51
|
Chen Y, Jiang Y, Kong X, Zhao C, Zhong S, Yang L, Feng T, Peng S, Bi Y, Corbetta M, Gong G. Common and unique structural plasticity after left and right hemisphere stroke. J Cereb Blood Flow Metab 2021; 41:3350-3364. [PMID: 34415210 PMCID: PMC8669287 DOI: 10.1177/0271678x211036606] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Strokes to the left and right hemisphere lead to distinctive behavioral profiles. Are left and right hemisphere strokes (LHS and RHS) associated with distinct or common poststroke neuroplasticity patterns? Understanding this issue would reveal hemispheric neuroplasticity mechanisms in response to brain damage. To this end, we investigated poststroke structural changes (2 weeks to 3 months post-onset) using longitudinal MRI data from 69 LHS and 55 RHS patients and 31 demographic-matched healthy control participants. Both LHS and RHS groups showed statistically common plasticity independent of the lesioned hemisphere, including 1) gray matter (GM) expansion in the ipsilesional and contralesional precuneus, and contralesional superior frontal gyrus; 2) GM shrinkage in the ipsilesional medial orbital frontal gyrus and middle cingulate cortex. On the other hand, only RHS patients had significant GM expansion in the ipsilesional medial superior and orbital frontal cortex. Importantly, these common and unique GM changes post-stroke largely overlapped with highly-connected cortical hub regions in healthy individuals. Moreover, they correlated with behavioral recovery, indicating that post-stroke GM volumetric changes in cortical hubs reflect compensatory rather than maladaptive mechanisms. These results highlight the importance of structural neuroplasticity in hub regions of the cortex, along with the hemispheric specificity, for stroke recovery.
Collapse
Affiliation(s)
- Yijun Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yaya Jiang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xiangyu Kong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Chenxi Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Suyu Zhong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Liyuan Yang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Tao Feng
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China.,Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shaoling Peng
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yanchao Bi
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Maurizio Corbetta
- Department of Neuroscience, Neurology Clinic, University of Padua, Padua, Italy.,Padova Neuroscience Center, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy.,Department of Neurology, Radiology, and Neuroscience, Washington University in St. Louis, St. Louis, USA
| | - Gaolang Gong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| |
Collapse
|
52
|
Loiola RA, García-Gabilondo M, Grayston A, Bugno P, Kowalska A, Duban-Deweer S, Rizzi E, Hachani J, Sano Y, Shimizu F, Kanda T, Mysiorek C, Mazurek MP, Rosell A, Gosselet F. Secretome of endothelial progenitor cells from stroke patients promotes endothelial barrier tightness and protects against hypoxia-induced vascular leakage. Stem Cell Res Ther 2021; 12:552. [PMID: 34702368 PMCID: PMC8549346 DOI: 10.1186/s13287-021-02608-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Cell-based therapeutic strategies have been proposed as an alternative for brain repair after stroke, but their clinical application has been hampered by potential adverse effects in the long term. The present study was designed to test the effect of the secretome of endothelial progenitor cells (EPCs) from stroke patients (scCM) on in vitro human models of angiogenesis and vascular barrier. METHODS Two different scCM batches were analysed by mass spectrometry and a proteome profiler. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used for designing angiogenesis studies (proliferation, migration, and tubulogenesis) or in vitro models of EC monolayer (confluent monolayer ECs-CMECs) and blood-brain barrier (BBB; brain-like ECs-BLECs). Cells were treated with scCM (5 μg/mL) or protein-free endothelial basal medium (scEBM-control). CMECs or BLECs were exposed (6 h) to oxygen-glucose deprivation (OGD) conditions (1% oxygen and glucose-free medium) or normoxia (control-5% oxygen, 1 g/L of glucose) and treated with scCM or scEBM during reoxygenation (24 h). RESULTS The analysis of different scCM batches showed a good reproducibility in terms of protein yield and composition. scCM increased CD34+-EC proliferation, tubulogenesis, and migration compared to the control (scEBM). The proteomic analysis of scCM revealed the presence of growth factors and molecules modulating cell metabolism and inflammatory pathways. Further, scCM decreased the permeability of CMECs and upregulated the expression of the junctional proteins such as occludin, VE-cadherin, and ZO-1. Such effects were possibly mediated through the activation of the interferon pathway and a moderate downregulation of Wnt signalling. Furthermore, OGD increased the permeability of both CMECs and BLECs, while scCM prevented the OGD-induced vascular leakage in both models. These effects were possibly mediated through the upregulation of junctional proteins and the regulation of MAPK/VEGFR2 activity. CONCLUSION Our results suggest that scCM promotes angiogenesis and the maturation of newly formed vessels while restoring the BBB function in ischemic conditions. In conclusion, our results highlight the possibility of using EPC-secretome as a therapeutic alternative to promote brain angiogenesis and protect from ischemia-induced vascular leakage.
Collapse
Affiliation(s)
| | - Miguel García-Gabilondo
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Alba Grayston
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Paulina Bugno
- Pure Biologics S.A., Duńska 11, 54-427, Wroclaw, Poland
| | | | - Sophie Duban-Deweer
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Eleonora Rizzi
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Johan Hachani
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Caroline Mysiorek
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | | | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Fabien Gosselet
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France.
- Laboratory of the Blood-Brain Barrier, Sciences Faculty Jean Perrin, Artois University, Lens, France.
| |
Collapse
|
53
|
Proangiogenic functions of osteopontin-derived synthetic peptide RSKSKKFRR in endothelial cells and postischemic brain. Neuroreport 2021; 32:1248-1254. [PMID: 34494989 DOI: 10.1097/wnr.0000000000001722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the potential therapeutic effects of a newly discovered osteopontin-derived synthetic peptide "RSKKFRR" in a rat model of ischemic stroke. METHODS A total of 24 male SD rats were randomly divided into three groups. The model of ischemic stroke was made up of the middle cerebral artery occlusion (MACO). The rats were divided into sham operation group (Sham), control group (MACO + PBS) and treatment group (MACO + OPNpt9), eight rats in each group. In the control group and the treatment group, PBS or OPNpt9 was injected into the nasal cavity after MACO once a day, and the area of new blood vessels and the recovery of nerve function were observed 14 days later. Whether the proliferation, migration and tube formation of HUVECs were promoted by OPNpt9 was tested. The expression levels of related proangiogenic factors were also detected. RESULTS OPNpt9 was found to contribute to cerebral microvascular remodeling and neurological improvement in ischemic rats while promoting endothelial cell migration, proliferation and tube formation in vitro. These effects were mediated by activation of the p-ERK/MMP-9/VEGF pathway. CONCLUSION In conclusion, OPNpt9 promotes angiogenesis and neurological recovery after ischemic stroke.
Collapse
|
54
|
Cui Q, Ma YH, Yu HY, Zhang YL, Qin XD, Ge SQ, Zhang GW. Systematic analysis of the mechanism of hydroxysafflor yellow A for treating ischemic stroke based on network pharmacology technology. Eur J Pharmacol 2021; 908:174360. [PMID: 34302817 DOI: 10.1016/j.ejphar.2021.174360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 01/18/2023]
Abstract
In this study, we analyzed the mechanism of hydroxysafflor yellow A (HSYA) for treating ischemic stroke (IS) based on network pharmacology tools, and verified the kernel targets via animal experiments. The targets of HSYA were collected via PharmMapper server and the IS-related targets were searched using Genecards, Online Mendelian Inheritance in Man, Therapeutic Target, and Disgenet databases. The targets identified from the above two steps were overlapped to acquire candidate targets involved in the effects of HSYA for treating IS. Subsequently, the Database for Annotation, Visualization, and Integrated Discovery was used for gene ontology analysis and the Kyoto encyclopedia of genes and genomes pathway analysis. Cytoscape 3.7.1 was applied to establish the component-target-pathway network. Potential core targets were obtained by protein-protein interaction analysis. Furthermore, Autodock Vina was used to identify core genes, and animal experiments was used to verify the expression level of core genes. On the basis of the modified neurologic severity score and the results of 2,3,5-Triphenyltetrazolium chloride and Hematoxylin-eosin staining, we confirmed that HSYA reduced the infarct volume in rats and protected neuronal cells in the hippocampal region after IS. Western blot and immunohistochemical staining showed that HSYA increased the expression of epidermal growth factor receptor, hypoxia inducible factor 1 alpha, and endothelial nitric oxide synthase (P < 0.05). The effects of HSYA on IS are mediated through several targets and pathways related to the regulation of oxidative stress and the renewal of cell and blood vessels while improving post-ischemic brain impairment.
Collapse
Affiliation(s)
- Qian Cui
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, 071002, China
| | - Yu-Hui Ma
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China
| | - Hao-Yu Yu
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, 071002, China
| | - Yu-Liang Zhang
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, 071002, China
| | - Xiu-de Qin
- Shenzhen TCM Hospital, Shenzhen, Guangdong, 518000, China
| | - Shao-Qin Ge
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, 071002, China
| | - Guo-Wei Zhang
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, 071002, China.
| |
Collapse
|
55
|
Sun P, Ma F, Xu Y, Zhou C, Stetler RA, Yin KJ. Genetic deletion of endothelial microRNA-15a/16-1 promotes cerebral angiogenesis and neurological recovery in ischemic stroke through Src signaling pathway. J Cereb Blood Flow Metab 2021; 41:2725-2742. [PMID: 33910400 PMCID: PMC8504951 DOI: 10.1177/0271678x211010351] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral angiogenesis is tightly controlled by specific microRNAs (miRs), including the miR-15a/16-1 cluster. Recently, we reported that endothelium-specific conditional knockout of the miR-15a/16-1 cluster (EC-miR-15a/16-1 cKO) promotes post-stroke angiogenesis and improves long-term neurological recovery by increasing protein levels of VEGFA, FGF2, and their respective receptors VEGFR2 and FGFR1. Herein, we further investigated the underlying signaling mechanism of these pro-angiogenic factors after ischemic stroke using a selective Src family inhibitor AZD0530. EC-miR-15a/16-1 cKO and age- and sex-matched wild-type littermate (WT) mice were subjected to 1 h middle cerebral artery occlusion (MCAO) and 28d reperfusion. AZD0530 was administered daily by oral gavage to both genotypes of mice 3-21d after MCAO. Compared to WT, AZD0530 administration exacerbated spatial cognitive impairments and brain atrophy in EC-miR-15a/16-1 cKO mice following MCAO. AZD0530 also attenuated long-term recovery of blood flow and inhibited the formation of new microvessels, including functional vessels with blood circulation, in the penumbra of stroked cKO mice. Moreover, AZD0530 blocked the Src signaling pathway by downregulating phospho-Src and its downstream mediators (p-Stat3, p-Akt, p-FAK, p-p44/42 MAPK, p-p38 MAPK) in post-ischemic brains. Collectively, our data demonstrated that endothelium-targeted deletion of the miR-15a/16-1 cluster promotes post-stroke angiogenesis and improves long-term neurological recovery via activating Src signaling pathway.
Collapse
Affiliation(s)
- Ping Sun
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chao Zhou
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
56
|
Fang J, Chopp M, Xin H, Zhang L, Wang F, Golembieski W, Zhang ZG, He L, Liu Z. Plasminogen deficiency causes reduced angiogenesis and behavioral recovery after stroke in mice. J Cereb Blood Flow Metab 2021; 41:2583-2592. [PMID: 33853408 PMCID: PMC8504962 DOI: 10.1177/0271678x211007958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Plasminogen is involved in the process of angiogenesis; however, the underlying mechanism is unclear. Here, we investigated the potential contribution of plasmin/plasminogen in mediating angiogenesis and thereby contributing to functional recovery post-stroke. Wild-type plasminogen naive (Plg+/+) mice and plasminogen knockout (Plg-/-) mice were subjected to unilateral permanent middle cerebral artery occlusion (MCAo). Blood vessels were labeled with FITC-dextran. Functional outcomes, and cerebral vessel density were compared between Plg+/+ and Plg-/- mice at different time points after stroke. We found that Plg-/- mice exhibited significantly reduced functional recovery, associated with significantly decreased vessel density in the peri-infarct area in the ipsilesional cortex compared with Plg+/+ mice. In vitro, cerebral endothelial cells harvested from Plg-/- mice exhibited significantly reduced angiogenesis assessed using tube formation assay, and migration, as evaluated using Scratch assays, compared to endothelial cells harvested from Plg+/+ mice. In addition, using Western blots, expression of thrombospondin (TSP)-1 and TSP-2 were increased after MCAo in the Plg-/- group compared to Plg+/+ mice, especially in the ipsilesional side of brain. Taken together, our data suggest that plasmin/plasminogen down-regulates the expression level of TSP-1 and TSP-2, and thereby promotes angiogenesis in the peri-ischemic brain tissue, which contributes to functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Jinghuan Fang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Neurology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Hongqi Xin
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Fengjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | | | - Li He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
57
|
Chen J, Li X, Ni R, Chen Q, Yang Q, He J, Luo L. Acute brain vascular regeneration occurs via lymphatic transdifferentiation. Dev Cell 2021; 56:3115-3127.e6. [PMID: 34562378 DOI: 10.1016/j.devcel.2021.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
Acute ischemic stroke damages the regional brain blood vessel (BV) network. Acute recovery of basic blood flows, which is carried out by the earliest regenerated BVs, are critical to improve clinical outcomes and minimize lethality. Although the late-regenerated BVs form via growing along the meninge-derived ingrown lymphatic vessels (iLVs), mechanisms underlying the early, acute BV regeneration remain elusive. Using zebrafish cerebrovascular injury models, we show that the earliest regenerated BVs come from lymphatic transdifferentiation, a hitherto unappreciated process in vertebrates. Mechanistically, the LV-to-BV transdifferentiation occurs exclusively in the stand-alone iLVs through Notch activation. In the track iLVs adhered by late-regenerated BVs, transdifferentiation never occurs because the BV-expressing EphrinB2a paracellularly activates the iLV-expressing EphB4a to inhibit Notch activation. Suppression of LV-to-BV transdifferentiation blocks acute BV regeneration and becomes lethal. These results demonstrate that acute BV regeneration occurs via lymphatic transdifferentiation, suggesting this process and key regulatory molecules EphrinB2a/EphB4a/Notch as new postischemic therapeutic targets.
Collapse
Affiliation(s)
- Jingying Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China; University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei 400714, Chongqing, China
| | - Xiuhua Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Qi Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Qifen Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China; University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei 400714, Chongqing, China.
| |
Collapse
|
58
|
Delayed rFGF21 Administration Improves Cerebrovascular Remodeling and White Matter Repair After Focal Stroke in Diabetic Mice. Transl Stroke Res 2021; 13:311-325. [PMID: 34523038 DOI: 10.1007/s12975-021-00941-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a major comorbidity exacerbating ischemic brain injury and impairing post-stroke recovery. Our previous study suggested that recombinant human fibroblast growth factor (rFGF) 21 might be a potent therapeutic targeting multiple aspects of pathophysiology in T2DM stroke. This study aims to evaluate the potential effects of rFGF21 on cerebrovascular remodeling after T2DM stroke. Permanent distal middle cerebral artery occlusion was performed in heterozygous non-diabetic db/ + and homozygous diabetic db/db mice. Daily rFGF21 administration was initiated 1 week after stroke induction and maintained for up to 2 weeks thereafter. Multiple markers associated with post-stroke recovery, including angiogenesis, oligodendrogenesis, white matter integrity, and neurogenesis, were assessed up to 3 weeks after stroke. Our results showed an impairment in post-stroke vascular remodeling under T2DM condition, reflected by the decreased expression of trophic factors in brain microvessels and impairments of angiogenesis. The defected cerebrovascular remodeling was accompanied by the decreased oligodendrogenesis and neurogenesis. However, delayed rFGF21 administration normalized post-stroke hyperglycemia and improved neurological outcomes, which may partially be via the promotion of pro-angiogenic trophic factor expression in brain microvessels and cerebrovascular remodeling. The better cerebrovascular remodeling may also contribute to oligodendrogenesis, white matter integrity, and neurogenesis after T2DM stroke. Therefore, delayed rFGF21 administration may improve neurological outcomes in T2DM stroke mice, at least in part by normalizing the metabolic abnormalities and promoting cerebrovascular remodeling and white matter repair.
Collapse
|
59
|
Schlecht A, Vallon M, Wagner N, Ergün S, Braunger BM. TGFβ-Neurotrophin Interactions in Heart, Retina, and Brain. Biomolecules 2021; 11:biom11091360. [PMID: 34572573 PMCID: PMC8464756 DOI: 10.3390/biom11091360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic insults to the heart and brain, i.e., myocardial and cerebral infarction, respectively, are amongst the leading causes of death worldwide. While there are therapeutic options to allow reperfusion of ischemic myocardial and brain tissue by reopening obstructed vessels, mitigating primary tissue damage, post-infarction inflammation and tissue remodeling can lead to secondary tissue damage. Similarly, ischemia in retinal tissue is the driving force in the progression of neovascular eye diseases such as diabetic retinopathy (DR) and age-related macular degeneration (AMD), which eventually lead to functional blindness, if left untreated. Intriguingly, the easily observable retinal blood vessels can be used as a window to the heart and brain to allow judgement of microvascular damages in diseases such as diabetes or hypertension. The complex neuronal and endocrine interactions between heart, retina and brain have also been appreciated in myocardial infarction, ischemic stroke, and retinal diseases. To describe the intimate relationship between the individual tissues, we use the terms heart-brain and brain-retina axis in this review and focus on the role of transforming growth factor β (TGFβ) and neurotrophins in regulation of these axes under physiologic and pathologic conditions. Moreover, we particularly discuss their roles in inflammation and repair following ischemic/neovascular insults. As there is evidence that TGFβ signaling has the potential to regulate expression of neurotrophins, it is tempting to speculate, and is discussed here, that cross-talk between TGFβ and neurotrophin signaling protects cells from harmful and/or damaging events in the heart, retina, and brain.
Collapse
|
60
|
Asgari Taei A, Nasoohi S, Hassanzadeh G, Kadivar M, Dargahi L, Farahmandfar M. Enhancement of angiogenesis and neurogenesis by intracerebroventricular injection of secretome from human embryonic stem cell-derived mesenchymal stem cells in ischemic stroke model. Biomed Pharmacother 2021; 140:111709. [PMID: 34020250 DOI: 10.1016/j.biopha.2021.111709] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
It is well accepted that the success of mesenchymal stem cells (MSCs) therapy against experimental stroke is mainly due to cellular paracrine manners rather than to replace lost tissue per se. Given such "bystander" effects, cell-free therapeutics manifest as a promising approach in regenerative medicine. Here we aimed at evaluating the effect of conditioned medium (CM) derived from human embryonic MSCs (hESC-MSC) on the neurological deficit, neurogenesis, and angiogenesis in experimental stroke. Adult male Wistar rats subjected to middle cerebral artery occlusion (MCAO), were treated with intracerebroventricular CM either one time (1 h post MCAO) or three times (1, 24, and 48 h post MCAO). Motor performance was assessed by the cylinder test on days 3 and 7. Cerebral samples were obtained for infarct size and molecular analysis on day 7 post-injury. Neurogenesis was evaluated by probing Nestin, Ki67, DCX, and Reelin transcripts and protein levels in the striatum, cortex, subventricular zone, and corpus callosum. The mRNA and protein expression of CD31 were also assessed in the striatum and cortical region to estimate angiogenesis post MCAO. Our findings demonstrate that CM treatment could significantly ameliorate neurological deficits and infarct volume in MCAO rats. Furthermore, ischemic stroke was associated with higher levels of neurogenesis and angiogenesis markers. Following treatment with CM, these markers were further potentiated in the brain regions. This study suggests that the therapeutic benefits of CM obtained from hESC-MSCs at least partly are mediated through improved neurogenesis and angiogenesis to accelerate the recovery of cerebral ischemia insult.
Collapse
Affiliation(s)
- Afsaneh Asgari Taei
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Kadivar
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Farahmandfar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
61
|
Przykaza Ł, Kozniewska E. Ligands of the Neuropeptide Y Y2 Receptors as a Potential Multitarget Therapeutic Approach for the Protection of the Neurovascular Unit Against Acute Ischemia/Reperfusion: View from the Perspective of the Laboratory Bench. Transl Stroke Res 2021; 13:12-24. [PMID: 34292517 PMCID: PMC8766383 DOI: 10.1007/s12975-021-00930-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Ischemic stroke is the third leading cause of death and disability worldwide, with no available satisfactory prevention or treatment approach. The current treatment is limited to the use of “reperfusion methods,” i.e., an intravenous or intra-arterial infusion of a fibrinolytic agent, mechanical removal of the clot by thrombectomy, or a combination of both methods. It should be stressed, however, that only approximately 5% of all acute strokes are eligible for fibrinolytic treatment and fewer than 10% for thrombectomy. Despite the tremendous progress in understanding of the pathomechanisms of cerebral ischemia, the promising results of basic research on neuroprotection are not currently transferable to human stroke. A possible explanation for this failure is that experiments on in vivo animal models involve healthy young animals, and the experimental protocols seldom consider the importance of protecting the whole neurovascular unit (NVU), which ensures intracranial homeostasis and is seriously damaged by ischemia/reperfusion. One of the endogenous protective systems activated during ischemia and in neurodegenerative diseases is represented by neuropeptide Y (NPY). It has been demonstrated that activation of NPY Y2 receptors (Y2R) by a specific ligand decreases the volume of the postischemic infarction and improves performance in functional tests of rats with arterial hypertension subjected to middle cerebral artery occlusion/reperfusion. This functional improvement suggests the protection of the NVU. In this review, we focus on NPY and discuss the potential, multidirectional protective effects of Y2R agonists against acute focal ischemia/reperfusion injury, with special reference to the NVU.
Collapse
Affiliation(s)
- Łukasz Przykaza
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland
| | - Ewa Kozniewska
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
62
|
Extracellular Vesicle Application as a Novel Therapeutic Strategy for Ischemic Stroke. Transl Stroke Res 2021; 13:171-187. [PMID: 33982152 DOI: 10.1007/s12975-021-00915-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
Ischemic stroke (IS) accounts for most of the cases of stroke onset, and due to short therapeutic time window for thrombolysis and numerous limited treatment measures and contraindications, lots of patients cannot receive satisfying therapeutic effects resulting in high disability and mortality worldly. In recent years, extracellular vesicles (EVs), as nanosized membrane-structured vesicles secreted from almost all cells, especially from stem/progenitor cells, have been reported to exert significant beneficial effects on IS from multiple approaches and notably ameliorate neurological outcome. Moreover, based on nano-size and lipid bilayer structure, EVs can easily penetrate the blood-brain barrier and migrate into the brain. In this review, we mainly systematically summarize the therapeutic effects of EVs on IS and explore their potential applications. Simultaneously, we also discuss administration routines, dosages, experimental observation time, and some key issues of EV application during IS treatment. It contributes to a comprehensive understanding of the progress of EV treatment for IS and providing confident evidence for further EV clinical application widely.
Collapse
|
63
|
Marquez-Curtis LA, Bokenfohr R, McGann LE, Elliott JAW. Cryopreservation of human cerebral microvascular endothelial cells and astrocytes in suspension and monolayers. PLoS One 2021; 16:e0249814. [PMID: 33852594 PMCID: PMC8046249 DOI: 10.1371/journal.pone.0249814] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/24/2021] [Indexed: 01/19/2023] Open
Abstract
The blood-brain barrier (BBB) keeps pathogens and toxins out of the brain but also impedes the entry of pharmaceuticals. Human cerebral microvascular endothelial cells (hCMECs) and astrocytes are the main functional cell components of the BBB. Although available commercially as cryopreserved cells in suspension, improvements in their cryopreservation and distribution as cryopreserved monolayers could enhance BBB in vitro studies. Here, we examined the response to slow cooling and storage in liquid nitrogen of immortalized hCMEC/D3 cells and human primary astrocytes in suspension and in monolayers. HCMEC/D3 cells in suspension cryopreserved in 5% dimethyl sulfoxide (DMSO) and 95% fetal bovine serum or in 5% DMSO and 6% hydroxyethyl starch (HES) showed post-thaw membrane integrities above 90%, similar to unfrozen control. Cryopreservation did not affect the time-dependent ability of hCMEC/D3 cells to form tubes on Matrigel. Primary astrocytes in suspension cryopreserved in the presence of 5% DMSO and 6% HES had improved viability over those cryopreserved in 10% DMSO. Monolayers of single cultures or co-cultures of hCMEC/D3 cells and astrocytes on fibronectin-coated Rinzl coverslips retained membrane integrities and metabolic function, after freezing in 5% DMSO, 6% HES, and 2% chondroitin sulfate, that were comparable to those of unfrozen controls even after overnight incubation. Rinzl is better than glass or Thermanox as an underlying solid substrate for cryopreserving hCMEC/D3 monolayers. Cryopreserved hCMEC/D3 monolayers expressed the junction proteins ZO-1 and claudin-5 similar to their unfrozen counterparts. Hence, we describe improved cryopreservation protocols for hCMEC/D3 cells and astrocytes in suspension, and a novel protocol for the cryopreservation of monolayers of hCMEC/D3 cells and astrocytes as single cultures or co-cultures that could expand their distribution for research on disease modeling, drug screening, and targeted therapy pertaining to the BBB.
Collapse
Affiliation(s)
- Leah A. Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Reid Bokenfohr
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Locksley E. McGann
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Janet A. W. Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
64
|
Biomarkers of Angiogenesis and Neuroplasticity as Promising Clinical Tools for Stroke Recovery Evaluation. Int J Mol Sci 2021; 22:ijms22083949. [PMID: 33920472 PMCID: PMC8068953 DOI: 10.3390/ijms22083949] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
Several key issues impact the clinical practice of stroke rehabilitation including a patient’s medical history, stroke experience, the potential for recovery, and the selection of the most effective type of therapy. Until clinicians have answers to these concerns, the treatment and rehabilitation are rather intuitive, with standard procedures carried out based on subjective estimations using clinical scales. Therefore, there is a need to find biomarkers that could predict brain recovery potential in stroke patients. This review aims to present the current state-of-the-art stroke recovery biomarkers that could be used in clinical practice. The revision of biochemical biomarkers has been developed based on stroke recovery processes: angiogenesis and neuroplasticity. This paper provides an overview of the biomarkers that are considered to be ready-to-use in clinical practice and others, considered as future tools. Furthermore, this review shows the utility of biomarkers in the development of the concept of personalized medicine. Enhancing brain neuroplasticity and rehabilitation facilitation are crucial concerns not only after stroke, but in all central nervous system diseases.
Collapse
|
65
|
Hsu TW, Lu YJ, Lin YJ, Huang YT, Hsieh LH, Wu BH, Lin YC, Chen LC, Wang HW, Chuang JC, Fang YQ, Huang CC. Transplantation of 3D MSC/HUVEC spheroids with neuroprotective and proangiogenic potentials ameliorates ischemic stroke brain injury. Biomaterials 2021; 272:120765. [PMID: 33780686 DOI: 10.1016/j.biomaterials.2021.120765] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/02/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022]
Abstract
Ischemic stroke, and the consequent brain cell death, is a common cause of death and disability worldwide. Current treatments that primarily aim to relieve symptoms are relatively inefficient in achieving brain tissue regeneration and functional recovery, and thus novel therapeutic options are urgently needed. Although cell-based therapies have shown promise for treating the infarcted brain, a recurring challenge is the inadequate retention and engraftment of transplanted cells at the target tissue, thereby limiting the ultimate therapeutic efficacy. Here, we show that transplantation of preassembled three-dimensional (3D) spheroids of mesenchymal stem cells (MSCs) and vascular endothelial cells (ECs) results in significantly improved cell retention and survival compared with conventional mixed-cell suspensions. The transplanted 3D spheroids exhibit notable neuroprotective, proneurogenic, proangiogenic and anti-scarring potential as evidenced by clear extracellular matrix structure formation and paracrine factor expression and secretion; this ultimately results in increased structural and motor function recovery in the brain of an ischemic stroke mouse model. Therefore, transplantation of MSCs and ECs using the 3D cell spheroid configuration not only reduces cell loss during cell harvesting/administration but also enhances the resultant therapeutic benefit, thus providing important proof-of-concept for future clinical translation.
Collapse
Affiliation(s)
- Ting-Wei Hsu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, 33305, Taiwan; Centre for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan; Centre for Biomedical Science and Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Jie Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Ting Huang
- College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Li-Hung Hsieh
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Bing-Huan Wu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Li-Chi Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hsin-Wen Wang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jui-Che Chuang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Qiao Fang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
66
|
Guo Z, Wu X, Fan W. Clarifying the effects of diabetes on the cerebral circulation: Implications for stroke recovery and beyond. Brain Res Bull 2021; 171:67-74. [PMID: 33662495 DOI: 10.1016/j.brainresbull.2021.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
Given the sheer increased number of victims per year and the availability of only one effective treatment, acute ischemic stroke (AIS) remains to be one of the most under-treated serious diseases. Diabetes not only increases the incidence of ischemic stroke, but amplifies the ischemic damage, upon which if patients with diabetes suffer from stroke, he/she will confront increased risks of long-term functional deficits. The grim reality makes it a pressing need to intensify efforts at the basic science level to understand how diabetes impairs stroke recovery. This review retrospects the clinical and experimental studies in order to elucidate the detrimental effect of diabetes on cerebrovascular circulation including the major arteries/arterioles, collateral circulation, and neovascularization to shed light on further exploration of novel strategies for cerebral circulation protection before and after AIS in patients with diabetes.
Collapse
Affiliation(s)
- Zhihui Guo
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Xuqing Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Wei Fan
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
67
|
Fei Y, Zhao B, Zhu J, Fang W, Li Y. XQ-1H promotes cerebral angiogenesis via activating PI3K/Akt/GSK3β/β-catenin/VEGF signal in mice exposed to cerebral ischemic injury. Life Sci 2021; 272:119234. [PMID: 33607158 DOI: 10.1016/j.lfs.2021.119234] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Stroke still ranks as a most lethal disease worldwide. Angiogenesis during the chronic phase of ischemic stroke can alleviate ischemic injury and attenuate neurological deficit. XQ-1H is a new compound derived from the structure modification of ginkgolide B, which exerts anti-inflammation and neuroprotection against cerebral ischemic injury during the acute or subacute phase. However, whether XQ-1H facilitates angiogenesis and neural functional recovery during the chronic phase remains unclear. This research was designed to explore whether XQ-1H promotes angiogenesis after ischemic stroke and to preliminarily elucidate the mechanism. In vitro, XQ-1H was found to facilitate proliferation, migration and tube formation in bEnd.3 cells. In vivo, XQ-1H raised the CD31 positive microvessel number and increased focal cerebral blood flow in mice exposed to cerebral ischemic injury, and improved the neurological function. Mechanism studies revealed that XQ-1H exerted angiogenesis promoting effect via the PI3K/Akt/GSK3β/β-catenin/VEGF signal pathway, which was reversed by LY294002 (the specific inhibitor of PI3K/Akt). In conclusion, XQ-1H exerts angiogenetic effect both in vivo and in vitro, which is a potential agent against ischemic stroke during chronic phase.
Collapse
Affiliation(s)
- Yuxiang Fei
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bo Zhao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jianping Zhu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
68
|
Zhong J, Li RW, Wang J, Wang Y, Ge HF, Xian JS, Feng H, Tan L. Neuroprotection by cattle encephalon glycoside and ignotin beyond the time window of thrombolysis in ischemic stroke. Neural Regen Res 2021; 16:312-318. [PMID: 32859790 PMCID: PMC7896241 DOI: 10.4103/1673-5374.290899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/02/2019] [Accepted: 03/03/2020] [Indexed: 01/01/2023] Open
Abstract
Cattle encephalon glycoside and ignotin (CEGI) injection is known as a multi-target neuroprotective drug that contains numerous liposoluble molecules, such as polypeptides, monosialotetrahexosyl ganglioside (GM-1), free amino acids, hypoxanthine and carnosine. CEGI has been approved by the Chinese State Food and Drug Administration and widely used in the treatments of various diseases, such as stroke and Alzheimer's disease. However, the neuroprotective effects of CEGI beyond the time window of thrombolysis (within 4.5 hours) on acute ischemic stroke remain unclear. This study constructed a rat middle cerebral artery occlusion model by suture-occluded method to simulate ischemic stroke. The first daily dose was intraperitoneally injected at 8 hours post-surgery and the CEGI treatments continued for 14 days. Results of the modified five-point Bederson scale, beam balance test and rotameric test showed the neurological function of ischemic stroke rats treated with 4 mL/kg/d CEGI improved significantly, but the mortality within 14 days did not change significantly. Brain MRI and 2,3,5-triphenyltetrazolium chloride staining confirmed that the infarct size in the 4 mL/kg/d CEGI-treated rats was significantly reduced compared with ischemic insult only. The results of transmission electron microscopy and double immunofluorescence staining showed that the hippocampal neuronal necrosis in the ischemic penumbra decreased whereas the immunopositivity of new neuronal-specific protein doublecortin and the percentage of Ki67/doublecortin positive cells increased in CEGI-treated rats compared with untreated rats. Our results suggest that CEGI has an effective neuroprotective effect on ischemic stroke when administered after the time window of thrombolysis. The study was approved by the Animal Ethics Committee of The Third Military Medical University, China.
Collapse
Affiliation(s)
- Jun Zhong
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Rong-Wei Li
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
- Department of Neurosurgery, Hanzhong Central Hospital, Hanzhong, Shaanxi Province, China
| | - Ju Wang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Wang
- Department of Oncology, Hanzhong Central Hospital, Hanzhong, Shaanxi Province, China
| | - Hong-Fei Ge
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Ji-Shu Xian
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, School of Electrical Engineering, Chongqing University, Chongqing, China
| |
Collapse
|
69
|
da Costa A, Metais T, Mouthon F, Kerkovich D, Charvériat M. Evaluating and modulating TFEB in the control of autophagy: toward new treatments in CNS disorders. Fundam Clin Pharmacol 2020; 35:539-551. [PMID: 33259088 DOI: 10.1111/fcp.12634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/15/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
TFEB is a mammalian transcription factor that binds directly to the CLEAR consensus sequence (5'-GTCACGTGAC-3') present in the regulatory regions of genes inducing autophagosome formation, autophagosome-lysosome fusion, hydrolase enzyme expression, and lysosomal exocytosis. By modulating these activities, TFEB coordinates on-demand control over each cell's degradation pathway. Thus, a nuclear signaling pathway regulates cellular energy metabolism through TFEB. Our growing understanding of the role of TFEB and CLEAR in the promotion of healthy clearance together with in vitro and in vivo preclinical findings in various animal models of disease supports the conclusion that the pharmacological activation of TFEB could clear toxic proteins to treat both rare and common forms of neurodegenerative disease.
Collapse
|
70
|
Jiang Z, Alamuri TT, Muir ER, Choi DW, Duong TQ. Longitudinal multiparametric MRI study of hydrogen-enriched water with minocycline combination therapy in experimental ischemic stroke in rats. Brain Res 2020; 1748:147122. [DOI: 10.1016/j.brainres.2020.147122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
|
71
|
Diabetes Mellitus/Poststroke Hyperglycemia: a Detrimental Factor for tPA Thrombolytic Stroke Therapy. Transl Stroke Res 2020; 12:416-427. [PMID: 33140258 DOI: 10.1007/s12975-020-00872-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Intravenous administration of tissue-type plasminogen activator (IV tPA) therapy has long been considered a mainstay in ischemic stroke management. However, patients respond to IV tPA therapy unequally with some subsets of patients having worsened outcomes after treatment. In particular, diabetes mellitus (DM) is recognized as a clinically important vascular comorbidity that leads to lower recanalization rates and increased risks of hemorrhagic transformation (HT). In this short-review, we summarize the recent advances in understanding of the underlying mechanisms involved in post-IV tPA worsening of outcome in diabetic stroke. Potential pathologic factors that are related to the suboptimal tPA recanalization in diabetic stroke include higher plasma plasminogen activator inhibitor (PAI)-1 level, diabetic atherogenic vascular damage, glycation of the tPA receptor annexin A2, and alterations in fibrin clot density. While factors contributing to the exacerbation of HT in diabetic stroke include hyperglycemia, vascular oxidative stress, and inflammation, tPA neurovascular toxicity and imbalance in extracellular proteolysis are discussed. Besides, impaired collaterals in DM also compromise the efficacy of IV tPA therapy. Additionally, several tPA combination approaches developed from experimental studies that may help to optimize IV tPA therapy are also briefly summarized. In summary, more research efforts are needed to improve the safety and efficacy of IV tPA therapy in ischemic stroke patients with DM/poststroke hyperglycemia.
Collapse
|
72
|
Sapkota A, Park SJ, Choi JW. Inhibition of LPA 5 Activity Provides Long-Term Neuroprotection in Mice with Brain Ischemic Stroke. Biomol Ther (Seoul) 2020; 28:512-518. [PMID: 33024060 PMCID: PMC7585638 DOI: 10.4062/biomolther.2020.159] [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] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is a leading cause of long-term disability in ischemic survivors who are suffering from motor, cognitive, and memory impairment. Previously, we have reported suppressing LPA5 activity with its specific antagonist can attenuate acute brain injuries after ischemic stroke. However, it is unclear whether suppressing LPA5 activity can also attenuate chronic brain injuries after ischemic stroke. Here, we explored whether effects of LPA5 antagonist, TCLPA5, could persist a longer time after brain ischemic stroke using a mouse model challenged with tMCAO. TCLPA5 was administered to mice every day for 3 days, starting from the time immediately after reperfusion. TCLPA5 administration improved neurological function up to 21 days after tMCAO challenge. It also reduced brain tissue loss and cell apoptosis in mice at 21 days after tMCAO challenge. Such long-term neuroprotection of TCLPA5 was associated with enhanced neurogenesis and angiogenesis in post-ischemic brain, along with upregulated expression levels of vascular endothelial growth factor. Collectively, results of the current study indicates that suppressing LPA5 activity can provide long-term neuroprotection to mice with brain ischemic stroke.
Collapse
Affiliation(s)
- Arjun Sapkota
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Sung Jean Park
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Ji Woong Choi
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
73
|
Zong X, Li Y, Liu C, Qi W, Han D, Tucker L, Dong Y, Hu S, Yan X, Zhang Q. Theta-burst transcranial magnetic stimulation promotes stroke recovery by vascular protection and neovascularization. Theranostics 2020; 10:12090-12110. [PMID: 33204331 PMCID: PMC7667689 DOI: 10.7150/thno.51573] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/26/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: The integrity and function of the blood-brain barrier (BBB) is compromised after stroke. The current study was performed to examine potential beneficial effects and underlying mechanisms of repetitive transcranial magnetic stimulation (rTMS) on angiogenesis and vascular protection, function, and repair following stroke, which are largely unknown. Methods: Using a rat photothrombotic (PT) stroke model, continuous theta-burst rTMS was administered once daily to the infarcted hemisphere for 5 min, beginning 3 h after PT stroke. This treatment was applied for 6 days. BBB integrity, blood flow, vascular associated proteins, angiogenesis, integrity of neuronal morphology and structure, and behavioral outcome were measured and analyzed at 6 and/or 22 days after PT stroke. Results: We report that rTMS significantly mitigated BBB permeabilization and preserved important BBB components ZO-1, claudin-5, occludin, and caveolin-1 from PT-induced degradation. Damage to vascular structure, morphology, and perfusion was ameliorated by rTMS, resulting in improved local tissue oxygenation. This was accompanied with robust protection of critical vascular components and upregulation of regulatory factors. A complex cytokine response was induced by PT, particularly at the late phase. Application of rTMS modulated this response, ameliorating levels of cytokines related to peripheral immune cell infiltration. Further investigation revealed that rTMS promoted and sustained post-ischemic angiogenesis long-term and reduced apoptosis of newborn and existing vascular endothelial cells. Application of rTMS also inhibited PT-induced excessive astrocyte-vasculature interactions and stimulated an A1 to A2 shift in vessel-associated astrocytes. Mechanistic studies revealed that rTMS dramatically increased levels of PDGFRβ associated with A2 astrocytes and their adjacent vasculature. As well, A2 astrocytes displayed marked amplification of the angiogenesis-related factors VEGF and TGFβ. PT induced a rise in vessel-associated expression of HIF-1α that was starkly intensified by rTMS treatment. Finally, rTMS preserved neuronal morphology, synaptic structure integrity and behavioral outcome. Conclusions: These results indicate that rTMS can exert powerful protective and restorative effects on the peri-infarct microvasculature after PT stroke by, in part, promoting HIF-1α signaling and shifting vessel-associated astrocytic polarization to the A2 phenotype. This study provides further support for the potent protective effects of rTMS in the context of ischemic stroke, and these findings implicate vascular repair and protection as an important underlying phenomenon.
Collapse
|
74
|
Allegra Mascaro AL, Conti E, Lai S, Di Giovanna AP, Spalletti C, Alia C, Panarese A, Scaglione A, Sacconi L, Micera S, Caleo M, Pavone FS. Combined Rehabilitation Promotes the Recovery of Structural and Functional Features of Healthy Neuronal Networks after Stroke. Cell Rep 2020; 28:3474-3485.e6. [PMID: 31553915 DOI: 10.1016/j.celrep.2019.08.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 06/19/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022] Open
Abstract
Rehabilitation is considered the most effective treatment for promoting the recovery of motor deficits after stroke. One of the most challenging experimental goals is to unambiguously link brain rewiring to motor improvement prompted by rehabilitative therapy. Previous work showed that robotic training combined with transient inactivation of the contralesional cortex promotes a generalized recovery in a mouse model of stroke. Here, we use advanced optical imaging and manipulation tools to study cortical remodeling induced by this rehabilitation paradigm. We show that the stabilization of peri-infarct synaptic contacts accompanies increased vascular density induced by angiogenesis. Furthermore, temporal and spatial features of cortical activation recover toward pre-stroke conditions through the progressive formation of a new motor representation in the peri-infarct area. In the same animals, we observe reinforcement of inter-hemispheric connectivity. Our results provide evidence that combined rehabilitation promotes the restoration of structural and functional features distinctive of healthy neuronal networks.
Collapse
Affiliation(s)
- Anna Letizia Allegra Mascaro
- Neuroscience Institute, National Research Council, Pisa 56124, Italy; European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy.
| | - Emilia Conti
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Stefano Lai
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy
| | | | | | - Claudia Alia
- Neuroscience Institute, National Research Council, Pisa 56124, Italy
| | - Alessandro Panarese
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy
| | - Alessandro Scaglione
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Leonardo Sacconi
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; National Institute of Optics, National Research Council, Sesto Fiorentino 50019, Italy
| | - Silvestro Micera
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy; Bertarelli Foundation Chair in Translational NeuroEngineering, Centre for Neuroprosthetics and Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Matteo Caleo
- Neuroscience Institute, National Research Council, Pisa 56124, Italy; Department of Biomedical Sciences, University of Padua, Padova 35131, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy; National Institute of Optics, National Research Council, Sesto Fiorentino 50019, Italy
| |
Collapse
|
75
|
Fan J, Saft M, Sadanandan N, Gonzales-Portillo B, Park YJ, Sanberg PR, Borlongan CV, Luo Y. LncRNAs Stand as Potent Biomarkers and Therapeutic Targets for Stroke. Front Aging Neurosci 2020; 12:594571. [PMID: 33192490 PMCID: PMC7604318 DOI: 10.3389/fnagi.2020.594571] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is a major public health problem worldwide with a high burden of neurological disability and mortality. Long noncoding RNAs (lncRNAs) have attracted much attention in the past decades because of their newly discovered roles in pathophysiological processes in many diseases. The abundance of lncRNAs in the nervous system indicates that they may be part of a complex regulatory network governing physiology and pathology of the brain. In particular, lncRNAs have been shown to play pivotal roles in the pathogenesis of stroke. In this article, we provide a review of the multifaceted functions of lncRNAs in the pathogenesis of ischemic stroke and intracerebral hemorrhage, highlighting their promising use as stroke diagnostic biomarkers and therapeutics. To this end, we discuss the potential of stem cells in aiding lncRNA applications in stroke.
Collapse
Affiliation(s)
- Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Madeline Saft
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Bella Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - You Jeong Park
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Paul R Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
76
|
Giordano M, Trotta MC, Ciarambino T, D’Amico M, Galdiero M, Schettini F, Paternosto D, Salzillo M, Alfano R, Andreone V, Malatino LS, Biolo G, Paolisso G, Adinolfi LE. Circulating MiRNA-195-5p and -451a in Diabetic Patients with Transient and Acute Ischemic Stroke in the Emergency Department. Int J Mol Sci 2020; 21:E7615. [PMID: 33076256 PMCID: PMC7593949 DOI: 10.3390/ijms21207615] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Circulating micro-RNAs (miRNAs) modulate the expression of molecules in diabetes. We evaluated the expression of serum miRNA-195-5p and -451a in diabetic patients with ischemic stroke and correlated them with two markers of brain tissue integrity. (2) Methods: Seventy-eight subjects with acute ischemic stroke (AIS) or transient ischemic attack (TIA) (40 with diabetes) were enrolled. Serum miRNA levels, brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor A (VEGF-A) were assessed at admission and 24 and 72 h after a post-ischemic stroke, and were compared to 20 controls. (3) Results: Both circulating miRNAs were two-fold up-regulated in diabetic AIS and TIA patients compared to non-diabetics. Their levels progressively decreased at 24 and 72 h in both AIS and TIA patients. Interestingly, in the non-diabetic TIA group, both circulating miRNAs, although higher than the controls, tended to achieve a complete decay after 72 h. Furthermore, miRNA-195-5p and miRNA-451a levels inversely correlated with both BDNF and VEGF-A serum levels. (4) Conclusions: These data show a different profile of both micro-RNAs in diabetic versus non-diabetic patients after acute ischemic stroke, suggesting their pivotal role in cerebrovascular ischemic attack.
Collapse
Affiliation(s)
- Mauro Giordano
- Department of Advanced Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.G.); (F.S.); (G.P.); (L.E.A.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (M.G.); (R.A.)
| | - Tiziana Ciarambino
- Department of Internal Medicine, Hospital of Marcianise, ASL, 81025 Caserta, Italy;
| | - Michele D’Amico
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (M.G.); (R.A.)
| | - Marilena Galdiero
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (M.G.); (R.A.)
| | - Federico Schettini
- Department of Advanced Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.G.); (F.S.); (G.P.); (L.E.A.)
| | - Diego Paternosto
- Department of Emergency Medicine, AORN Sant’Anna e San Sebastiano,81100 Caserta, Italy; (D.P.); (M.S.); (V.A.)
| | - Marta Salzillo
- Department of Emergency Medicine, AORN Sant’Anna e San Sebastiano,81100 Caserta, Italy; (D.P.); (M.S.); (V.A.)
| | - Roberto Alfano
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.C.T.); (M.G.); (R.A.)
| | - Vincenzo Andreone
- Department of Emergency Medicine, AORN Sant’Anna e San Sebastiano,81100 Caserta, Italy; (D.P.); (M.S.); (V.A.)
| | | | - Gianni Biolo
- Department of Medical and Surgical Sciences, University of Trieste, 34100 Trieste, Italy;
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.G.); (F.S.); (G.P.); (L.E.A.)
| | - Luigi Elio Adinolfi
- Department of Advanced Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.G.); (F.S.); (G.P.); (L.E.A.)
| |
Collapse
|
77
|
Pawelec P, Ziemka-Nalecz M, Sypecka J, Zalewska T. The Impact of the CX3CL1/CX3CR1 Axis in Neurological Disorders. Cells 2020; 9:cells9102277. [PMID: 33065974 PMCID: PMC7600611 DOI: 10.3390/cells9102277] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Fractalkine (FKN, CX3CL1) is a transmembrane chemokine expressed by neurons in the central nervous system (CNS). CX3CL1 signals through its unique receptor, CX3CR1, that is expressed in microglia. Within the CNS, fractalkine acts as a regulator of microglia activation in response to brain injury or inflammation. During the last decade, there has been a growing interest in the roles that the CX3CL1/CX3CR1 signaling pathway plays in the neuropathology of a diverse array of brain disorders. However, the reported results have proven controversial, indicating that a disruption of the CX3CL1 axis induces a disease-specific microglial response that may have either beneficial or detrimental effects. Therefore, it has become clear that the understanding of neuron-to-glia signals mediated by CX3CL1/CX3CR1 at different stages of diseases could provide new insight into potential therapeutic targets. Hence, the aim of this review is to provide a summary of the literature on the emerging role of CX3CL1 in animal models of some brain disorders.
Collapse
|
78
|
Ng H, La Heij EC, Andrinopoulou ER, van Meurs JC, Vermeer KA. Smaller Foveal Avascular Zone in Deep Capillary Plexus Is Associated with Better Visual Acuity in Patients after Macula-off Retinal Detachment Surgery. Transl Vis Sci Technol 2020; 9:25. [PMID: 33024618 PMCID: PMC7521173 DOI: 10.1167/tvst.9.10.25] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/26/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose To associate the change in the foveal avascular zone (FAZ) and vessel density (VD) with final best corrected visual acuity (BCVA) in eyes after macula-off rhegmatogenous retinal detachment surgery, and to investigate the evolution of FAZ and VD during 12 months of follow-up. Methods We prospectively evaluated 47 patients with macula-off rhegmatogenous retinal detachment and healthy fellow eyes. At 1.5, 3.0, 6.0, and 12.0 months postoperatively, optical coherence tomography angiography scans were obtained from both eyes on a 3.0 × 3.0 mm macula-centered grid. En face images of the superficial vascular plexus, intermediate capillary plexus and deep capillary plexus were used to quantify FAZ and VD. BCVA was assessed with ETDRS-charts (logarithm of the minimal angle of resolution). At 12 months postoperatively, the association between the change in optical coherence tomography angiography parameters and visual function in study eyes was evaluated using the Spearman correlation coefficient. We calculated the BCVA difference and the percentage difference of FAZ and VD between the study and control eye. The evolution of FAZ and VD was investigated with linear mixed-effects models with nested random effects (eyes nested within patients). Results At 12 months postoperatively, FAZ difference of the deep capillary plexus and BCVA difference were correlated (P = 0.0004, rs = 0.5). Furthermore, there was no evidence that FAZ and VD changed during follow-up. Conclusions Although FAZ and VD remained stable during 12 months after surgery for macula-off rhegmatogenous retinal detachment, a smaller FAZ in the deep capillary plexus is associated with better BCVA. Translational relevance Reduction in FAZ area may be caused by angiogenesis to counteract ischemia, therefore therapeutic stimulation of angiogenesis could be beneficial to visual recovery.
Collapse
Affiliation(s)
- Heijan Ng
- Rotterdam Ophthalmic Institute, Rotterdam, the Netherlands
| | - Ellen C La Heij
- The Rotterdam Eye Hospital, Vitreoretinal Surgery, Rotterdam, the Netherlands
| | | | - Jan C van Meurs
- The Rotterdam Eye Hospital, Vitreoretinal Surgery, Rotterdam, the Netherlands.,Erasmus Medical Center, Department of Ophthalmology, Rotterdam, the Netherlands
| | | |
Collapse
|
79
|
Williamson MR, Franzen RL, Fuertes CJA, Dunn AK, Drew MR, Jones TA. A Window of Vascular Plasticity Coupled to Behavioral Recovery after Stroke. J Neurosci 2020; 40:7651-7667. [PMID: 32873722 PMCID: PMC7531554 DOI: 10.1523/jneurosci.1464-20.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/03/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke causes remodeling of vasculature surrounding the infarct, but whether and how vascular remodeling contributes to recovery are unclear. We established an approach to monitor and compare changes in vascular structure and blood flow with high spatiotemporal precision after photothrombotic infarcts in motor cortex using longitudinal 2-photon and multiexposure speckle imaging in mice of both sexes. A spatially graded pattern of vascular structural remodeling in peri-infarct cortex unfolded over the first 2 weeks after stroke, characterized by vessel loss and formation, and selective stabilization of a subset of new vessels. This vascular structural plasticity was coincident with transient activation of transcriptional programs relevant for vascular remodeling, reestablishment of peri-infarct blood flow, and large improvements in motor performance. Local vascular plasticity was strongly predictive of restoration of blood flow, which was in turn predictive of behavioral recovery. These findings reveal the spatiotemporal evolution of vascular remodeling after stroke and demonstrate that a window of heightened vascular plasticity is coupled to the reestablishment of blood flow and behavioral recovery. Our findings support that neovascularization contributes to behavioral recovery after stroke by restoring blood flow to peri-infarct regions. These findings may inform strategies for enhancing recovery from stroke and other types of brain injury.SIGNIFICANCE STATEMENT An improved understanding of neural repair could inform strategies for enhancing recovery from stroke and other types of brain injury. Stroke causes remodeling of vasculature surrounding the lesion, but whether and how the process of vascular remodeling contributes to recovery of behavioral function have been unclear. Here we used longitudinal in vivo imaging to track vascular structure and blood flow in residual peri-infarct cortex after ischemic stroke in mice. We found that stroke created a restricted period of heightened vascular plasticity that was associated with restoration of blood flow, which was in turn predictive of recovery of motor function. Therefore, our findings support that vascular remodeling facilitates behavioral recovery after stroke by restoring blood flow to peri-infarct cortex.
Collapse
Affiliation(s)
| | | | | | - Andrew K Dunn
- Institute for Neuroscience
- Department of Biomedical Engineering
| | - Michael R Drew
- Institute for Neuroscience
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712
| | | |
Collapse
|
80
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
81
|
Zhai X, Wang X, Wang L, Xiu L, Wang W, Pang X. Treating Different Diseases With the Same Method-A Traditional Chinese Medicine Concept Analyzed for Its Biological Basis. Front Pharmacol 2020; 11:946. [PMID: 32670064 PMCID: PMC7332878 DOI: 10.3389/fphar.2020.00946] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/10/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction The fundamental theory of traditional Chinese medicine (TCM) implies that when different diseases have the same pathogen, the syndromes of these individual diseases will be the same. “Treating different diseases with the same method” is a TCM principle suggesting that when different diseases have similar pathological changes during different stages of their development, the same method of treatment can be applied. Our study aims to analyze the concept “treating different diseases with the same method” from a molecular perspective, in order to clarify its biological basis and to objectively standardize future TCM syndrome research. Objective The TCM syndromes Qi deficiency and blood stasis have similar pathogenesis in relation to coronary heart disease (CHD) and stroke. We aim to use big data technology and complex network theory to mine the genes specifically relevant to these TCM syndromes. This study aims to explore the correlation between the biological indicators of CHD and stroke from a scientific perspective. Methods Mining the relevant neuroendocrine-immune (NEI) genes by means of gene entity recognition, complex network construction, network integration, and decomposition to categorize relevant syndrome terms and establish a digital dictionary of gene specifically related to individual diseases. We analyzed the biological basis of “treating different diseases with the same method” from a molecular level using the TCMIP v2.0 platform in order to categorize the TCM syndromes most relevant to CHD and stroke. Results We found 46 genes were involved in the TCM syndromes of Qi deficiency and blood stasis of CHD and stroke. The same genes and their molecular mechanism also appeared to be in close relation to inflammatory response, apoptosis, and proliferation. Conclusion By using information extraction and complex network technology, we discovered the biological indicators of the TCM syndromes Qi deficiency and blood stasis of CHD and stroke. In the era of big data, our results can provide a new method for the researchers of TCM syndrome differentiation, as well as an effective and specific methodology for standardization of TCM.
Collapse
Affiliation(s)
- Xing Zhai
- School of Management, Beijing University of Chinese Medicine, Beijing, China
| | - Xi Wang
- College of Humanities,Beijing University of Chinese Medicine, Beijing, China
| | - Li Wang
- School of Management, Beijing University of Chinese Medicine, Beijing, China
| | - Linlin Xiu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weilu Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohan Pang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
82
|
Zhang ZY, Fang YJ, Luo YJ, Lenahan C, Zhang JM, Chen S. The role of medical gas in stroke: an updated review. Med Gas Res 2020; 9:221-228. [PMID: 31898607 PMCID: PMC7802415 DOI: 10.4103/2045-9912.273960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Medical gas is a large class of bioactive gases used in clinical medicine and basic scientific research. At present, the role of medical gas in neuroprotection has received growing attention. Stroke is a leading cause of death and disability in adults worldwide, but current treatment is still very limited. The common pathological changes of these two types of stroke may include excitotoxicity, free radical release, inflammation, cell death, mitochondrial disorder, and blood-brain barrier disruption. In this review, we will discuss the pathological mechanisms of stroke and the role of two medical gases (hydrogen and hydrogen sulfide) in stroke, which may potentially provide a new insight into the treatment of stroke.
Collapse
Affiliation(s)
- Ze-Yu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yuan-Jian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yu-Jie Luo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM; Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jian-Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
83
|
Abate M, Pisanti S, Caputo M, Citro M, Vecchione C, Martinelli R. 3-Hydroxytyrosol Promotes Angiogenesis In Vitro by Stimulating Endothelial Cell Migration. Int J Mol Sci 2020; 21:E3657. [PMID: 32455901 PMCID: PMC7279499 DOI: 10.3390/ijms21103657] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases, followed by strokes, represent the leading cause of mortality worldwide. Despite its success in preventing cardiovascular diseases, the therapeutic potential of 3-Hydroxytyrosol (HT) for treating ischemic diseases is yet to be investigated in detail, especially with regard to ischemic heart disease, which is a major challenge for humans. We assessed that low concentrations (1-5 µM) of HT, generally achieved after the ingestion of olive oil, stimulate endothelial cells migration and angiogenesis in an in vitro model. At early time points (1-6 h), HT induces the expression of different proteins such as proto-oncogene tyrosine-protein kinase Src (Src), rho-associated protein kinase (ROCK) and matrix metalloproteinase-2 (MMP-2) protein influencing cell adhesion, cytoskeletal dynamics and cell migration. We observed that at the same time, HT induces prominent vascular formation in the tube formation assay, accompanied by an increase in the expression of the vascular endothelial growth factor receptor (VEGF-R2) and PI3K-Akt-eNOS protein pathways, which are recognized for their central role in angiogenesis. Therefore, in addition to the proven capability of HT to regulate reactive oxygen species (ROS) levels, through both direct scavenging properties and indirect antioxidant efficacy, our results revealed that HT promotes angiogenesis, arguing in favor of great pharma-nutritional potential in ischemic injuries.
Collapse
Affiliation(s)
- Mario Abate
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
| | - Simona Pisanti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
| | - Mariella Caputo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
| | - Marianna Citro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
- Vascular Pathophysiology Unit, IRCCS Neuromed, Via Atinense, Pozzilli, 86077 Isernia, Italy
| | - Rosanna Martinelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, Baronissi, 84081 Salerno, Italy; (M.A.); (S.P.); (M.C.); (M.C.); (C.V.)
| |
Collapse
|
84
|
Phosphodiesterase 10A Inhibition Leads to Brain Region-Specific Recovery Based on Stroke Type. Transl Stroke Res 2020; 12:303-315. [PMID: 32378029 PMCID: PMC7644574 DOI: 10.1007/s12975-020-00819-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
Stroke is the leading cause of adult disability. Recovery of function after stroke involves signaling events that are mediated by cAMP and cGMP pathways, such as axonal sprouting, neurogenesis, and synaptic plasticity. cAMP and cGMP are degraded by phosphodiesterases (PDEs), which are differentially expressed in brain regions. PDE10A is highly expressed in the basal ganglia/striatum. We tested a novel PDE10A inhibitor (TAK-063) for its effects on functional recovery. Stroke was produced in mice in the cortex or the striatum. Behavioral recovery was measured to 9 weeks. Tissue outcome measures included analysis of growth factor levels, angiogenesis, neurogenesis, gliogenesis, and inflammation. TAK-063 improved motor recovery after striatal stroke in a dose-related manner, but not in cortical stroke. Recovery of motor function correlated with increases in striatal brain-derived neurotrophic factor. TAK-063 treatment also increased motor system axonal connections. Stroke affects distinct brain regions, with each comprising different cellular and molecular elements. Inhibition of PDE10A improved recovery of function after striatal but not cortical stroke, consistent with its brain localization. This experiment is the first demonstration of brain region-specific enhanced functional recovery after stroke, and indicates that differential molecular signaling between brain regions can be exploited to improve recovery based on stroke subtype.
Collapse
|
85
|
Xie H, Zhang Q, Zhou N, Li C, Yu K, Liu G, Wu J, Jiang C, Hu R, Wu Y. Environmental enrichment enhances post-ischemic cerebral blood flow and functional hyperemia in the ipsilesional somatosensory cortex. Brain Res Bull 2020; 160:91-97. [PMID: 32388014 DOI: 10.1016/j.brainresbull.2020.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 11/16/2022]
Abstract
Environmental enrichment has been reported to promote functional recovery in an ischemic stroke. However, the underlying mechanism remains unclear. This study aimed to investigate the effect of environmental enrichment treatment on post-ischemic cerebral blood flow and functional hyperemia in the ipsilesional primary somatosensory cortex of rats. With laser speckle imaging, we were able to monitor the resting cerebral blood flow alteration in the middle cerebral artery occlusion model. Both 3- and 28-day post-ischemic infarct volumes were then examined with triphenyltetrazolium chloride and cresyl violet staining, respectively. We found that an exposure to environmental enrichment was associated with higher post-ischemic cerebral blood flow and less brain tissue loss in the ipsilesional primary somatosensory cortex compared with the standard cage environment. Furthermore, environmental enrichment also enhanced the cerebral blood flow response to whisker stimulation in the ipsilesional barrel cortex when measured 28 days after the middle cerebral artery occlusion. Together, the data suggested that an exposure to environmental enrichment promoted the restoration of cerebral blood flow in the ipsilesional cortex and contributed to a better coupling between functional activation and cerebral blood flow change, which might be the possible mechanisms underlying the neuroprotective effects of EE after ischemia.
Collapse
Affiliation(s)
- Hongyu Xie
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qun Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Naiyun Zhou
- Department of Biomedical Engineering, Stony Brook University, New York, USA
| | - Ce Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Kewei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Junfa Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Congyu Jiang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Ruiping Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
86
|
Kim Y, Lee S, Zhang H, Lee S, Kim H, Kim Y, Won MH, Kim YM, Kwon YG. CLEC14A deficiency exacerbates neuronal loss by increasing blood-brain barrier permeability and inflammation. J Neuroinflammation 2020; 17:48. [PMID: 32019570 PMCID: PMC7001304 DOI: 10.1186/s12974-020-1727-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/28/2020] [Indexed: 12/17/2022] Open
Abstract
Background Ischemic stroke is a main cause of mortality. Blood-brain barrier (BBB) breakdown appears to play a critical role in inflammation in patients with ischemic stroke and acceleration of brain injury. The BBB has a protective function and is composed of endothelial cells, pericytes, and astrocytes. In ischemic stroke treatments, regulation of vascular endothelial growth factor (VEGF)-A and vascular endothelial growth factor receptor (VEGFR)-2 is a crucial target despite adverse effects. Our previous study found that loss of C-type lectin family 14 member A (CLEC14A) activated VEGF-A/VEGFR-2 signaling in developmental and tumoral angiogenesis. Here, we evaluate the effects of BBB impairment caused by CLEC14A deficiency in ischemia-reperfusion injury. Methods In vitro fluorescein isothiocyanate (FITC)-dextran permeability, transendothelial electrical resistance (TEER) assay, and immunostaining were used to evaluate endothelial integrity. BBB permeability was assessed using Evans blue dye and FITC-dextran injection in Clec14a−/− (CLEC14A-KO) mice and wild-type mice. Middle cerebral artery occlusion surgery and behavioral assessments were performed to evaluate the neurologic damage. The change of tight junctional proteins, adhesion molecules, pro-inflammatory cytokines, and microglial were confirmed by immunofluorescence staining, Western blotting, and quantitative reverse transcription polymerase chain reaction of brain samples. Results In endothelial cells, knockdown of CLEC14A increased FITC-dextran permeability and decreased transendothelial electrical resistance; the severity of this effect increased with VEGF treatment. Immunofluorescence staining revealed that tight junctional proteins were attenuated in the CLEC14A knockdown endothelial cells. Consistent with the in vitro results, CLEC14A-KO mice that were injected with Evans blue dye had cerebral vascular leakage at postnatal day 8; wild-type mice had no leakage. We used a middle cerebral artery occlusion model and found that CLEC14A-KO mice had severe infarcted brain and neurological deficits with upregulated VEGFR-2 expression. FITC-dextran leakage was present in CLEC14A-KO mice after ischemia-reperfusion, and the numbers of tight junctional molecules were significantly decreased. Loss of CLEC14A increased the pro-inflammatory response through adhesion molecule expression, and glial cells were activated. Conclusions These results suggest that activation of VEGFR-2 in CLEC14A-KO mice aggravates ischemic stroke by exacerbating cerebral vascular leakage and increasing neuronal inflammation after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yeomyeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Sungwoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Haiying Zhang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Sunghye Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Hyejeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Yeaji Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea.
| |
Collapse
|
87
|
Growth Hormone Promotes Motor Function after Experimental Stroke and Enhances Recovery-Promoting Mechanisms within the Peri-Infarct Area. Int J Mol Sci 2020; 21:ijms21020606. [PMID: 31963456 PMCID: PMC7013985 DOI: 10.3390/ijms21020606] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/31/2022] Open
Abstract
Motor impairment is the most common and widely recognised clinical outcome after stroke. Current clinical practice in stroke rehabilitation focuses mainly on physical therapy, with no pharmacological intervention approved to facilitate functional recovery. Several studies have documented positive effects of growth hormone (GH) on cognitive function after stroke, but surprisingly, the effects on motor function remain unclear. In this study, photothrombotic occlusion targeting the motor and sensory cortex was induced in adult male mice. Two days post-stroke, mice were administered with recombinant human GH or saline, continuing for 28 days, followed by evaluation of motor function. Three days after initiation of the treatment, bromodeoxyuridine was administered for subsequent assessment of cell proliferation. Known neurorestorative processes within the peri-infarct area were evaluated by histological and biochemical analyses at 30 days post-stroke. This study demonstrated that GH treatment improves motor function after stroke by 50%–60%, as assessed using the cylinder and grid walk tests. Furthermore, the observed functional improvements occurred in parallel with a reduction in brain tissue loss, as well as increased cell proliferation, neurogenesis, increased synaptic plasticity and angiogenesis within the peri-infarct area. These findings provide new evidence about the potential therapeutic effects of GH in stroke recovery.
Collapse
|
88
|
Heydari E, Alishahi M, Ghaedrahmati F, Winlow W, Khoshnam SE, Anbiyaiee A. The role of non-coding RNAs in neuroprotection and angiogenesis following ischemic stroke. Metab Brain Dis 2020; 35:31-43. [PMID: 31446548 DOI: 10.1007/s11011-019-00485-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022]
Abstract
Stroke is the leading cause of death and physical disability worldwide. Non-coding RNAs (ncRNAs) are endogenous molecules that play key roles in the pathophysiology and retrieval processes following ischemic stroke. The potential of ncRNAs, especially microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in neuroprotection and angiogenesis highlights their potential as targets for therapeutic intervention. In this review, we document the miRNAs and lncRNAs that have been reported to exert regulatory actions in neuroprotective and angiogenic processes through different mechanisms involving their interaction with target coding genes. We believe that exploration of the expression profiles and the possible functions of ncRNAs during the recovery processes will help comprehension of the molecular mechanisms responsible for neuroprotection and angiogenesis, and may also contribute to find biomarkers and targets for future stroke intervention.
Collapse
Affiliation(s)
- Elaheh Heydari
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Farhoodeh Ghaedrahmati
- Immunology Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - William Winlow
- Dipartimento di Biologia, Università degli Studi di Napoli, Federico II, Via Cintia 26, 80126, Napoli, Italy
- Honorary Research Fellow, Institute of Ageing and Chronic Diseases, University of Liverpool, The APEX building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 6135715794, Iran.
| | - Amir Anbiyaiee
- Department of Obstetrics & Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran.
| |
Collapse
|
89
|
Regenhardt RW, Takase H, Lo EH, Lin DJ. Translating concepts of neural repair after stroke: Structural and functional targets for recovery. Restor Neurol Neurosci 2020; 38:67-92. [PMID: 31929129 PMCID: PMC7442117 DOI: 10.3233/rnn-190978] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stroke is among the most common causes of adult disability worldwide, and its disease burden is shifting towards that of a long-term condition. Therefore, the development of approaches to enhance recovery and augment neural repair after stroke will be critical. Recovery after stroke involves complex interrelated systems of neural repair. There are changes in both structure (at the molecular, cellular, and tissue levels) and function (in terms of excitability, cortical maps, and networks) that occur spontaneously within the brain. Several approaches to augment neural repair through enhancing these changes are under study. These include identifying novel drug targets, implementing rehabilitation strategies, and developing new neurotechnologies. Each of these approaches has its own array of different proposed mechanisms. Current investigation has emphasized both cellular and circuit-based targets in both gray and white matter, including axon sprouting, dendritic branching, neurogenesis, axon preservation, remyelination, blood brain barrier integrity, blockade of extracellular inhibitory signals, alteration of excitability, and promotion of new brain cortical maps and networks. Herein, we review for clinicians recovery after stroke, basic elements of spontaneous neural repair, and ongoing work to augment neural repair. Future study requires alignment of basic, translational, and clinical research. The field continues to grow while becoming more clearly defined. As thrombolysis changed stroke care in the 1990 s and thrombectomy in the 2010 s, the augmentation of neural repair and recovery after stroke may revolutionize care for these patients in the coming decade.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Hajime Takase
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Eng H Lo
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - David J Lin
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
90
|
Peng H, Yang H, Xiang X, Li S. ΜicroRNA-221 participates in cerebral ischemic stroke by modulating endothelial cell function by regulating the PTEN/PI3K/AKT pathway. Exp Ther Med 2019; 19:443-450. [PMID: 31885694 PMCID: PMC6913279 DOI: 10.3892/etm.2019.8263] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/27/2019] [Indexed: 12/17/2022] Open
Abstract
An effective method to improve the blood supply of brain tissue is by angiogenesis, which is crucial for the prognosis of patients with cerebral ischemic stroke (CIS). Therefore, angiogenesis has been a focus of CIS research in recent years. The present study aimed to investigate the expression of microRNA (miR)-221 in patients with CIS and to explore the effect of miR-221 on endothelial cell function. The level of miR-221 was detected using reverse transcription-quantitative PCR (RT-qPCR). The relationship between miR-221 and phosphatase and tensin homolog (PTEN) was predicted and confirmed by bioinformatics and dual luciferase reporter assay. Cell viability, migration and invasion, and cell apoptosis were determined using MTT assay, Transwell assay and flow cytometry respectively. Tube formation in human umbilical vein endothelial cells (HUVECs) was determined by performing the tube formation assay. In addition, protein levels were measured using western blot analysis. The results of the current study indicated that miR-221 levels were significantly decreased in the peripheral blood of patients with CIS. PTEN was confirmed to be a direct target of miR-221. Downregulation of miR-221 significantly inhibited the function of HUVECs as evidenced by the decreased cell viability, migration and invasion with increased cell apoptosis and tube formation inhibition. miR-221 upregulation produced the reverse effects, whilst all the effects of miR-221 upregulation on HUVECs were reversed by PTEN overexpression. The PI3K/AKT pathway was identified to be involved in the regulation of miR-221 on HUVECs. In conclusion, miR-221 was downregulated in CIS patients, and it promoted the function of HUVECs by regulating the PTEN/PI3K/AKT pathway in vitro, suggesting the ability to promote angiogenesis. Therefore, miR-221 may be a novel and promising therapeutic target for CIS treatment.
Collapse
Affiliation(s)
- Han Peng
- Department of Pathology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Xin Xiang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Shenggang Li
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
91
|
Beckmann L, Zhang X, Nadkarni NA, Cai Z, Batra A, Sullivan DP, Muller WA, Sun C, Kuranov R, Zhang HF. Longitudinal deep-brain imaging in mouse using visible-light optical coherence tomography through chronic microprism cranial window. BIOMEDICAL OPTICS EXPRESS 2019; 10:5235-5250. [PMID: 31646044 PMCID: PMC6788609 DOI: 10.1364/boe.10.005235] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 05/02/2023]
Abstract
We longitudinally imaged both the superficial and deep cortical microvascular networks in brains of healthy mice and in a mouse model of stroke in vivo using visible-light optical coherence tomography (vis-OCT). We surgically implanted a microprism in mouse brains sealed by a chronic cranial window. The microprism enabled vis-OCT to image the entire depth of the mouse cortex. Following microprism implantation, we imaged the mice for 28 days and found that that it took around 15 days for both the superficial and deep cortical microvessels to recover from the implantation surgery. After the brains recovered, we introduced ischemic strokes by transient middle cerebral artery occlusion (tMCAO). We monitored the strokes for up to 60 days and observed different microvascular responses to tMCAO at different cortical depths in both the acute and chronic phases of the stroke. This work demonstrates that the combined microprism and cranial window is well-suited for longitudinal investigation of cortical microvascular disorders using vis-OCT.
Collapse
Affiliation(s)
- Lisa Beckmann
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208, USA
- These authors contributed equally to this work
| | - Xian Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208, USA
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan, Hubei, China
- These authors contributed equally to this work
| | - Neil A. Nadkarni
- Department of Neurology, Northwestern University, Chicago IL 60611, USA
| | - Zhen Cai
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208, USA
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan, Hubei, China
| | - Ayush Batra
- Department of Neurology, Northwestern University, Chicago IL 60611, USA
| | - David P. Sullivan
- Department of Pathology, Northwestern University, Chicago IL 60611, USA
| | - William A. Muller
- Department of Pathology, Northwestern University, Chicago IL 60611, USA
| | - Cheng Sun
- Department of Mechanical Engineering, Northwestern University, Evanston IL 60208, USA
| | - Roman Kuranov
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208, USA
- Opticent Health, Evanston IL, Evanston IL 60201, USA
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208, USA
| |
Collapse
|
92
|
Conflicting Roles of 20-HETE in Hypertension and Stroke. Int J Mol Sci 2019; 20:ijms20184500. [PMID: 31514409 PMCID: PMC6770042 DOI: 10.3390/ijms20184500] [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: 07/17/2019] [Revised: 08/28/2019] [Accepted: 09/08/2019] [Indexed: 12/15/2022] Open
Abstract
Hypertension is the most common modifiable risk factor for stroke, and understanding the underlying mechanisms of hypertension and hypertension-related stroke is crucial. 20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid (20-HETE), which plays an important role in vasoconstriction, autoregulation, endothelial dysfunction, angiogenesis, inflammation, and blood-brain barrier integrity, has been linked to hypertension and stroke. 20-HETE can promote hypertension by potentiating the vascular response to vasoconstrictors; it also can reduce blood pressure by inhibition of sodium transport in the kidney. The production of 20-HETE is elevated after the onset of both ischemic and hemorrhagic strokes; on the other hand, subjects with genetic variants in CYP4F2 and CYP4A11 that reduce 20-HETE production are more susceptible to stroke. This review summarizes recent genetic variants in CYP4F2, and CYP4A11 influencing 20-HETE production and discusses the role of 20-HETE in hypertension and the susceptibility to the onset, progression, and prognosis of ischemic and hemorrhagic strokes.
Collapse
|
93
|
Matboli M, Shafei AE, Agwa SH, Elzahy SS, Anwar AK, Mansour AR, Gaber AI, Said AE, Lwis P, Hamdy M. Identification of Novel Molecular Network Expression in Acute Myocardial Infarction. Curr Genomics 2019; 20:340-348. [PMID: 32476991 PMCID: PMC7235391 DOI: 10.2174/1389202920666190820142043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In the current study, we aimed to analyze the hypothesis that human myocardial-specific extracellular RNAs expression could be used for acute myocardial injury(AMI) diagnosis. METHODOLOGY We used bioinformatics' analysis to identify RNAs linked to ubiquitin system and specific to AMI, named, (lncRNA-RP11-175K6.1), (LOC101927740), microRNA-106b-5p (miR-106b-5p) and Anaphase, promoting complex 11 (ANapc11mRNA). We measured the serum expression of the chosen RNAs in 69 individuals with acute coronary syndromes, 31 individuals with angina pectoris without MI and non-cardiac chest pain and 31 healthy control individuals by real-time reverse-transcription PCR. RESULTS Our study revealed a significant decrease in both lncRNA-RP11-175K6.1 and ANapc11mRNA expression of in the sera samples of AMI patients compared to that of the two control groups alongside with significant upregulation of miR-106b-5p. CONCLUSION Of note, the investigated serum RNAs decrease the false discovery rate of AMI to 3.2%.
Collapse
Affiliation(s)
- Marwa Matboli
- Medicinal Biochemistry and Molecular Biology Department, Ain Shams University, Faculty of Medicine, Cairo, Egypt
| | - Ayman E. Shafei
- Biomedical Research Department, Military Medical Academy, Cairo, Egypt
- Biomedical Research Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Sara H.A. Agwa
- Clinical Pathology, Medical Ain Shams Research Institute (MASRI), Cairo, Egypt
| | - Sherif Sammir Elzahy
- Cardiovascular Medicine Department, Ain Shams University, Faculty of Medicine, Cairo, Egypt
| | | | | | | | | | - Paula Lwis
- Armed Forces College of Medicine, Cairo, Egypt
| | - Marwa Hamdy
- Medicinal Biochemistry and Molecular Biology Department, Ain Shams University, Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
94
|
Poster Viewing Sessions PA00-A01 to PA00-A49. J Cereb Blood Flow Metab 2019; 39:124-166. [PMID: 31265792 PMCID: PMC6610576 DOI: 10.1177/0271678x19851017] [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: 11/17/2022]
|
95
|
Nogo-A targeted therapy promotes vascular repair and functional recovery following stroke. Proc Natl Acad Sci U S A 2019; 116:14270-14279. [PMID: 31235580 DOI: 10.1073/pnas.1905309116] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stroke is a major cause of serious disability due to the brain's limited capacity to regenerate damaged tissue and neuronal circuits. After ischemic injury, a multiphasic degenerative and inflammatory response is coupled with severely restricted vascular and neuronal repair, resulting in permanent functional deficits. Although clinical evidence indicates that revascularization of the ischemic brain regions is crucial for functional recovery, no therapeutics that promote angiogenesis after cerebral stroke are currently available. Besides vascular growth factors, guidance molecules have been identified to regulate aspects of angiogenesis in the central nervous system (CNS) and may provide targets for therapeutic angiogenesis. In this study, we demonstrate that genetic deletion of the neurite outgrowth inhibitor Nogo-A or one of its corresponding receptors, S1PR2, improves vascular sprouting and repair and reduces neurological deficits after cerebral ischemia in mice. These findings were reproduced in a therapeutic approach using intrathecal anti-Nogo-A antibodies; such a therapy is currently in clinical testing for spinal cord injury. These results provide a basis for a therapeutic blockage of inhibitory guidance molecules to improve vascular and neural repair after ischemic CNS injuries.
Collapse
|
96
|
Leech T, Chattipakorn N, Chattipakorn SC. The beneficial roles of metformin on the brain with cerebral ischaemia/reperfusion injury. Pharmacol Res 2019; 146:104261. [PMID: 31170502 DOI: 10.1016/j.phrs.2019.104261] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Cerebral ischaemia/reperfusion (I/R) injury is the transient loss, followed by rapid return, of blood flow to the brain. This condition is often caused by strokes and heart attacks. The underlying mechanisms resulting in brain damage during cerebral I/R injury include mitochondrial dysregulation, increased oxidative stress/reactive oxygen species, blood-brain-barrier breakdown, inflammation of the brain, and increased neuronal apoptosis. Metformin is the first-line antidiabetic drug which has recently been shown to be capable of acting through the aforementioned pathways to improve recovery following cerebral I/R injury. However, some studies have suggested that metformin therapy may have no effect or even worsen recovery following cerebral I/R injury. The present review will compile and examine the available in vivo, in vitro, and clinical data concerning the neuroprotective effects of metformin following cerebral I/R injury. Any contradictory evidence will also be assessed and presented to determine the actual effectiveness of metformin treatment in stroke recovery.
Collapse
Affiliation(s)
- Tom Leech
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, United Kingdom; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
97
|
Uric acid treatment after stroke modulates the Krüppel-like factor 2-VEGF-A axis to protect brain endothelial cell functions: Impact of hypertension. Biochem Pharmacol 2019; 164:115-128. [DOI: 10.1016/j.bcp.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 12/29/2022]
|
98
|
Shen J, Zhao Z, Shang W, Liu C, Zhang B, Xu Z, Cai H. Fabrication and evaluation a transferrin receptor targeting nano-drug carrier for cerebral infarction treatment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:192-200. [PMID: 30663409 DOI: 10.1080/21691401.2018.1548471] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
After cerebral infarction, the regeneration of microvascular played an important role in the recovery. Ginsenoside Rg1 (Rg1) had good effects on promoting angiogenesis and neuro-protection in cerebral infarction treatment. However, the blood-brain barrier (BBB) restricted Rg1 to enter into cerebral tissue. Transferrin receptor (TfR) was over-expressed in the BBB. In this study, we fabricated a TfR targeting nano-carrier (PATRC) to penetrate the BBB for treatment of cerebral infarction. A TfR targeted peptide was conjugated with the nano-carrier wrapped hydrophobic Rg1. The nanoscale size (132 ± 12 nm), polydispersity index (PDI =0.29) and the zeta potential (-38mv) were tested with dynamic light scattering optical system. Surface morphology (ellipse, mean diameter 122 ± 26 nm) was detected by transmission electron microscope (TEM). PATRC implement cell targeting ability on rat brain microvascular endothelial cells RBE4 in vitro detected by immunofluorescence and flow cytometry methods. Comparing with Rg1 threated group, the PATRC exhibited more prominent ability on the tube formation ability (p < .05) in vitro. Comparing with the Rg1 treated group, PATRC penetrated BBB in vivo detected by HPLC, decreased the brain infarction volume tested with TTC staining and promoted regeneration of microvascular in infarction zone detected by CD31 immunofluorescence. PATRC has great potentiality for wide application in clinic.
Collapse
Affiliation(s)
- Junyi Shen
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Zhiming Zhao
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Wei Shang
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Chunli Liu
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Beibei Zhang
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Zihan Xu
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| | - Hui Cai
- a Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine , Nanjing University , Nanjing , China
| |
Collapse
|
99
|
Liu L, Anderson GA, Fernandez TG, Doré S. Efficacy and Mechanism of Panax Ginseng in Experimental Stroke. Front Neurosci 2019; 13:294. [PMID: 31068769 PMCID: PMC6491687 DOI: 10.3389/fnins.2019.00294] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/13/2019] [Indexed: 12/30/2022] Open
Abstract
Stroke is one of the leading causes of death and long-term disability worldwide. However, effective therapeutic approaches are still limited. The disruption of blood supply triggers complicated temporal and spatial events involving hemodynamic, biochemical, and neurophysiologic changes, eventually leading to pathological disturbance and diverse clinical symptoms. Ginseng (Panax ginseng), a popular herb distributed in East Asia, has been extensively used as medicinal and nutritional supplements for a variety of disorders worldwide. In recent years, ginseng has displayed attractive beneficial effects in distinct neurological disorders including stroke, involving multiple protective mechanisms. In this article, we reviewed the literature on ginseng studies in the experimental stroke field, particularly focusing on the in vivo evidence on the preventive or therapeutic efficacy and mechanisms of ginseng and ginsenosides in various stroke models of mice and rats. We also summarized the efficacy and underlying mechanisms of ginseng and ginsenosides on short- and long-term stroke outcomes.
Collapse
Affiliation(s)
- Lei Liu
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Gigi A Anderson
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Tyler G Fernandez
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Departments of Neurology, Psychiatry, Pharmaceutics, and Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
100
|
Gu N, Wang J, Di Z, Liu Z, Jia X, Yan Y, Chen X, Zhang Q, Qian Y. The Effects of Intelectin-1 on Antioxidant and Angiogenesis in HUVECs Exposed to Oxygen Glucose Deprivation. Front Neurol 2019; 10:383. [PMID: 31040819 PMCID: PMC6477047 DOI: 10.3389/fneur.2019.00383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Objective: Ischemic stroke leads to cellular death and tissue damage by depriving the areas of glucose and oxygen supplies. The effective treatment of stroke remains a challenge for modern medicine. This study used an oxygen-glucose deprivation (OGD) model of human umbilical vein endothelial cells (HUVECs) to mimic ischemic injuries and explored the role and mechanism of intelectin-1. Methods: Intelectin-1 was transduced into the HUVECs using a lentiviral vector. The PI3K/Akt signaling was examined in intelectin-induced eNOS phosphorylation. The PI3K inhibitor LY294002 was dealed in HUVECs. Results: Our results demonstrated an increase in capillary density, decrease in apoptotic cells, and increase in HIF-1α protein expression following intelectin-1 treatment. Real-time PCR and Western blotting revealed the increased intelectin-1 expression alongside eNOS and Akt phosphorylation with enhanced bcl-2 expression under OGD. Capillary density decreased significantly after LY294002 treatment. Conclusion: These results suggest intelectin-1 promotes angiogenesis, inhibits oxidative stress and reduces apoptosis by stimulating the Akt-eNOS signaling pathway in response to ischemia in vitro.
Collapse
Affiliation(s)
- Naibing Gu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Jun Wang
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Zhengli Di
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Zhiqin Liu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xiaotao Jia
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yu'e Yan
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Xiaoshan Chen
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Quanzeng Zhang
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|