51
|
Yang F, Yang MY, Le JQ, Luo BY, Yin MD, Chao-Li, Jiang JL, Fang YF, Shao JW. Protective Effects and Therapeutics of Ginsenosides for Improving Endothelial Dysfunction: From Therapeutic Potentials, Pharmaceutical Developments to Clinical Trials. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:749-772. [PMID: 35450513 DOI: 10.1142/s0192415x22500318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The endothelium covers the internal lumen of the entire circulatory system and plays an important modulatory role in vascular homeostasis. Endothelium dysfunction, characterized by a vasoconstrictive, pro-inflammatory, and pro-coagulant state, usually manifests as a significant pathological process of vascular diseases, including hypertension, atherosclerosis (AS), stroke, diabetes mellitus, coronary artery disease, and cancer. Therefore, there is an urgent necessity to seek promising therapeutic drugs or remedies to ameliorate endothelial dysfunction-induced vascular ailments and complications. Recently, much attention has been attached to ginsenosides, the most significant active components of ginseng, which have always been referred to as "all-healing" and widely used for its extensively medicinal value. Surprisingly, ginsenosides have diverse biological activity which might be related to inflammation, apoptosis, oxidative stress, and angiogenesis. In this review, a brief introduction about endothelial dysfunction and ginsenosides was demonstrated, and the emphasis was put on summarizing multi-faceted pharmacological effects and underlying molecular mechanisms of ginsenosides on the endothelium, including vasorelaxation, anti-oxidation, anti-inflammation, and angio-modulation. Beyond that, nanotechnology to improve efficacy and the existing clinical trials of ginsenosides were concluded. Hopefully, our work will give suggestions for promoting clinical application of traditional Chinese medicine, e.g., hypertension, AS, diabetes, ischemic stroke, and cancer. This review provides a comprehensive base of knowledge for ginsenosides to prevention and treatment of vascular injury- related diseases with clinical significance.
Collapse
Affiliation(s)
- Fang Yang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Ming-Yue Yang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Qing Le
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bang-Yue Luo
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Meng-Die Yin
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Chao-Li
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jia-Li Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yi-Fan Fang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Wei Shao
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
52
|
Liu W, Zhou Y, Zhang YX, Yang KL, Liu YL, Wu FH, Gao YR. Connexin 43 mediated the angiogenesis of buyang huanwu decoction via vascular endothelial growth factor and angiopoietin-1 after ischemic stroke. CHINESE J PHYSIOL 2022; 65:72-79. [DOI: 10.4103/cjp.cjp_94_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
53
|
Zhou F, Dai O, Peng C, Xiong L, Ao H, Liu F, Zhou QM. Pro-Angiogenic Effects of Essential Oil from Perilla frutescens and Its Main Component (Perillaldehyde) on Zebrafish Embryos and Human Umbilical Vein Endothelial Cells. Drug Des Devel Ther 2021; 15:4985-4999. [PMID: 34924753 PMCID: PMC8674578 DOI: 10.2147/dddt.s336826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Perilla frutescens (L.) Britt., a traditional edible-medicinal herb in China, has been used to treat cardiovascular and cerebrovascular (cardio-cerebrovascular) diseases for thousands of years. However, knowledge of the mechanisms underlying the effects of essential oil from P. frutescens (EOPF) in the treatment of cardio-cerebrovascular diseases is lacking. The promotion of angiogenesis is beneficial in the treatment of ischemic cardio-cerebrovascular diseases. The current study investigated the pro-angiogenic role of EOPF and its main component perillaldehyde in sunitinib-injured transgenic Tg (flk1:EGFP) zebrafish embryos and human umbilical vein endothelial cells (HUVECs) for the first time. Materials and Methods The pro-angiogenic effects of EOPF and perillaldehyde were observed in vivo using transgenic Tg (flk1:EGFP) zebrafish embryos and in vitro using HUVECs. Cell viability, proliferation, migration, tube formation, and protein levels were detected by MTT, EdU staining, wound healing, transwell chamber, and Western blot assays, respectively. Results EOPF and perillaldehyde exerted a significant stimulatory effect on the formation of zebrafish intersegmental vessels (ISVs). Moreover, EOPF and perillaldehyde promoted proliferation, migration, and tube formation in sunitinib-treated HUVECs. Additionally, our findings uncovered that the pro-angiogenic effects of EOPF and perillaldehyde were mediated by increases in the expression ratios of p-ERK1/2 to ERK1/2 and Bcl-2 to Bax. Conclusion The present study is the first report to provide clear evidence that EOPF and perillaldehyde promote angiogenesis by stimulating repair of sunitinib-injured ISVs in zebrafish embryos and promoting proliferation, migration, and tube formation in sunitinib-injured HUVECs. The underlying mechanisms are related to increased p-ERK1/2 to ERK1/2 and Bcl-2 to Bax expression ratios. EOPF and perillaldehyde may be used in the treatment of cardio-cerebrovascular diseases, which is consistent with the traditional application of P. frutescens.
Collapse
Affiliation(s)
- Fei Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Ou Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Liang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Fei Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Qin-Mei Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Institute of Innovative Medicine Ingredients of Southwest Specialty Medicinal Materials, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| |
Collapse
|
54
|
Li J, Li C, Loreno EG, Miriyala S, Panchatcharam M, Lu X, Sun H. Chronic Low-Dose Alcohol Consumption Promotes Cerebral Angiogenesis in Mice. Front Cardiovasc Med 2021; 8:681627. [PMID: 34869620 PMCID: PMC8635527 DOI: 10.3389/fcvm.2021.681627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/27/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic alcohol consumption dose-dependently affects the incidence and prognosis of ischemic stroke. We determined the influence of chronic alcohol consumption on cerebral angiogenesis under physiological conditions and following ischemic stroke. In in vitro studies, acute exposure to low-concentration ethanol significantly increased angiogenic capability and upregulated vascular endothelial growth factor A (VEGF-A) and vascular endothelial growth factor receptor 2 (VEGFR2) in C57BL/6J mouse brain microvascular endothelial cells (MBMVECs). The increased angiogenic capability was abolished in the presence of a VEGFR2 inhibitor. In addition, the increased angiogenic capability and upregulated VEGF-A and VEGFR2 remained in chronically low-concentration ethanol-exposed MBMVECs. In in vivo studies, 8-week gavage feeding with low-dose ethanol significantly increased vessel density and vessel branches and upregulated VEGF-A and VEGFR2 in the cerebral cortex under physiological conditions. Furthermore, vessel density, vessel branches, and expression of VEGF-A and VEGFR2 in the peri-infarct cortex were significantly greater in low-dose ethanol-fed mice at 72 h of reperfusion. Although low-dose ethanol did not alter cerebral vasoreactivity and regional cerebral blood flow (rCBF) either before or during ischemia, it significantly augmented post-ischemic hyperemia during reperfusion. In contrast, exposure to high-concentration ethanol and 8-week gavage feeding with high-dose ethanol only had a mild inhibitory effect on angiogenic capability and cerebral angiogenesis, respectively. We conclude that heavy alcohol consumption may not dramatically alter cerebral angiogenesis, whereas light alcohol consumption significantly promotes cerebral angiogenesis.
Collapse
Affiliation(s)
- Jiyu Li
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chun Li
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Ethyn G Loreno
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Xiaohong Lu
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Hong Sun
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
55
|
Rohden F, Teixeira LV, Bernardi LP, Ferreira PCL, Colombo M, Teixeira GR, de Oliveira FDS, Cirne Lima EO, Guma FCR, Souza DO. Functional Recovery Caused by Human Adipose Tissue Mesenchymal Stem Cell-Derived Extracellular Vesicles Administered 24 h after Stroke in Rats. Int J Mol Sci 2021; 22:12860. [PMID: 34884665 PMCID: PMC8657917 DOI: 10.3390/ijms222312860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability, intensely demanding innovative and accessible therapeutic strategies. Approaches presenting a prolonged period for therapeutic intervention and new treatment administration routes are promising tools for stroke treatment. Here, we evaluated the potential neuroprotective properties of nasally administered human adipose tissue mesenchymal stem cell (hAT-MSC)-derived extracellular vesicles (EVs) obtained from healthy individuals who underwent liposuction. After a single intranasal EV (200 µg/kg) administered 24 h after a focal permanent ischemic stroke in rats, a higher number of EVs, improvement of the blood-brain barrier, and re-stabilization of vascularization were observed in the recoverable peri-infarct zone, as well as a significant decrease in infarct volume. In addition, EV treatment recovered long-term motor (front paws symmetry) and behavioral impairment (short- and long-term memory and anxiety-like behavior) induced by ischemic stroke. In line with these findings, our work highlights hAT-MSC-derived EVs as a promising therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Francieli Rohden
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luciele Varaschini Teixeira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luis Pedro Bernardi
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Faculty of Biomedicine, Universidade Federal de Ciências da Saúde de Porto Alegre—UFCSPA, Porto Alegre 90050-170, Brazil
| | - Pamela Cristina Lukasewicz Ferreira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Mariana Colombo
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil;
| | - Geciele Rodrigues Teixeira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fernanda dos Santos de Oliveira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Elizabeth Obino Cirne Lima
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fátima Costa Rodrigues Guma
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Diogo Onofre Souza
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| |
Collapse
|
56
|
Sun P, Ma F, Xu Y, Zhou C, Stetler RA, Yin KJ. Genetic deletion of endothelial microRNA-15a/16-1 promotes cerebral angiogenesis and neurological recovery in ischemic stroke through Src signaling pathway. J Cereb Blood Flow Metab 2021; 41:2725-2742. [PMID: 33910400 PMCID: PMC8504951 DOI: 10.1177/0271678x211010351] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral angiogenesis is tightly controlled by specific microRNAs (miRs), including the miR-15a/16-1 cluster. Recently, we reported that endothelium-specific conditional knockout of the miR-15a/16-1 cluster (EC-miR-15a/16-1 cKO) promotes post-stroke angiogenesis and improves long-term neurological recovery by increasing protein levels of VEGFA, FGF2, and their respective receptors VEGFR2 and FGFR1. Herein, we further investigated the underlying signaling mechanism of these pro-angiogenic factors after ischemic stroke using a selective Src family inhibitor AZD0530. EC-miR-15a/16-1 cKO and age- and sex-matched wild-type littermate (WT) mice were subjected to 1 h middle cerebral artery occlusion (MCAO) and 28d reperfusion. AZD0530 was administered daily by oral gavage to both genotypes of mice 3-21d after MCAO. Compared to WT, AZD0530 administration exacerbated spatial cognitive impairments and brain atrophy in EC-miR-15a/16-1 cKO mice following MCAO. AZD0530 also attenuated long-term recovery of blood flow and inhibited the formation of new microvessels, including functional vessels with blood circulation, in the penumbra of stroked cKO mice. Moreover, AZD0530 blocked the Src signaling pathway by downregulating phospho-Src and its downstream mediators (p-Stat3, p-Akt, p-FAK, p-p44/42 MAPK, p-p38 MAPK) in post-ischemic brains. Collectively, our data demonstrated that endothelium-targeted deletion of the miR-15a/16-1 cluster promotes post-stroke angiogenesis and improves long-term neurological recovery via activating Src signaling pathway.
Collapse
Affiliation(s)
- Ping Sun
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chao Zhou
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
57
|
Fang J, Chopp M, Xin H, Zhang L, Wang F, Golembieski W, Zhang ZG, He L, Liu Z. Plasminogen deficiency causes reduced angiogenesis and behavioral recovery after stroke in mice. J Cereb Blood Flow Metab 2021; 41:2583-2592. [PMID: 33853408 PMCID: PMC8504962 DOI: 10.1177/0271678x211007958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Plasminogen is involved in the process of angiogenesis; however, the underlying mechanism is unclear. Here, we investigated the potential contribution of plasmin/plasminogen in mediating angiogenesis and thereby contributing to functional recovery post-stroke. Wild-type plasminogen naive (Plg+/+) mice and plasminogen knockout (Plg-/-) mice were subjected to unilateral permanent middle cerebral artery occlusion (MCAo). Blood vessels were labeled with FITC-dextran. Functional outcomes, and cerebral vessel density were compared between Plg+/+ and Plg-/- mice at different time points after stroke. We found that Plg-/- mice exhibited significantly reduced functional recovery, associated with significantly decreased vessel density in the peri-infarct area in the ipsilesional cortex compared with Plg+/+ mice. In vitro, cerebral endothelial cells harvested from Plg-/- mice exhibited significantly reduced angiogenesis assessed using tube formation assay, and migration, as evaluated using Scratch assays, compared to endothelial cells harvested from Plg+/+ mice. In addition, using Western blots, expression of thrombospondin (TSP)-1 and TSP-2 were increased after MCAo in the Plg-/- group compared to Plg+/+ mice, especially in the ipsilesional side of brain. Taken together, our data suggest that plasmin/plasminogen down-regulates the expression level of TSP-1 and TSP-2, and thereby promotes angiogenesis in the peri-ischemic brain tissue, which contributes to functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Jinghuan Fang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Neurology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Hongqi Xin
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Fengjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | | | - Li He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
58
|
Tie2-expressing monocytes/macrophages promote cerebral revascularization in peri-infarct lesions upon ischemic insult. Signal Transduct Target Ther 2021; 6:295. [PMID: 34366430 PMCID: PMC8349906 DOI: 10.1038/s41392-021-00637-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 11/12/2022] Open
|
59
|
Przykaza Ł, Kozniewska E. Ligands of the Neuropeptide Y Y2 Receptors as a Potential Multitarget Therapeutic Approach for the Protection of the Neurovascular Unit Against Acute Ischemia/Reperfusion: View from the Perspective of the Laboratory Bench. Transl Stroke Res 2021; 13:12-24. [PMID: 34292517 PMCID: PMC8766383 DOI: 10.1007/s12975-021-00930-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Ischemic stroke is the third leading cause of death and disability worldwide, with no available satisfactory prevention or treatment approach. The current treatment is limited to the use of “reperfusion methods,” i.e., an intravenous or intra-arterial infusion of a fibrinolytic agent, mechanical removal of the clot by thrombectomy, or a combination of both methods. It should be stressed, however, that only approximately 5% of all acute strokes are eligible for fibrinolytic treatment and fewer than 10% for thrombectomy. Despite the tremendous progress in understanding of the pathomechanisms of cerebral ischemia, the promising results of basic research on neuroprotection are not currently transferable to human stroke. A possible explanation for this failure is that experiments on in vivo animal models involve healthy young animals, and the experimental protocols seldom consider the importance of protecting the whole neurovascular unit (NVU), which ensures intracranial homeostasis and is seriously damaged by ischemia/reperfusion. One of the endogenous protective systems activated during ischemia and in neurodegenerative diseases is represented by neuropeptide Y (NPY). It has been demonstrated that activation of NPY Y2 receptors (Y2R) by a specific ligand decreases the volume of the postischemic infarction and improves performance in functional tests of rats with arterial hypertension subjected to middle cerebral artery occlusion/reperfusion. This functional improvement suggests the protection of the NVU. In this review, we focus on NPY and discuss the potential, multidirectional protective effects of Y2R agonists against acute focal ischemia/reperfusion injury, with special reference to the NVU.
Collapse
Affiliation(s)
- Łukasz Przykaza
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland
| | - Ewa Kozniewska
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
60
|
Four Decades of Ischemic Penumbra and Its Implication for Ischemic Stroke. Transl Stroke Res 2021; 12:937-945. [PMID: 34224106 DOI: 10.1007/s12975-021-00916-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
The ischemic penumbra defined four decades ago has been the main battleground of ischemic stroke. The evolving ischemic penumbra concept has been providing insight for the development of vascular and cellular approaches as well as diagnostic tools for the treatment of ischemic stroke. rt-PA thrombolytic therapy to prevent the transition of ischemic penumbra to core has been approved for acute ischemic stroke within 3 h and was later recommended to extend to 4.5 h after symptom onset. Mechanical thrombectomy was introduced for the treatment of acute ischemic stroke with a therapeutic window of up to 24 h after stroke onset. Multiple modalities brain imaging techniques have been developed that provide guidance to define ischemic penumbra for reperfusion therapy in clinical practice. Cellular and molecular dissection of ischemic penumbra has been providing targets for the development of neuroprotective therapy for ischemic stroke. However, the dynamic nature of ischemic penumbra implicates that infarct core eventually expands into penumbra over time without reperfusion, dictating relative short therapeutic windows and limiting the impact of current reperfusion intervention. Entering the 5th decade since the introduction, ischemic penumbra remains the main focus of ischemic stroke research and clinical practice. In this review, we summarized the evolving ischemic penumbra concept and its implication in the development of vascular and cellular interventions as well as diagnostic tools for acute ischemic stroke. In addition, we discussed future perspectives on expansion of the campaign beyond ischemic penumbra to develop treatment for ischemic stroke.
Collapse
|
61
|
Takase H, Hamanaka G, Ohtomo R, Ishikawa H, Chung KK, Mandeville ET, Lok J, Fornage M, Herrup K, Tse KH, Lo EH, Arai K. Transcriptome Profiling of Mouse Corpus Callosum After Cerebral Hypoperfusion. Front Cell Dev Biol 2021; 9:685261. [PMID: 34222254 PMCID: PMC8248229 DOI: 10.3389/fcell.2021.685261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 02/01/2023] Open
Abstract
White matter damage caused by cerebral hypoperfusion is a major hallmark of subcortical ischemic vascular dementia (SIVD), which is the most common subtype of vascular cognitive impairment and dementia (VCID) syndrome. In an aging society, the number of SIVD patients is expected to increase; however, effective therapies have yet to be developed. To understand the pathological mechanisms, we analyzed the profiles of the cells of the corpus callosum after cerebral hypoperfusion in a preclinical SIVD model. We prepared cerebral hypoperfused mice by subjecting 2-month old male C57BL/6J mice to bilateral carotid artery stenosis (BCAS) operation. BCAS-hypoperfusion mice exhibited cognitive deficits at 4 weeks after cerebral hypoperfusion, assessed by novel object recognition test. RNA samples from the corpus callosum region of sham- or BCAS-operated mice were then processed using RNA sequencing. A gene set enrichment analysis using differentially expressed genes between sham and BCAS-operated mice showed activation of oligodendrogenesis pathways along with angiogenic responses. This database of transcriptomic profiles of BCAS-hypoperfusion mice will be useful for future studies to find a therapeutic target for SIVD.
Collapse
Affiliation(s)
- Hajime Takase
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Kelly K Chung
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Human Genetics Center, Division of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karl Herrup
- Department of Neurobiology and ADRC, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kai-Hei Tse
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Eng H Lo
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ken Arai
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
62
|
Yu R, Kim NS, Li Y, Jeong JY, Park SJ, Zhou B, Oh WJ. Vascular Sema3E-Plexin-D1 Signaling Reactivation Promotes Post-stroke Recovery through VEGF Downregulation in Mice. Transl Stroke Res 2021; 13:142-159. [PMID: 33978913 PMCID: PMC8766426 DOI: 10.1007/s12975-021-00914-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022]
Abstract
Post-stroke vascular remodeling, including angiogenesis, facilitates functional recovery. Proper vascular repair is important for efficient post-stroke recovery; however, the underlying mechanisms coordinating the diverse signaling pathways involved in vascular remodeling remain largely unknown. Recently, axon guidance molecules were revealed as key players in injured vessel remodeling. One such molecule, Semaphorin 3E (Sema3E), and its receptor, Plexin-D1, control vascular development by regulating vascular endothelial growth factor (VEGF) signaling. In this study, using a mouse model of transient brain infarction, we aimed to investigate whether Sema3E-Plexin-D1 signaling was involved in cerebrovascular remodeling after ischemic injury. We found that ischemic damage rapidly induced Sema3e expression in the neurons of peri-infarct regions, followed by Plexin-D1 upregulation in remodeling vessels. Interestingly, Plexin-D1 reemergence was concurrent with brain vessels entering an active angiogenic process. In line with this, Plxnd1 ablation worsened neurological deficits, infarct volume, neuronal survival rate, and blood flow recovery. Furthermore, reduced and abnormal vascular morphogenesis was caused by aberrantly increased VEGF signaling. In Plxnd1 knockout mice, we observed significant extravasation of intravenously administered tracers in the brain parenchyma, junctional protein downregulation, and mislocalization in regenerating vessels. This suggested that the absence of Sema3E-Plexin-D1 signaling is associated with blood–brain barrier (BBB) impairment. Finally, the abnormal behavioral performance, aberrant vascular phenotype, and BBB breakdown defects in Plxnd1 knockout mice were restored following the inhibition of VEGF signaling during vascular remodeling. These findings demonstrate that Sema3E-Plexin-D1 signaling can promote functional recovery by downregulating VEGF signaling in the injured adult brain.
Collapse
Affiliation(s)
- Ri Yu
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.,College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Nam-Suk Kim
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Yan Li
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jin-Young Jeong
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea
| | - Sang-Joon Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Bin Zhou
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Won-Jong Oh
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| |
Collapse
|
63
|
Patabendige A, Singh A, Jenkins S, Sen J, Chen R. Astrocyte Activation in Neurovascular Damage and Repair Following Ischaemic Stroke. Int J Mol Sci 2021; 22:4280. [PMID: 33924191 PMCID: PMC8074612 DOI: 10.3390/ijms22084280] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
Transient or permanent loss of tissue perfusion due to ischaemic stroke can lead to damage to the neurovasculature, and disrupt brain homeostasis, causing long-term motor and cognitive deficits. Despite promising pre-clinical studies, clinically approved neuroprotective therapies are lacking. Most studies have focused on neurons while ignoring the important roles of other cells of the neurovascular unit, such as astrocytes and pericytes. Astrocytes are important for the development and maintenance of the blood-brain barrier, brain homeostasis, structural support, control of cerebral blood flow and secretion of neuroprotective factors. Emerging data suggest that astrocyte activation exerts both beneficial and detrimental effects following ischaemic stroke. Activated astrocytes provide neuroprotection and contribute to neurorestoration, but also secrete inflammatory modulators, leading to aggravation of the ischaemic lesion. Astrocytes are more resistant than other cell types to stroke pathology, and exert a regulative effect in response to ischaemia. These roles of astrocytes following ischaemic stroke remain incompletely understood, though they represent an appealing target for neurovascular protection following stroke. In this review, we summarise the astrocytic contributions to neurovascular damage and repair following ischaemic stroke, and explore mechanisms of neuroprotection that promote revascularisation and neurorestoration, which may be targeted for developing novel therapies for ischaemic stroke.
Collapse
Affiliation(s)
- Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2321, Australia;
- Priority Research Centre for Stroke and Brain Injury, and Priority Research Centre for Brain & Mental Health, University of Newcastle, Callaghan, NSW 2321, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- Institute of Infection & Global Health, University of Liverpool, Liverpool L7 3EA, UK
| | - Ayesha Singh
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Neural Tissue Engineering: Keele (NTEK), Keele University, Staffordshire ST5 5BG, UK
| | - Jon Sen
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Clinical Informatics and Neurosurgery Fellow, The Cleveland Clinic, 33 Grosvenor Square, London SW1X 7HY, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
64
|
Zhou G, Wang Y, Gao S, Fu X, Cao Y, Peng Y, Zhuang J, Hu J, Shao A, Wang L. Potential Mechanisms and Perspectives in Ischemic Stroke Treatment Using Stem Cell Therapies. Front Cell Dev Biol 2021; 9:646927. [PMID: 33869200 PMCID: PMC8047216 DOI: 10.3389/fcell.2021.646927] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke (IS) remains one of the major causes of death and disability due to the limited ability of central nervous system cells to regenerate and differentiate. Although several advances have been made in stroke therapies in the last decades, there are only a few approaches available to improve IS outcome. In the acute phase of IS, mechanical thrombectomy and the administration of tissue plasminogen activator have been widely used, while aspirin or clopidogrel represents the main therapy used in the subacute or chronic phase. However, in most cases, stroke patients fail to achieve satisfactory functional recovery under the treatments mentioned above. Recently, cell therapy, especially stem cell therapy, has been considered as a novel and potential therapeutic strategy to improve stroke outcome through mechanisms, including cell differentiation, cell replacement, immunomodulation, neural circuit reconstruction, and protective factor release. Different stem cell types, such as mesenchymal stem cells, marrow mononuclear cells, and neural stem cells, have also been considered for stroke therapy. In recent years, many clinical and preclinical studies on cell therapy have been carried out, and numerous results have shown that cell therapy has bright prospects in the treatment of stroke. However, some cell therapy issues are not yet fully understood, such as its optimal parameters including cell type choice, cell doses, and injection routes; therefore, a closer relationship between basic and clinical research is needed. In this review, the role of cell therapy in stroke treatment and its mechanisms was summarized, as well as the function of different stem cell types in stroke treatment and the clinical trials using stem cell therapy to cure stroke, to reveal future insights on stroke-related cell therapy, and to guide further studies.
Collapse
Affiliation(s)
- Guoyang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yucong Peng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junwen Hu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
65
|
Delayed Exercise-induced Upregulation of Angiogenic Proteins and Recovery of Motor Function after Photothrombotic Stroke in Mice. Neuroscience 2021; 461:57-71. [PMID: 33667592 DOI: 10.1016/j.neuroscience.2021.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 01/02/2023]
Abstract
Treatments promoting post-stroke functional recovery continue to be an unmet therapeutic problem with physical rehabilitation being the most reproduced intervention in preclinical and clinical studies. Unfortunately, physiotherapy is typically effective at high intensity and early after stroke - requirements that are hardly attainable by stroke survivors. The aim of this study was to directly evaluate and compare the dose-dependent effect of delayed physical rehabilitation (daily 5 h or overnight voluntary wheel running; initiated on post-stroke day 7 and continuing through day 21) on recovery of motor function in the mouse photothrombotic model of ischemic stroke and correlate it with angiogenic potential of the brain. Our observations indicate that overnight but not 5 h access to running wheels facilitates recovery of motor function in mice in grid-walking test. Western blotting and immunofluorescence microscopy experiments evaluating the expression of angiogenesis-associated proteins VEGFR2, doppel and PDGFRβ in the peri-infarct and corresponding contralateral motor cortices indicate substantial upregulation of these proteins (≥2-fold) in the infarct core and surrounding cerebral cortex in the overnight running mice on post-stroke day 21. These findings indicate that there is a dose-dependent relationship between the extent of voluntary exercise, motor recovery and expression of angiogenesis-associated proteins in this expert-recommended mouse ischemic stroke model. Notably, our observations also point out to enhanced angiogenesis and presence of pericytes within the infarct core region during the chronic phase of stroke, suggesting a potential contribution of this tissue area in the mechanisms governing post-stroke functional recovery.
Collapse
|
66
|
Chen Z, Duan Y, Wang H, Tang H, Wang S, Wang X, Liu J, Fang X, Ouyang K. Atypical protein kinase C is essential for embryonic vascular development in mice. Genesis 2021; 59:e23412. [PMID: 33547760 DOI: 10.1002/dvg.23412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 11/10/2022]
Abstract
The atypical PKC (aPKC) subfamily constitutes PKCζ and PKCλ in mice, and both aPKC isoforms have been proposed to be involved in regulating various endothelial cell (EC) functions. However, the physiological function of aPKC in ECs during embryonic development has not been well understood. To address this question, we utilized Tie2-Cre to delete PKCλ alone (PKCλ-SKO) or both PKCλ and PKCζ (DKO) in ECs, and found that all DKO mice died at around the embryonic day 11.5 (E11.5), whereas a small proportion of PKCλ-SKO mice survived till birth. PKCλ-SKO embryos also exhibited less phenotypic severity than DKO embryos at E10.5 and E11.5, suggesting a potential compensatory role of PKCζ for PKCλ in embryonic ECs. We then focused on DKO embryos and investigated the effects of aPKC deficiency on embryonic vascular development. At E9.5, deletion of both aPKC isoforms reduced the diameters of vitelline artery and vein, and decreased branching from both vitelline vessels in yolk sac. Ablation of both aPKC isoforms also disrupted embryonic angiogenesis in head and trunk at the same stage, increasing apoptosis of both ECs and non-ECs. Taken together, our results demonstrated that aPKC in ECs plays an essential role in regulating cell apoptosis, angiogenesis, and embryonic survival.
Collapse
Affiliation(s)
- Zee Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yaoyun Duan
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Hong Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Huayuan Tang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Shijia Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xinru Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Xi Fang
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
67
|
Li Y, Tang Y, Yang GY. Therapeutic application of exosomes in ischaemic stroke. Stroke Vasc Neurol 2021; 6:483-495. [PMID: 33431513 PMCID: PMC8485240 DOI: 10.1136/svn-2020-000419] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Ischaemic stroke is a leading cause of long-term disability in the world, with limited effective treatments. Increasing evidence demonstrates that exosomes are involved in ischaemic pathology and exhibit restorative therapeutic effects by mediating cell–cell communication. The potential of exosome therapy for ischaemic stroke has been actively investigated in the past decade. In this review, we mainly discuss the current knowledge of therapeutic applications of exosomes from different cell types, different exosomal administration routes, and current advances of exosome tracking and targeting in ischaemic stroke. We also briefly summarised the pathology of ischaemic stroke, exosome biogenesis, exosome profile changes after stroke as well as registered clinical trials of exosome-based therapy.
Collapse
Affiliation(s)
- Yongfang Li
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China .,Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| |
Collapse
|
68
|
Treatment with Atorvastatin During Vascular Remodeling Promotes Pericyte-Mediated Blood-Brain Barrier Maturation Following Ischemic Stroke. Transl Stroke Res 2021; 12:905-922. [PMID: 33423214 DOI: 10.1007/s12975-020-00883-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
We previously showed that newly formed vessels in ischemic rat brain have high blood-brain barrier (BBB) permeability at 3 weeks after stroke due to a lack of major endothelial tight junction proteins (TJPs), which may exacerbate edema in stroke patients. Atorvastatin was suggested a dose-dependent pro-angiogenic effect and ameliorating BBB permeability beyond its cholesterol-lowering effects. This study examined our hypothesis that, during vascular remodeling after stroke, treatment with atorvastatin could facilitate BBB maturation in remodeling vasculature in ischemic brain. Adult spontaneously hypertensive rats underwent middle cerebral artery occlusion with reperfusion (MCAO/RP). Atorvastatin, at dose of 3 mg/kg, was delivered daily starting at 14 days after MCAO/RP onset for 7 days. The rats were studied at multiple time points up to 8 weeks with multimodal-MRI, behavior tests, immunohistochemistry, and biochemistry. The delayed treatment of atorvastatin significantly reduced infarct size and BBB permeability, restored cerebral blood flow, and improved the neurological outcome at 8 weeks after MCAO/RP. Postmortem studies showed that atorvastatin promoted angiogenesis and stabilized the newly formed vessels in peri-infarct areas. Importantly, atorvastatin facilitated maturation of BBB properties in the new vessels by promoting endothelial tight junction (TJ) formation. Further in vivo and in vitro studies demonstrated that proliferating peri-vascular pericytes expressing neural-glial antigen 2 (NG2) mediated the role of atorvastatin on BBB maturation through regulating endothelial TJ strand formations. Our results suggested a therapeutic potential of atorvastatin in facilitating a full BBB integrity and functional stroke recovery, and an essential role for pericyte-mediated endothelial TJ formation in remodeling vasculature.
Collapse
|
69
|
Nemchek V, Haan EM, Mavros R, Macuiba A, Kerr AL. Voluntary exercise ameliorates the good limb training effect in a mouse model of stroke. Exp Brain Res 2021; 239:687-697. [PMID: 33388904 DOI: 10.1007/s00221-020-05994-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
Stroke is the leading cause of long-term disability in the United States, making research on rehabilitation imperative. Stroke rehabilitation typically focuses on recovery of the impaired limb, although this process is tedious. Compensatory use of the intact limb after stroke is more efficient, but it is known to negatively impact the impaired limb. Exercise may help with this problem; research has shown that exercise promotes neuronal growth and prevents cell death. This study used a mouse model to investigate if post-stroke exercise could prevent deterioration of the function of the impaired limb despite compensatory training of the intact limb. Results showed that mice that exercised, in combination with intact limb training, demonstrated improved functional outcome compared to mice that received no training or compensatory limb training only. These findings suggest that exercise can prevent the deterioration of impaired limb functional outcome that is typically seen with intact limb use.
Collapse
Affiliation(s)
- Victoria Nemchek
- Neuroscience Program, Illinois Wesleyan University, Bloomington, IL, USA
| | - Emma M Haan
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Rachel Mavros
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Amanda Macuiba
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Abigail L Kerr
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA.
- Neuroscience Program, Illinois Wesleyan University, Bloomington, IL, USA.
| |
Collapse
|
70
|
Xu X, Zhu L, Xue K, Liu J, Wang J, Wang G, Gu J, Zhang Y, Li X. Ultrastructural studies of the neurovascular unit reveal enhanced endothelial transcytosis in hyperglycemia‐enhanced hemorrhagic transformation after stroke. CNS Neurosci Ther 2021. [PMCID: PMC7804894 DOI: 10.1111/cns.13571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aims Pre‐existing hyperglycemia (HG) aggravates the breakdown of blood–brain barrier (BBB) and increases the risk of hemorrhagic transformation (HT) after acute ischemic stroke in both animal models and patients. To date, HG‐induced ultrastructural changes of brain microvascular endothelial cells (BMECs) and the mechanisms underlying HG‐enhanced HT after ischemic stroke are poorly understood. Methods We used a mouse model of mild brain ischemia/reperfusion to investigate HG‐induced ultrastructural changes of BMECs that contribute to the impairment of BBB integrity after stroke. Adult male mice received systemic glucose administration 15 min before middle cerebral artery occlusion (MCAO) for 20 min. Ultrastructural characteristics of BMECs were evaluated using two‐dimensional and three‐dimensional electron microscopy and quantitatively analyzed. Results Mice with acute HG had exacerbated BBB disruption and larger brain infarcts compared to mice with normoglycemia (NG) after MCAO and 4 h of reperfusion, as assessed by brain extravasation of the Evans blue dye and microtubule‐associated protein 2 immunostaining. Electron microscopy further revealed that HG mice had more endothelial vesicles in the striatal neurovascular unit than NG mice, which may account for their deterioration of BBB impairment. In contrast with enhanced endothelial transcytosis, paracellular tight junction ultrastructure was not disrupted after this mild ischemia/reperfusion insult or altered upon HG. Consistent with the observed increase of endothelial vesicles, transcytosis‐related proteins caveolin‐1, clathrin, and hypoxia‐inducible factor (HIF)‐1α were upregulated by HG after MCAO and reperfusion. Conclusion Our study provides solid structural evidence to understand the role of endothelial transcytosis in HG‐elicited BBB hyperpermeability. Enhanced transcytosis occurs prior to the physical breakdown of BMECs and is a promising therapeutic target to preserve BBB integrity.
Collapse
Affiliation(s)
- Xiaomin Xu
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
- Qidong Women's and Children's Health Qidong China
| | - Liuqi Zhu
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Ke Xue
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Jiayi Liu
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Jian Wang
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Guohua Wang
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Jin‐hua Gu
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Yunfeng Zhang
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| | - Xia Li
- Institute of Special Environmental Medicine and Department of Neurology of Affiliated Hospital Co‐innovation Center of Neuroregeneration Nantong University Nantong China
| |
Collapse
|
71
|
Otsu Y, Namekawa M, Toriyabe M, Ninomiya I, Hatakeyama M, Uemura M, Onodera O, Shimohata T, Kanazawa M. Strategies to prevent hemorrhagic transformation after reperfusion therapies for acute ischemic stroke: A literature review. J Neurol Sci 2020; 419:117217. [PMID: 33161301 DOI: 10.1016/j.jns.2020.117217] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/09/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Reperfusion therapies by tissue plasminogen activator (tPA) and mechanical thrombectomy (MT) have ushered in a new era in the treatment of acute ischemic stroke (AIS). However, reperfusion therapy-related HT remains an enigma. AIM To provide a comprehensive review focused on emerging concepts of stroke and therapeutic strategies, including the use of protective agents to prevent HT after reperfusion therapies for AIS. METHODS A literature review was performed using PubMed and the ClinicalTrials.gov database. RESULTS Risk of HT increases with delayed initiation of tPA treatment, higher baseline glucose level, age, stroke severity, episode of transient ischemic attack within 7 days of stroke onset, and hypertension. At a molecular level, HT that develops after thrombolysis is thought to be caused by reactive oxygen species, inflammation, remodeling factor-mediated effects, and tPA toxicity. Modulation of these pathophysiological mechanisms could be a therapeutic strategy to prevent HT after tPA treatment. Clinical mechanisms underlying HT after MT are thought to involve smoking, a low Alberta Stroke Program Early CT Score, use of general anesthesia, unfavorable collaterals, and thromboembolic migration. However, the molecular mechanisms are yet to be fully investigated. Clinical trials with MT and protective agents have also been planned and good outcomes are expected. CONCLUSION To fully utilize the easily accessible drug-tPA-and the high recanalization rate of MT, it is important to reduce bleeding complications after recanalization. A future study direction could be to investigate the recovery of neurological function by combining reperfusion therapies with cell therapies and/or use of pleiotropic protective agents.
Collapse
Affiliation(s)
- Yutaka Otsu
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Namekawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masafumi Toriyabe
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan; Department of Medical Technology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
| | - Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiro Uemura
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
72
|
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: 2.0] [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
|
73
|
Gaire BP, Sapkota A, Choi JW. BMS-986020, a Specific LPA 1 Antagonist, Provides Neuroprotection against Ischemic Stroke in Mice. Antioxidants (Basel) 2020; 9:antiox9111097. [PMID: 33171697 PMCID: PMC7695306 DOI: 10.3390/antiox9111097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/29/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022] Open
Abstract
Stroke is a leading cause of death. Stroke survivors often suffer from long-term functional disability. This study demonstrated neuroprotective effects of BMS-986020 (BMS), a selective lysophosphatidic acid receptor 1 (LPA1) antagonist under clinical trials for lung fibrosis and psoriasis, against both acute and sub-acute injuries after ischemic stroke by employing a mouse model with transient middle cerebral artery occlusion (tMCAO). BMS administration immediately after reperfusion significantly attenuated acute brain injuries including brain infarction, neurological deficits, and cell apoptosis at day 1 after tMCAO. Neuroprotective effects of BMS were preserved even when administered at 3 h after reperfusion. Neuroprotection by BMS against acute injuries was associated with attenuation of microglial activation and lipid peroxidation in post-ischemic brains. Notably, repeated BMS administration daily for 14 days after tMCAO exerted long-term neuroprotection in tMCAO-challenged mice, as evidenced by significantly attenuated neurological deficits and improved survival rate. It also attenuated brain tissue loss and cell apoptosis in post-ischemic brains. Mechanistically, it significantly enhanced neurogenesis and angiogenesis in injured brains. A single administration of BMS provided similar long-term neuroprotection except survival rate. Collectively, BMS provided neuroprotection against both acute and sub-acute injuries of ischemic stroke, indicating that BMS might be an appealing therapeutic agent to treat ischemic stroke.
Collapse
|
74
|
Zhou F, Liu F, Liu J, He YL, Zhou QM, Guo L, Peng C, Xiong L. Stachydrine promotes angiogenesis by regulating the VEGFR2/MEK/ERK and mitochondrial-mediated apoptosis signaling pathways in human umbilical vein endothelial cells. Biomed Pharmacother 2020; 131:110724. [DOI: 10.1016/j.biopha.2020.110724] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
|
75
|
Zhang L, Wang H. FTY720 in CNS injuries: Molecular mechanisms and therapeutic potential. Brain Res Bull 2020; 164:75-82. [DOI: 10.1016/j.brainresbull.2020.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/23/2020] [Accepted: 08/15/2020] [Indexed: 12/25/2022]
|
76
|
Cell Therapies under Clinical Trials and Polarized Cell Therapies in Pre-Clinical Studies to Treat Ischemic Stroke and Neurological Diseases: A Literature Review. Int J Mol Sci 2020; 21:ijms21176194. [PMID: 32867222 PMCID: PMC7503631 DOI: 10.3390/ijms21176194] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke remains a major cause of serious disability because the brain has a limited capacity to regenerate. In the last two decades, therapies for stroke have dramatically changed. However, half of the patients cannot achieve functional independence after treatment. Presently, cell-based therapies are being investigated to improve functional outcomes. This review aims to describe conventional cell therapies under clinical trial and outline the novel concept of polarized cell therapies based on protective cell phenotypes, which are currently in pre-clinical studies, to facilitate functional recovery after post-reperfusion treatment in patients with ischemic stroke. In particular, non-neuronal stem cells, such as bone marrow-derived mesenchymal stem/stromal cells and mononuclear cells, confer no risk of tumorigenesis and are safe because they do not induce rejection and allergy; they also pose no ethical issues. Therefore, recent studies have focused on them as a cell source for cell therapies. Some clinical trials have shown beneficial therapeutic effects of bone marrow-derived cells in this regard, whereas others have shown no such effects. Therefore, more clinical trials must be performed to reach a conclusion. Polarized microglia or peripheral blood mononuclear cells might provide promising therapeutic strategies after stroke because they have pleiotropic effects. In traumatic injuries and neurodegenerative diseases, astrocytes, neutrophils, and T cells were polarized to the protective phenotype in pre-clinical studies. As such, they might be useful therapeutic targets. Polarized cell therapies are gaining attention in the treatment of stroke and neurological diseases.
Collapse
|
77
|
Chen L, Fu C, Zhang Q, He C, Zhang F, Wei Q. The role of CD44 in pathological angiogenesis. FASEB J 2020; 34:13125-13139. [PMID: 32830349 DOI: 10.1096/fj.202000380rr] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is required for normal development and occurs as a pathological step in a variety of disease settings, such as cancer, ocular diseases, and ischemia. Recent studies have revealed the role of CD44, a widely expressed cell surface adhesion molecule, in promoting pathological angiogenesis and the development of its associated diseases through its regulation of diverse function of endothelial cells, such as proliferation, migration, adhesion, invasion, and communication with the microenvironment. Conversely, the absence of CD44 expression or inhibition of its function impairs pathological angiogenesis and disease progression. Here, we summarize the current understanding of the roles of CD44 in pathological angiogenesis and the underlying cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Li Chen
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
78
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
79
|
PPAR- γ Mediates Ta-VNS-Induced Angiogenesis and Subsequent Functional Recovery after Experimental Stroke in Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8163789. [PMID: 32775443 PMCID: PMC7396041 DOI: 10.1155/2020/8163789] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 02/05/2023]
Abstract
Background Neoangiogenesis after cerebral ischemia in mammals is insufficient to restore neurological function, illustrating the need to design better strategies for improving outcomes. Our previous study has suggested that transcutaneous auricular vagus nerve stimulation (ta-VNS) induced angiogenesis and improved neurological functions in a rat model of cerebral ischemia/reperfusion (I/R) injury. However, the mechanisms involved need further exploration. Peroxisome proliferator-activated receptor-γ (PPAR-γ), well known as a ligand-modulated nuclear transcription factor, plays a crucial role in the regulation of cerebrovascular structure and function. Hence, the present study was designed to explore the role of PPAR-γ in ta-VNS-mediated angiogenesis and uncover the possible molecular mechanisms against ischemic stroke. Methods Adult male Sprague-Dawley rats were transfected with either PPAR-γ small interfering RNA (siRNA) or lentiviral vector without siRNA prior to surgery and subsequently received ta-VNS treatment. The expression and localization of PPAR-γ in the ischemic boundary after ta-VNS treatment were examined. Subsequently, neurological deficit scores, neuronal damage, and infarct volume were all evaluated. Additionally, microvessel density, endothelial cell proliferation condition, and the expression of angiogenesis-related molecules in the peri-infarct cortex were measured. Results We found that the expression of PPAR-γ in the peri-infarct cortex increased at 14 d and reached normal levels at 28 d after reperfusion. Ta-VNS treatment further upregulated PPAR-γ expression in the ischemic cortex. PPAR-γ was mainly expressed in neurons and astrocytes. Furthermore, ta-VNS-treated I/R rats showed better neurobehavioral recovery, alleviated neuronal injury, reduced infarct volume, and increased angiogenesis, as indicated by the elevated levels of brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), and phosphorylated endothelial nitric oxide synthase (P-eNOS). Surprisingly, the beneficial effects of ta-VNS were weakened after PPAR-γ silencing. Conclusions Our results suggest that PPAR-γ is a potential mediator of ta-VNS-induced angiogenesis and neuroprotection against cerebral I/R injury.
Collapse
|
80
|
Puig B, Yang D, Brenna S, Altmeppen HC, Magnus T. Show Me Your Friends and I Tell You Who You Are: The Many Facets of Prion Protein in Stroke. Cells 2020; 9:E1609. [PMID: 32630841 PMCID: PMC7407975 DOI: 10.3390/cells9071609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke belongs to the leading causes of mortality and disability worldwide. Although treatments for the acute phase of stroke are available, not all patients are eligible. There is a need to search for therapeutic options to promote neurological recovery after stroke. The cellular prion protein (PrPC) has been consistently linked to a neuroprotective role after ischemic damage: it is upregulated in the penumbra area following stroke in humans, and animal models of stroke have shown that lack of PrPC aggravates the ischemic damage and lessens the functional outcome. Mechanistically, these effects can be linked to numerous functions attributed to PrPC: (1) as a signaling partner of the PI3K/Akt and MAPK pathways, (2) as a regulator of glutamate receptors, and (3) promoting stem cell homing mechanisms, leading to angio- and neurogenesis. PrPC can be cleaved at different sites and the proteolytic fragments can account for the manifold functions. Moreover, PrPC is present on extracellular vesicles (EVs), released membrane particles originating from all types of cells that have drawn attention as potential therapeutic tools in stroke and many other diseases. Thus, identification of the many mechanisms underlying PrPC-induced neuroprotection will not only provide further understanding of the physiological functions of PrPC but also new ideas for possible treatment options after ischemic stroke.
Collapse
Affiliation(s)
- Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Denise Yang
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | | | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| |
Collapse
|
81
|
Ren C, Li N, Gao C, Zhang W, Yang Y, Li S, Ji X, Ding Y. Ligustilide provides neuroprotection by promoting angiogenesis after cerebral ischemia. Neurol Res 2020; 42:683-692. [PMID: 32584207 DOI: 10.1080/01616412.2020.1782122] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Changhong Ren
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Ning Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Chen Gao
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Wei Zhang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Yang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Sijie Li
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
| | - Xunming Ji
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
82
|
Abstract
Stroke remains a major cause of serious disability due to the brain's limited capacity to regenerate. Current treatments focus on acute recanalization of the occluded blood vessels; however, currently there are no approved therapy options to regenerate neural circuits and reduce stroke-related disability. To promote recovery, therapeutic angiogenesis has been proposed as a promising target. Although restoration of blood vessels providing oxygen and nutrients to the peri-infarct regions may be beneficial, newly generated capillaries may also carry pathophysiological risk factors that need to be considered. One major concern are adverse effects including edema formation and haemorrhagic transformation due to the comprised endothelial barrier function during vascular remodelling. This brief opinion article will discuss the challenges and the newest advancements of angiogenesis as a therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
83
|
Zhang Y, Zhao Y, Song X, Luo H, Sun J, Han C, Gu X, Li J, Cai G, Zhu Y, Liu Z, Wei L, Wei ZZ. Modulation of Stem Cells as Therapeutics for Severe Mental Disorders and Cognitive Impairments. Front Psychiatry 2020; 11:80. [PMID: 32425815 PMCID: PMC7205035 DOI: 10.3389/fpsyt.2020.00080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Severe mental illnesses (SMI) such as schizophrenia and bipolar disorder affect 2-4% of the world population. Current medications and diagnostic methods for mental illnesses are not satisfying. In animal studies, stem cell therapy is promising for some neuropsychiatric disorders and cognitive/social deficits, not only treating during development (targeting modulation and balancing) but also following neurodegeneration (cell replacement and regenerating support). We believe that novel interventions such as modulation of particular cell populations to develop cell-based treatment can improve cognitive and social functions in SMI. With pathological synaptic/myelin damage, oligodendrocytes seem to play a role. In this review, we have summarized oligodendrogenesis mechanisms and some related calcium signals in neural cells and stem/progenitor cells. The related benefits from endogenous stem/progenitor cells within the brain and exogenous stem cells, including multipotent mesenchymal-derived stromal cells (MSC), fetal neural stem cells (NSC), pluripotent stem cells (PSC), and differentiated progenitors, are discussed. These also include stimulating mechanisms of oligodendrocyte proliferation, maturation, and myelination, responsive to the regenerative effects by both endogenous stem cells and transplanted cells. Among the mechanisms, calcium signaling regulates the neuronal/glial progenitor cell (NPC/GPC)/oligodendrocyte precursor cell (OPC) proliferation, migration, and differentiation, dendrite development, and synaptic plasticity, which are involved in many neuropsychiatric diseases in human. On the basis of numerous protein annotation and protein-protein interaction databases, a total of 119 calcium-dependent/activated proteins that are related to neuropsychiatry in human are summarized in this investigation. One of the advanced methods, the calcium/cation-channel-optogenetics-based stimulation of stem cells and transplanted cells, can take advantage of calcium signaling regulations. Intranasal-to-brain delivery of drugs and stem cells or local delivery with the guidance of brain imaging techniques may provide a unique new approach for treating psychiatric disorders. It is also expected that preconditioning stem cell therapy following precise brain imaging as pathological confirmation has high potential if translated to cell clinic use. Generally, modulable cell transplantation followed by stimulations should provide paracrine protection, synaptic modulation, and myelin repair for the brain in SMI.
Collapse
Affiliation(s)
- Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingying Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Xiaopeng Song
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Hua Luo
- Emory Critical Care Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Jinmei Sun
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Chunyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jun Li
- Department of Biological Psychiatry, Peking University Sixth Hospital, Beijing, China
- Department of Biological Psychiatry, Peking University Institute of Mental Health, Beijing, China
- Department of Biological Psychiatry, NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Department of Biological Psychiatry, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Guilan Cai
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanbing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhandong Liu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
84
|
Zou Y, Hu J, Huang W, Ye S, Han F, Du J, Shao M, Guo R, Lin J, Zhao Y, Xiong Y, Wang X. Non-Mitogenic Fibroblast Growth Factor 1 Enhanced Angiogenesis Following Ischemic Stroke by Regulating the Sphingosine-1-Phosphate 1 Pathway. Front Pharmacol 2020; 11:59. [PMID: 32194396 PMCID: PMC7063943 DOI: 10.3389/fphar.2020.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes account for about 80% of all strokes and are associated with a high risk of mortality. Angiogenesis of brain microvascular endothelial cells may contribute to functional restoration following ischemia. Fibroblast growth factor 1 (FGF1), a member of FGF superfamily, involved in embryonic development, angiogenesis, wound healing, and neuron survival. However, the mitogenic activity of FGF1 is known to contribute to several human pathologies, thereby questioning the safety of its clinical applications. Here, we explored the effects and mechanism of action of non-mitogenic FGF1 (nmFGF1) on angiogenesis in mice after ischemia stroke and an oxygen-glucose deprivation (OGD)-induced human brain microvascular endothelial cells (HBMECs) injury model. We found that intranasal administration nmFGF1 significantly promoted angiogenesis in mice after stroke, and significantly increased the formation of matrigel tube and promoted scratch migration in a dose-dependent manner in OGD-induced HBMECs in vitro. However, the co-administration of an FGF receptor 1 (FGFR1)-specific inhibitor PD173074 significantly reversed the effects of nmFGF1 in vitro, suggesting that nmFGF1 functions via FGFR1 activation. Moreover, nmFGF1 activated sphingosine-1-phosphate receptor 1 (S1PR1, S1P1) in mice after stroke in vivo. S1P1 protein antagonist VPC23019 and agonist FTY720 were used to confirm that nmFGF1 promotes angiogenesis in vitro partially through the S1P1 pathway. OGD induced downregulation of S1P1 expression. The S1P1 antagonist VPC23019 blocked the stimulatory effects of nmFGF1, whereas the S1P1 agonist FTY720 exerted effects comparable with those of nmFGF1. Furthermore, PD173074 reversed the effect of nmFGF1 on upregulating S1P1 signaling. In conclusion, nmFGF1 enhanced angiogenesis in mice following stroke and OGD-induced HBMECs through S1P1 pathway regulation mediated via FGFR1 activation. This new discovery suggests the potential therapeutic role of nmFGF1 for the treatment of ischemic strokes.
Collapse
Affiliation(s)
- Yuchi Zou
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mingjie Shao
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, China
| | - Ye Xiong
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
85
|
Chabriat H, Bassetti CL, Marx U, Audoli-Inthavong ML, Sors A, Lambert E, Wattez M, Hermann DM, ALTHAUS K, AMARO S, BAE HJ, BAK Z, BARBARINI L, BASSI P, BAZAN R, BERECZKI D, BERKOWICZ T, BERROUSCHOT J, BLACQUIERE D, BROLA W, BUTCHER K, CARDONA P, CHA JK, CLOUD G, COHEN D, CORDONNIER C, CSANYI A, CZLONKOWSKA A, DAVIS S, DAWSON J, DE KLIPPEL N, DENIER C, DESFONTAINES P, DIENER HC, DIOSZEGHY P, DIPPEL DW, DORADO L, FOLYOVICH A, FREITAS GR, FRIEDRICH MA, FRYZE W, GAGLIARDI RJ, GOTTSCHAL M, GRIMLEY R, GROND M, GRÖSCHEL K, HOSSEINI H, HWANG Y, KALLMUENZER B, KHAN U, KIM JS, KLEINIG T, KOVES A, LAGO MARTIN A, LASEK-BAL A, LEMBO G, LEMMENS R, LINDERT R, PORCELLO MARRONE LC, MARTINEZ ZABALETA M, MAS JL, MASJUAN VALLEJO J, MAZIGHI M, MINELLI C, MISTRI A, MOLINA C, MONICHE ALVAREZ F, CABRAL MORO CH, MULLENERS W, NABAVI D, NEAU JP, O'BRIEN B, OVARY C, PANCZEL G, PARK MS, PHAN T, RAGAB S, REJDAK K, RODRIGUEZ DE FREITAS G, ROFFE C, ROQUER GONZALEZ J, ROVER L, SAMPAIO SILVA G, SCHELLINGER P, SEGURA MARTIN T, SHAW L, SIBON I, SKODA O, SMADJA D, SOBOLEWSKI P, SODA H, SPRIGG N, SWIAT M, SZAPARY L, SZEGEDI N, TONI D, VALIKOVICS A, VANHOOREN G, VECSEI L, WEIN T, WONG A, XIMENEZ CARRILLO A. Safety and efficacy of GABAA α5 antagonist S44819 in patients with ischaemic stroke: a multicentre, double-blind, randomised, placebo-controlled trial. Lancet Neurol 2020; 19:226-233. [DOI: 10.1016/s1474-4422(20)30004-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/16/2019] [Accepted: 01/08/2020] [Indexed: 10/25/2022]
|
86
|
Neuroprotective effect of salvianolate on cerebral ischaemia-reperfusion injury in rats by inhibiting the Caspase-3 signal pathway. Eur J Pharmacol 2020; 872:172944. [PMID: 31978424 DOI: 10.1016/j.ejphar.2020.172944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
Salvianolate has been widely used for the treatment of cerebrovascular diseases. However, the detailed molecular mechanism of how it alleviates cerebral ischaemia-reperfusion injury is not well understood. In the present study, we investigated the neuroprotective effects of salvianolate in acute cerebral infarction using the PC12 cell oxygen-glucose deprivation (OGD) model in vitro and the rat transient middle cerebral artery occlusion (MCAO) model in vivo. The results showed that the salvianolate significantly reduced the level of reactive oxygen species and inhibited the Caspase-3 signalling pathway in vitro; at the same time, in vivo experiments showed that salvianolate obviously reduced the infarct area (12.9%) and repaired cognitive function compared with the model group (28.28%). In conclusion, our data demonstrated that the salvianolate effectively alleviated cerebral ischaemia-reperfusion injury via suppressing the Caspase-3 signalling pathway.
Collapse
|
87
|
Abstract
Increased microvessel density in the peri-infarct region has been reported and has been correlated with longer survival times in ischemic stroke patients and has improved outcomes in ischemic animal models. This raises the possibility that enhancement of angiogenesis is one of the strategies to facilitate functional recovery after ischemic stroke. Blood vessels and neuronal cells communicate with each other using various mediators and contribute to the pathophysiology of cerebral ischemia as a unit. In this mini-review, we discuss how angiogenesis might couple with axonal outgrowth/neurogenesis and work for functional recovery after cerebral ischemia. Angiogenesis occurs within 4 to 7 days after cerebral ischemia in the border of the ischemic core and periphery. Post-ischemic angiogenesis may contribute to neuronal remodeling in at least two ways and is thought to contribute to functional recovery. First, new blood vessels that are formed after ischemia are thought to have a role in the guidance of sprouting axons by vascular endothelial growth factor and laminin/β1-integrin signaling. Second, blood vessels are thought to enhance neurogenesis in three stages: 1) Blood vessels enhance proliferation of neural stem/progenitor cells by expression of several extracellular signals, 2) microvessels support the migration of neural stem/progenitor cells toward the peri-infarct region by supplying oxygen, nutrients, and soluble factors as well as serving as a scaffold for migration, and 3) oxygenation induced by angiogenesis in the ischemic core is thought to facilitate the differentiation of migrated neural stem/progenitor cells into mature neurons. Thus, the regions of angiogenesis and surrounding tissue may be coupled, representing novel treatment targets.
Collapse
Affiliation(s)
- Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
88
|
Hatakeyama M, Kanazawa M, Ninomiya I, Omae K, Kimura Y, Takahashi T, Onodera O, Fukushima M, Shimohata T. A novel therapeutic approach using peripheral blood mononuclear cells preconditioned by oxygen-glucose deprivation. Sci Rep 2019; 9:16819. [PMID: 31728010 PMCID: PMC6856386 DOI: 10.1038/s41598-019-53418-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Cell therapies that invoke pleiotropic mechanisms may facilitate functional recovery in patients with stroke. Based on previous experiments using microglia preconditioned by oxygen-glucose deprivation, we hypothesized that the administration of peripheral blood mononuclear cells (PBMCs) preconditioned by oxygen-glucose deprivation (OGD-PBMCs) to be a therapeutic strategy for ischemic stroke. Here, OGD-PBMCs were identified to secrete remodelling factors, including the vascular endothelial growth factor and transforming growth factor-β in vitro, while intra-arterial administration of OGD-PBMCs at 7 days after focal cerebral ischemia prompted expression of such factors in the brain parenchyma at 28 days following focal cerebral ischemia in vivo. Furthermore, administration of OGD-PBMCs induced an increasing number of stage-specific embryonic antigen-3-positive cells both in vitro and in vivo. Finally, it was found to prompt angiogenesis and axonal outgrowth, and functional recovery after cerebral ischemia. In conclusion, the administration of OGD-PBMCs might be a novel therapeutic strategy against ischemic stroke.
Collapse
Affiliation(s)
- Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata, 951-8585, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata, 951-8585, Japan.
| | - Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata, 951-8585, Japan
| | - Kaoru Omae
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Kobe, 650-0047, Japan
| | - Yasuko Kimura
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Kobe, 650-0047, Japan
| | - Tetsuya Takahashi
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata, 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata, 951-8585, Japan
| | - Masanori Fukushima
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Kobe, 650-0047, Japan
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| |
Collapse
|
89
|
Kanazawa M, Takahashi T, Kawamura K, Shimohata T. [VEGF-A therapeutic target against hemorrhagic transformation after t-PA treatment]. Rinsho Shinkeigaku 2019; 59:699-706. [PMID: 31656268 DOI: 10.5692/clinicalneurol.cn-001346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tissue plasminogen activator (t-PA) treatment is beneficial for patients with ischemic stroke within 4.5 h of stroke onset, because the risk of intracerebral hemorrhagic transformation (HT) increases with delayed t-PA treatment. The benefits of t-PA thrombolysis are heavily dependent on time to treatment. Development of vasoprotective drugs that attenuate HT after delayed t-PA treatment might improve the prognosis of stroke patients and extend the therapeutic time window of t-PA and endovascular thrombolysis. An angiogenic factor, vascular endothelial growth factor (VEGF), might be associated with the blood-brain barrier (BBB) disruption after focal cerebral ischemia. By using a rat thromboembolic model, delayed t-PA treatment at 4 h after ischemia promoted expression of VEGF in BBB, matrix metalloproteinase-9 (MMP-9) activation, degradation of BBB components, and HT. We demonstrated that HT was inhibited by intravenous administration of an anti-VEGF neutralizing antibody/VEGF receptor antagonist. In addition, for clinical application, reverse translation studies, a path from bedside to bench, are necessary.
Collapse
Affiliation(s)
- Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University
| | - Tetsuya Takahashi
- Department of Neurology, National Hospital Organization Nishiniigata Chuo Hospital
| | | | | |
Collapse
|
90
|
Ginsenoside Rg1 promotes cerebral angiogenesis via the PI3K/Akt/mTOR signaling pathway in ischemic mice. Eur J Pharmacol 2019; 856:172418. [DOI: 10.1016/j.ejphar.2019.172418] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022]
|