Affiliation(s)
- Evelyn M.R. Lake
- Department of Radiology and Biomedical Imaging; Yale University; New Haven Connecticut USA
- Department of Medical Biophysics; University of Toronto; Ontario Canada
| | - James Mester
- Department of Medical Biophysics; University of Toronto; Ontario Canada
| | - Lynsie AM Thomason
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
| | - Conner Adams
- Department of Medical Biophysics; University of Toronto; Ontario Canada
| | - Paolo Bazzigaluppi
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
- Fundamental Neurobiology, Toronto Western Research Institute; Toronto Ontario Canada
| | - Margaret Koletar
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
| | - Rafal Janik
- Department of Medical Biophysics; University of Toronto; Ontario Canada
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
| | - Peter Carlen
- Fundamental Neurobiology, Toronto Western Research Institute; Toronto Ontario Canada
| | - JoAnne McLaurin
- Biological Science, Sunnybrook Research Institute; Toronto Ontario Canada
- Department of Laboratory Medicine and Pathobiology; University of Toronto; Ontario Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery; Ottawa Ontario Canada
| | - Greg J Stanisz
- Department of Medical Biophysics; University of Toronto; Ontario Canada
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
- Department of Neurosurgery and Pediatric Neurosurgery; Medical University; Lublin Poland
| | - Bojana Stefanovic
- Department of Medical Biophysics; University of Toronto; Ontario Canada
- Physical Sciences, Sunnybrook Research Institute; Toronto Ontario Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery; Ottawa Ontario Canada
| |
Collapse
|
10
|
Hawkes CA, Michalski D, Anders R, Nissel S, Grosche J, Bechmann I, Carare RO, Härtig W. Stroke-induced opposite and age-dependent changes of vessel-associated markers in co-morbid transgenic mice with Alzheimer-like alterations. Exp Neurol 2013; 250:270-81. [DOI: 10.1016/j.expneurol.2013.09.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 09/17/2013] [Accepted: 09/21/2013] [Indexed: 10/26/2022]
|
11
|
Prakash R, Li W, Qu Z, Johnson MA, Fagan SC, Ergul A. Vascularization pattern after ischemic stroke is different in control versus diabetic rats: relevance to stroke recovery. Stroke 2013; 44:2875-82. [PMID: 23920018 DOI: 10.1161/strokeaha.113.001660] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND PURPOSE Pre-existing diabetes mellitus worsens brain functionality in ischemic stroke. We have previously shown that type 2 diabetic rats exhibit enhanced dysfunctional cerebral neovascularization and when these rats are subjected to cerebral ischemic reperfusion injury develop hemorrhagic transformation and greater neurological deficits. However, our knowledge of vascular and functional plasticity during the recovery phase of diabetic stroke is limited. This study tested the hypothesis that vascular repair is impaired in the poststroke period in diabetes mellitus, and this is associated with poor sensorimotor and cognitive function. We further hypothesized that glycemic control prevents impaired vascularization and improves functional outcome in diabetes mellitus. METHODS Vascularization was assessed in the ipsilateral and contralateral hemispheres in control, diabetes mellitus and diabetes mellitus plus metformin groups 14 days after ischemic reperfusion injury, as well as in respective sham controls. Three-dimensional reconstruction of the fluorescein isothiocyanate (FITC)-stained vasculature was achieved by confocal microscopy, and stereological parameters, including vascular volume and surface area, were measured. Astrogliosis was determined by glial fibrillary acidic protein staining. The relative rates of sensorimotor recovery, cognitive decline, and spontaneous activity were assessed. RESULTS Vascular density in the peri-infarct area was significantly reduced in diabetes mellitus, whereas there was reparative neovascularization in control rats. Astroglial swelling and reactivity were more pronounced in diabetic stroke compared with control stroke. Diabetes mellitus blunted sensorimotor recovery and also exacerbated anxiety-like symptoms and cognitive deficits. Glycemic control started after stroke partially prevented these changes. CONCLUSIONS Diabetes mellitus impairs poststroke reparative neovascularization and impedes the recovery. Glycemic control after stroke can improve neurovascular repair and improve functional outcome.
Collapse
Affiliation(s)
- Roshini Prakash
- From the Department of Physiology, Charlie Norwood Veterans Administration Medical Center, Augusta, GA (R.P., W.L., S.C.F., A.E.); Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA (R.P., S.C.F., A.E.); and Departments of Physiology (W.L., Z.Q., A.E.), and Biostatistics (M.A.J.), Georgia Regents University, Augusta, GA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
It is being increasingly suggested that the microcirculation, which is known to be in a large part responsible for maintaining an adequate and constant microenvironment for function of the central nervous system, functions as part of a neurovascular unit. The neurovascular unit includes neurons, astrocytes and elements of capillaries. The cerebral circulation exhibits unique functional characteristics and critical elements for the pathogenesis of cerebrovascular disease. For example, the blood-brain barrier formed by epithelial-like high resistance tight junctions within the endothelium is a key feature of microvessels of the central nervous system. Alterations in the microcirculation after ischemia/reperfusion include disruption of the blood-brain barrier, edema and swelling of perivascular astrocyte foot processes, decrease in arteriole endothelium-dependent relaxation and reduced inwardly-rectifying potassium channel function, altered expression of proteases and matrix metalloproteinases, increased inflammatory mediators and inflammation. Experiments studying the microcirculation in ischemia are few compared with those examining neuroprotection, although the two overlap because protection of the microcirculation might achieve some degree of neuroprotection and both processes may be mediated by at least some mechanisms in common.
Collapse
Affiliation(s)
- Masataka Takahashi
- Section of Neurosurgery, Department of Surgery, University of Chicago Medical Center and Pritzker School of Medicine, Chicago, IL 60637, USA
| | | |
Collapse
|
13
|
Bosomtwi A, Chopp M, Zhang L, Zhang ZG, Lu M, Jiang Q. Mean microvessel segment length and radius after embolic stroke: Comparison of magnetic resonance imaging (MRI) and laser scanning confocal microscopy (LSCM). Brain Res 2011; 1381:217-27. [PMID: 21237138 DOI: 10.1016/j.brainres.2011.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 10/18/2022]
Abstract
We offer a new means of noninvasively assessing mean microvessel segment length and radius after ischemic stroke. This method involves measuring changes in T₂ and T₂⁎ after injecting an intravascular superparamagnetic iron oxide contrast agent and result was verified using laser scanning confocal microscopy (LSCM) of both normal brain tissue and the ischemic recovery region. Embolic stroke was induced in 8 male Wistar rats and magnetic resonance imaging (MRI) performed 1 day and 6 weeks later. On MRI taken at 6 weeks, MRI of the recovery region revealed a significant increase in mean vessel size index (VSI) (5.75 ± 0.54 vs 4.81 ± 0.3 9μm; p < 0.001) and decrease in mean segment length (MSL) (16.61 ± 2.33 vs 26.52 ± 3.20 μm; p < 0.001) compared to the normal contralateral hemisphere, comparable with published values. There was also a significant correlation between MSL and VSI measured by MRI vs LSCM in the recovery region and normal contralateral hemisphere (p < 0.001). Our data suggest that a) morphological changes in the microvasculature can be measured noninvasively using MRI, and b) both MRI and LSCM give comparable information about both of these important parameters.
Collapse
Affiliation(s)
- Asamoah Bosomtwi
- Imaging Center, Yerkes National Primate Center, Emory University, Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
14
|
Zhang P, Li J, Liu Y, Chen X, Lu H, Kang Q, Li W, Gao M. Human embryonic neural stem cell transplantation increases subventricular zone cell proliferation and promotes peri-infarct angiogenesis after focal cerebral ischemia. Neuropathology 2010; 31:384-91. [PMID: 21175862 DOI: 10.1111/j.1440-1789.2010.01182.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neurogenesis and angiogenesis are two important processes that may contribute to the repair of brain injury after stroke. This study was designed to investigate whether transplantation of human embryonic neural stem cells (NSCs) into cortical peri-infarction 24h after ischemia effects cell proliferation in the subventricular zone (SVZ) and angiogenesis in the peri-infarct zone. NSCs were prepared from embryonic human brains at 8 weeks gestation. Focal cerebral ischemia was induced by permanent occlusion of the middle cerebral artery of adult rats. Animals were randomly divided into two groups (n=30, each) at 24h after ischemia: NSC-grafted and medium-grafted groups. Toluidine blue staining and 5'-bromo-2'-deoxyuridine (BrdU) or von Willebrand factor (vWF) immunohistochemistry were performed at 7, 14 and 28 days after transplantation. NSC transplantation increased the number of BrdU-positive cells in the ischemic ipsilateral SVZ compared with the medium control at 7 days (P<0.01). This difference in SVZ cell proliferation persisted at 14 days (P<0.01), but was not significant at 28 days (P>0.05). In addition, angiogenesis, as indicated by BrdU and vWF staining in cortical peri-infarct regions, was augmented by 46% and 65% in NSC-grafted rats versus medium-grafted rats at 7 and 14 days, respectively (P<0.05). However, this increase became non-significant at 28 days (P>0.05). Our results indicate that NSC transplantation enhances endogenous cell proliferation in the SVZ and promotes angiogenesis in the peri-infarct zone, even if it is performed in the acute phase of ischemic injury.
Collapse
Affiliation(s)
- Pengbo Zhang
- Department of Anesthesiology, Second Affiliated Hospital Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Mostany R, Chowdhury TG, Johnston DG, Portonovo SA, Carmichael ST, Portera-Cailliau C. Local hemodynamics dictate long-term dendritic plasticity in peri-infarct cortex. J Neurosci 2010; 30:14116-26. [PMID: 20962232 PMCID: PMC6634780 DOI: 10.1523/jneurosci.3908-10.2010] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 08/16/2010] [Accepted: 08/19/2010] [Indexed: 11/21/2022] Open
Abstract
Changes in dendritic spine turnover are a major mechanism of experience-dependent plasticity in the adult neocortex. Dendritic spine plasticity may also contribute to functional recovery after stroke, but in that setting its expression may be complicated by alterations in local tissue perfusion, especially around the infarct. Using adult Thy-1 GFP-M mice, we simultaneously recorded long-term spine dynamics in apical dendrites from layer 5 pyramidal cells and blood flow from surrounding capillaries with in vivo two-photon microscopy in peri-infarct cortex before and after unilateral middle cerebral artery occlusion. Blood flow in peri-infarct cortex decreased significantly immediately after stroke and improved gradually over time, in a distance-dependent manner from the epicenter of the infarct. However, local tissue perfusion was never fully restored even after a 3 month recovery period. On average, surviving layer 5 pyramidal neurons experienced a ∼20% decrease in spine density acutely after stroke but eventually recovered. The dynamics of this improvement were different depending on the degree of tissue perfusion acutely after arterial occlusion. Cells in ischemic areas closer to the infarct returned to normal spine density levels slowly by retaining spines, while cells in more remote regions with preserved blood flow recovered faster by adding more spines, eventually surpassing baseline spine density by 15%. Our data suggest that maintaining tissue perfusion in the area surrounding the infarct could hasten or augment synaptic plasticity and functional recovery after stroke.
Collapse
Affiliation(s)
- Ricardo Mostany
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Guo X, Liu L, Zhang M, Bergeron A, Cui Z, Dong JF, Zhang J. Correlation of CD34+ cells with tissue angiogenesis after traumatic brain injury in a rat model. J Neurotrauma 2010; 26:1337-44. [PMID: 19226208 DOI: 10.1089/neu.2008.0733] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Increasing evidence suggests that circulating endothelial progenitor cells, which are a subpopulation of hematopoietic progenitor CD34(+) cells, play a critical role in neovascularization and tissue repair. We have tested the hypothesis that traumatic brain injury (TBI) could mobilize CD34(+) cells to peripheral blood and brain tissue, a process critical for vascular repair, in a rat model of TBI. Male Wistar rats were subjected to controlled fluid percussion. Blood and brain tissue were collected before and after TBI to measure the levels of CD34(+) cells in peripheral blood and to detect their accumulation in the damaged cerebral tissue. Compared with surgery controls, CD34(+) cells significantly increased in the peripheral blood and accumulated in the brain tissue of TBI rats. Immunohistochemistry detected new vessels with incomplete CD34(+) endothelial-like cell lining and an increased number of microvessels in the injured and surrounding tissue. The results demonstrate a close correlation between an increase in circulating CD34(+) cells in response to traumatic injury and angiogenesis in TBI rat brain. They also suggest that transplantation of CD34(+) cells or augmentation of endogenous CD34(+) cells may be a novel therapeutic approach for patients with TBI.
Collapse
Affiliation(s)
- Xinbin Guo
- Department of Neurosurgery, Tianjin Medical University, Laboratory of TJIVR, Tianjin Neurological Institute, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Zhu HF, Wan D, Luo Y, Zhou JL, Chen L, Xu XY. Catalpol increases brain angiogenesis and up-regulates VEGF and EPO in the rat after permanent middle cerebral artery occlusion. Int J Biol Sci 2010; 6:443-53. [PMID: 20827397 PMCID: PMC2935667 DOI: 10.7150/ijbs.6.443] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 08/01/2010] [Indexed: 11/05/2022] Open
Abstract
To investigate the role and mechanism of catalpol in brain angiogenesis in a rat model of stroke, the effect of catalpol (5 mg/kg; i.p) or vehicle administered 24 hours after permanent middle cerebral artery occlusion (pMCAO) on behavior, angiogenesis, ultra-structural integrity of brain capillary endothelial cells, and expression of EPO and VEGF were assessed. Repeated treatments with Catalpol reduced neurological deficits and significantly improved angiogenesis, while significantly increasing brain levels of EPO and VEGF without worsening BBB edema. These results suggested that catalpol might contribute to infarcted-brain angiogenesis and ameliorate the edema of brain capillary endothelial cells (BCECs) by upregulating VEGF and EPO coordinately.
Collapse
Affiliation(s)
- Hui-Feng Zhu
- School of Pharmaceutical Sciences & School of Chinese Medicine, Southwest University, Chongqing 400715, China.
| | | | | | | | | | | |
Collapse
|
18
|
Zhu M, Bi X, Jia Q, Shangguan S. The possible mechanism for impaired angiogenesis after transient focal ischemia in type 2 diabetic GK rats: Different expressions of angiostatin and vascular endothelial growth factor. Biomed Pharmacother 2010; 64:208-13. [DOI: 10.1016/j.biopha.2009.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 08/24/2009] [Indexed: 01/22/2023] Open
|
19
|
Yang JP, Liu HJ, Wang ZL, Cheng SM, Cheng X, Xu GL, Liu XF. The dose-effectiveness of intranasal VEGF in treatment of experimental stroke. Neurosci Lett 2009; 461:212-6. [PMID: 19559076 DOI: 10.1016/j.neulet.2009.06.060] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 05/26/2009] [Accepted: 06/19/2009] [Indexed: 01/19/2023]
Abstract
The aim of the present study was to assess the dose-effectiveness of intranasal (IN) vascular endothelial growth factor (VEGF)in the treatment of experimental stroke. Sprague-Dawley rats were randomized into four groups as IN low (100 microg/ml), IN middle (200 microg/ml) and IN high (500 microg/ml) VEGF-treated group, and IN saline-treated group (n=12), given recombinant human VEGF 165 or saline intranasally. Focal cerebral ischemia was induced by transient (90 min) middle cerebral artery occlusion (MCAO) method. Behavioral neurological deficits were assessed 1, 7 and 14 d after the onset of MCAO. Rats were sacrificed at 14 d, the brain sections were stained and an image analysis system was used to calculate the infarct volume. Microvessels were labeled by FITC-dextran and the segment lengths, diameters and number of microvessels were measured by Image Pro-Plus Version 6.0 software. Fourteen days post MCAO, infarct volume significantly reduced (P<0.01) in rats which received the middle dose of IN VEGF when compared to IN saline. And middle dose of VEGF significantly improved behavioral recovery (P<0.01). No significant difference in the behavioral recovery and infarct volume was observed between the saline-treated group and the low or high VEGF-treated groups (P>0.05). Compared to IN saline, middle and high doses of VEGF significantly increased the segment length, diameter and number of microvessels (P<0.01). No significant difference in the segment length, diameter and number of microvessels was observed between the IN saline-treated group and the low VEGF-treated group (P>0.05). The middle dose of IN VEGF was most effective on reducing infarct volume, improving behavioral recovery and enhancing angiogenesis in stroke brain, which can be used in the following treatments to further evaluate the effect of VEGF.
Collapse
Affiliation(s)
- Ji-Ping Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Wang F, Keimig T, He Q, Ding J, Zhang Z, Pourabdollah-Nejad S, Yang XP. Augmented healing process in female mice with acute myocardial infarction. ACTA ACUST UNITED AC 2008; 4:230-47. [PMID: 18022590 DOI: 10.1016/s1550-8579(07)80043-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND It is well established that premenopausal women are protected from cardiovascular disease. This gender difference in favor of females is also demonstrated in animal studies. Our research group previously found that female mice had much lower incidence of cardiac rupture and mortality than did males during the acute phase of myocardial infarction (MI); however, the mechanisms responsible for such protection are not fully understood. OBJECTIVE The aim of this study was to determine whether the favorable cardiac effect observed in female mice with MI is due to an augmented healing process that includes less inflammation, reduced matrix degradation, and enhanced neovascularization. METHODS Twelve-week-old male and female C57BL/6J mice were subjected to MI by ligating the left anterior descending coronary artery and then euthanized at 1, 4, 7, or 14 days post-MI. Inflammatory cell infiltration and myofibroblast transformation, matrix metalloproteinase (MMP)-2 and MMP-9 activity, tissue inhibitor of metalloproteinase (TIMP)-I expression, and neovascularization were examined by immunohistochemistry, zymography, Western blot, and laser scanning confocal microscopy, respectively. Cardiac function was evaluated by echocardiography on day 14. RESULTS We found that: (1) neutrophil infiltration during the early phase of MI (1-4 days) was much lower in females than in males and was associated with lower MMP-9 activity and higher TIMP-1 protein expression, indicating less-exaggerated inflammation and extracellular matrix degradation in females; (2) myofibroblast transformation, as indicated by expression of alpha-smooth muscle actin, was significantly greater in females than in males at day 7 of MI (P<0.05), indicating facilitated collagen deposition and scar formation; and (3) neovascularization (vascular area in the infarct border) was markedly increased in females, and was associated with better preserved cardiac function and less left ventricular dilatation. CONCLUSION Our data suggest that less-exaggerated early inflammation and augmented reparative fibrotic response, indicated by enhanced myofibroblast transformation, may contribute greatly to low rupture rates in females during the acute and subacute phases of MI, whereas enhanced neovascularization may lead to better preserved cardiac function post-MI.
Collapse
Affiliation(s)
- Fangfei Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, and Department of Biology, Wayne State University, Detroit, Michigan 48202-2689, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Tanaka Y, Marumo T, Omura T, Yoshida S. Quantitative assessments of cerebral vascular damage with a silicon rubber casting method in photochemically-induced thrombotic stroke rat models. Life Sci 2007; 81:1381-8. [PMID: 17936852 DOI: 10.1016/j.lfs.2007.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 09/11/2007] [Accepted: 09/12/2007] [Indexed: 11/16/2022]
Abstract
Previous studies have described microvascular disturbances downstream of occluded large vessels arising during the acute phase (several hours) following cerebral ischemic insult. Prolonged microvascular disturbances may cause delayed neuronal cell death in ischemic penumbral regions, leading to expanded brain infarctions and poor neurological and functional outcomes. The lack of simple and quantitative methods for investigating this microcirculation failure suggests the need to develop a new method for clarifying the precise distribution and persistence of post-ischemic microvascular disturbances. The present study used a silicone rubber casting method in quantitative analyses of microvascular conditions in photochemically-induced thromboembolic (PIT) stroke rat models. After the casting procedure in rats with PIT stroke, a 6 microm-thick coronal section was obtained, and quantitative analyses of microvascular density and measurements of the infarct area in the serial section were performed. The major findings of the present study are as follows: (1) Silicone rubber casting techniques can be applied to precise quantitative analyses of microvessels in the same individual in whom brain infarct volume was measured; (2) the persistence and spatial distribution of microvascular disturbances assessed at the ischemic core, ischemic penumbra, and non-ischemic regions strongly suggest that microvascular disturbances affect brain infarct expansion; (3) the current method demonstrated the protective effects of MK-801 on microvessels, indicating that the technique may be useful in investigating factors that provide vascular protection. The experimental procedure introduced here would facilitate future evaluations of vascular protective agents.
Collapse
Affiliation(s)
- Yu Tanaka
- Molecular Function and Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., Saitama 331-9530, Japan.
| | | | | | | |
Collapse
|
22
|
Gertz K, Priller J, Kronenberg G, Fink KB, Winter B, Schröck H, Ji S, Milosevic M, Harms C, Böhm M, Dirnagl U, Laufs U, Endres M. Physical Activity Improves Long-Term Stroke Outcome via Endothelial Nitric Oxide Synthase–Dependent Augmentation of Neovascularization and Cerebral Blood Flow. Circ Res 2006; 99:1132-40. [PMID: 17038638 DOI: 10.1161/01.res.0000250175.14861.77] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Physical activity upregulates endothelial nitric oxide synthase (eNOS), improves endothelium function, and protects from vascular disease. Here, we tested whether voluntary running would enhance neovascularization and long-term recovery following mild brain ischemia. Wild-type mice were exposed to 30 minutes of middle-cerebral artery occlusion (MCAo) and reperfusion. Continuous voluntary running on wheels conferred long-term upregulation of eNOS in the vasculature and of endothelial progenitor cells (EPCs) in the spleen and bone marrow (BM). This was associated with higher numbers of circulating EPCs in the blood and enhanced neovascularization. Moreover, engraftment of TIE2/LacZ-positive BM-derived cells was increased in the ischemic brain. Four weeks after the insult, trained animals showed higher numbers of newly generated cells in vascular sites, increased density of perfused microvessels and sustained augmentation of cerebral blood flow within the ischemic striatum. Moreover, running conferred tissue sparing and improved functional outcome at 4 weeks. The protective effects of running on angiogenesis and outcome were completely abolished when animals were treated with a NOS inhibitor or the antiangiogenic compound endostatin after brain ischemia, and in animals lacking eNOS expression. Voluntary physical activity improves long-term stroke outcome by eNOS-dependent mechanisms related to improved angiogenesis and cerebral blood flow.
Collapse
Affiliation(s)
- Karen Gertz
- Klinik und Poliklinik für Neurologie, Charité Campus Mitte, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nakano T, Hozumi Y, Ali H, Saino-Saito S, Kamii H, Sato S, Kayama T, Watanabe M, Kondo H, Goto K. Diacylglycerol kinase ζ is involved in the process of cerebral infarction. Eur J Neurosci 2006; 23:1427-35. [PMID: 16553606 DOI: 10.1111/j.1460-9568.2006.04685.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Diacylglycerol kinase (DGK) is an enzyme that phosphorylates a second messenger diacylglycerol (DG) and is involved in a variety of pathophysiological cellular responses. We have previously reported that DGKzeta may be involved in the selective vulnerability of hippocampal CA1 neurons in transient forebrain ischemia. In this study we aimed to further elucidate functional implications of DGK isozymes in the cerebral cortex suffering from infarction using a focal ischemic model. In the early phase of 90 min of middle cerebral artery occlusion, DGKzeta-immunoreactivity is reduced rapidly in the nucleus of cortical neurons in the ischemic core, while DGKiota and other neuronal proteins such as MAP-2 and NeuN remain intact. This suggests that rapid disappearance of DGKzeta in ischemic neurons is a quite early event precedent to neuronal degeneration in response to ischemia. Furthermore, in the late inflammatory phase of infarction DGKzeta-immunoreactivity is detected in non-neuronal cells including factor VIII-positive endothelial cells and ED-1-positive phagocytic cells. The present study suggests that DGKzeta may play roles in various processes of ischemic brain damage including neuronal death and non-neuronal inflammatory response.
Collapse
Affiliation(s)
- Tomoyuki Nakano
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-nishi 2-2-2, Yamagata 990-9585, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Theodorsson A, Holm L, Theodorsson E. Modern anesthesia and peroperative monitoring methods reduce per- and postoperative mortality during transient occlusion of the middle cerebral artery in rats. ACTA ACUST UNITED AC 2005; 14:181-90. [PMID: 15795172 DOI: 10.1016/j.brainresprot.2005.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 01/25/2005] [Accepted: 01/27/2005] [Indexed: 11/15/2022]
Abstract
Mortality and morbidity during and after occlusion of the middle cerebral artery in rats are important confounding factors which may be minimized by improved anesthesia and peroperative monitoring techniques. We describe state of the art techniques for inducing anesthesia, endotracheal intubation, ventilation and monitoring peroperatively in this context. Introducing the subtemporal approach of Tamura et al. in our laboratory 5 years ago, we experienced 25% peroperative and 24 h postoperative rat mortality when performing temporary clipping of the middle cerebral artery. This prompted us to abandon intraperitoneal anesthesia by chloral hydrate and ventilation by tracheotomy in favor of endotracheal intubation and isoflurane anesthesia (1% isoflurane in 30%:70% O(2)/N(2)O). These anesthetic techniques in combination with improved surgical skills have reduced our initial 25% peroperative- and 24 h postoperative mortality to 2.7% (1.8% peroperatively and 0.9% 24 h postoperatively). Furthermore, the following 14 days postoperative mortality rate was 1.8%. A total number of 203 rats have been operated with this method in different studies where a focal reperfusion stroke model combined with extended periods of observations were the cornerstone.
Collapse
MESH Headings
- Anesthesia, Inhalation/methods
- Anesthetics, Inhalation/pharmacology
- Animals
- Disease Models, Animal
- Female
- Infarction, Middle Cerebral Artery/mortality
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Intubation, Intratracheal/instrumentation
- Intubation, Intratracheal/methods
- Isoflurane/pharmacology
- Monitoring, Intraoperative/methods
- Postoperative Complications/mortality
- Postoperative Complications/prevention & control
- Rats
- Rats, Sprague-Dawley
- Surgical Instruments
Collapse
Affiliation(s)
- Annette Theodorsson
- Department of Biomedicine and Surgery, Division of Clinical Chemistry, University Hospital, SE-581 85 Linkoping, Sweden.
| | | | | |
Collapse
|