1
|
Menet R, Nasrallah L, Bernard M, Allain A, ElAli A. VEGF-E Attenuates Injury After Ischemic Stroke by Promoting Reparative Revascularization. Eur J Neurosci 2025; 61:e70114. [PMID: 40277075 PMCID: PMC12023035 DOI: 10.1111/ejn.70114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
The angiogenic response after stroke correlates with mild injury and an improved recovery. Stimulation of post-stroke angiogenesis using vascular endothelial growth factor (VEGF)-A is associated with an increased risk of vascular destabilization, leading to life-threatening complications. The non-mammalian VEGF-A homolog, VEGF-E, stimulates stable cutaneous vascularization and promotes wound healing. Herein, we posit that VEGF-E represents a potential mediator of reparative revascularization after ischemic stroke. C57BL6/J wildtype mice were subjected to experimental stroke, and VEGF-E or VEGF-A were intranasally delivered during the subacute phase. Our results indicate that VEGF-E improves neurological recovery and increases vascular density without compromising permeability, more efficiently than VEGF-A. We show that VEGF-E-mediated revascularization correlates with normal restoration of brain perfusion, whereas VEGF-A induces cerebral hyperperfusion, indicative of vascular dysfunction. Furthermore, VEGF-E reduces microvascular stalls, increases the density of angiogenic vasculature, and improves the interaction of brain endothelial cell with pericytes, which is critical for vascular stabilization. Using cell-based assays, we demonstrate that stimulation of brain endothelial cells with VEGF-E, but not with VEGF-A, increases the expression of platelet-derived growth factor (PDGF)-D, a potent ligand of PDGFRβ that plays critical roles in regulating the survival and functions of perivascular cells, including pericytes. These effects are associated with activation of extracellular signal-regulated kinase (ERK)1/2 and P38 mitogen-activated protein kinase (MAPK). Finally, we confirm that the secretome of VEGF-E-stimulated brain endothelial cells ameliorates pericyte migration required for vascular recruitment. Our study indicates that VEGF-E promotes a stable and functional revascularization after ischemic stroke, outlining its promises for therapeutic purposes.
Collapse
Affiliation(s)
- Romain Menet
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Leila Nasrallah
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Maxime Bernard
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Anne‐Sophie Allain
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Ayman ElAli
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| |
Collapse
|
2
|
Phelps J, Hart DA, Mitha AP, Duncan NA, Sen A. Extracellular Vesicles Generated by Mesenchymal Stem Cells in Stirred Suspension Bioreactors Promote Angiogenesis in Human-Brain-Derived Endothelial Cells. Int J Mol Sci 2024; 25:5219. [PMID: 38791256 PMCID: PMC11121007 DOI: 10.3390/ijms25105219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Interrupted blood flow in the brain due to ischemic injuries such as ischemic stroke or traumatic brain injury results in irreversible brain damage, leading to cognitive impairment associated with inflammation, disruption of the blood-brain barrier (BBB), and cell death. Since the BBB only allows entry to a small class of drugs, many drugs used to treat ischemia in other tissues have failed in brain-related disorders. The administration of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) has shown promise in improving the functional recovery of the brain following cerebral ischemia by inducing blood vessel formation. To facilitate such a treatment approach, it is necessary to develop bioprocesses that can produce therapeutically relevant MSC-EVs in a reproducible and scalable manner. This study evaluated the feasibility of using stirred suspension bioreactors (SSBs) to scale-up the serum-free production of pro-angiogenic MSC-EVs under clinically relevant physioxic conditions. It was found that MSCs grown in SSBs generated EVs that stimulated angiogenesis in cerebral microvascular endothelial cells, supporting the use of SSBs to produce MSC-EVs for application in cerebral ischemia. These properties were impaired at higher cell confluency, outlining the importance of considering the time of harvest when developing bioprocesses to manufacture EV populations.
Collapse
Affiliation(s)
- Jolene Phelps
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada;
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; (D.A.H.); (A.P.M.)
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB T2N 4Z6, Canada;
| | - David A. Hart
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; (D.A.H.); (A.P.M.)
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB T2N 4Z6, Canada;
- Department of Surgery, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB T2N 4N1, Canada
- Faculty of Kinesiology, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada
| | - Alim P. Mitha
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; (D.A.H.); (A.P.M.)
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, 1403 29 Street N.W., Calgary, AB T2N 2T9, Canada
| | - Neil A. Duncan
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB T2N 4Z6, Canada;
- Department of Surgery, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB T2N 4N1, Canada
- Department of Civil Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada;
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada; (D.A.H.); (A.P.M.)
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB T2N 4Z6, Canada;
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB T2N 1N4, Canada
| |
Collapse
|
3
|
Díaz-Pérez A, Pérez B, Manich G, García-Aranda J, Navarro X, Penas C, Jiménez-Altayó F. Histone deacetylase inhibition by suberoylanilide hydroxamic acid during reperfusion promotes multifaceted brain and vascular protection in spontaneously hypertensive rats with transient ischaemic stroke. Biomed Pharmacother 2024; 172:116287. [PMID: 38382328 DOI: 10.1016/j.biopha.2024.116287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024] Open
Abstract
Hypertension is the most prevalent modifiable risk factor for stroke and is associated with worse functional outcomes. Pharmacological inhibition of histone deacetylases by suberoylanilide hydroxamic acid (SAHA) modulates gene expression and has emerged as a promising therapeutic approach to reduce ischaemic brain injury. Here, we have tested the therapeutic potential of SAHA administered during reperfusion in adult male spontaneously hypertensive (SHR) rats subjected to transient middle cerebral artery occlusion (tMCAO; 90 min occlusion/24 h reperfusion). Animals received a single dose of SAHA (50 mg/kg) or vehicle i.p. at 1, 4, or 6 h after reperfusion onset. The time-course of brain histone H3 acetylation was studied. After tMCAO, drug brain penetrance and beneficial effects on behavioural outcomes, infarct volume, oedema, angiogenesis, blood-brain barrier integrity, cerebral artery oxidative stress and remodelling, and brain and vascular inflammation were evaluated. SAHA increased brain histone H3 acetylation from 1 to 6 h after injection, reaching the ischaemic brain administered during reperfusion. Treatment given at 4 h after reperfusion onset improved neurological score, reduced infarct volume and oedema, attenuated microglial activation, prevented exacerbated MCA angiogenic sprouting and blood-brain barrier breakdown, normalised MCA oxidative stress and remodelling, and modulated brain and cerebrovascular cytokine expression. Overall, we demonstrate that SAHA administered during early reperfusion exerts robust brain and vascular protection after tMCAO in hypertensive rats. These findings are aligned with previous research in ischaemic normotensive mice and help pave the way to optimise the design of clinical trials assessing the effectiveness and safety of SAHA in ischaemic stroke.
Collapse
Affiliation(s)
- Andrea Díaz-Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Gemma Manich
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Human Anatomy and Embriology Unit, Department of Morphological Sciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Julián García-Aranda
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Red Española de Terapias Avanzadas (RED-TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Penas
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Red Española de Terapias Avanzadas (RED-TERAV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Furman M, Sihotsky V, Virag M, Kopolovets I, Nemethova M, Mucha R. Quantitative analysis of selected genetic markers of induced brain stroke ischemic tolerance detected in human blood. Brain Res 2023; 1821:148590. [PMID: 37739332 DOI: 10.1016/j.brainres.2023.148590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
A brain stroke is a serious disease and the second leading cause of death in the European Union. Carotid stenosis accounts for 15% of all ischemic cerebral strokes. However, there is currently no effective screening for carotid disease. Analysis of the DNA from peripheral blood is increasingly being used for several disease diagnoses. The potentially beneficial therapeutic method of inducing tissue tolerance to ischemia has so far been studied mainly in animal models. The aim of this study is to investigate changes in the gene expression of selected markers of brain ischemia during carotid endarterectomy, considered in this study as an activator of ischemic tolerance. During the carotid endarterectomy, there is a short-term occlusion of the internal carotid artery. Using the RT-qPCR method, we detected changes in the early identified gene markers of brain ischemia (ADM, CDKN1A, GADD45G, IL6, TM4SF1) in peripheral blood during sub lethal cerebral ischemia caused by carotid endarterectomy. Patients underwenting surgical procedure were divided into three groups: asymptomatic, symptomatic, and those who underwent carotid endarterectomy after an acute stroke. The results were compared to a negative/control group. Carotid endarterectomy had an impact on the expression of all monitored biomarkers. We observed statistically significant changes (p value 0.05-0.001) when comparing the groups among themselves, as well as the presence of ischemic tolerance of brain tissue to ischemic attacks. In conclusion, ADM, GADD45G, and TM4SF1 were affected in symptomatic patients, GADD45G and IL6 in acute patients, and CDKN1A and ADM in asymptomatic group after application of carotid endarterectomy.
Collapse
Affiliation(s)
- Marek Furman
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Vladimir Sihotsky
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Michal Virag
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Ivan Kopolovets
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Miroslava Nemethova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Rastislav Mucha
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia.
| |
Collapse
|
5
|
Rejdak K, Sienkiewicz-Jarosz H, Bienkowski P, Alvarez A. Modulation of neurotrophic factors in the treatment of dementia, stroke and TBI: Effects of Cerebrolysin. Med Res Rev 2023; 43:1668-1700. [PMID: 37052231 DOI: 10.1002/med.21960] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophic factors (NTFs) are involved in the pathophysiology of neurological disorders such as dementia, stroke and traumatic brain injury (TBI), and constitute molecular targets of high interest for the therapy of these pathologies. In this review we provide an overview of current knowledge of the definition, discovery and mode of action of five NTFs, nerve growth factor, insulin-like growth factor 1, brain derived NTF, vascular endothelial growth factor and tumor necrosis factor alpha; as well as on their contribution to brain pathology and potential therapeutic use in dementia, stroke and TBI. Within the concept of NTFs in the treatment of these pathologies, we also review the neuropeptide preparation Cerebrolysin, which has been shown to resemble the activities of NTFs and to modulate the expression level of endogenous NTFs. Cerebrolysin has demonstrated beneficial treatment capabilities in vitro and in clinical studies, which are discussed within the context of the biochemistry of NTFs. The review focuses on the interactions of different NTFs, rather than addressing a single NTF, by outlining their signaling network and by reviewing their effect on clinical outcome in prevalent brain pathologies. The effects of the interactions of these NTFs and Cerebrolysin on neuroplasticity, neurogenesis, angiogenesis and inflammation, and their relevance for the treatment of dementia, stroke and TBI are summarized.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | | | | | - Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, Coruña, Spain
| |
Collapse
|
6
|
Phelps J, Hart DA, Mitha AP, Duncan NA, Sen A. Physiological oxygen conditions enhance the angiogenic properties of extracellular vesicles from human mesenchymal stem cells. Stem Cell Res Ther 2023; 14:218. [PMID: 37612731 PMCID: PMC10463845 DOI: 10.1186/s13287-023-03439-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/01/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Following an ischemic injury to the brain, the induction of angiogenesis is critical to neurological recovery. The angiogenic benefits of mesenchymal stem cells (MSCs) have been attributed at least in part to the actions of extracellular vesicles (EVs) that they secrete. EVs are membrane-bound vesicles that contain various angiogenic biomolecules capable of eliciting therapeutic responses and are of relevance in cerebral applications due to their ability to cross the blood-brain barrier (BBB). Though MSCs are commonly cultured under oxygen levels present in injected air, when MSCs are cultured under physiologically relevant oxygen conditions (2-9% O2), they have been found to secrete higher amounts of survival and angiogenic factors. There is a need to determine the effects of MSC-EVs in models of cerebral angiogenesis and whether those from MSCs cultured under physiological oxygen provide greater functional effects. METHODS Human adipose-derived MSCs were grown in clinically relevant serum-free medium and exposed to either headspace oxygen concentrations of 18.4% O2 (normoxic) or 3% O2 (physioxic). EVs were isolated from MSC cultures by differential ultracentrifugation and characterized by their size, concentration of EV specific markers, and their angiogenic protein content. Their functional angiogenic effects were evaluated in vitro by their induction of cerebral microvascular endothelial cell (CMEC) proliferation, tube formation, and angiogenic and tight junction gene expressions. RESULTS Compared to normoxic conditions, culturing MSCs under physioxic conditions increased their expression of angiogenic genes SDF1 and VEGF, and subsequently elevated VEGF-A content in the EV fraction. MSC-EVs demonstrated an ability to induce CMEC angiogenesis by promoting tube formation, with the EV fraction from physioxic cultures having the greatest effect. The physioxic EV fraction further upregulated the expression of CMEC angiogenic genes FGF2, HIF1, VEGF and TGFB1, as well as genes (OCLN and TJP1) involved in BBB maintenance. CONCLUSIONS EVs from physioxic MSC cultures hold promise in the generation of a cell-free therapy to induce angiogenesis. Their positive angiogenic effect on cerebral microvascular endothelial cells demonstrates that they may have utility in treating ischemic cerebral conditions, where the induction of angiogenesis is critical to improving recovery and neurological function.
Collapse
Affiliation(s)
- Jolene Phelps
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - David A Hart
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB, T2N 4N1, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - Alim P Mitha
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, 3300 Hospital Drive N.W., Calgary, AB, T2N 4N1, Canada
| | - Neil A Duncan
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, AB, T2N 4N1, Canada
- Department of Civil Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- Musculoskeletal Mechanobiology and Multiscale Mechanics Bioengineering Lab, Department of Civil Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada.
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada.
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, AB, T2N 1N4, Canada.
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada.
| |
Collapse
|
7
|
Hasan MY, Siran R, Mahadi MK. The Effects of Vagus Nerve Stimulation on Animal Models of Stroke-Induced Injury: A Systematic Review. BIOLOGY 2023; 12:biology12040555. [PMID: 37106754 PMCID: PMC10136363 DOI: 10.3390/biology12040555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Ischemic stroke is one of the leading causes of death worldwide, and poses a great burden to society and the healthcare system. There have been many recent advances in the treatment of ischemic stroke, which usually results from the interruption of blood flow to a particular part of the brain. Current treatments for ischemic stroke mainly focus on revascularization or reperfusion of cerebral blood flow to the infarcted tissue. Nevertheless, reperfusion injury may exacerbate ischemic injury in patients with stroke. In recent decades, vagus nerve stimulation (VNS) has emerged as an optimistic therapeutic intervention. Accumulating evidence has demonstrated that VNS is a promising treatment for ischemic stroke in various rat models through improved neural function, cognition, and neuronal deficit scores. We thoroughly examined previous evidence from stroke-induced animal studies using VNS as an intervention until June 2022. We concluded that VNS yields stroke treatment potential by improving neurological deficit score, infarct volume, forelimb strength, inflammation, apoptosis, and angiogenesis. This review also discusses potential molecular mechanisms underlying VNS-mediated neuroprotection. This review could help researchers conduct additional translational research on patients with stroke.
Collapse
Affiliation(s)
- Mohammad Yusuf Hasan
- Centre for Drug Herbal and Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Rosfaiizah Siran
- Neuroscience Research Group (NRG), Faculty of Medicine, Jalan Hospital, Universiti Teknologi MARA, Sungai Buloh Campus, Sungai Buloh 47000, Malaysia
| | - Mohd Kaisan Mahadi
- Centre for Drug Herbal and Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
8
|
The immunolocalization of cluster of differentiation 31, phalloidin and alpha smooth muscle actin on vascular network of normal and ischemic rat brain. Sci Rep 2022; 12:22288. [PMID: 36566295 PMCID: PMC9789995 DOI: 10.1038/s41598-022-26831-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Cluster of differentiation 31 (CD31), phalloidin and alpha smooth muscle actin (α-SMA) have been widely applied to label the cerebral blood vessels in the past years. Although CD31 is mainly used as endothelial marker in determining the cerebral capillaries, it seems likely that its labeling efficiency is closely correlated with the antibodies from the polyclonal or monoclonal one, as well as the conditions of blood vessels. In order to test this phenomenon, we compared the labeling characteristics of goat polyclonal anti-CD31 (gP-CD31) and mouse monoclonal anti-CD31 (mM-CD31) with those of phalloidin and α-SMA on the rat brain in health and ischemia/reperfusion (I/R) with the middle cerebral artery occlusion. By multiple immunofluorescence staining, it was found that gP-CD31 labeling expressed extensively on the cerebral capillaries forming the vascular networks on the normal and ischemic regions, but mM-CD31 labeling mainly presented on the capillaries in the ischemic region. In contrast to the vascular labeling with gP-CD31, phalloidin and α-SMA were mainly expressed on the wall of cortical penetrating arteries, and less on that of capillaries. By three-dimensional reconstruction analysis, it was clearly shown that gP-CD31 labeling was mainly located on the lumen side of vascular wall and was surrounded by phalloidin labeling and α-SMA labeling. These results indicate that gP-CD31 is more sensitive than mM-CD31 for labeling the cerebral vasculature, and is highly compatible with phalloidin and α-SMA for evaluating the cerebral vascular networks under the physiological and pathological conditions.
Collapse
|
9
|
Lee MJ, Zhu J, An JH, Lee SE, Kim TY, Oh E, Kang YE, Chung W, Heo JY. A transcriptomic analysis of cerebral microvessels reveals the involvement of Notch1 signaling in endothelial mitochondrial-dysfunction-dependent BBB disruption. Fluids Barriers CNS 2022; 19:64. [PMID: 36028880 PMCID: PMC9414148 DOI: 10.1186/s12987-022-00363-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background Endothelial cells (ECs) in cerebral vessels are considered the primary targets in acute hemorrhagic brain injuries. EC dysfunction can aggravate neuronal injuries by causing secondary inflammatory responses and blood–brain barrier (BBB) disruption. Previous studies have reported that enhancement of mitochondrial function within ECs may reduce BBB disruption and decrease the severity of acute brain injuries. However, the molecular signaling pathways through which enhanced EC mitochondrial function is enhanced to exert this BBB protective effect have not been fully elucidated. Methods To identify signaling pathways involved in linking EC-specific mitochondrial dysfunction and BBB disruption, we first performed RNA sequencing using isolated cerebral vessels from TEKCRIF1 KO mice, a mouse strain that displays EC-specific mitochondrial dysfunction. After identification, we assessed the significance of candidate signaling pathways using an intracerebral hemorrhage (ICH) mouse model. BBB integrity was assessed using an IgG leakage assay, and symptomatic changes were evaluated using behavioral assays. Results Transcriptome analyses of the TEKCRIF1 KO mouse revealed significant changes in Notch1 signaling, a pathway intimately involved in BBB maintenance. We also observed a decrease in Notch1 signaling and expression of the mitochondrial oxidative phosphorylation (OxPhos) complex in the ICH mouse model, which also exhibits BBB disruption. To further assess the function of Notch1 signaling in relation to BBB disruption, we injected ICH model mice with adropin, a protein that interacts with the Notch1 ligand NB-3 and activates Notch1 signaling. We found that adropin prevented BBB disruption and reduced the extent (area) of the injury compared with that in vehicle controls, in association with alteration of mitochondrial function. Conclusion These results suggest that the Notch1 signaling pathway acts as an upstream regulator of DEGs and can be a target to regulate the changes involved with endothelial mitochondrial dysfunction-dependent BBB disruption. Thus, treatment methods that activate Notch1 may be beneficial in acute brain injuries by protecting BBB integrity. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00363-7.
Collapse
Affiliation(s)
- Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jiebo Zhu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jong Hun An
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Disease, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University School of Medicine, Deajeon, 35015, Republic of Korea
| | - Tae Yeon Kim
- Bio-Synergy Research Center, Daejeon, 34141, Republic of Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Yea Eun Kang
- Research Center for Endocrine and Metabolic Disease, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University School of Medicine, Deajeon, 35015, Republic of Korea.
| | - Woosuk Chung
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea.
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea. .,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
10
|
Chen J, He J, Luo L. Brain vascular damage-induced lymphatic ingrowth is directed by Cxcl12b/Cxcr4a. Development 2022; 149:275687. [PMID: 35694896 DOI: 10.1242/dev.200729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022]
Abstract
After ischemic stroke, promotion of vascular regeneration without causing uncontrolled vessel growth appears to be the major challenge for pro-angiogenic therapies. The molecular mechanisms underlying how nascent blood vessels (BVs) are correctly guided into the post-ischemic infarction area remain unknown. Here, using a zebrafish cerebrovascular injury model, we show that chemokine signaling provides crucial guidance cues to determine the growing direction of ingrown lymphatic vessels (iLVs) and, in turn, that of nascent BVs. The chemokine receptor Cxcr4a is transcriptionally activated in the iLVs after injury, whereas its ligand Cxcl12b is expressed in the residual central BVs, the destinations of iLV ingrowth. Mutant and mosaic studies indicate that Cxcl12b/Cxcr4a-mediated chemotaxis is necessary and sufficient to determine the growing direction of iLVs and nascent BVs. This study provides a molecular basis for how the vessel directionality of cerebrovascular regeneration is properly determined, suggesting potential application of Cxcl12b/Cxcr4a in the development of post-ischemic pro-angiogenic therapies.
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
| | - 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
|
11
|
Yanev P, van Tilborg GA, van der Toorn A, Kong X, Stowe AM, Dijkhuizen RM. Prolonged release of VEGF and Ang1 from intralesionally implanted hydrogel promotes perilesional vascularization and functional recovery after experimental ischemic stroke. J Cereb Blood Flow Metab 2022; 42:1033-1048. [PMID: 34986707 PMCID: PMC9125493 DOI: 10.1177/0271678x211069927] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Injectable hydrogels can generate and support pro-repair environments in injured tissue. Here we used a slow-releasing drug carrying in situ-forming hydrogel to promote post-stroke recovery in a rat model. Release kinetics were measured in vitro and in vivo with MRI, using gadolinium-labeled albumin (Galbumin), which demonstrated prolonged release over multiple weeks. Subsequently, this hydrogel was used for long-term delivery of vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang1) (Gel VEGF + Ang1, n = 14), in a photothrombotically induced cortical stroke lesion in rats. Control stroke animals were intralesionally injected with saline (Saline, n = 10), non-loaded gel (Gel, n = 10), or a single bolus of VEGF + Ang1 in saline (Saline VEGF + Ang1, n = 10). MRI was executed to guide hydrogel injection. Functional recovery was assessed with sensorimotor function tests, while tissue status and vascularization were monitored by serial in vivo MRI. Significant recovery from sensorimotor deficits from day 28 onwards was only measured in the Gel VEGF + Ang1 group. This was accompanied by significantly increased vascularization in the perilesional cortex. Histology confirmed (re)vascularization and neuronal sparing in perilesional areas. In conclusion, intralesional injection of in situ-forming hydrogel loaded with pro-angiogenic factors can support prolonged brain tissue regeneration and promote functional recovery in the chronic phase post-stroke.
Collapse
Affiliation(s)
- Pavel Yanev
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Geralda Af van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Xiangmei Kong
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| | - Ann M Stowe
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| |
Collapse
|
12
|
Myagmar BO, Chen R, Zhang X, Xu R, Jiang W, Cao W, Ji H, Zhang X. Cerebroprotein hydrolysate injection is involved in promoting long-term angiogenesis, vessel diameter and density after cerebral ischemia in mice. Life Sci 2022; 300:120568. [PMID: 35489566 DOI: 10.1016/j.lfs.2022.120568] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/09/2022]
Abstract
AIMS In this study, we aimed investigate the impacts of CH-I on angiogenesis, effects for vascular structure changes and long-term neurological recovery after ischemic stroke as well as the potential mechanisms. MAIN METHODS Young male mice subjected to intraluminal middle cerebral artery occlusion were administrated with CH-I once daily from day 1 to day 14 after stroke. The infarct volume was evaluated by TTC staining at day 7 after stroke. Neurological deficits were measured 1 to 28 days after stroke. Microvascular density, astrocyte coverage, and angiogenesis were assessed by IF, qRT-PCR, and WB at regular intervals after stroke. LSCI and TPMI measured changes in blood flow and vascular density and width from the day after stroke to day 28. KEY FINDINGS Compared with the dMCAO group, CH-I treatment significantly improved neurological recovery and reduced the infarct at day 7 after stroke. CH-I treatment increased the expression of the CD31, BrdU+/CD31+ microvessels and GFAP positive vessels in the peri-infarct cortex at day 7 to 28 after stroke. The expression of protein and gene were enhanced in CH-I group. CH-I significantly improved cerebral blood flow at day 7 after stroke. CH-I increased the vascular density and vascular width at day 14 after stroke. SIGNIFICANCE CH-I has been shown to restore nerve function, reduce the rate of cerebral infarction, increase microvascular density, and promote angiogenesis. CH-I improved cerebral blood flow, protected blood vessels from postoperative stenosis, and improved vascular plasticity.
Collapse
Affiliation(s)
- Bat-Otgon Myagmar
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiao Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wei Jiang
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wen Cao
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Hui Ji
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
13
|
Wang L, Astone M, Alam SK, Zhu Z, Pei W, Frank DA, Burgess SM, Hoeppner LH. Suppressing STAT3 activity protects the endothelial barrier from VEGF-mediated vascular permeability. Dis Model Mech 2021; 14:272222. [PMID: 34542605 PMCID: PMC8592016 DOI: 10.1242/dmm.049029] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/10/2021] [Indexed: 12/27/2022] Open
Abstract
Vascular permeability triggered by inflammation or ischemia promotes edema, exacerbates disease progression and impairs tissue recovery. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability. VEGF plays an integral role in regulating vascular barrier function physiologically and in pathologies, including cancer, stroke, cardiovascular disease, retinal conditions and COVID-19-associated pulmonary edema, sepsis and acute lung injury. Understanding temporal molecular regulation of VEGF-induced vascular permeability will facilitate developing therapeutics to inhibit vascular permeability, while preserving tissue-restorative angiogenesis. Here, we demonstrate that VEGF signals through signal transducer and activator of transcription 3 (STAT3) to promote vascular permeability. We show that genetic STAT3 ablation reduces vascular permeability in STAT3-deficient endothelium of mice and VEGF-inducible zebrafish crossed with CRISPR/Cas9-generated Stat3 knockout zebrafish. Intercellular adhesion molecule 1 (ICAM-1) expression is transcriptionally regulated by STAT3, and VEGF-dependent STAT3 activation is regulated by JAK2. Pyrimethamine, an FDA-approved antimicrobial agent that inhibits STAT3-dependent transcription, substantially reduces VEGF-induced vascular permeability in zebrafish, mouse and human endothelium. Collectively, our findings suggest that VEGF/VEGFR-2/JAK2/STAT3 signaling regulates vascular barrier integrity, and inhibition of STAT3-dependent activity reduces VEGF-induced vascular permeability. This article has an associated First Person interview with the first author of the paper. Summary: Genetic STAT3 ablation in mice and VEGF-inducible zebrafish reveals that VEGF signals through STAT3 to promote vascular permeability. Pyrimethamine reduces VEGF-induced permeability in animal models.
Collapse
Affiliation(s)
- Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
14
|
Xu J, Zhang P, Chen Y, Xu Y, Luan P, Zhu Y, Zhang J. Sodium tanshinone IIA sulfonate ameliorates cerebral ischemic injury through regulation of angiogenesis. Exp Ther Med 2021; 22:1122. [PMID: 34504576 PMCID: PMC8383733 DOI: 10.3892/etm.2021.10556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular remodeling and neuroprotection are two major adaptable methods for treating ischemic stroke. Edaravone is a protective agent for the treatment of stroke and was used as a positive control in the present study. Sodium tanshinone IIA sulfonate (STS) has demonstrated therapeutic clinical effects in cerebral infarction in China, while its mechanisms of action in ischemic stroke have remained elusive. The angiogenesis and neuroprotective effects of STS were evaluated in a rat model induced by middle cerebral artery occlusion and 3 days of reperfusion. When used at the same dose, the magnitude of the therapeutic effect of STS was similar to that of edaravone in terms of decreased blood-brain barrier damage as indicated by reduced Evans blue leakage, improved neurological deficits, alleviated cerebral edema and inhibition of histopathological changes caused by ischemia/reperfusion. The TUNEL assay demonstrated that the ability of STS to inhibit neuronal apoptosis was equivalent to that of edaravone. Immunofluorescence detection of CD31 and α-smooth muscle actin indicated that the vascular density was significantly reduced in the vehicle group compared with that in the sham operation group, STS increased the microvessel density in the ischemic area. Furthermore, in the vehicle group the protein expression of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR) as determined by fluorescence microscopy and immunohistochemistry was significantly reduced compared with that in the sham group. However, STS promoted their expression compared to the vehicle group respectively, and increaed the mRNA expression of VEGF, VEGFR, CD31 and angiopoietin-1 as determined by reverse transcription-quantitative PCR, but these changes were not significant or not present for edaravone apart from Ang-1. In conclusion, STS protected against ischemic brain injury by promoting angiogenesis in ischemic areas and inhibiting neuronal apoptosis. These results provide a potential treatment for stroke recovery.
Collapse
Affiliation(s)
- Jiazhen Xu
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Pei Zhang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yao Chen
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yulan Xu
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Pengwei Luan
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuying Zhu
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jiange Zhang
- Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
15
|
Pericyte hypoxia-inducible factor-1 (HIF-1) drives blood-brain barrier disruption and impacts acute ischemic stroke outcome. Angiogenesis 2021; 24:823-842. [PMID: 34046769 PMCID: PMC8487886 DOI: 10.1007/s10456-021-09796-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/04/2021] [Indexed: 12/19/2022]
Abstract
Pericytes play essential roles in blood-brain barrier integrity and their dysfunction is implicated in neurological disorders such as stroke although the underlying mechanisms remain unknown. Hypoxia-inducible factor-1 (HIF-1), a master regulator of injury responses, has divergent roles in different cells especially during stress scenarios. On one hand HIF-1 is neuroprotective but on the other it induces vascular permeability. Since pericytes are critical for barrier stability, we asked if pericyte HIF-1 signaling impacts barrier integrity and injury severity in a mouse model of ischemic stroke. We show that pericyte HIF-1 loss of function (LoF) diminishes ischemic damage and barrier permeability at 3 days reperfusion. HIF-1 deficiency preserved barrier integrity by reducing pericyte death thereby maintaining vessel coverage and junctional protein organization, and suppressing vascular remodeling. Importantly, considerable improvements in sensorimotor function were observed in HIF-1 LoF mice indicating that better vascular functionality post stroke improves outcome. Thus, boosting vascular integrity by inhibiting pericytic HIF-1 activation and/or increasing pericyte survival may be a lucrative option to accelerate recovery after severe brain injury.
Collapse
|
16
|
Babaei M, Rezaie J. Application of stem cell-derived exosomes in ischemic diseases: opportunity and limitations. J Transl Med 2021; 19:196. [PMID: 33964940 PMCID: PMC8106139 DOI: 10.1186/s12967-021-02863-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic diseases characterized by an insufficient blood flow that leads to a decrease in oxygen and nutrient uptake by cells have emerged as an important contributor to both disability and death worldwide. Up-regulation of angiogenesis may be a key factor for the improvement of ischemic diseases. This article searched articles in PubMed with the following keywords: stem cells, exosomes, angiogenesis, ischemic diseases either alone or in grouping form. The most relevant selected items were stem cell-derived exosomes and ischemic diseases. A growing body of evidence indicates that stem cells produce exosomes, which is the novel emerging approach to cell-to-cell communication and offers a new standpoint on known therapeutic strategies of ischemic diseases. Exosomes transport biological molecules such as many types of proteins, RNAs, DNA fragments, signaling molecules, and lipids between cells. Different stem cells release exosomes representing beneficial effects on ischemic diseases as they promote angiogenesis both in vitro and in vivo experiments. Application of exosomes for therapeutic angiogenesis opened new opportunities in the regenerative medicine, however, some limitations regarding exosomes isolation and application remain concerned. In addition, most of the experiments were conducted in preclinical and therefore translation of these results from bench to bed requires more effort in this field. Exosomes from stem cells are a promising tool for the treatment of ischemic diseases. In addition, translation of pre-clinic results into clinic needs further studies in this field.
Collapse
Affiliation(s)
- Majid Babaei
- Social Determinants of Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
17
|
Shindo A, Takase H, Hamanaka G, Chung KK, Mandeville ET, Egawa N, Maki T, Borlongan M, Takahashi R, Lok J, Tomimoto H, Lo EH, Arai K. Biphasic roles of pentraxin 3 in cerebrovascular function after white matter stroke. CNS Neurosci Ther 2020; 27:60-70. [PMID: 33314664 PMCID: PMC7804900 DOI: 10.1111/cns.13510] [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: 08/30/2020] [Revised: 10/24/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022] Open
Abstract
Recent clinical studies suggest that pentraxin 3 (PTX3), which is known as an acute-phase protein that is produced rapidly at local sites of inflammation, may be a new biomarker of disease risk for central nervous system disorders, including stroke. However, the effects of PTX3 on cerebrovascular function in the neurovascular unit (NVU) after stroke are mostly unknown, and the basic research regarding the roles of PTX3 in NVU function is still limited. In this reverse translational study, we prepared mouse models of white matter stroke by vasoconstrictor (ET-1 or L-Nio) injection into the corpus callosum region to examine the roles of PTX3 in the pathology of cerebral white matter stroke. PTX3 expression was upregulated in GFAP-positive astrocytes around the affected region in white matter for at least 21 days after vasoconstrictor injection. When PTX3 expression was reduced by PTX3 siRNA, blood-brain barrier (BBB) damage at day 3 after white matter stroke was exacerbated. In contrast, when PTX3 siRNA was administered at day 7 after white matter stroke, compensatory angiogenesis at day 21 was promoted. In vitro cell culture experiments confirmed the inhibitory effect of PTX3 in angiogenesis, that is, recombinant PTX3 suppressed the tube formation of cultured endothelial cells in a Matrigel-based in vitro angiogenesis assay. Taken together, our findings may support a novel concept that astrocyte-derived PTX3 plays biphasic roles in cerebrovascular function after white matter stroke; additionally, it may also provide a proof-of-concept that PTX3 could be a therapeutic target for white matter-related diseases, including stroke.
Collapse
Affiliation(s)
- Akihiro Shindo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kelly K Chung
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Naohiro Egawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mia Borlongan
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
18
|
Activation of endothelial Wnt/β-catenin signaling by protective astrocytes repairs BBB damage in ischemic stroke. Prog Neurobiol 2020; 199:101963. [PMID: 33249091 DOI: 10.1016/j.pneurobio.2020.101963] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/02/2020] [Accepted: 11/19/2020] [Indexed: 01/04/2023]
Abstract
The role of astrocytes in dysregulation of blood-brain barrier (BBB) function following ischemic stroke is not well understood. Here, we investigate the effects of restoring the repair properties of astrocytes on the BBB after ischemic stroke. Mice deficient for NHE1, a pH-sensitive Na+/H+ exchanger 1, in astrocytes have reduced BBB permeability after ischemic stroke, increased angiogenesis and cerebral blood flow perfusion, in contrast to wild-type mice. Bulk RNA-sequencing transcriptome analysis of purified astrocytes revealed that ∼177 genes were differentially upregulated in mutant astrocytes, with Wnt7a mRNA among the top genes. Using a Wnt reporter line, we confirmed that the pathway was upregulated in cerebral vessels of mutant mice after ischemic stroke. However, administration of the Wnt/β-catenin inhibitor, XAV-939, blocked the reparative effects of Nhe1-deficient astrocytes. Thus, astrocytes lacking pH-sensitive NHE1 protein are transformed from injurious to "protective" by inducing Wnt production to promote BBB repair after ischemic stroke.
Collapse
|
19
|
Wang L, Astone M, Alam SK, Zhu Z, Pei W, Frank DA, Burgess SM, Hoeppner LH. Suppressing STAT3 activity protects the endothelial barrier from VEGF-mediated vascular permeability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33140053 PMCID: PMC7605565 DOI: 10.1101/2020.10.27.358374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular permeability triggered by inflammation or ischemia promotes edema, exacerbates disease progression, and impairs tissue recovery. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability. VEGF plays an integral role in regulating vascular barrier function physiologically and in pathologies, such as cancer, ischemic stroke, cardiovascular disease, retinal conditions, and COVID-19-associated pulmonary edema and sepsis, which often leads to acute lung injury, including acute respiratory distress syndrome. However, after initially stimulating permeability, VEGF subsequently mediates angiogenesis to repair damaged tissue. Consequently, understanding temporal molecular regulation of VEG-Finduced vascular permeability will facilitate developing therapeutics that achieve the delicate balance of inhibiting vascular permeability while preserving tissue repair. Here, we demonstrate that VEGF signals through signal transducer and activator of transcription 3 (STAT3) to promote vascular permeability. Specifically, we show that genetic STAT3 ablation reduces vascular permeability in STAT3-deficient endothelium of mice and VEGF-inducible zebrafish crossed with CRISPR/Cas9 generated genomic STAT3 knockout zebrafish. Importantly, STAT3 deficiency does not impair vascular development and function in vivo. We identify intercellular adhesion molecule 1 (ICAM-1) as a STAT3-dependent transcriptional regulator and show VEGF-dependent STAT3 activation is regulated by JAK2. Pyrimethamine, an FDA-approved antimicrobial agent that inhibits STAT3-dependent transcription, substantially reduces VEGF-induced vascular permeability in zebrafish, mouse, and human endothelium. Indeed, pharmacologically targeting STAT3 increases vascular barrier integrity using two additional compounds, atovaquone and C188-9. Collectively, our findings suggest that the VEGF, VEGFR-2, JAK2, and STAT3 signaling cascade regulates vascular barrier integrity, and inhibition of STAT3-dependent activity reduces VEGF-induced vascular permeability in vertebrate models.
Collapse
Affiliation(s)
- Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
Kim SW, Lee HK, Seol SI, Davaanyam D, Lee H, Lee JK. Ninjurin 1 dodecamer peptide containing the N-terminal adhesion motif (N-NAM) exerts proangiogenic effects in HUVECs and in the postischemic brain. Sci Rep 2020; 10:16656. [PMID: 33028854 PMCID: PMC7542178 DOI: 10.1038/s41598-020-73340-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nerve injury-induced protein 1 (Ninjurin 1, Ninj1) is a cell adhesion molecule responsible for cell-to-cell interactions between immune cells and endothelial cells. In our previous paper, we have shown that Ninj1 plays an important role in the infiltration of neutrophils in the postischemic brain and that the dodecamer peptide harboring the Ninj1 N-terminal adhesion motif (N-NAM, Pro26-Asn37) inhibits infiltration of neutrophils in the postischemic brain and confers robust neuroprotective and anti-inflammatory effects. In the present study, we examinedt the pro-angiogenic effect of N-NAM using human umbilical vein endothelial cells (HUVECs) and rat MCAO (middle cerebral artery occlusion) model of stroke. We found that N-NAM promotes proliferation, migration, and tube formation of HUVECs and demonstrate that the suppression of endogenous Ninj1 is responsible for the N-NAM-mediated pro-angiogenic effects. Importantly, a pull-down assay revealed a direct binding between exogenously delivered N-NAM and endogenous Ninj1 and it is N-terminal adhesion motif dependent. In addition, N-NAM activated the Ang1-Tie2 and AKT signaling pathways in HUVECs, and blocking those signaling pathways with specific inhibitors suppressed N-NAM-induced tube formation, indicating critical roles of those signaling pathways in N-NAM-induced angiogenesis. Moreover, in a rat MCAO model, intranasal administration of N-NAM beginning 4 days post-MCAO (1.5 µg daily for 3 days) augmented angiogenesis in the penumbra of the ipsilateral hemisphere of the brain and significantly enhanced total vessel lengths, vessel densities, and pro-angiogenic marker expression. These results demonstrate that the 12-amino acid Ninj1 peptide, which contains the N-terminal adhesion motif of Ninj1, confers pro-angiogenic effects and suggest that those effects might contribute to its neuroprotective effects in the postischemic brain.
Collapse
Affiliation(s)
- Seung-Woo Kim
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea.,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea.,Department of Biomedical Sciences, Inha University School of Medicine, Inchon, Republic of Korea
| | - Hye-Kyung Lee
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea.,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea
| | - Song-I Seol
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea.,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea
| | - Dashdulam Davaanyam
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea.,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea
| | - Hahnbie Lee
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea.,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Medical Research Center, Inha University School of Medicine, Inharo 100, Inchon, 22202, Republic of Korea. .,Medical Research Center, Inha University School of Medicine, Inchon, Republic of Korea.
| |
Collapse
|
21
|
Edwards DN, Salmeron K, Lukins DE, Trout AL, Fraser JF, Bix GJ. Integrin α5β1 inhibition by ATN-161 reduces neuroinflammation and is neuroprotective in ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1695-1708. [PMID: 31575337 PMCID: PMC7370357 DOI: 10.1177/0271678x19880161] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Stroke remains a leading cause of death and disability with limited therapeutic options. Endothelial cell β1 integrin receptors play a direct role in blood-brain barrier (BBB) dysfunction through regulation of tight junction proteins and infiltrating leukocytes, potentially mediated by β1 integrins. Following tandem transient common carotid artery/middle cerebral artery occlusion on wild-type mice, we administered the integrin a5b1 inhibitor, ATN-161, intraperitoneal (IP) injection at 1 mg/kg acutely after reperfusion, on post-stroke day (PSD)1 and PSD2. Systemic changes (heart rate, pulse distension, and body temperature) were determined. Additionally, infarct volume and edema were determined by 2,3-triphenyltetrazolium chloride and magnetic resonance imaging, while neurological changes were evaluated using an 11-point Neuroscore. Brain immunohistochemistry was performed for claudin-5, α5β1, IgG, and CD45 + cells, and quantitative polymerase chain reaction (qPCR) was performed for matrix metalloproteinase-9 (MMP-9), interleukin (IL)-1β, collagen IV, and CXCL12. ATN-161 significantly reduced integrin α5β1 expression in the surrounding peri-infarct region with no systemic changes. Infarct volume, edema, and functional deficit were significantly reduced in ATN-161-treated mice. Furthermore, ATN-161 treatment reduced IgG extravasation into the parenchyma through conserved claudin-5, collagen IV, CXCL12 while reducing MMP-9 transcription. Additionally, IL-1β and CD45 + cells were reduced in the ipsilateral cortex following ATN-161 administration. Collectively, ATN-161 may be a promising novel stroke therapy by reducing post-stroke inflammation and BBB permeability.
Collapse
Affiliation(s)
| | - Kathleen Salmeron
- Department of Neuroscience, University of Kentucky, Lexington, USA.,Department of Physiology, University of Kentucky, Lexington, USA
| | | | - Amanda L Trout
- Department of Neurology, University of Kentucky, Lexington, USA
| | - Justin F Fraser
- Department of Neuroscience, University of Kentucky, Lexington, USA.,Department of Radiology, University of Kentucky, Lexington, USA.,Department of Neurology, University of Kentucky, Lexington, USA.,Department of Neurosurgery, University of Kentucky, Lexington, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, USA
| | - Gregory J Bix
- Department of Neuroscience, University of Kentucky, Lexington, USA.,Department of Neurology, University of Kentucky, Lexington, USA.,Department of Neurosurgery, University of Kentucky, Lexington, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
| |
Collapse
|
22
|
Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci 2020; 30:839-855. [PMID: 31203262 DOI: 10.1515/revneuro-2019-0002] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of injury-related mortality and morbidity in the USA and around the world. The survivors may suffer from cognitive and memory deficits, vision and hearing loss, movement disorders, and different psychological problems. The primary insult causes neuronal damage and activates astrocytes and microglia which evokes immune responses causing further damage to the brain. Clinical trials of drugs to recover the neuronal loss are not very successful. Regenerative approaches for TBI using mesenchymal stem cells (MSCs) seem promising. Results of preclinical research have shown that transplantation of MSCs reduced secondary neurodegeneration and neuroinflammation, promoted neurogenesis and angiogenesis, and improved functional outcome in the experimental animals. The functional improvement is not necessarily related to cell engraftment; rather, immunomodulation by molecular factors secreted by MSCs is responsible for the beneficial effects of this therapy. However, MSC therapy has a few drawbacks including tumor formation, which can be avoided by the use of MSC-derived exosomes. This review has focused on the research works published in the field of regenerative therapy using MSCs after TBI and its future direction.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
23
|
Empagliflozin alleviates neuronal apoptosis induced by cerebral ischemia/reperfusion injury through HIF-1α/VEGF signaling pathway. Arch Pharm Res 2020; 43:514-525. [PMID: 32436127 DOI: 10.1007/s12272-020-01237-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 05/14/2020] [Indexed: 12/19/2022]
Abstract
Ischemic stroke is a serious condition associated with severe functional disability and high mortality, however; effective therapy remains elusive. Empagliflozin, a sodium-glucose cotransporter 2 inhibitor, has been shown to exert additional non-glycemic benefits including anti-apoptotic effects in different disease settings. Thereby, this study was designed to investigate the ameliorative effect of empagliflozin on the neuronal apoptosis exhibited in cerebral ischemia/reperfusion (I/R) in a rat model targeting HIF-1α/VEGF signaling which is involved in this insult. Global cerebral I/R injury was induced in male Wistar rats through occlusion of the bilateral common carotid arteries for 30 min followed by one-hour reperfusion. Empagliflozin doses of 1 and 10 mg/kg were administered 1 and 24 h after reperfusion. In I/R-injured rats, empagliflozin treatments significantly reduced infarct size and enhanced neurobehavioral functions in a dose-dependent manner. The drug alleviated neuronal death and cerebral injury inflicted by global ischemia as it suppressed neuronal caspase-3 protein expression. In parallel, protein expressions of HIF-1α and its downstream mediator VEGF were upregulated in the ischemic brain following empagliflozin treatment. The results indicated that empagliflozin attenuates cerebral I/R-induced neuronal death via the HIF-1α/VEGF cascade.
Collapse
|
24
|
Liu Q, Tan Y, Qu T, Zhang J, Duan X, Xu H, Mu Y, Ma H, Wang F. Therapeutic mechanism of human neural stem cell-derived extracellular vesicles against hypoxia-reperfusion injury in vitro. Life Sci 2020; 254:117772. [PMID: 32437794 DOI: 10.1016/j.lfs.2020.117772] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
AIMS This study aimed to explore that the human neural stem cell derived extracellular vesicles (hNSC-EVs) have therapeutic effect on neuronal hypoxia-reperfusion (H/R) injured neurons in vitro by mediating the nuclear translocation of NF-E2-related factor 2 (Nrf2) to regulate the expression of downstream oxidative kinases. MAIN METHODS The neuroprotective effects of hNSC-EVs were evaluated in an in vitro neuronal H/R model. Three parameters of hNSC-EVs, structure, phenotype and particle size, were characterized. At the cellular level, a human neuron cerebral ischemic reperfusion (CIR) injury model was constructed. Cell viability, apoptosis, and the amount of reactive oxygen species (ROS) were detected using real-time cell analysis (RTCA), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and dichloro-dihydro-fluorescein diacetate (DCFH-DA), respectively. The neuronal axonal elongation was assessed by Opera Phenix™ screening system. The angiogenesis of human umbilical vein endothelial cells (HUVECs) was evaluated by co-culturing HUVECs with hNSC-EVs in Matrigel. The expression of apoptosis and oxidative stress-related proteins in cells and the nuclear transfer of Nrf2 following hypoxia-reperfusion (H/R) was verified by Western-blotting. KEY FINDINGS We found that the hNSC-EVs can promote the survival of post-H/R injury neurons, inhibit neuronal apoptosis, and enhance nuclear transfer of Nrf2, in response to oxidative stress. We also found the hNSC-EVs can promote the elongation of neuronal axons and the angiogenesis of HUVECs. SIGNIFICANCE At present, there is no effective therapy for CIR injury. We suggest that the hNSC-EVs could be considered a new strategy to achieve nerve repair for the treatment of neurological diseases, especially stroke.
Collapse
Affiliation(s)
- Qingyue Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhuaxi Road, Jinan 250012, China
| | - Yi Tan
- Qilu Cell Therapy Technology Co., Ltd, Jinan 250000, China; Shandong Yinfeng Life Science Research Institute, Jinan, 250000, Shandong, China
| | - Tingyu Qu
- Shandong Yinfeng Life Science Research Institute, Jinan, 250000, Shandong, China; R & D of Cell and Tissue Bank, Qilu Stem Cell Engineering Company of Shandong Province, Jinan 250000, Shandong, China
| | - Jianhui Zhang
- Qilu Cell Therapy Technology Co., Ltd, Jinan 250000, China
| | - Xuexia Duan
- Maternal and Child Health Care Hospital of Shandong Province, Jinan, 250014, Shandong, China
| | - Hongpeng Xu
- Qilu Cell Therapy Technology Co., Ltd, Jinan 250000, China
| | - Yue Mu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhuaxi Road, Jinan 250012, China
| | - Heran Ma
- Qilu Cell Therapy Technology Co., Ltd, Jinan 250000, China; Shandong Yinfeng Life Science Research Institute, Jinan, 250000, Shandong, China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhuaxi Road, Jinan 250012, China.
| |
Collapse
|
25
|
Abdel-Rahman RF, El Awdan SA, Hegazy RR, Mansour DF, Ogaly HA, Abdelbaset M. Neuroprotective effect of Crocus sativus against cerebral ischemia in rats. Metab Brain Dis 2020; 35:427-439. [PMID: 31728890 DOI: 10.1007/s11011-019-00505-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022]
Abstract
The present study aimed to investigate the role of vascular endothelial growth factor (VEGF) in the neuroprotective effect of Crocus sativus (saffron) against cerebral ischemia/reperfusion injury (I/R) in rats. Four groups of a total forty I/R rats with 60-min occlusion followed by 48 h reperfusion or sham surgery were used. The sham and left-brain I/R control groups where treated with normal saline. The rats of the other two groups received saffron extract (100 or 200 mg/kg, ip, respectively) for 3 successive weeks prior to left-brain I/R. Other four doses of saffron extract were received by the rats of the last 2 groups 60 min prior to operation, during the surgery, and on days 1 and 2 following reperfusion. I/R group showed marked neurobehavioral, neurochemical and histopathological alterations. The results revealed a significant reduction in neurological deficit scores in the saffron-treated rats at both doses. Saffron significantly attenuated lipid peroxidation, decreased NO and brain natriuretic peptide (BNP) contents in I/R-brain tissue. On the other hand, saffron reversed the depletion of GSH in the injured brain. Moreover, saffron treatment evidently reduced apoptosis as revealed by a decrease in caspase-3 and Bax protein expression with a marked decrease in the apoptotic neuronal cells compared to I/R group. In addition, saffron administration effectively upregulated the expression of VEGF in I/R-brain tissue. In conclusion, saffron treatment offers significant neuroprotection against I/R damage possibly through diminishing oxidative stress and apoptosis and enhancement of VEGF.
Collapse
Affiliation(s)
| | | | - Rehab R Hegazy
- Pharmacology Department, National Research Centre, Giza, Egypt
| | - Dina F Mansour
- Pharmacology Department, National Research Centre, Giza, Egypt
| | - H A Ogaly
- Chemistry Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Bu F, Min JW, Munshi Y, Lai YJ, Qi L, Urayama A, McCullough LD, Li J. Activation of endothelial ras-related C3 botulinum toxin substrate 1 (Rac1) improves post-stroke recovery and angiogenesis via activating Pak1 in mice. Exp Neurol 2019; 322:113059. [PMID: 31499064 DOI: 10.1016/j.expneurol.2019.113059] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE Long-term disability after stroke is common yet the mechanisms of post-stroke recovery are far from clear. It has been suggested that Ras-related C3 botulinum toxin substrate 1 (Rac1) contributes to functional recovery after ischemic stroke in mice. As Rac1 activation plays diverse roles in multiple cell types after central nervous system (CNS) injury, we herein examined the functional role of endothelial Rac1 in post-stroke recovery and angiogenesis. METHODS Transient middle cerebral artery occlusion (MCAO) in mice and oxygen-glucose deprivation (OGD) in human brain endothelial cell line-5i (HEBC 5i) were performed to mimic ischemic stroke. Lentivirus vectors encoding Rac1 with GFP and endothelial promotor ENG were injected into the animal's brain after stroke to overexpress Rac1. After injection, stroke recovery was tested by multiple behavioral tests including novel object recognition, adhesive removal and single pellet reaching tests. Endothelial regeneration in the peri-infarct zone was detected by immunohistochemistry (IHC). In the vitro model, the effect of Rac1 and Pak1 inhibitors to cell proliferation and migration was examined by CCK-8 and wound healing assays after OGD. The cellular protein level of brain-derived neurotrophic factor (BDNF), phosphorylated cAMP response element-binding protein (CREB), extracellular signal-regulated kinase (ERK) 1/2 and mitogen-activated protein kinase kinase (MEK) 1/2 were detected by western blots. RESULTS Delayed overexpression of endothelial Rac1 after MCAO improved cognitive and sensorimotor recovery from day 14 to 21 after stroke, increased vascular density and the protein level of pericytes in the peri-infarct zone without altering tissue loss in mice. Consistently, inhibition of Rac1 prevented endothelial proliferation and migration after OGD. Pak1 inhibition exerted a similar effect on endothelial cells. However, co-incubation of Rac1 and Pak1 inhibitors with cells did not lead to additive effects when compared with either inhibitor alone. Moreover, individual inhibition of Rac1 or Pak1 suppressed OGD-induced activation of pro-regenerative molecules, including CREB, MEK1/2 and ERK1/2, as well as the production of BDNF in vitro. The level of these proteins did not further decrease if both Rac1 and Pak1 were simultaneously inhibited. CONCLUSIONS We conclude that activation of endothelial Rac1 improves functional recovery and angiogenesis after stroke, and this process is mediated by Pak1 signaling. This study provides novel insight for Rac1 in the mechanism of long-term stroke recovery.
Collapse
Affiliation(s)
- Fan Bu
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Jia-Wei Min
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Yashasvee Munshi
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Yun-Ju Lai
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Li Qi
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Jun Li
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
29
|
Bian X, Ma K, Zhang C, Fu X. Therapeutic angiogenesis using stem cell-derived extracellular vesicles: an emerging approach for treatment of ischemic diseases. Stem Cell Res Ther 2019; 10:158. [PMID: 31159859 PMCID: PMC6545721 DOI: 10.1186/s13287-019-1276-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ischemic diseases, which are caused by a reduction of blood supply that results in reduced oxygen transfer and nutrient uptake, are becoming the leading cause of disabilities and deaths. Therapeutic angiogenesis is key for the treatment of these diseases. Stem cells have been used in animal models and clinical trials to treat various ischemic diseases. Recently, the efficacy of stem cell therapy has increasingly been attributed to exocrine functions, particularly extracellular vesicles. Extracellular vesicles are thought to act as intercellular communication vehicles to transport informational molecules including proteins, mRNA, microRNAs, DNA fragments, and lipids. Studies have demonstrated that extracellular vesicles promote angiogenesis in cellular experiments and animal models. Herein, recent reports on the use of extracellular vesicles for therapeutic angiogenesis during ischemic diseases are presented and discussed. We believe that extracellular vesicles-based therapeutics will be an ideal treatment method for patients with ischemic diseases.
Collapse
Affiliation(s)
- Xiaowei Bian
- Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.,Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 100048, Beijing, People's Republic of China
| | - Kui Ma
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 100048, Beijing, People's Republic of China
| | - Cuiping Zhang
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 100048, Beijing, People's Republic of China.
| | - Xiaobing Fu
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 100048, Beijing, People's Republic of China.
| |
Collapse
|
30
|
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]
|
31
|
Roth M, Gaceb A, Enström A, Padel T, Genové G, Özen I, Paul G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke. FASEB J 2019; 33:8990-8998. [PMID: 31039042 PMCID: PMC6662981 DOI: 10.1096/fj.201900153r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Poststroke recovery requires multiple repair mechanisms, including vascular remodeling and blood-brain barrier (BBB) restoration. Brain pericytes are essential for BBB repair and angiogenesis after stroke, but they also give rise to scar-forming platelet-derived growth factor receptor β (PDGFR-β)–expressing cells. However, many of the molecular mechanisms underlying this pericyte response after stroke still remain unknown. Regulator of G-protein signaling 5 (RGS5) has been associated with pericyte detachment from the vascular wall, but whether it regulates pericyte function and vascular stabilization in the chronic phase of stroke is not known. Using RGS5–knockout (KO) mice, we study how loss of RGS5 affects the pericyte response and vascular remodeling in a stroke model at 7 d after ischemia. Loss of RGS5 leads to a shift toward an increase in the number of perivascular pericytes and reduction in the density of parenchymal PDGFR-β–expressing cells associated with normalized PDGFR-β activation after stroke. The redistribution of pericytes resulted in higher pericyte coverage, increased vascular density, preservation of vessel lengths, and a significant reduction in vascular leakage in RGS5-KO mice compared with controls. Our study demonstrates RGS5 in pericytes as an important target to enhance vascular remodeling.—Roth, M., Gaceb, A., Enström, A., Padel, T., Genové, G., Özen, I., Paul, G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke.
Collapse
Affiliation(s)
- Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Thomas Padel
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Guillem Genové
- Department of Medicine, Integrated Cardio Metabolic Centre, Karolinska Institute, Huddinge, Sweden
| | - Ilknur Özen
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Lund University, Lund, Sweden.,Department of Neurology, Scania University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Abdul Y, Abdelsaid M, Li W, Webb RC, Sullivan JC, Dong G, Ergul A. Inhibition of Toll-Like Receptor-4 (TLR-4) Improves Neurobehavioral Outcomes After Acute Ischemic Stroke in Diabetic Rats: Possible Role of Vascular Endothelial TLR-4. Mol Neurobiol 2019; 56:1607-1617. [PMID: 29909454 PMCID: PMC6295357 DOI: 10.1007/s12035-018-1184-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/07/2018] [Indexed: 12/11/2022]
Abstract
Diabetes increases the risk of occurrence and poor functional recovery after ischemic stroke injury. Previously, we have demonstrated greater hemorrhagic transformation (HT), edema, and more severe functional deficits after stroke in diabetic animals that also presented with cerebral vasoregression and endothelial cell death in the recovery period. Given that Toll-like receptor 4 (TLR-4) activation in microvascular endothelial cells triggers a robust inflammatory response, we hypothesized that inhibition of TLR-4 signaling prevents endothelial cell death and improves outcomes after stroke. Animals were treated with vehicle or TLR-4 inhibitor TAK242 (3 mg/kg; i.p.) following middle cerebral artery occlusion (MCAO). Neurobehavioral deficits were measured at baseline and day 3 after ischemic stroke. Primary brain microvascular endothelial cells (BMVECs) from diabetic animals were subjected to oxygen glucose deprivation re-oxygenation (OGDR) and treated with 0.1 mM iron(III)sulfate hydrate (iron) (to mimic the post-stroke bleeding) and TLR-4 inhibitors. Ischemic stroke increased the expression of TLR-4 in both hemispheres and in the microvasculature of diabetic animals. Cerebral infarct, edema, HT, and functional deficits were greater in diabetic compared to control animals. Inhibition of TLR-4 significantly reduced the neurovascular injury and improved functional outcomes. OGDR and iron reduced the cell viability and increased the expression of TLR-4 associated proteins (RIP3, MyD88, phospho-NF-kB, and release of IL-6) in BMVECs from diabetic animals. In conclusion, TLR-4 is highly upregulated in the microvasculature and that beneficial effects of TLR-4 inhibition are more profound in diabetes. This suggests that inhibition of vascular TLR-4 may provide therapeutic benefits for stroke recovery in diabetes.
Collapse
Affiliation(s)
- Yasir Abdul
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA
| | | | - Weiguo Li
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA
| | - Jennifer C Sullivan
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA
| | - Guangkuo Dong
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA.
- Department of Physiology, Augusta University, 1120 15th Street CA-2094, Augusta, GA, 30912, USA.
| |
Collapse
|
33
|
Edwards DN, Bix GJ. Roles of blood-brain barrier integrins and extracellular matrix in stroke. Am J Physiol Cell Physiol 2018; 316:C252-C263. [PMID: 30462535 DOI: 10.1152/ajpcell.00151.2018] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemicstroke is a leading cause of death and disability in the United States, but recent advances in treatments [i.e., endovascular thrombectomy and tissue plasminogen activator (t-PA)] that target the stroke-causing blood clot, while improving overall stroke mortality rates, have had much less of an impact on overall stroke morbidity. This may in part be attributed to the lack of therapeutics targeting reperfusion-induced injury after the blood clot has been removed, which, if left unchecked, can expand injury from its core into the surrounding at risk tissue (penumbra). This occurs in two phases of increased permeability of the blood-brain barrier, a physical barrier that under physiologic conditions regulates brain influx and efflux of substances and consists of tight junction forming endothelial cells (and transporter proteins), astrocytes, pericytes, extracellular matrix, and their integrin cellular receptors. During, embryonic development, maturity, and following stroke reperfusion, cerebral vasculature undergoes significant changes including changes in expression of integrins and degradation of surrounding extracellular matrix. Integrins, heterodimers with α and β subunits, and their extracellular matrix ligands, a collection of proteoglycans, glycoproteins, and collagens, have been modestly studied in the context of stroke compared with other diseases (e.g., cancer). In this review, we describe the effect that various integrins and extracellular matrix components have in embryonic brain development, and how this changes in both maturity and in the poststroke environment. Particular focus will be on how these changes in integrins and the extracellular matrix affect blood-brain barrier components and their potential as diagnostic and therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Danielle N Edwards
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,Department of Neuroscience, University of Kentucky , Lexington, Kentucky
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,Department of Neuroscience, University of Kentucky , Lexington, Kentucky.,Department of Neurology, University of Kentucky , Lexington, Kentucky.,Department of Neurosurgery, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
34
|
Li M, Qian M, Kyler K, Xu J. Endothelial-Vascular Smooth Muscle Cells Interactions in Atherosclerosis. Front Cardiovasc Med 2018; 5:151. [PMID: 30406116 PMCID: PMC6207093 DOI: 10.3389/fcvm.2018.00151] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory process that can eventually lead to cardiovascular disease (CVD). Despite available treatment, the prevalence of atherosclerotic CVD, which has become the leading cause of death worldwide, persists. Identification of new mechanisms of atherogenesis are highly needed in order to develop an effective therapeutic treatment. The blood vessels contain two primary major cell types: endothelial cells (EC) and vascular smooth muscle cells (VSMC). Each of these performs an essential function in sustaining vascular homeostasis. EC-VSMC communication is essential not only to development, but also to the homeostasis of mature blood vessels. Aberrant EC-VSMC interaction could promote atherogenesis. Identification of the mode of EC-VSMC crosstalk that regulates vascular functionality and sustains homeostasis may offer strategic insights for prevention and treatment of atherosclerotic CVD. Here we will review the molecular mechanisms underlying the interplay between EC and VSMC that could contribute to atherosclerosis. We also highlight open questions for future research directions.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
35
|
Zhao M, Wang J, Xi X, Tan N, Zhang L. SNHG12 Promotes Angiogenesis Following Ischemic Stroke via Regulating miR-150/VEGF Pathway. Neuroscience 2018; 390:231-240. [PMID: 30193860 DOI: 10.1016/j.neuroscience.2018.08.029] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/21/2018] [Accepted: 08/26/2018] [Indexed: 12/21/2022]
Abstract
The promotion of angiogenesis is a promising therapeutic strategy for ischemic stroke. Many long noncoding RNAs (lncRNAs) are related to angiogenesis following ischemic stroke. LncRNA small nucleolar RNA host gene 12 (SNHG12) was upregulated in oxygen-glucose deprivation (OGD)-exposed primary brain microvascular endothelial cells and in microvessel from middle cerebral artery occlusion (MCAO) animal brains. However, the role and underlying mechanism of SNHG12 in ischemic stroke especially associated with angiogenesis process remain unknown. The expression of SNHG12 and miR-150 was determined in OGD-stimulated mouse brain microvascular endothelial (bEnd.3) cells. The role and mechanism of SNHG12 in the angiogenesis after ischemic stroke were investigated using gain- and loss-of function approaches both in OGD-exposed bEnd.3 cells and in MCAO mouse models. We found SNHG12 expression was elevated, whereas miR-150 reduced in OGD-exposed bEnd.3 cells. Upregulation of SNHG12 elevated, and SNHG12 knockdown suppressed the capillary-like tube formation, viability, migration, and VEGF expression in OGD-injured bEnd.3 cells. miR-150 mimic reversed, whereas anti-miR-150 further strengthened the effect of SNHG12 upregulation on the angiogenesis in bEnd.3 cells. Furthermore, we found that SNHG12 functioned as a competing endogenous RNA for miR-150 to regulate VEGF expression. Additionally, overexpression of SNHG12 improved the recovery of neurological function, reduced infarct volume and miR-150 expression, increased vascular density and VEGF expression in the infarct border zone of MCAO mice. In conclusion, SNHG12 promotes the angiogenesis following ischemic stroke via miR-150/VEGF pathway, which further clarified the mechanism of angiogenesis after ischemic stroke and provides a target for the treatment of this disease.
Collapse
Affiliation(s)
- Mian Zhao
- The Clinical Laboratory of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Jun Wang
- The Clinical Laboratory of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Xinlong Xi
- The Cardiac Intervention Room of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Nan Tan
- Department of Cadre's Ward of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Li Zhang
- Department of Clinical Laboratory, Shaanxi Friendship Hospital, Xi'an 710068, Shaanxi, China.
| |
Collapse
|
36
|
Rajkovic O, Potjewyd G, Pinteaux E. Regenerative Medicine Therapies for Targeting Neuroinflammation After Stroke. Front Neurol 2018; 9:734. [PMID: 30233484 PMCID: PMC6129611 DOI: 10.3389/fneur.2018.00734] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a major pathological event following ischemic stroke that contributes to secondary brain tissue damage leading to poor functional recovery. Following the initial ischemic insult, post-stroke inflammatory damage is driven by initiation of a central and peripheral innate immune response and disruption of the blood-brain barrier (BBB), both of which are triggered by the release of pro-inflammatory cytokines and infiltration of circulating immune cells. Stroke therapies are limited to early cerebral blood flow reperfusion, and whilst current strategies aim at targeting neurodegeneration and/or neuroinflammation, innovative research in the field of regenerative medicine aims at developing effective treatments that target both the acute and chronic phase of inflammation. Anti-inflammatory regenerative strategies include the use of nanoparticles and hydrogels, proposed as therapeutic agents and as a delivery vehicle for encapsulated therapeutic biological factors, anti-inflammatory drugs, stem cells, and gene therapies. Biomaterial strategies-through nanoparticles and hydrogels-enable the administration of treatments that can more effectively cross the BBB when injected systemically, can be injected directly into the brain, and can be 3D-bioprinted to create bespoke implants within the site of ischemic injury. In this review, these emerging regenerative and anti-inflammatory approaches will be discussed in relation to ischemic stroke, with a perspective on the future of stroke therapies.
Collapse
Affiliation(s)
- Olivera Rajkovic
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Geoffrey Potjewyd
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
37
|
Zhou YF, Li PC, Wu JH, Haslam JA, Mao L, Xia YP, He QW, Wang XX, Lei H, Lan XL, Miao QR, Yue ZY, Li YN, Hu B. Sema3E/PlexinD1 inhibition is a therapeutic strategy for improving cerebral perfusion and restoring functional loss after stroke in aged rats. Neurobiol Aging 2018; 70:102-116. [PMID: 30007159 DOI: 10.1016/j.neurobiolaging.2018.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 05/21/2018] [Accepted: 06/04/2018] [Indexed: 01/19/2023]
Abstract
Brain tissue survival and functional recovery after ischemic stroke greatly depend on cerebral vessel perfusion and functional collateral circulation in the ischemic area. Semaphorin 3E (Sema3E), one of the class 3 secreted semaphorins, has been demonstrated to be a critical regulator in embryonic and postnatal vascular formation via binding to its receptor PlexinD1. However, whether Sema3E/PlexinD1 signaling is involved in poststroke neovascularization remains unknown. To determine the contribution of Sema3E/PlexinD1 signaling to poststroke recovery, aged rats (18 months) were subjected to a transient middle cerebral artery occlusion. We found that depletion of Sema3E/PlexinD1 signaling with lentivirus-mediated PlexinD1-specific-shRNA improves tissue survival and functional outcome. Sema3E/PlexinD1 inhibition not only increases cortical perfusion but also ameliorates blood-brain barrier damage, as determined by positron emission tomography and magnetic resonance imaging. Mechanistically, we demonstrated that Sema3E suppresses endothelial cell proliferation and angiogenic capacity. More importantly, Sema3E/PlexinD1 signaling inhibits recruitment of pericytes by decreasing production of platelet derived growth factor-BB in endothelial cells. Overall, our study revealed that inhibition of Sema3E/PlexinD1 signaling in the ischemic penumbra, which increases both endothelial angiogenic capacity and recruitment of pericytes, contributed to functional neovascularization and blood-brain barrier integrity in the aged rats. Our findings imply that Sema3E/PlexinD1 signaling is a novel therapeutic target for improving brain tissue survival and functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng-Cheng Li
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie-Hong Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - James Andrew Haslam
- Swansea College of Medicine, Swansea University, Singleton Park, Swansea, UK
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan-Wei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu-Xia Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Hao Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xiao-Li Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Robert Miao
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhen-Yu Yue
- Department of Neurology, Department of Neuroscience, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ya-Nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
38
|
Abstract
Stroke is a cerebrovascular disorder that affects many people worldwide. In addition to the well-established functions of astrocytes and microglia in stroke pathogenesis, pericytes also play an important role in stroke progression and recovery. As perivascular multi-potent cells and an important component of the blood–brain barrier (BBB), pericytes have been shown to exert a large variety of functions, including serving as stem/progenitor cells and maintaining BBB integrity. Here in this review, we summarize the roles of pericytes in stroke pathogenesis, with a focus on their effects in cerebral blood flow, BBB integrity, angiogenesis, immune responses, scar formation and fibrosis.
Collapse
Affiliation(s)
- Jyoti Gautam
- 1 Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Yao Yao
- 1 Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| |
Collapse
|
39
|
Li W, Kandhare AD, Mukherjee AA, Bodhankar SL. Hesperidin, a plant flavonoid accelerated the cutaneous wound healing in streptozotocin-induced diabetic rats: Role of TGF-ß/Smads and Ang-1/Tie-2 signaling pathways. EXCLI JOURNAL 2018; 17:399-419. [PMID: 29805347 PMCID: PMC5962903 DOI: 10.17179/excli2018-1036] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022]
Abstract
Background: Delayed wound healing is a diverse, multifactorial, complex and inter-related complication of diabetes resulting in significant clinical morbidity. Hesperidin possesses potent antidiabetic and wound healing activity. Aim: To evaluate the potential of hesperidin against experimentally induced diabetes foot ulcers. Methods: Diabetes was induced experimentally by streptozotocin (STZ, 55 mg/kg, i.p.) in Sprague Dawley rats (180-220 g) and wounds were created on the dorsal surface of the hind paw of rats. Hesperidin (25, 50 and 100 mg/kg, p.o.) was administered for 21 days after wound stabilization. Various biochemical, molecular and histopathological parameters were evaluated in wound tissue. Results: STZ-induced decrease in body weight and increase in blood glucose, food, and water intake was significantly (p < 0.05) inhibited by hesperidin (50 and 100 mg/kg) treatment. It showed a significant increase (p < 0.05) in percent wound closure and serum insulin level. The STZ-induced decrease in SOD and GSH level, as well as elevated MDA and NO levels, were significantly (p < 0.05) attenuated by hesperidin (50 and 100 mg/kg) treatment. Intraperitoneal administration of STZ caused significant down-regulation in VEGF-c, Ang-1, Tie-2, TGF-β and Smad 2/3 mRNA expression in wound tissues whereas hesperidin (50 and 100 mg/kg) treatment showed significant up-regulation in these mRNA expressions. STZ-induced alteration in would architecture was also attenuated by hesperidin (50 and 100 mg/kg) treatment. Conclusion: Together, treatment with hesperidin accelerate angiogenesis and vasculogenesis via up-regulation of VEGF-c, Ang-1/Tie-2, TGF-β and Smad-2/3 mRNA expression to enhance wound healing in chronic diabetic foot ulcers.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Dermatology, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, Shaanxi, 710003, China
| | - Amit D Kandhare
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Paud Road, Pune-411 038, India.,Jalan Universiti Bandar Barat, 31900, Kampar, Perak, Malaysia
| | - Anwesha A Mukherjee
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Paud Road, Pune-411 038, India
| | - Subhash L Bodhankar
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Paud Road, Pune-411 038, India
| |
Collapse
|
40
|
Abstract
Stroke-induced endothelial cell injury leads to destruction of cerebral microvasculature and significant damage to the brain tissue. A subacute phase of cerebral ischemia is associated with regeneration involving the activation of vascular remodeling, neuroplasticity, neurogenesis, and neuroinflammation processes. Effective restoration and improvement of blood supply to the damaged brain tissue offers a potential therapy for stroke. microRNAs (miRNAs) are recently identified small RNA molecules that regulate gene expression and significantly influence the essential cellular processes associated with brain repair following stroke. A number of specific miRNAs are implicated in regulating the development and propagation of the ischemic tissue damage as well as in mediating post-stroke regeneration. In this review, I discuss the functions of the miRNA miR-155 and the effect of its in vivo inhibition on brain recovery following experimental cerebral ischemia. The article introduces new and unexplored approach to cerebral regeneration: regulation of brain tissue repair through a direct modulation of specific miRNA activity.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosurgery, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
41
|
Popa-Wagner A, Glavan DG, Olaru A, Olaru DG, Margaritescu O, Tica O, Surugiu R, Sandu RE. Present Status and Future Challenges of New Therapeutic Targets in Preclinical Models of Stroke in Aged Animals with/without Comorbidities. Int J Mol Sci 2018; 19:ijms19020356. [PMID: 29370078 PMCID: PMC5855578 DOI: 10.3390/ijms19020356] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 01/02/2023] Open
Abstract
The aging process, comorbidities, and age-associated diseases are closely dependent on each other. Cerebral ischemia impacts a wide range of systems in an age-dependent manner. However, the aging process has many facets which are influenced by the genetic background and epigenetic or environmental factors, which can explain why some people age differently than others. Therefore, there is an urgent need to identify age-related changes in body functions or structures that increase the risk for stroke and which are associated with a poor outcome. Multimodal imaging, electrophysiology, cell biology, proteomics, and transcriptomics, offer a useful approach to link structural and functional changes in the aging brain, with or without comorbidities, to post-stroke rehabilitation. This can help us to improve our knowledge about senescence firstly, and in this context, aids in elucidating the pathophysiology of age-related diseases that allows us to develop therapeutic strategies or prevent diseases. These processes, including potential therapeutical interventions, need to be studied first in relevant preclinical models using aged animals, with and without comorbidities. Therefore, preclinical research on ischemic stroke should consider age as the most important risk factor for cerebral ischemia. Furthermore, the identification of effective therapeutic strategies, corroborated with successful translational studies, will have a dramatic impact on the lives of millions of people with cerebrovascular diseases.
Collapse
Affiliation(s)
- Aurel Popa-Wagner
- Griffith University School of Medicine, Gold Coast Campus, QLD, Queensland Eye Institute, Brisbane, QLD 4101, Australia.
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Daniela-Gabriela Glavan
- Psychiatry Clinic Hospital, University of Medicine and Pharmacy of Craiova, Petru Rares Street 2, 200349 Craiova, Romania.
| | - Andrei Olaru
- Department of Ophthalmology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | | | - Otilia Margaritescu
- Department of Neurosurgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Oana Tica
- Department of "Mother and Child", University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Roxana Surugiu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Raluca Elena Sandu
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| |
Collapse
|
42
|
Manuel GE, Johnson T, Liu D. Therapeutic angiogenesis of exosomes for ischemic stroke. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2017; 9:188-191. [PMID: 29348795 PMCID: PMC5770515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/15/2017] [Indexed: 06/07/2023]
Abstract
Angiogenesis is the process through which new blood vessels are formed, while therapeutic angiogenesis aims to promote and control the angiogenic response. Ischemia results from the lack of blood flow with oxygen and nutrients. Therapeutic angiogenesis is crucial in preserving brain tissue and bodily functions after ischemic stroke. Various approaches have been proposed to promote angiogenesis in ischemic diseases. Traditional protein/gene and subsequent stem/progenitor cell approaches have not shown consistent efficacy for ischemic diseases in clinical trials. Exosomes are microparticles secreted from cells and conduct cell-cell communication including stem cell or cancer cell induced pro-angiogenesis. Utilization of exogenous exosomes for the treatment of ischemic diseases is an emerging approach which may prevent certain disadvantages such as easy degradation and tumor formation happened in other strategies. This review highlights recent reports on the use of exosomes as a therapeutic agent to promote angiogenesis in ischemic stroke.
Collapse
Affiliation(s)
- Gygeria E Manuel
- Department of Chemistry & Biochemistry, Spelman CollegeAtlanta, GA 30310, USA
| | - Takerra Johnson
- Cardiovascular Research Institute, Morehouse School of MedicineAtlanta, GA 30310, USA
| | - Dong Liu
- Cardiovascular Research Institute, Morehouse School of MedicineAtlanta, GA 30310, USA
- Department of Physiology, Morehouse School of MedicineAtlanta, GA 30310, USA
| |
Collapse
|
43
|
Wang Y, Zhang R, Xing X, Guo J, Xie F, Zhang G, Qin X. Repulsive guidance molecule a suppresses angiogenesis after ischemia/reperfusion injury of middle cerebral artery occlusion in rats. Neurosci Lett 2017; 662:318-323. [PMID: 29061393 DOI: 10.1016/j.neulet.2017.10.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/26/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Abstract
Repulsive guidance molecule a (RGMa) has now emerged as a molecule with pleiotropic roles, including repulsion, adhesion, migration and differentiation in the nervous system. In this study, adult male Sprague-Dawley (SD) rats received 90-min middle cerebral artery occlusion (MCAO) to observe RGMa/neogenin expression sites after ischemia/reperfusion injury and changes in angiogenesis after treatment with RNA interference using RGMa-specific recombinant adenovirus rAd5-shRNA-RGMa (rAd-shRGMa). To clarify how RGMa mediates angiogenesis, the RGMa function-blocking peptide six fibronectin type III (6FNIII) was also administered, and corresponding changes in vascular endothelial growth factor (VEGF), angiopoietin-2 (Ang2), angiopoietin-1 (Ang1), and brain derived neurotrophic factor (BDNF) were determined by western blotting. Both RGMa and its receptor neogenin were expressed in neurons and vessel endothelial cells after ischemia/reperfusion injury, and angiogenesis, coupled with functional recovery, was enhanced after RNA interference against RGMa compared with the vehicle groups. VEGF, Ang2, Ang1 and BDNF expression levels were significantly increased after intervention with rAd-shRGMa or 6FNIII. Thus, RGMa might suppress angiogenesis via VEGF, Ang2, Ang1 and BDNF after cerebral ischemia/reperfusion injury, which has therapeutic potential by reducing these endogenous detrimental mechanisms.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rongrong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiangfeng Xing
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jia Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fei Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Gang Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
44
|
Ramli Y, Alwahdy AS, Kurniawan M, Juliandi B, Wuyung PE, Bilianti Susanto YD. Intravenous Versus Intraarterial Transplantation of Human Umbilical Cord Blood Mononuclear Cells for Brain Ischemia in Rats. HAYATI JOURNAL OF BIOSCIENCES 2017. [DOI: 10.1016/j.hjb.2017.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
45
|
Ji Y, Yan X, Hu Y, Xue H, Sun J, Chen H, Zhang J, Wang L, Xue B, Sun L. DhHP‑6 attenuates cerebral ischemia‑reperfusion injury in rats through the inhibition of apoptosis. Mol Med Rep 2017; 16:7229-7236. [PMID: 28944912 PMCID: PMC5865850 DOI: 10.3892/mmr.2017.7569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 05/08/2017] [Indexed: 11/06/2022] Open
Abstract
As a novel reactive oxygen species (ROS) scavenger, deuterohemin His peptide‑6 (DhHP‑6) has been demonstrated to prolong the lifespan of Caenorhabditis elegans and has also exhibited protective effects in myocardial ischemia‑reperfusion injury. Whether similar effects occur during cerebral ischemia‑reperfusion (CIR) injury remains to be elucidated. The present study evaluated the function of DhHP‑6 and its underlying mechanisms in a middle cerebral artery occlusion (MCAO) model in rats. The focal transient MCAO model was implemented using the Longa method of ischemia for 2 h followed by reperfusion for 22 h in male Wistar rats. DhHP‑6 was administered at the onset of reperfusion via intraperitoneal injection. The infarct volume, brain edema, brain apoptosis and neurological function were evaluated 24 h following stroke. To further determine the role of DhHP‑6 in CIR injury, the levels of ROS and malondialdehyde (MDA), the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH‑Px), and the protein expression levels of B‑cell lymphoma 2 (Bcl‑2)‑associated X protein (Bax), cleaved caspase‑3, cytochrome c, Bcl‑2 and phosphorylated‑Akt/Akt were measured in ischemic cortex tissues. The results demonstrated that DhHP‑6 significantly improved infarct volume, brain edema and neurological deficits, and reduced the percentage of TUNEL‑positive cells. The levels of ROS and MDA were decreased, whereas no significant changes in the activities of SOD, CAT and GSH‑Px were observed. The levels of Bax, cleaved caspase‑3, and cytochrome c were downregulated, whereas the levels of Bcl‑2 and p‑Akt/Akt were upregulated. The results of the present study indicated that DhHP‑6 may offer therapeutic potential for cerebral ischemia. The neuroprotective effects of DhHP‑6 maybe mediated by its anti‑oxidative properties, anti‑apoptotic activities, or activation of the phosphoinositide 3‑kinase/Akt survival pathway.
Collapse
Affiliation(s)
- Yingshi Ji
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin Yan
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Hu
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huan Xue
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jianfeng Sun
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huaqiu Chen
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiayu Zhang
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liping Wang
- Department of Biological Chemistry, College of Life Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Baigong Xue
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li Sun
- Department of Neurology, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
46
|
Chen CK, Hsu PY, Wang TM, Miao ZF, Lin RT, Juo SHH. TRPV4 Activation Contributes Functional Recovery from Ischemic Stroke via Angiogenesis and Neurogenesis. Mol Neurobiol 2017; 55:4127-4135. [PMID: 28597396 DOI: 10.1007/s12035-017-0625-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
The endothelial transient receptor potential cation channel subfamily V member 4 (TRPV4) plays a crucial role in vascular remodeling; however, TRPV4-mediated angiogenesis after ischemic neuronal death as a neurorestorative strategy has not yet been thoroughly examined. In this study, we first tested whether TRPV4 activation can improve functional recovery in rats subjected to transient brain ischemia. The possible mechanisms for TRPV4 activation-promoted functional recovery were explored. A TRPV4 agonist, 4α-phorbol 12,13-didecanoate (4α-PDD), was intravenously injected via the tail vein at 6 h and 1, 2, 3, 4 days after ischemic stroke. The treatment reduced infarct volume by almost 50% (14.7 ± 3.7 vs. 29.2 ± 6.2%; p < 0.0001) and improved functional outcomes (p = 0.03) on day 5. To explore the therapeutic mechanism, we measured endothelial nitric oxide synthase (eNOS) expression and phosphorylation, vascular endothelial growth factor A (VEGFA) signaling, and neural stem/progenitor cells (NPCs). TRPV4 activation significantly increased eNOS expression and phosphorylation (serine 1177) by more than 2-fold in the ischemic region. The expressions of VEGFA and VEGF receptor-2 were significantly higher in the treated animals, especially an increase of the proangiogenic VEGFA164a isoform while a decrease of the antiangiogenic VEGFA165b isoform. We evaluated angiogenesis by detecting microvessel density in ischemic region. Using the immunohistochemistry staining, we found that 4α-PDD treatment caused a 3.4-fold increase of microvessel density (p < 0.0001). In addition, NPC proliferation and migration in the ischemic hemisphere were increased by 3-fold and 5-fold, respectively. In conclusion, our data suggest that TRPV4 activation by 4α-PDD may improve poststroke functional improvement through angiogenesis and neurogenesis.
Collapse
Affiliation(s)
- Chun-Kai Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Collage of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Yuan Hsu
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ming Wang
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Zhi-Feng Miao
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ruey-Tay Lin
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Suh-Hang H Juo
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan. .,Institute of New Drug Development, China Medical University, Taichung, Taiwan. .,Brain Disease Research Center, China Medical University, Taichung, Taiwan. .,Center for Myopia and Eye Disease, China Medical University, Taichung, Taiwan.
| |
Collapse
|
47
|
Machado-Pereira M, Santos T, Ferreira L, Bernardino L, Ferreira R. Challenging the great vascular wall: Can we envision a simple yet comprehensive therapy for stroke? J Tissue Eng Regen Med 2017; 12:e350-e354. [PMID: 28182332 DOI: 10.1002/term.2427] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 09/27/2016] [Accepted: 02/06/2017] [Indexed: 12/26/2022]
Abstract
Stroke is a leading cause of death in adult life, closely behind ischemic heart disease, and causes a significant and abiding socioeconomic burden. However, current therapies are not able to ensure full neurologic and/or sequelae-free recovery to all stroke survivors. We believe treatment efficacy and patient rehabilitation could be enhanced significantly by targeting blood-brain barrier (BBB) deregulation and inflammation-induced barrier loss that occurs after stroke. In this pathological context, bone marrow-derived endothelial progenitor cells (EPC) enter the bloodstream towards the lesion site, but their insufficient numbers and impaired angiogenic ability compromise neurovascular regeneration. In this context, cell-based therapies have become increasingly appealing since treating patients with large numbers of mesenchymal or hematopoietic stem/progenitor cells alone may boost repair. However, this approach could be met with several challenges in terms of logistics and cost; hence, the development of a drug delivery system suitable for intravenous administration and functionalized for selective uptake by circulating EPC could enhance their restorative potential without perceived complications. The ability to encapsulate proangiogenic and anti-inflammatory agents, such as retinoic acid, and to safely and easily deliver them systemically may open new therapeutic perspectives for the treatment of cerebrovascular disorders. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Marta Machado-Pereira
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Tiago Santos
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Lino Ferreira
- Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal.,CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Raquel Ferreira
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
48
|
Mechanisms of Acupuncture Therapy for Cerebral Ischemia: an Evidence-Based Review of Clinical and Animal Studies on Cerebral Ischemia. J Neuroimmune Pharmacol 2017; 12:575-592. [DOI: 10.1007/s11481-017-9747-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 04/13/2017] [Indexed: 12/20/2022]
|
49
|
Soleimannejad K, Rahmani A, Hatefi M, Khataminia M, Hafezi Ahmadi MR, Asadollahi K. Effects of Nigella sativa Extract on Markers of Cerebral Angiogenesis after Global Ischemia of Brain in Rats. J Stroke Cerebrovasc Dis 2017; 26:1514-1520. [PMID: 28396188 DOI: 10.1016/j.jstrokecerebrovasdis.2017.02.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/15/2017] [Accepted: 02/25/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reduction of permanent or transient cerebral blood flow may lead to some structural and functional changes of the brain, causing high mortality and morbidity. The aim of this experimental study was to investigate the effects of hydroalcoholic extract of Nigella sativa (NS) on markers of cerebral angiogenesis in rats induced by global brain ischemia. METHODS Thirty-two male Wistar rats (250 ± 20 g) were randomly divided into 4 groups: group 1, control group receiving only normal saline; group 2, sham group undergoing surgery and stroke induction without treatment; and groups 3 and 4 treated with 10 and 20 mg/kg NS, respectively, after induction of stroke. Global ischemia was induced by ligation of the right carotid artery for 20 minutes. RESULTS According to the results of this study, brain edema and infarct volume were significantly decreased in the group treated with 20 mg/kg NS compared with the group treated with 10 mg/kg NS (P < .05). Global ischemia caused a significant reduction in gene expression of vasoactive endothelial growth factor (VEGF) and hypoxia-inducible factor (HIF) in the sham group compared with the control group (P < .05), but NS groups, in led to a significant increase in gene expression of VEGF and HIF compared with the sham group (P < .05). In addition, the activity level of matrix metallopeptidase-9 was decreased among NS groups compared with the control group (P < .05). CONCLUSIONS Application of NS extract among rats with brain ischemia is associated with increase of VEGF and HIF as angiogenic markers and inhibition of matrix metallopeptidase-9 activities.
Collapse
Affiliation(s)
- Koroush Soleimannejad
- Department of Cardiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Asghar Rahmani
- Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Masoud Hatefi
- Department of Neurosurgery, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Masoud Khataminia
- Department of Pharmacology, Faculty of Pharmacy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Khairollah Asadollahi
- Department of Social Medicine, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
50
|
Vascular Endothelial Growth Factor Isoform-B Stimulates Neurovascular Repair After Ischemic Stroke by Promoting the Function of Pericytes via Vascular Endothelial Growth Factor Receptor-1. Mol Neurobiol 2017; 55:3611-3626. [PMID: 28290152 DOI: 10.1007/s12035-017-0478-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
Ischemic stroke triggers endogenous angiogenic mechanisms, which correlates with longer survival in patients. As such, promoting angiogenesis appears to be a promising approach. Experimental studies investigated mostly the potent angiogenic factor vascular endothelial growth factor isoform-A (VEGF-A). However, VEGF-A increases the risk of destabilizing the brain microvasculature, thus hindering the translation of its usage in clinics. An attractive alternative VEGF isoform-B (VEGF-B) was recently reported to act as a survival factor rather than a potent angiogenic factor. In this study, we investigated the therapeutic potential of VEGF-B in ischemic stroke using different in vivo and in vitro approaches. We showed that the delayed intranasal administration of VEGF-B reduced neuronal damage and inflammation. Unexpectedly, VEGF-B stimulated the formation of stable brain microvasculature within the injured region by promoting the interaction between endothelial cells and pericytes. Our data indicate that the effects of VEGF-B were mediated via its specific receptor VEGF receptor-1 (VEGFR-1) that is predominately expressed in brain pericytes. Importantly, VEGF-B promoted the survival of pericytes, and not brain endothelial cells, by inducing expression of the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) and the main protein involved in energy homeostasis AMP-activated protein kinase α (AMPKα). Moreover, we showed that VEGF-B stimulated the pericytic release of factors stimulating a "reparative angiogenesis" that does not compromise microvasculature stability. Our study unraveled hitherto unknown role of VEGF-B/VEGFR-1 signaling in regulating the function of pericytes. Furthermore, our findings suggest that brain microvasculature stabilization via VEGF-B constitutes a safe therapeutic approach for ischemic stroke.
Collapse
|