51
|
Correlation of Plasma Vascular Endothelial Growth Factor and Endostatin Levels with Symptomatic Intra- and Extracranial Atherosclerotic Stenosis in a Chinese Han Population. J Stroke Cerebrovasc Dis 2017; 26:1061-1070. [PMID: 28189572 DOI: 10.1016/j.jstrokecerebrovasdis.2016.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/16/2016] [Accepted: 12/24/2016] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Symptomatic intracranial atherosclerotic stenosis (ICAS) and extracranial atherosclerotic stenosis (ECAS) are different in many aspects. Here, we explored the association between the location or severity of atherosclerotic stenosis and pro- or antiangiogenic factors, specifically vascular endothelial growth factor (VEGF) and endostatin (ES). METHODS We evaluated 198 consecutive patients with acute ischemia stroke: 132 with large-artery atherosclerosis (LAA) and 66 with small-artery occlusion (small-vessel occlusion). The LAA group was subclassified into 102 patients with ICAS and 30 with ECAS. Independent associations of VEGF, ES levels, and VEGF/ES ratio with the location of cerebral stenosis and the severity or short-term prognosis (14th day modified Rankin Scale) of ICAS were evaluated. RESULTS Plasma concentrations of VEGF and ES were lower (P < .05) in ICAS (38.07, 32.76-46.28 pg/mL and 58.95, 55.04-59.77 ng/mL) than those in ECAS (45.00, 34.30-83.34 pg/mL and 140.74, 85.63-231.21 ng/mL). Logistic regression analysis showed that VEGF concentrations and dyslipidemia were independently associated with ICAS, with odds ratios of .987 [95% CI = (.976, .998)] and .265 [95% CI = (.103, .792)], respectively. Moreover, plasmatic VEGF levels increased gradually along with the severity of ICAS (P = .003), and lower levels of ES (P = .040) or a higher VEGF/ES ratio (P = .048) were related to unfavorable short-term prognosis of ICAS. CONCLUSION Lower VEGF levels are associated with the presence of symptomatic ICAS, but not with ECAS. Furthermore, the severity of ICAS is positively correlated with the levels of VEGF, and lower ES levels or a predominance of VEGF over ES are predictors of poor short-term prognosis of ICAS.
Collapse
|
52
|
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine 2016; 11:5381-5414. [PMID: 27799765 PMCID: PMC5077137 DOI: 10.2147/ijn.s117210] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review summarizes articles that have been reported in literature on liposome-based strategies for effective drug delivery across the blood–brain barrier. Due to their unique physicochemical characteristics, liposomes have been widely investigated for their application in drug delivery and in vivo bioimaging for the treatment and/or diagnosis of neurological diseases, such as Alzheimer’s, Parkinson’s, stroke, and glioma. Several strategies have been used to deliver drug and/or imaging agents to the brain. Covalent ligation of such macromolecules as peptides, antibodies, and RNA aptamers is an effective method for receptor-targeting liposomes, which allows their blood–brain barrier penetration and/or the delivery of their therapeutic molecule specifically to the disease site. Additionally, methods have been employed for the development of liposomes that can respond to external stimuli. It can be concluded that the development of liposomes for brain delivery is still in its infancy, although these systems have the potential to revolutionize the ways in which medicine is administered.
Collapse
Affiliation(s)
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
53
|
Jiang Y, Li L, Ma J, Zhang L, Niu F, Feng T, Li C. Auricular vagus nerve stimulation promotes functional recovery and enhances the post-ischemic angiogenic response in an ischemia/reperfusion rat model. Neurochem Int 2016; 97:73-82. [DOI: 10.1016/j.neuint.2016.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 12/22/2022]
|
54
|
Paeoniflorin Promotes Angiogenesis in A Vascular Insufficiency Model of Zebrafish in vivo and in Human Umbilical Vein Endothelial Cells in vitro. Chin J Integr Med 2016; 24:494-501. [DOI: 10.1007/s11655-016-2262-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
|
55
|
XU AILING, ZHENG GUANYI, WANG ZHIJIAN, CHEN XIAODONG, JIANG QIONG. Neuroprotective effects of Ilexonin A following transient focal cerebral ischemia in rats. Mol Med Rep 2016; 13:2957-66. [PMID: 26936330 PMCID: PMC4805093 DOI: 10.3892/mmr.2016.4921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 12/30/2015] [Indexed: 11/06/2022] Open
Abstract
Ilexonin A is a compound isolated from the root of a plant commonly used in traditional Chinese medicine. The aim of the present study was to investigate the possible protective mechanism of Ilexonin A in rats subjected to occlusion of the middle cerebral artery (MCAO). Transient focal cerebral ischemia was induced by 2 h of MCAO, followed by reperfusion. Ilexonin A at doses of 20, 40 and 80 mg/kg were administered via intraperitoneal injection immediately following ischemia/reperfusion. The expression levels of glial fibrillary acidic protein (GFAP), ionized calcium‑binding adapter molecule‑1 (Iba‑1), vascular endothelial growth factor (VEGF), fetal liver kinase‑1 (Flk‑1) and Nestin were examined using immunostaining and Western blot analysis of the peri‑infarct region following ischemia/reperfusion. Ilexonin A significantly decreased the infarct volume and improved neurological deficits in a dose‑dependent manner. The expression levels of VEGF, Flk‑1 and Nestin were significantly increased in the rats treated with Ilexonin A, compared with the rats administered with saline. Following treatment with Ilexonin A, a higher number of GFAP‑positive astrocytes were found in the Ilexonin A‑treated rats at 1, 3 and 7 days, compared with the rats exposed to ischemia only, however, there were fewer astrocytes at 14 days, compared with the ischemia group. Ilexonin A significantly decreased the protein expression of Iba‑1. The results of the present study suggested that the protective effects of Ilexonin A were associated with revascularization, neuronal regeneration, and the regulation of astrocyte and microglia cell activation.
Collapse
Affiliation(s)
- AI-LING XU
- Department of Traditional Chinese Medicine, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Neonatal Department, The People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350004, P.R. China
| | - GUAN-YI ZHENG
- Department of Traditional Chinese Medicine, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - ZHI-JIAN WANG
- Department of Traditional Chinese Medicine, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Neurology, Fuzhou Neuro-Psychiatric Hospital, Fuzhou, Fujian 350000, P.R. China
| | - XIAO-DONG CHEN
- Burns Institute of the Affliated Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - QIONG JIANG
- Burns Institute of the Affliated Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
56
|
Abeysinghe HCS, Phillips EL, Chin-Cheng H, Beart PM, Roulston CL. Modulating Astrocyte Transition after Stroke to Promote Brain Rescue and Functional Recovery: Emerging Targets Include Rho Kinase. Int J Mol Sci 2016; 17:288. [PMID: 26927079 PMCID: PMC4813152 DOI: 10.3390/ijms17030288] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/26/2016] [Accepted: 02/05/2016] [Indexed: 01/13/2023] Open
Abstract
Stroke is a common and serious condition, with few therapies. Whilst previous focus has been directed towards biochemical events within neurons, none have successfully prevented the progression of injury that occurs in the acute phase. New targeted treatments that promote recovery after stroke might be a better strategy and are desperately needed for the majority of stroke survivors. Cells comprising the neurovascular unit, including blood vessels and astrocytes, present an alternative target for supporting brain rescue and recovery in the late phase of stroke, since alteration in the unit also occurs in regions outside of the lesion. One of the major changes in the unit involves extensive morphological transition of astrocytes resulting in altered energy metabolism, decreased glutamate reuptake and recycling, and retraction of astrocyte end feed from both blood vessels and neurons. Whilst globally inhibiting transitional change in astrocytes after stroke is reported to result in further damage and functional loss, we discuss the available evidence to suggest that the transitional activation of astrocytes after stroke can be modulated for improved outcomes. In particular, we review the role of Rho-kinase (ROCK) in reactive gliosis and show that inhibiting ROCK after stroke results in reduced scar formation and improved functional recovery.
Collapse
Affiliation(s)
- Hima Charika S Abeysinghe
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
- Department of Surgery, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| | - Ellie L Phillips
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Heung Chin-Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Insitute, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Philip M Beart
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, Parkville, VIC 3010, Australia.
| | - Carli L Roulston
- Neurotrauma Research, Department of Medicine, St Vincent's Campus, University of Melbourne, Parkville, VIC 3065, Australia.
| |
Collapse
|
57
|
Mammoto T, Chen Z, Jiang A, Jiang E, Ingber DE, Mammoto A. Acceleration of Lung Regeneration by Platelet-Rich Plasma Extract through the Low-Density Lipoprotein Receptor-Related Protein 5-Tie2 Pathway. Am J Respir Cell Mol Biol 2016; 54:103-13. [PMID: 26091161 PMCID: PMC5455682 DOI: 10.1165/rcmb.2015-0045oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis, the growth of new blood vessels, plays a key role in organ development, homeostasis, and regeneration. The cooperation of multiple angiogenic factors, rather than a single factor, is required for physiological angiogenesis. Recently, we have reported that soluble platelet-rich plasma (PRP) extract, which contains abundant angiopoietin-1 and multiple other angiogenic factors, stimulates angiogenesis and maintains vascular integrity in vitro and in vivo. In this report, we have demonstrated that mouse PRP extract increases phosphorylation levels of the Wnt coreceptor low-density lipoprotein receptor-related protein 5 (LRP5) and thereby activates angiogenic factor receptor Tie2 in endothelial cells (ECs) and accelerates EC sprouting and lung epithelial cell budding in vitro. PRP extract also increases phosphorylation levels of Tie2 in the mouse lungs and accelerates compensatory lung growth and recovery of exercise capacity after unilateral pneumonectomy in mice, whereas soluble Tie2 receptor or Lrp5 knockdown attenuates the effects of PRP extract. Because human PRP extract is generated from autologous peripheral blood and can be stored at -80°C, our findings may lead to the development of novel therapeutic interventions for various angiogenesis-related lung diseases and to the improvement of strategies for lung regeneration.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhao Chen
- Department of Medicine, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Amanda Jiang
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Elisabeth Jiang
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Donald E. Ingber
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts; and
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts
| | - Akiko Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
58
|
Abstract
A multifunctional microRNA, miR-155, has been recently recognized as an important modulator of numerous biological processes. In our previous in vitro studies, miR-155 was identified as a potential regulator of the endothelial morphogenesis. The present study demonstrates that in vivo inhibition of miR-155 supports cerebral vasculature after experimental stroke. Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Microvasculature in peri-infarct area, infarct size, and animal functional recovery were assessed at 1, 2, and 3 weeks after dMCAO. Using in vivo two-photon microscopy, we detected improved blood flow and microvascular integrity in the peri-infarct area of miR-155 inhibitor-injected mice. Electron microscopy revealed that, in contrast to the control group, these animals demonstrated well preserved capillary tight junctions (TJs). Western blot analysis data indicate that improved TJ integrity in the inhibitor-injected animals could be associated with stabilization of the TJ protein ZO-1 and mediated by the miR-155 target protein Rheb. MRI analysis showed significant (34%) reduction of infarct size in miR-155 inhibitor-injected animals at 21 d after dMCAO. Reduced brain injury was confirmed by electron microscopy demonstrating decreased neuronal damage in the peri-infarct area of stroke. Preservation of brain tissue was reflected in efficient functional recovery of inhibitor-injected animals. Based on our findings, we propose that in vivo miR-155 inhibition after ischemia supports brain microvasculature, reduces brain tissue damage, and improves the animal functional recovery. Significance statement: In the present study, we investigated an effect of the in vivo inhibition of a microRNA, miR-155, on brain recovery after experimental cerebral ischemia. To our knowledge, this is the first report describing the efficiency of intravenous anti-miRNA injections in a mouse model of ischemic stroke. The role of miRNAs in poststroke revascularization has been unexplored and in vivo regulation of miRNAs during the subacute phase of stroke has not yet been proposed. Our investigation introduces a new and unexplored approach to cerebral regeneration: regulation of poststroke angiogenesis and recovery through direct modulation of specific miRNA activity. We expect that our findings will lead to the development of novel strategies for regulating neurorestorative processes in the postischemic brain.
Collapse
|
59
|
Lei C, Wu B, Cao T, Liu M, Hao Z. Brain recovery mediated by toll-like receptor 4 in rats after intracerebral hemorrhage. Brain Res 2015; 1632:1-8. [PMID: 26657742 DOI: 10.1016/j.brainres.2015.11.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/07/2015] [Accepted: 11/27/2015] [Indexed: 02/05/2023]
Abstract
Activation of the immune system via toll-like receptor 4 (TLR4) is implicated in both negative and positive processes in the central nervous system, including inflammation and angiogenesis. Whether TLR4 also participates in brain recovery following intracerebral hemorrhage (ICH) has not been investigated. We used the rat model of collagenase-induced ICH to determine whether TLR4 acts as a key regulator of brain recovery in the late phase of injury. After ICH, TLR4 levels in the ipsilateral striatum were significantly higher in the ICH group than in the Sham group on days 1, 3, 7 and 14 after ICH induction. By 14 d, the ICH group showed significantly higher levels of vascular endothelial growth factor, brain-derived neurotrophic factor, and MMP-9 than the Sham group, as well as greater numbers of vessels and BrdU- and DCX-positive cells. All these ICH-induced increases were significantly smaller in the TAK-242 group. The TLR4 antagonist also inhibited the recovery of neurological function after ICH. A TLR4 antagonist reduced ICH-induced neurogenesis and angiogenesis in a rat. These findings suggest that TLR4 may promote brain repair in the late phase of ICH.
Collapse
Affiliation(s)
- Chunyan Lei
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - Bo Wu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy and Ministry of Education, West China Hospital, Sichuan University, PR China.
| | - Tian Cao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - Ming Liu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy and Ministry of Education, West China Hospital, Sichuan University, PR China
| | - Zilong Hao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| |
Collapse
|
60
|
Seto SW, Kiat H, Lee SMY, Bensoussan A, Sun YT, Hoi MPM, Chang D. Zebrafish models of cardiovascular diseases and their applications in herbal medicine research. Eur J Pharmacol 2015; 768:77-86. [PMID: 26494630 DOI: 10.1016/j.ejphar.2015.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/02/2015] [Accepted: 10/16/2015] [Indexed: 01/12/2023]
Abstract
The zebrafish (Danio rerio) has recently become a powerful animal model for cardiovascular research and drug discovery due to its ease of maintenance, genetic manipulability and ability for high-throughput screening. Recent advances in imaging techniques and generation of transgenic zebrafish have greatly facilitated in vivo analysis of cellular events of cardiovascular development and pathogenesis. More importantly, recent studies have demonstrated the functional similarity of drug metabolism systems between zebrafish and humans, highlighting the clinical relevance of employing zebrafish in identifying lead compounds in Chinese herbal medicine with potential beneficial cardiovascular effects. This paper seeks to summarise the scope of zebrafish models employed in cardiovascular studies and the application of these research models in Chinese herbal medicine to date.
Collapse
Affiliation(s)
- Sai-Wang Seto
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Hosen Kiat
- Faculty of Medicine, University of New South Wales, NSW, Australia; School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW, Australia; Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Simon M Y Lee
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Alan Bensoussan
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Yu-Ting Sun
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Maggie P M Hoi
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia.
| |
Collapse
|
61
|
Ruan L, Wang B, ZhuGe Q, Jin K. Coupling of neurogenesis and angiogenesis after ischemic stroke. Brain Res 2015; 1623:166-73. [PMID: 25736182 PMCID: PMC4552615 DOI: 10.1016/j.brainres.2015.02.042] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 01/19/2023]
Abstract
Stroke is a leading cause of mortality and severe long-term disability worldwide. Development of effective treatment or new therapeutic strategies for ischemic stroke patients is therefore crucial. Ischemic stroke promotes neurogenesis by several growth factors including FGF-2, IGF-1, BDNF, VEGF and chemokines including SDF-1, MCP-1. Stroke-induced angiogenesis is similarly regulated by many factors most notably, eNOS and CSE, VEGF/VEGFR2, and Ang-1/Tie2. Important findings in the last decade have revealed that neurogenesis is not the stand-alone consideration in the fight for full functional recovery from stroke. Angiogenesis has been also shown to be critical in improving post-stroke neurological functional recovery. More than that, recent evidence has shown a highly possible interplay or dependence between stroke-induced neurogenesis and angiogenesis. Moving forward, elucidating the underlying mechanisms of this coupling between stroke-induced neurogenesis and angiogenesis will be of great importance, which will provide the basis for neurorestorative therapy. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Linhui Ruan
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA; Institute of Aging and Alzheimer׳s Disease Research, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Qichuan ZhuGe
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kunlin Jin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA; Institute of Aging and Alzheimer׳s Disease Research, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| |
Collapse
|
62
|
Ergul A, Valenzuela JP, Fouda AY, Fagan SC. Cellular connections, microenvironment and brain angiogenesis in diabetes: Lost communication signals in the post-stroke period. Brain Res 2015; 1623:81-96. [PMID: 25749094 PMCID: PMC4743654 DOI: 10.1016/j.brainres.2015.02.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/18/2015] [Accepted: 02/23/2015] [Indexed: 12/16/2022]
Abstract
Diabetes not only increases the risk but also worsens the motor and cognitive recovery after stroke, which is the leading cause of disability worldwide. Repair after stroke requires coordinated communication among various cell types in the central nervous system as well as circulating cells. Vascular restoration is critical for the enhancement of neurogenesis and neuroplasticity. Given that vascular disease is a major component of all complications associated with diabetes including stroke, this review will focus on cellular communications that are important for vascular restoration in the context of diabetes. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - John Paul Valenzuela
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA
| | - Abdelrahman Y Fouda
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Susan C Fagan
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| |
Collapse
|
63
|
Feng J, Huang T, Huang Q, Chen H, Li Y, He W, Wang GB, Zhang L, Xia J, Zhang N, Liu Y. Pro‑angiogenic microRNA‑296 upregulates vascular endothelial growth factor and downregulates Notch1 following cerebral ischemic injury. Mol Med Rep 2015; 12:8141-7. [PMID: 26500043 DOI: 10.3892/mmr.2015.4436] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 06/03/2015] [Indexed: 11/06/2022] Open
Abstract
The present study examined the association between microRNA (miR)‑296 and angiogenesis following cerebral ischemic injury, and the underlying mechanisms. A cerebral ischemic model was established in rats via right middle cerebral artery occlusion. The animals were randomly divided into four groups (baseline, 1 day, 3 day and 7 day). Quantitative polymerase chain reaction and western blot analyses were performed to examine the expression levels of miR‑296 and hepatocyte growth factor‑regulated tyrosine kinase substrate (HGS), respectively. Angiogenesis was assessed by examining microvessel density. The results demonstrated that miR‑296 and angiogenesis were significantly upregulated, while HGS was significantly downregulated following ischemic injury. Adenovirus‑mediated overexpression of miR‑296 markedly enhanced the formation of capillary‑like structures in human umbilical vein endothelial cells, parallel with significantly increased expression levels of vascular endothelial growth factor (VEGF) and VEGF receptor 2, and reduced expression levels of DLL4 and Notch1. The results of the present study provided in vivo and in vitro evidence suggesting that miR‑296 promotes angiogenesis in the ischemic brain through upregulating VEGF and downregulating Notch1 following cerebral ischemic injury.
Collapse
Affiliation(s)
- Jie Feng
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Tianxiang Huang
- Institute of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qing Huang
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hua Chen
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu Li
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei He
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Gui-Bin Wang
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Le Zhang
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jian Xia
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ning Zhang
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yunhai Liu
- Institute of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
64
|
Esposito E, Hayakawa K, Maki T, Arai K, Lo EH. Effects of Postconditioning on Neurogenesis and Angiogenesis During the Recovery Phase After Focal Cerebral Ischemia. Stroke 2015; 46:2691-4. [PMID: 26243221 DOI: 10.1161/strokeaha.115.009070] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE Postconditioning may be a clinically feasible way to protect the brain after a stroke. However, its effects during the recovery phase post stroke remain to be fully elucidated. Here, we examine the hypothesis that ischemic postconditioning amplifies neurogenesis and angiogenesis during stroke recovery. METHODS Male Sprague-Dawley rats were subjected to 100-minute transient middle cerebral artery occlusion (MCAO) or postconditioning (100-minute middle cerebral artery occlusion plus 10-minute reperfusion plus 10-minute reocclusion). After 2 weeks, infarct volumes, behavioral outcomes, and immunohistochemical markers of neurogenesis and angiogenesis were quantified. RESULTS Postconditioning significantly reduced infarction and improved neurological outcomes. Concomitantly, brains subjected to postconditioning showed an increase in doublecortin/BrdU and collagen-IV/Ki67-positive cells. CONCLUSIONS These results suggest that therapeutic effects of postconditioning may involve the promotion of neurogenesis and angiogenic remodeling during the recovery phase after focal cerebral ischemia.
Collapse
Affiliation(s)
- Elga Esposito
- From the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kazuhide Hayakawa
- From the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Takakuni Maki
- From the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ken Arai
- From the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Eng H Lo
- From the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
65
|
Soliman S, Ishrat T, Fouda AY, Patel A, Pillai B, Fagan SC. Sequential Therapy with Minocycline and Candesartan Improves Long-Term Recovery After Experimental Stroke. Transl Stroke Res 2015; 6:309-22. [PMID: 26004281 DOI: 10.1007/s12975-015-0408-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/07/2023]
Abstract
Minocycline and candesartan have both shown promise as candidate therapeutics in ischemic stroke, with multiple, and somewhat contrasting, molecular mechanisms. Minocycline is an anti-inflammatory, antioxidant, and anti-apoptotic agent and a known inhibitor of matrix metalloproteinases (MMPs). Yet, minocycline exerts antiangiogenic effects both in vivo and in vitro. Candesartan promotes angiogenesis and activates MMPs. Aligning these therapies with the dynamic processes of injury and repair after ischemia is likely to improve success of treatment. In this study, we hypothesize that opposing actions of minocycline and candesartan on angiogenesis, when administered simultaneously, will reduce the benefit of candesartan treatment. Therefore, we propose a sequential combination treatment regimen to yield a better outcome and preserve the proangiogenic potential of candesartan. In vitro angiogenesis was assessed using human brain endothelial cells. In vivo, Wistar rats subjected to 90-min middle cerebral artery occlusion (MCAO) were randomized into four groups: saline, candesartan, minocycline, and sequential combination of minocycline and candesartan. Neurobehavioral tests were performed 1, 3, 7, and 14 days after stroke. Brain tissue was collected on day 14 for assessment of infarct size and vascular density. Minocycline, when added simultaneously, decreased the proangiogenic effect of candesartan treatment in vitro. Sequential treatment, however, preserved the proangiogenic potential of candesartan both in vivo and in vitro, improved neurobehavioral outcome, and reduced infarct size. Sequential combination therapy with minocycline and candesartan improves long-term recovery and maintains candesartan's proangiogenic potential.
Collapse
|
66
|
Vitamin D prevents hypoxia/reoxygenation-induced blood-brain barrier disruption via vitamin D receptor-mediated NF-kB signaling pathways. PLoS One 2015; 10:e0122821. [PMID: 25815722 PMCID: PMC4376709 DOI: 10.1371/journal.pone.0122821] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/21/2015] [Indexed: 11/19/2022] Open
Abstract
Maintaining blood-brain barrier integrity and minimizing neuronal injury are critical components of any therapeutic intervention following ischemic stroke. However, a low level of vitamin D hormone is a risk factor for many vascular diseases including stroke. The neuroprotective effects of 1,25(OH)2D3 (vitamin D) after ischemic stroke have been studied, but it is not known whether it prevents ischemic injury to brain endothelial cells, a key component of the neurovascular unit. We analyzed the effect of 1,25(OH)2D3 on brain endothelial cell barrier integrity and tight junction proteins after hypoxia/reoxygenation in a mouse brain endothelial cell culture model that closely mimics many of the features of the blood-brain barrier in vitro. Following hypoxic injury in bEnd.3 cells, 1,25(OH)2D3 treatment prevented the decrease in barrier function as measured by transendothelial electrical resistance and permeability of FITC-dextran (40 kDa), the decrease in the expression of the tight junction proteins zonula occludin-1, claudin-5, and occludin, the activation of NF-kB, and the increase in matrix metalloproteinase-9 expression. These responses were blocked when the interaction of 1,25(OH) )2D3 with the vitamin D receptor (VDR) was inhibited by pyridoxal 5'-phosphate treatment. Our findings show a direct, VDR-mediated, protective effect of 1,25(OH) )2D3 against ischemic injury-induced blood-brain barrier dysfunction in cerebral endothelial cells.
Collapse
|
67
|
Lei C, Zhang S, Cao T, Tao W, Liu M, Wu B. HMGB1 may act via RAGE to promote angiogenesis in the later phase after intracerebral hemorrhage. Neuroscience 2015; 295:39-47. [PMID: 25813710 DOI: 10.1016/j.neuroscience.2015.03.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/15/2015] [Accepted: 03/17/2015] [Indexed: 02/05/2023]
Abstract
Following intracerebral hemorrhage (ICH), high-mobility group box 1 protein (HMGB1) may promote vascular remodeling. Whether HMGB1 supports angiogenesis after ICH is unclear, as are the receptors and downstream signaling pathway(s) involved. We used the rat model of collagenase-induced ICH to determine whether HMGB1 acts via the receptor for advanced glycation end-products (RAGE) to upregulate vascular endothelial growth factor (VEGF), a potent mitogen of endothelial cells and key regulator of normal and abnormal angiogenesis in the late phase of injury. At 3d after ICH induction, rats were treated with saline, ethyl pyruvate (EP) or N-benzyl-4-chloro-N-cyclohexylbenzamide (FPS-ZM1). ICH induced the movement of HMGB1 from the nucleus into the cytoplasm. Levels of HMGB1 and RAGE in the ipsilateral striatum increased within a few days of induction and continued to rise for 7-14d afterward. By 14d after induction, levels of VEGF and vessel density were higher than in the Sham group. Administering EP 3 days after ICH induction prevented much of the stroke-induced increases in vessel density and in expression of HMGB1, RAGE, and VEGF. Administering FPS-ZM1 after ICH blocked much of the stroke-induced increases in vessel density and VEGF expression. Our results suggest that after ICH, HMGB1 may upregulate VEGF in the ipsilateral striatum predominantly via RAGE. Hence, targeting the HMGB1/RAGE signaling pathway may help reduce inappropriate angiogenesis after ICH.
Collapse
Affiliation(s)
- C Lei
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - S Zhang
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China.
| | - T Cao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - W Tao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - M Liu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - B Wu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China.
| |
Collapse
|
68
|
Adamczak JM, Schneider G, Nelles M, Que I, Suidgeest E, van der Weerd L, Löwik C, Hoehn M. In vivo bioluminescence imaging of vascular remodeling after stroke. Front Cell Neurosci 2014; 8:274. [PMID: 25249937 PMCID: PMC4155794 DOI: 10.3389/fncel.2014.00274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/21/2014] [Indexed: 12/14/2022] Open
Abstract
Thrombolysis remains the only beneficial therapy for ischemic stroke, but is restricted to a short therapeutic window following the infarct. Currently research is focusing on spontaneous regenerative processes during the sub-acute and chronic phase. Angiogenesis, the formation of new blood vessels from pre-existing ones, was observed in stroke patients, correlates with longer survival and positively affects the formation of new neurons. Angiogenesis takes place in the border zones of the infarct, but further insight into the temporal profile is needed to fully apprehend its therapeutic potential and its relevance for neurogenesis and functional recovery. Angiogenesis is a multistep process, involving extracellular matrix degradation, endothelial cell proliferation, and, finally, new vessel formation. Interaction between vascular endothelial growth factor and its receptor 2 (VEGFR2) plays a central role in these angiogenic signaling cascades. In the present study we investigated non-invasively the dynamics of VEGFR2 expression following cerebral ischemia in a mouse model of middle cerebral artery occlusion (MCAO). We used a transgenic mouse expressing firefly luciferase under the control of the VEGFR2 promotor to non-invasively elucidate the temporal profile of VEGFR2 expression after stroke as a biomarker for VEGF/VEGFR2 signaling. We measured each animal repetitively up to 2 weeks after stroke and found increased VEGFR2 expression starting 3 days after the insult with peak values at 7 days. These were paralleled by increased VEGFR2 protein levels and increased vascular volume in peri-infarct areas at 14 days after the infarct, indicating that signaling via VEGFR2 leads to successful vascular remodeling. This study describes VEGFR2-related signaling is active at least up to 2 weeks after the infarct and results in increased vascular volume. Further, this study presents a novel strategy for the non-invasive evaluation of angiogenesis-based therapies.
Collapse
Affiliation(s)
- Joanna M Adamczak
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Gabriele Schneider
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Ivo Que
- Department of Endocrinology, Leiden University Medical Center Leiden, Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center Leiden, Netherlands ; Department of Human Genetics, Leiden University Medical Center Leiden, Netherlands
| | - Clemens Löwik
- Department of Endocrinology, Leiden University Medical Center Leiden, Netherlands ; Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany ; Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| |
Collapse
|
69
|
Ischemia/Reperfusion-induced neovascularization in the cerebral cortex of the ovine fetus. J Neuropathol Exp Neurol 2014; 73:495-506. [PMID: 24806298 DOI: 10.1097/nen.0000000000000071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Information on the effects of injury on neovascularization in the immature brain is limited. We investigated the effects of ischemia on cerebral cortex neovascularization after the exposure of fetuses to 30 minutes of cerebral ischemia followed by 48 hours of reperfusion (I/R-48), 30 minutes of cerebral ischemia followed by 72 hours of reperfusion (I/R-72), or sham control treatment (Non-I/R). Immunohistochemical and morphometric analyses of cerebral cortex sections included immunostaining for glial fibrillary acidic protein and collagen type IV (a molecular component of the vascular basal lamina) to determine the glial vascular network in fetal brains and Ki67 as a proliferation marker. Cerebral cortices from I/R-48 and I/R-72 fetuses exhibited general responses to ischemia, including reactive astrocyte morphology, which was not observed in Non-I/R fetuses. Cell bodies of reactive proliferating astrocytes, along with large end-feet, surrounded the walls of cerebral cortex microvessels in addition to the thick collagen type IV-enriched basal lamina. Morphometric analysis of the Non-I/R group with the I/R-48 and I/R-72 groups revealed increased collagen type IV density in I/R-72 cerebral cortex microvessels (p < 0.01), which also frequently displayed a sprouting appearance characterized by growing tip cells and activated pericytes. Increases in cerebral cortex basic fibroblast growth factor were associated with neovascularization. We conclude that increased neovascularization in fetal cerebral cortices occurs within 72 hours of ischemia.
Collapse
|
70
|
Li J, Tang Y, Wang Y, Tang R, Jiang W, Yang GY, Gao WQ. Neurovascular recovery via co-transplanted neural and vascular progenitors leads to improved functional restoration after ischemic stroke in rats. Stem Cell Reports 2014; 3:101-14. [PMID: 25068125 PMCID: PMC4110773 DOI: 10.1016/j.stemcr.2014.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 12/23/2022] Open
Abstract
The concept of the “neurovascular unit,” emphasizing the interactions between neural and vascular components in the brain, raised the notion that neural progenitor cell (NPC) transplantation therapy aimed at neural repair may be insufficient for the treatment of ischemic stroke. Here, we demonstrate that enhanced neurovascular recovery via cotransplantation of NPCs and embryonic stem cell-derived vascular progenitor cells (VPCs) in a rat stroke model is correlated with improved functional recovery after stroke. We found that cotransplantation promoted the survival, migration, differentiation, and maturation of neuronal and vascular cells derived from the cotransplanted progenitors. Furthermore, it triggered an increased generation of VEGF-, BDNF-, and IGF1-expressing neural cells derived from the grafted NPCs. Consistently, compared with transplantation of NPCs alone, cotransplantation more effectively improved the neurobehavioral deficits and attenuated the infarct volume. Thus, cotransplantation of NPCs and VPCs represents a more effective therapeutic strategy for the treatment of stroke than transplantation of NPCs alone. Neural and vascular progenitor cell cotransplantation therapy for ischemic stroke Better neurovascular recovery by cotransplanted progenitor cells in the infarct area Better infarction reduction and functional restoration by cotransplantation Neurovascular recovery likely mediated by neural production of growth/trophic factors
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China ; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongting Wang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Rongbiao Tang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Weifang Jiang
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200030, China
| | - Guo-Yuan Yang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China ; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
71
|
Soliman S, Ishrat T, Pillai A, Somanath PR, Ergul A, El-Remessy AB, Fagan SC. Candesartan induces a prolonged proangiogenic effect and augments endothelium-mediated neuroprotection after oxygen and glucose deprivation: role of vascular endothelial growth factors A and B. J Pharmacol Exp Ther 2014; 349:444-57. [PMID: 24681872 PMCID: PMC4019323 DOI: 10.1124/jpet.113.212613] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/27/2014] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is a key component of recovery after stroke. Angiotensin II receptor blocker (ARB) treatment improves neurobehavioral outcome and is associated with enhanced angiogenesis after stroke. The purpose of this study is to investigate the temporal pattern of the ARB proangiogenic effect in the ischemic brain and its association with vascular endothelial growth factors VEGF-A and VEGF-B. Wistar rats were exposed to 90-minute middle cerebral artery occlusion and treated with candesartan (1 mg/kg) at reperfusion. The proangiogenic potential of the cerebrospinal fluid was determined at 8, 24, 48, and 72 hours using an in vitro Matrigel tube formation assay. In addition, the expression of VEGF-A and VEGF-B was measured in brain homogenates using Western blotting at the same time points. A single candesartan dose induced a prolonged proangiogenic effect and a prolonged upregulation of VEGF-A and VEGF-B in vivo. In the ischemic hemisphere, candesartan treatment was associated with stabilization of hypoxia-inducible factor-1α and preservation of angiopoietin-1. The effect of ARB treatment on endothelial cells was studied in vitro. Our results identified brain endothelial cells as one target for the action of ARBs and a source of the upregulated VEGF-A and VEGF-B, which exerted an autocrine angiogenic response, in addition to a paracrine neuroprotective effect. Taken together, this study highlights the potential usefulness of augmenting the endogenous restorative capacity of the brain through the administration of ARBs.
Collapse
Affiliation(s)
- Sahar Soliman
- Program in Clinical and Experimental Therapeutics, University of Georgia, Augusta, Georgia (S.S., T.I., P.R.S., A.E., A.B.E., S.C.F.); Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia (S.S., T.I., P.R.S., A.E., A.B.E., S.C.F.); and Departments of Psychiatry (A.P.), Medicine (P.R.S.), Physiology (A.E.), Pharmacology and Toxicology (A.B.E.), and Neurology (S.C.F.), Georgia Regents University, Augusta, Georgia
| | | | | | | | | | | | | |
Collapse
|
72
|
Camós S, Gubern C, Sobrado M, Rodríguez R, Romera V, Moro M, Lizasoain I, Serena J, Mallolas J, Castellanos M. The high-mobility group I-Y transcription factor is involved in cerebral ischemia and modulates the expression of angiogenic proteins. Neuroscience 2014; 269:112-30. [DOI: 10.1016/j.neuroscience.2014.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/07/2014] [Accepted: 03/18/2014] [Indexed: 12/24/2022]
|
73
|
Liu J, Wang Y, Akamatsu Y, Lee CC, Stetler RA, Lawton MT, Yang GY. Vascular remodeling after ischemic stroke: mechanisms and therapeutic potentials. Prog Neurobiol 2013; 115:138-56. [PMID: 24291532 DOI: 10.1016/j.pneurobio.2013.11.004] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 11/14/2013] [Accepted: 11/16/2013] [Indexed: 12/18/2022]
Abstract
The brain vasculature has been increasingly recognized as a key player that directs brain development, regulates homeostasis, and contributes to pathological processes. Following ischemic stroke, the reduction of blood flow elicits a cascade of changes and leads to vascular remodeling. However, the temporal profile of vascular changes after stroke is not well understood. Growing evidence suggests that the early phase of cerebral blood volume (CBV) increase is likely due to the improvement in collateral flow, also known as arteriogenesis, whereas the late phase of CBV increase is attributed to the surge of angiogenesis. Arteriogenesis is triggered by shear fluid stress followed by activation of endothelium and inflammatory processes, while angiogenesis induces a number of pro-angiogenic factors and circulating endothelial progenitor cells (EPCs). The status of collaterals in acute stroke has been shown to have several prognostic implications, while the causal relationship between angiogenesis and improved functional recovery has yet to be established in patients. A number of interventions aimed at enhancing cerebral blood flow including increasing collateral recruitment are under clinical investigation. Transplantation of EPCs to improve angiogenesis is also underway. Knowledge in the underlying physiological mechanisms for improved arteriogenesis and angiogenesis shall lead to more effective therapies for ischemic stroke.
Collapse
Affiliation(s)
- Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA.
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai 200030, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yosuke Akamatsu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurological Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Chih Cheng Lee
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael T Lawton
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai 200030, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| |
Collapse
|
74
|
Bednar MM, Perry A. Neurorestoration therapeutics for neurodegenerative and psychiatric disease. Neurol Res 2013; 34:129-42. [DOI: 10.1179/1743132811y.0000000069] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
75
|
Mammoto T, Jiang A, Jiang E, Mammoto A. Platelet rich plasma extract promotes angiogenesis through the angiopoietin1-Tie2 pathway. Microvasc Res 2013; 89:15-24. [PMID: 23660186 DOI: 10.1016/j.mvr.2013.04.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/23/2013] [Accepted: 04/28/2013] [Indexed: 12/18/2022]
Abstract
Development and regeneration of tissues and organs require precise coordination among endothelial, epithelial and mesenchymal morphogenesis. Angiogenesis plays key roles in normal development, wound healing, recovery from ischemic disease, and organ regeneration. It has been recognized that the combination of various angiogenic factors in an appropriate physiological ratio is critical for long-term functional blood vessel formation. Here we show that mouse soluble platelet-rich-plasma (PRP) extract, which includes abundant angiopoetin-1 (Ang1) and other angiogenic factors, stimulates endothelial cell growth, migration and differentiation in cultured human dermal microvascular endothelial cells in vitro and neonatal mouse retinal angiogenesis in vivo. Mouse platelet rich fibrin (PRF) matrix, the three-dimensional fibrin matrix that releases angiogenic factors with similar concentrations and proportions to the PRP extract, also recapitulates robust angiogenesis inside the matrix when implanted subcutaneously on the living mouse. Inhibition of Ang1-Tie2 signaling suppresses PRP extract-induced angiogenesis in vitro and angiogenic ability of the PRF matrix in vivo. Since human PRP extract and PRF matrix can be prepared from autologous peripheral blood, our findings may lead to the development of novel therapeutic interventions for various angiogenesis-related diseases as well as to the improvement of strategies for tissue engineering and organ regeneration.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
76
|
Exercise therapy augments the ischemia-induced proangiogenic state and results in sustained improvement after stroke. Int J Mol Sci 2013; 14:8570-84. [PMID: 23598418 PMCID: PMC3645762 DOI: 10.3390/ijms14048570] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/11/2013] [Accepted: 04/11/2013] [Indexed: 01/19/2023] Open
Abstract
The induction of angiogenesis will stimulate endogenous recovery mechanisms, which are involved in the long-term repair and restoration process of the brain after an ischemic event. Here, we tested whether exercise influences the pro-angiogenic factors and outcomes after cerebral infarction in rats. Wistar rats were exposed to two hours of middle-cerebral artery occlusion and reperfusion. Different durations of treadmill training were performed on the rats. The expression of matrix metalloproteinase 2 (MMP2) and vascular endothelial growth factor (VEGF)-related genes and proteins were higher over time post-ischemia, and exercise enhanced their expression. Sixteen days post-ischemia, the regional cerebral blood flow in the ischemic striatum was significantly increased in the running group over the sedentary. Although no difference was seen in infarct size between the running and sedentary groups, running evidently improved the neurobehavioral score. The effects of running on MMP2 expression, regional cerebral blood flow and outcome were abolished when animals were treated with bevacizumab (BEV), a VEGF-targeting antibody. Exercise therapy improves long-term stroke outcome by MMP2-VEGF-dependent mechanisms related to improved cerebral blood flow.
Collapse
|
77
|
Alhusban A, Kozak A, Ergul A, Fagan SC. AT1 receptor antagonism is proangiogenic in the brain: BDNF a novel mediator. J Pharmacol Exp Ther 2013; 344:348-59. [PMID: 23211364 PMCID: PMC3558823 DOI: 10.1124/jpet.112.197483] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 12/03/2012] [Indexed: 12/12/2022] Open
Abstract
Candesartan is an angiotensin II type 1 receptor blocker (ARB) that has been to shown to limit ischemic stroke and improve stroke outcome. In experimental stroke, candesartan induces a proangiogenic effect that is partly attributable to vascular endothelial growth factor. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family that has been reported to have angiogenic effects and play an important role in recovery after stroke. The purpose of this investigation was to determine the role of BDNF in the proangiogenic effect of candesartan in the brain under hypertensive conditions. Accordingly, spontaneously hypertensive rats were treated with candesartan, and brain tissue samples were collected for quantification of BDNF expression. In addition, human cerebromicrovascular endothelial cells were treated with either low-dose (1 ƒM) or high-dose (1 µM) angiotensin II alone or in combination with candesartan (0.16 µM) to assess the effect of candesartan treatment and BDNF involvement in the behavior of endothelial cells. Candesartan significantly increased the expression of BDNF in the SHR (P < 0.05). In addition, candesartan reversed the antiangiogenic effect of the 1-µM dose of AngII (P = 0.0001). The observed effects of candesartan were ablated by neutralizing the effects of BDNF. Treatment with the AT2 antagonist PD-123319 significantly reduced tube-like formation in endothelial cells. AT2 stimulation induced the BDNF expression and migration (P < 0.05). In conclusion, candesartan exerts a proangiogenic effect on brain microvascular endothelial cells treated with angiotensin II. This response is attributable to increased BDNF expression and is mediated through stimulation of the AT2 receptor.
Collapse
Affiliation(s)
- Ahmed Alhusban
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | | | | | | |
Collapse
|
78
|
Effects of high-mobility group box1 on cerebral angiogenesis and neurogenesis after intracerebral hemorrhage. Neuroscience 2012; 229:12-9. [PMID: 23137544 DOI: 10.1016/j.neuroscience.2012.10.054] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 02/05/2023]
Abstract
Neural stem cells, which reside mainly in the subventricular and subgranular zones of the hippocampus, can regenerate new neuroblasts after various brain insults. Aided by vascular remodeling, these new neuroblasts migrate long distances to injured brain regions. Studies have suggested that high-mobility group box1 (HMGB1), a nonhistone nuclear DNA-binding protein, may stimulate such remodeling in the late phase of some types of brain injury, but it is unclear whether this is true for intracerebral hemorrhage (ICH). Here we used a rat model of collagenase-induced ICH to determine whether HMGB1 can promote neurogenesis and angiogenesis in the late phase of injury. Daily administration of ethyl pyruvate, which inhibited HMGB1 expression, reduced the recovery of neurological function, decreased vascular endothelial growth factor (VEGF) and nerve growth factor (NGF) levels in the ipsilateral striatum, and decreased the numbers of 5-bromo-2-deoxyuridine (BrdU)- and doublecortin (DCX)-positive cells around the hematoma. These findings suggest that HMGB1 may promote angiogenesis and neurogenesis in the late phase of ICH.
Collapse
|
79
|
Zhang Y, Huang S, Wang B, Sun B, Li W, Lu X, Ding X. Atorvastatin and whisker stimulation synergistically enhance angiogenesis in the barrel cortex of rats following focal ischemia. Neurosci Lett 2012; 525:135-9. [DOI: 10.1016/j.neulet.2012.07.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/25/2012] [Accepted: 07/26/2012] [Indexed: 12/14/2022]
|
80
|
Jiang Q, Thiffault C, Kramer BC, Ding GL, Zhang L, Nejad-Davarani SP, Li L, Arbab AS, Lu M, Navia B, Victor SJ, Hong K, Li QJ, Wang SY, Li Y, Chopp M. MRI detects brain reorganization after human umbilical tissue-derived cells (hUTC) treatment of stroke in rat. PLoS One 2012; 7:e42845. [PMID: 22900057 PMCID: PMC3416784 DOI: 10.1371/journal.pone.0042845] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/12/2012] [Indexed: 11/24/2022] Open
Abstract
Human umbilical tissue-derived cells (hUTC) represent an attractive cell source and a potential technology for neurorestoration and improvement of functional outcomes following stroke. Male Wistar rats were subjected to a transient middle cerebral artery occlusion (tMCAo) and were intravenously administered hUTC (N = 11) or vehicle (N = 10) 48 hrs after stroke. White matter and vascular reorganization was monitored over a 12-week period using MRI and histopathology. MRI results were correlated with neurological functional and histology outcomes to demonstrate that MRI can be a useful tool to measure structural recovery after stroke. MRI revealed a significant reduction in the ventricular volume expansion and improvement in cerebral blood flow (CBF) in the hUTC treated group compared to vehicle treated group. Treatment with hUTC resulted in histological and functional improvements as evidenced by enhanced expression of vWF and synaptophysin, and improved outcomes on behavioral tests. Significant correlations were detected between MRI ventricular volumes and histological lesion volume as well as number of apoptotic cells. A positive correlation was also observed between MRI CBF or cerebral blood volume (CBV) and histological synaptic density. Neurological functional tests were also significantly correlated with MRI ventricular volume and CBV. Our data demonstrated that MRI measurements can detect the effect of hUTC therapy on the brain reorganization and exhibited positive correlation with histological measurements of brain structural changes and functional behavioral tests after stroke. MRI ventricular volumes provided the most sensitive index in monitoring brain remodeling and treatment effects and highly correlated with histological and functional measurements.
Collapse
Affiliation(s)
- Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Marshall JF, O'Dell SJ. Methamphetamine influences on brain and behavior: unsafe at any speed? Trends Neurosci 2012; 35:536-45. [PMID: 22709631 DOI: 10.1016/j.tins.2012.05.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/09/2012] [Accepted: 05/16/2012] [Indexed: 11/30/2022]
Abstract
Methamphetamine damages monoamine-containing nerve terminals in the brains of both animals and human drug abusers, and the cellular mechanisms underlying this injury have been extensively studied. More recently, the growing evidence for methamphetamine influences on memory and executive function of human users has prompted studies of cognitive impairments in methamphetamine-exposed animals. After summarizing current knowledge about the cellular mechanisms of methamphetamine-induced brain injury, this review emphasizes research into the brain changes that underlie the cognitive deficits that accompany repeated methamphetamine exposure. Novel approaches to mitigating or reversing methamphetamine-induced brain and behavioral changes are described, and it is argued that the slow spontaneous reversibility of the injury produced by this drug may offer opportunities for novel treatment development.
Collapse
Affiliation(s)
- John F Marshall
- Department of Neurobiology and Behavior, Center for Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA.
| | | |
Collapse
|
82
|
Revealing the role of phospholipase Cβ3 in the regulation of VEGF-induced vascular permeability. Blood 2012; 120:2167-73. [PMID: 22674805 DOI: 10.1182/blood-2012-03-417824] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
VEGF induces vascular permeability (VP) in ischemic diseases and cancer, leading to many pathophysiological consequences. The molecular mechanisms by which VEGF acts to induce hyperpermeability are poorly understood and in vivo models that easily facilitate real-time, genetic studies of VP do not exist. In the present study, we report a heat-inducible VEGF transgenic zebrafish (Danio rerio) model through which VP can be monitored in real time. Using this approach with morpholino-mediated gene knock-down and knockout mice, we describe a novel role of phospholipase Cβ3 as a negative regulator of VEGF-mediated VP by regulating intracellular Ca2+ release. Our results suggest an important effect of PLCβ3 on VP and provide a new model with which to identify genetic regulators of VP crucial to several disease processes.
Collapse
|
83
|
Affiliation(s)
- Adviye Ergul
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912, USA
| | | | | |
Collapse
|
84
|
Abstract
OBJECTIVE Extensive research in the past decade has confirmed that the adult brain maintains some plasticity, including neural cell birth, migration and integration. Pre-clinical data strongly suggest that phosphodiesterase type 5 (PDE5) inhibitors promote cerebral neovascularization and neurogenesis. Animal studies of cerebral stroke suggest potential regenerative benefits following treatment with sildenafil citrate, a PDE5 inhibitor. This study reports a case in which compassionate use of sildenafil was investigated as a treatment to improve physical functioning, more than 4 decades after development of spastic quadriplegia during the 1st-2nd year of life. METHODS Sildenafil 100 mg was administered every 24 hours for 7 months. RESULTS Sildenafil treatment was associated with clinical (functional) improvement. CONCLUSIONS The activity of sildenafil on cerebral neovascularization and neurogenesis may be the mechanism for the observed functional benefits.
Collapse
Affiliation(s)
- Antonio Cocchiarella
- Clinical Rehabilitation Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
85
|
Liman TG, Endres M. New vessels after stroke: postischemic neovascularization and regeneration. Cerebrovasc Dis 2012; 33:492-9. [PMID: 22517438 DOI: 10.1159/000337155] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 02/08/2012] [Indexed: 12/30/2022] Open
Abstract
The formation of new blood vessels after acute ischemic stroke is one of the most promising approaches to future therapies in the emerging field of stroke medicine. Angiogenesis and postnatal vasculogenesis are the underlying mechanisms of the formation of new blood vessels. Bone marrow-derived endothelial progenitor cells (EPCs) are thought to play an important role in neovascularization and during the regenerative processes after a vascular injury as well as in the maintenance of endothelial integrity. This review summarizes possible mechanisms of angiogenesis, postischemic neovascularization and regeneration with a focus on the potential role of EPCs as a risk marker and as a therapeutic target in stroke medicine.
Collapse
Affiliation(s)
- T G Liman
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | |
Collapse
|
86
|
Gertz K, Kronenberg G, Kälin RE, Baldinger T, Werner C, Balkaya M, Eom GD, Hellmann-Regen J, Kröber J, Miller KR, Lindauer U, Laufs U, Dirnagl U, Heppner FL, Endres M. Essential role of interleukin-6 in post-stroke angiogenesis. ACTA ACUST UNITED AC 2012; 135:1964-80. [PMID: 22492561 DOI: 10.1093/brain/aws075] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ambivalent effects of interleukin-6 on the pathogenesis of ischaemic stroke have been reported. However, to date, the long-term actions of interleukin-6 after stroke have not been investigated. Here, we subjected interleukin-6 knockout (IL-6(-/-)) and wild-type control mice to mild brain ischaemia by 30-min filamentous middle cerebral artery occlusion/reperfusion. While ischaemic tissue damage was comparable at early time points, IL-6(-/-) mice showed significantly increased chronic lesion volumes as well as worse long-term functional outcome. In particular, IL-6(-/-) mice displayed an impaired angiogenic response to brain ischaemia with reduced numbers of newly generated endothelial cells and decreased density of perfused microvessels along with lower absolute regional cerebral blood flow and reduced vessel responsivity in ischaemic striatum at 4 weeks. Similarly, the early genomic activation of angiogenesis-related gene networks was strongly reduced and the ischaemia-induced signal transducer and activator of transcription 3 activation observed in wild-type mice was almost absent in IL-6(-/-) mice. In addition, systemic neoangiogenesis was impaired in IL-6(-/-) mice. Transplantation of interleukin-6 competent bone marrow into IL-6(-/-) mice (IL-6(chi)) did not rescue interleukin-6 messenger RNA expression or the early transcriptional activation of angiogenesis after stroke. Accordingly, chronic stroke outcome in IL-6(chi) mice recapitulated the major effects of interleukin-6 deficiency on post-stroke regeneration with significantly enhanced lesion volumes and reduced vessel densities. Additional in vitro experiments yielded complementary evidence, which showed that after stroke resident brain cells serve as the major source of interleukin-6 in a self-amplifying network. Treatment of primary cortical neurons, mixed glial cultures or immortalized brain endothelia with interleukin 6-induced robust interleukin-6 messenger RNA transcription in each case, whereas oxygen-glucose deprivation did not. However, oxygen-glucose deprivation of organotypic brain slices resulted in strong upregulation of interleukin-6 messenger RNA along with increased transcription of key angiogenesis-associated genes. In conclusion, interleukin-6 produced locally by resident brain cells promotes post-stroke angiogenesis and thereby affords long-term histological and functional protection.
Collapse
Affiliation(s)
- Karen Gertz
- Department of Neurology, Charité– Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Cellular Therapy for Ischemic Stroke. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|