151
|
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: 16] [Impact Index Per Article: 1.8] [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
|
152
|
Spinal cord injury repair by implantation of structured hyaluronic acid scaffold with PLGA microspheres in the rat. Cell Tissue Res 2015; 364:17-28. [DOI: 10.1007/s00441-015-2298-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/25/2015] [Indexed: 11/26/2022]
|
153
|
Meng YC, Ding ZY, Wang HQ, Ning LP, Wang C. Effect of microRNA-155 on angiogenesis after cerebral infarction of rats through AT1R/VEGFR2 pathway. ASIAN PAC J TROP MED 2015; 8:829-35. [DOI: 10.1016/j.apjtm.2015.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/20/2015] [Accepted: 09/15/2015] [Indexed: 10/23/2022] Open
|
154
|
Dalkara T, Alarcon-Martinez L. Cerebral microvascular pericytes and neurogliovascular signaling in health and disease. Brain Res 2015; 1623:3-17. [DOI: 10.1016/j.brainres.2015.03.047] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/10/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023]
|
155
|
MicroRNA-107 contributes to post-stroke angiogenesis by targeting Dicer-1. Sci Rep 2015; 5:13316. [PMID: 26294080 PMCID: PMC4543985 DOI: 10.1038/srep13316] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 07/21/2015] [Indexed: 01/27/2023] Open
Abstract
Previous studies have suggested that microRNA-107 (miR-107) regulates cell migration in tumor and promotes Hypoxia Inducible Factor 1α (HIF1α) regulated angiogenesis under hypoxia. We found that miR-107 was strongly expressed in ischemic boundary zone (IBZ) after permanent middle cerebral artery occlusion (pMCAO) in rats and inhibition of miR-107 could reduce capillary density in the IBZ after stroke. Such finding led us to hypothesize that miR-107 might regulate post-stroke angiogenesis and therefore serve as a therapeutic target. We also found that antagomir-107, a synthetic miR-107 inhibitor, decreased the number of capillaries in IBZ and increased overall infarct volume after pMCAO in rats. We demonstrated that miR-107 could directly down-regulate Dicer-1, a gene that encodes an enzyme essential for processing microRNA (miRNA) precursors. This resulted in translational desupression of VEGF (vascular endothelial growth factor) mRNA, thereby increasing expression of endothelial cell-derived VEGF (VEGF165/VEGF164), leading to angiogenesis after stroke. This process might be a protective mechanism for ischemia-induced cerebral injury and miR-107 might be used as a novel tool in stroke treatment.
Collapse
|
156
|
Abstract
Angiotensin (Ang) (1-7) is the main component of the depressor and protective arm of the renin-angiotensin system. Ang-(1-7) induces vasodilation, natriuresis and diuresis, cardioprotection, inhibits angiogenesis and cell growth and opposes the pressor, proliferative, profibrotic, and prothrombotic actions mediated by Ang II. Centrally, Ang-(1-7) induces changes in mean arterial pressure and this effect may be linked with its inhibitory neuromodulatory action on norepinephrine neurotransmission. The present review is focused on the role of Ang-(1-7) as a protective agent in the brain.
Collapse
Affiliation(s)
- Mariela M Gironacci
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113 Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
157
|
Praet J, Santermans E, Daans J, Le Blon D, Hoornaert C, Goossens H, Hens N, Van Der Linden A, Berneman Z, Ponsaerts P. Early Inflammatory Responses following Cell Grafting in the CNS Trigger Activation of the Subventricular Zone: A Proposed Model of Sequential Cellular Events. Cell Transplant 2015; 24:1481-92. [DOI: 10.3727/096368914x682800] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
While multiple rodent preclinical studies, and to a lesser extent human clinical trials, claim the feasibility, safety, and potential clinical benefit of cell grafting in the central nervous system (CNS), currently only little convincing knowledge exists regarding the actual fate of the grafted cells and their effect on the surrounding environment (or vice versa). Our preceding studies already indicated that only a minor fraction of the initially grafted cell population survives the grafting process, while the surviving cell population becomes invaded by highly activated microglia/macrophages and surrounded by reactive astrogliosis. In the current study, we further elaborate on early cellular and inflammatory events following syngeneic grafting of eGFP mouse embryonic fibroblasts (mEFs) in the CNS of immunocompetent mice. Based on obtained quantitative histological data, we here propose a detailed mathematically derived working model that sequentially comprises hypoxia-induced apoptosis of grafted mEFs, neutrophil invasion, neoangiogenesis, microglia/macrophage recruitment, astrogliosis, and eventually survival of a limited number of grafted mEFs. Simultaneously, we observed that the cellular events following mEF grafting activates the subventricular zone neural stem and progenitor cell compartment. This proposed model therefore further contributes to our understanding of cell graft-induced cellular responses and will eventually allow for successful manipulation of this intervention.
Collapse
Affiliation(s)
- Jelle Praet
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- BioImaging Laboratory, University of Antwerp, Wilrijk, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
| | - Jasmijn Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Debbie Le Blon
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Chloé Hoornaert
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
- Centre for Health Economic Research and Modeling Infectious Diseases (Chermid), University of Antwerp, Wilrijk, Belgium
| | | | - Zwi Berneman
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
158
|
Xu Y, Zhang J, Jiang W, Zhang S. Astaxanthin induces angiogenesis through Wnt/β-catenin signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:744-751. [PMID: 26141761 DOI: 10.1016/j.phymed.2015.05.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 05/24/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE In the present study, we sought to elucidate whether astaxanthin contributes to induce angiogenesis and its mechanisms. MATERIALS AND METHODS To this end, we examined the role of astaxanthin on human brain microvascular endothelial cell line (HBMEC) and rat aortic smooth muscle cell (RASMC) proliferation, invasion and tube formation in vitro. For study of mechanism, the Wnt/β-catenin signaling pathway inhibitor IWR-1-endo was used. HMBECs and RASMCs proliferation were tested by cell counting. Scratch adhesion test was used to assess the ability of invasion. A matrigel tube formation assay was performed to test capillary tube formation ability. The Wnt/β-catenin pathway activation in HMBECs and RASMCs were tested by Western blot. RESULTS Our data suggested that astaxanthin induces angiogenesis by increasing proliferation, invasion and tube formation in vitro. Wnt and β-catenin expression were increased by astaxanthin and counteracted by IWR-1-endo in HMBECs and RASMCs. Tube formation was increased by astaxanthin and counteracted by IWR-1-endo. CONCLUSIONS It may be suggested that astaxanthin induces angiogenesis in vitro via a programmed Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yangyang Xu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China
| | - Wanglin Jiang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China.
| | - Shuping Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China.
| |
Collapse
|
159
|
Neuroprotection of Early Locomotor Exercise Poststroke: Evidence From Animal Studies. Can J Neurol Sci 2015; 42:213-20. [PMID: 26041314 DOI: 10.1017/cjn.2015.39] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Early locomotor exercise after stroke has attracted a great deal of attention in clinical and animal research in recent years. A series of animal studies showed that early locomotor exercise poststroke could protect against ischemic brain injury and improve functional outcomes through the promotion of angiogenesis, inhibition of acute inflammatory response and neuron apoptosis, and protection of the blood-brain barrier. However, to date, the clinical application of early locomotor exercise poststroke was limited because some clinicians have little confidence in its effectiveness. Here we review the current progress of early locomotor exercise poststroke in animal models. We hope that a comprehensive awareness of the early locomotor exercise poststroke may help to implement early locomotor exercise more appropriately in treatment for ischemic stroke.
Collapse
|
160
|
Huang CH, Shih YYI, Siow TY, Hsu YH, Chen CCV, Lin TN, Jaw FS, Chang C. Temporal assessment of vascular reactivity and functionality using MRI during postischemic proangiogenenic vascular remodeling. Magn Reson Imaging 2015; 33:903-10. [PMID: 25944092 DOI: 10.1016/j.mri.2015.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 03/13/2015] [Accepted: 04/26/2015] [Indexed: 11/18/2022]
Abstract
Postischemic angiogenesis is an important recovery mechanism. Both arteries and veins are upregulated during angiogenesis, but eventually there are more angiogenic veins than arteries in terms of number and length. It is critical to understand how the veins are modulated after ischemia and then transitioned into angiogenic vessels during the proangiogenic stage to finally serve as a restorative strength to the injured area. Using a rat model of transient focal cerebral ischemia, the hypercapnic blood oxygen level-dependent (BOLD) response was used to evaluate vascular reactivity, while the hyperoxic BOLD and tissue oxygen level-dependent (TOLD) responses were used to evaluate the vascular functionality at 1, 3, and 7days after ischemia. Vessel-like venous signals appeared on R2* maps on days 3 and 7, but not on day 1. The large hypercapnic BOLD responses on days 3 and 7 indicated that these areas have high vascular reactivity. The temporal correlation between vascular reactivity and the immunoreactivity to desmin and VEGF further indicates that the integrity of vascular reactivity is associated with the pericyte coverage as regulated by the VEGF level. Vascular functionality remained low on days 1, 3, and 7, as reflected by the small hyperoxic BOLD and large hyperoxic TOLD responses, indicating the low oxygen consumption of the ischemic tissues. These functional changes in proangiogenic veins may be critical for angiogenesis.
Collapse
Affiliation(s)
- Chien-Hsiang Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Yen-Yu Ian Shih
- Experimental Neuroimaging Laboratory, Department of Neurology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Tiing-Yee Siow
- Department of Medical Imaging and Intervention, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Hua Hsu
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Chiao-Chi V Chen
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Fu-Shan Jaw
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Chen Chang
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan.
| |
Collapse
|
161
|
Abdelsaid M, Prakash R, Li W, Coucha M, Hafez S, Johnson MH, Fagan SC, Ergul A. Metformin treatment in the period after stroke prevents nitrative stress and restores angiogenic signaling in the brain in diabetes. Diabetes 2015; 64:1804-17. [PMID: 25524911 PMCID: PMC4407857 DOI: 10.2337/db14-1423] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/11/2014] [Indexed: 12/04/2022]
Abstract
Diabetes impedes vascular repair and causes vasoregression in the brain after stroke, but mechanisms underlying this response are still unclear. We hypothesized that excess peroxynitrite formation in diabetic ischemia/reperfusion (I/R) injury inactivates the p85 subunit of phosphoinositide 3-kinase (PI3K) by nitration and diverts the PI3K-Akt survival signal to the p38-mitogen-activated protein kinase apoptosis pathway. Nitrotyrosine (NY), Akt and p38 activity, p85 nitration, and caspase-3 cleavage were measured in brains from control, diabetic (GK), or metformin-treated GK rats subjected to sham or stroke surgery and in brain microvascular endothelial cells (BMVECs) from Wistar and GK rats subjected to hypoxia/reoxygenation injury. GK rat brains showed increased NY, caspase-3 cleavage, and p38 activation and decreased Akt activation. Metformin attenuated stroke-induced nitrative signaling in GK rats. GK rat BMVECs showed increased basal nitrative stress compared with controls. A second hit by hypoxia/reoxygenation injury dramatically increased the nitration of p85 and activation of p38 but decreased Akt. These effects were associated with impairment of angiogenic response and were restored by treatment with the peroxynitrite scavenger 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron III chloride or the nitration inhibitor epicatechin. Our results provide evidence that I/R-induced peroxynitrite inhibits survival, induces apoptosis, and promotes peroxynitrite as a novel therapeutic target for the improvement of reparative angiogenesis after stroke in diabetes.
Collapse
Affiliation(s)
- Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA
| | - Roshini Prakash
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | - Weiguo Li
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA
| | - Maha Coucha
- Department of Physiology, Georgia Regents University, Augusta, GA
| | - Sherif Hafez
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | | | - Susan C Fagan
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| |
Collapse
|
162
|
Feng N, Wang Z, Zhang Z, He X, Wang C, Zhang L. miR-487b promotes human umbilical vein endothelial cell proliferation, migration, invasion and tube formation through regulating THBS1. Neurosci Lett 2015; 591:1-7. [DOI: 10.1016/j.neulet.2015.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 01/28/2015] [Accepted: 02/01/2015] [Indexed: 12/31/2022]
|
163
|
Xu L, Guo R, Xie Y, Ma M, Ye R, Liu X. Caveolae: molecular insights and therapeutic targets for stroke. Expert Opin Ther Targets 2015; 19:633-50. [PMID: 25639269 DOI: 10.1517/14728222.2015.1009446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Caveolae are specialized plasma membrane micro-invaginations of most mammalian cell types. The organization and function of caveolae are carried out by their coat proteins, caveolins and adaptor proteins, cavins. Caveolae/caveolins physically interact with membrane-associated signaling molecules and function in cholesterol incorporation, signaling transduction and macromolecular transport/permeability. AREAS COVERED Recent investigations have implicated a check-and-balance role of caveolae in the pathophysiology of cerebral ischemia. Caveolin knockout mice displayed exacerbated ischemic injury, whereas caveolin peptide exerted remarkable protection against ischemia/reperfusion injury. This review attempts to provide a comprehensive synopsis of how caveolae/caveolins modulate blood-brain barrier permeability, pro-survival signaling, angiogenesis and neuroinflammation, and how this may contribute to a better understanding of the participation of caveolae in ischemic cascade. The role of caveolin in the preconditioning-induced tolerance against ischemia is also discussed. EXPERT OPINION Caveolae represent a novel target for cerebral ischemia. It remains open how to manipulate caveolin expression in a practical way to recapitulate the beneficial therapeutic outcomes. Caveolin peptides and associated antagomirs may be efficacious and deserve further investigations for their potential benefits for stroke.
Collapse
Affiliation(s)
- Lili Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing 210002 , China
| | | | | | | | | | | |
Collapse
|
164
|
Costacou T, Rosano C, Aizenstein H, Mettenburg JM, Nunley K, Ferrell RE, Orchard TJ. The haptoglobin 1 allele correlates with white matter hyperintensities in middle-aged adults with type 1 diabetes. Diabetes 2015; 64:654-9. [PMID: 25213335 PMCID: PMC4303969 DOI: 10.2337/db14-0723] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although the haptoglobin (Hp) 1-1 genotype is associated with a lower coronary artery disease (CAD) risk in diabetes, we recently reported an increased stroke incidence in type 1 diabetes with Hp 1-1. We, thus, evaluated differences in earlier brain vascular abnormality markers by Hp using neuroimaging. Neuroimaging was completed in 94 participants of the Pittsburgh Epidemiology of Diabetes Complications study with Hp genotyping available (mean age, 49; duration, 41 years). White matter hyperintensities (WMH) volume, lacunar infarcts, and gray matter atrophy were quantified. Sixteen percent were homozygous for Hp 1 and 43% for Hp 2. A significant trend toward increased WMH was observed with greater duration and the number of Hp 1 alleles. Associations were strongest for the interhemispheric connecting fibers of the corpus callosum. Allowing for duration, sex, waist-to-hip ratio, HbA1c, systolic blood pressure, and lipids in models with backward elimination, results were similar. No significant differences by Hp were noted for atrophy or lacunar infarcts. Consistent with its direct association with stroke, the Hp 1-1 genotype is associated with higher WMH in this population. Further, including mechanistic, studies on the role of the Hp genotype in cerebrovascular disease and the implications for worsening cognitive function are needed.
Collapse
Affiliation(s)
- Tina Costacou
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Caterina Rosano
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Howard Aizenstein
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Joseph M Mettenburg
- Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Karen Nunley
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Robert E Ferrell
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
| | - Trevor J Orchard
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
165
|
Lei C, Wu B, Cao T, Zhang S, Liu M. Activation of the High-Mobility Group Box 1 Protein-Receptor for Advanced Glycation End-Products Signaling Pathway in Rats During Neurogenesis After Intracerebral Hemorrhage. Stroke 2015; 46:500-6. [PMID: 25538203 DOI: 10.1161/strokeaha.114.006825] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background and Purpose—
Following intracerebral hemorrhage (ICH), high-mobility group box 1 protein (HMGB1) may promote neurogenesis that supports functional recovery. How HMGB1 regulates or participates in this process is unclear, as are the pattern recognition receptors and signaling pathways involved.
Methods—
ICH was induced by injection of collagenase in Sprague–Dawley rats, which were treated 3 days later with saline, with the HMGB1 inhibitor ethyl pyruvate or with FPS-ZM1, an antagonist of the receptor for advanced glycation end-products. A Sham group was treated with saline solution instead of collagenase and then treated 3 days later with saline again or with ethyl pyruvate or N-benzyl-4-chloro-N-cyclohexylbenzamide (FPS-ZM1). Expression of the following proteins was measured by Western blot, immunohistochemistry, or immunofluorescence: HMGB1, receptor for advanced glycation end-products, toll-like receptor (TLR)-2, TLR4, brain-derived neurotrophic factor, and matrix metalloproteinase-9. The number of cells positive for 5-bromo-2-deoxyuridine or doublecortin was determined by immunohistochemistry and immunofluorescence.
Results—
Levels of HMGB1, receptor for advanced glycation end-products, TLR4, TLR2, brain-derived neurotrophic factor, and matrix metalloproteinase-9 were significantly higher 14 days after ICH than at baseline, as were the numbers of 5-bromo-2-deoxyuridine- or doublecortin-positive cells. At the same time, HMGB1 moved from the nucleus into the cytoplasm. Administering ethyl pyruvate significantly reduced all these ICH-induced increases, except the increase in TLR4 and TLR2. Administering FPS-ZM1 reduced the ICH-induced increases in the expression of brain-derived neurotrophic factor and matrix metalloproteinase-9 and in the numbers of 5-bromo-2-deoxyuridine- or doublecortin-positive cells.
Conclusions—
These findings suggest that HMGB1 acts via the receptor for advanced glycation end-products signaling pathway to promote neurogenesis in later phases of ICH.
Collapse
Affiliation(s)
- Chunyan Lei
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Bo Wu
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Tian Cao
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Shuting Zhang
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Ming Liu
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
166
|
Detante O, Jaillard A, Moisan A, Barbieux M, Favre I, Garambois K, Barbier E, Hommel M. Fisiopatologia dell’ischemia cerebrale. Neurologia 2015. [DOI: 10.1016/s1634-7072(14)69823-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
167
|
Zhu J, Liu Q, Jiang Y, Wu L, Xu G, Liu X. Enhanced angiogenesis promoted by human umbilical mesenchymal stem cell transplantation in stroked mouse is Notch1 signaling associated. Neuroscience 2015; 290:288-99. [PMID: 25637797 DOI: 10.1016/j.neuroscience.2015.01.038] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/18/2014] [Accepted: 01/08/2015] [Indexed: 12/16/2022]
Abstract
Cellular therapy has provided hope for restoring neurological function post stroke through promoting endogenous neurogenesis, angiogenesis and synaptogenesis. The current study was based on the observation that transplantation of human umbilical cord mesenchymal stem cells (hUCMSCs) promoted the neurological function improvement in stroked mice and meanwhile enhanced angiogenesis in the stroked hemisphere. Grafted hUCMSCs secreted human vascular endothelial growth factor A (VEGF-A). Notch1 signaling was activated after stroke and also in the grafted hUCMSCs. To address the potential mechanism that might mediate such pro-angiogenic effect, we established a hUCMSC-neuron co-culture system. Neurons were subjected to oxygen glucose deprivation (OGD) injury before co-culturing to mimic the in vivo cell transplantation. Consistent with the in vivo data, co-culture medium claimed from hUCMSC-OGD neuron co-culture system significantly promoted the capillary-like tube formation of brain-derived endothelial cells. Moreover, coincident with our in vivo data, Notch 1 signaling activation was detected in hUCMSCs after co-cultured with OGD neurons as demonstrated by the up-regulation of key Notch1 signaling components Notch1 and Notch1 intercellular domain (NICD). In addition, OGD-neuron co-culture also increased the VEGF-A production by hUCMSCs. To verify whether Notch1 activation was involved in the pro-angiogenic effect, γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) was added into the co-culture medium before co-culture. It turned out that DAPT significantly prevented the Notch1 activation in hUCMSCs after co-culture with OGD neurons. More importantly, the pro-angiogenic effect of hUCMSCs was remarkably abolished by DAPT addition as demonstrated by inhibited capillary-like tube formation and less VEGF-A production. Regarding how Notch1 signaling was linked with VEGF-A secretion, we provided some clue that Notch1 effector Hes1 mRNA expression was significantly up-regulated by OGD-neuron co-culturing and down-regulated after additional treatment of DAPT. In summary, our data provided evidence that the VEGF-A secretion from hUCMSCs after being triggered by OGD neurons is Notch1 signaling associated. This might be a possible mechanism that contributes to the angiogenic effect of hUCMSC transplantation in stroked brain.
Collapse
Affiliation(s)
- J Zhu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - Q Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - Y Jiang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - L Wu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - G Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - X Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| |
Collapse
|
168
|
Detante O, Jaillard A, Moisan A, Barbieux M, Favre I, Garambois K, Hommel M, Remy C. Biotherapies in stroke. Rev Neurol (Paris) 2014; 170:779-98. [DOI: 10.1016/j.neurol.2014.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/29/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
|
169
|
Moniche F, Montaner J, Gonzalez-Marcos JR, Carmona M, Piñero P, Espigado I, Cayuela A, Escudero I, de la Torre-Laviana FJ, Boada C, Rosell A, Mayol A, Jimenez MD, Gil-Peralta A, Gonzalez A. Intra-arterial bone marrow mononuclear cell transplantation correlates with GM-CSF, PDGF-BB, and MMP-2 serum levels in stroke patients: results from a clinical trial. Cell Transplant 2014; 23 Suppl 1:S57-64. [PMID: 25335778 DOI: 10.3727/096368914x684934] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bone marrow mononuclear cell (BM-MNC) intra-arterial transplantation improves recovery in experimental models of ischemic stroke through secretion of cytokines and growth factors (GFs), enhancing neoangiogenesis, and enhancing neuroplasticity. In this study, we tested whether BM-MNC transplantation in stroke patients induces changes in serum levels of cytokines and GFs. A phase I/II trial was conducted in middle cerebral artery (MCA) stroke patients with autologous intra-arterial BM-MNC transplantation between 5 and 9 days after stroke. Follow-up was done for up to 6 months. Eight cases and nine controls were included, and the serum levels of granulocyte-macrophage colony-stimulating factor (GM-CSF), platelet-derived growth factor-BB (PDGF-BB), β nerve growth factor (β-NGF), and matrix metalloproteinases 2 (MMP-2) and 9 (MMP-9) were measured before and 4, 8, and 90 days after transplantation. The correlation of these serum levels with dose of cells and clinical outcomes was studied. A total of 1.59 × 10(8) (±1.21 × 10(8)) BM-MNCs were injected in cases; of them 3.38 × 10(6) (±2.33 × 10(6)) were CD34(+) cells. There was a positive correlation between total BM-MNCs injected and levels of GM-CSF and PDGF-BB at 90 days after transplantation (r = 0.929, p = 0.001 and r = 0.714, p = 0.047, respectively), and a negative correlation between total CD34(+) cells injected and MMP-2 levels at 4 days after transplantation (r = -0.786, p = 0.036). Lower plasma levels of MMP-2 at 4 days and higher levels of PDGF-BB at 90 days were associated with better functional outcomes during follow-up (p = 0.019 and p = 0.037, respectively). When administered intra-arterially in subacute MCA stroke patients, BM-MNCs seem to induce changes in serum levels of GM-CSF, PDGF-BB, and MMP-2, even 3 months after transplantation, which could be associated with better functional outcomes. This manuscript is published as part of the International Association of Neurorestoratology (IANR) special issue of Cell Transplantation.
Collapse
Affiliation(s)
- Francisco Moniche
- Department of Neurology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Chou CH, Sinden JD, Couraud PO, Modo M. In vitro modeling of the neurovascular environment by coculturing adult human brain endothelial cells with human neural stem cells. PLoS One 2014; 9:e106346. [PMID: 25187991 PMCID: PMC4154686 DOI: 10.1371/journal.pone.0106346] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 08/01/2014] [Indexed: 11/18/2022] Open
Abstract
Brain and vascular cells form a functionally integrated signalling network that is known as the neurovascular unit (NVU). The signalling (autocrine, paracrine and juxtacrine) between different elements of this unit, especially in humans, is difficult to disentangle in vivo. Developing representative in vitro models is therefore essential to better understand the cellular interactions that govern the neurovascular environment. We here describe a novel approach to assay these cellular interactions by combining a human adult cerebral microvascular endothelial cell line (hCMEC/D3) with a fetal ganglionic eminence-derived neural stem cell (hNSC) line. These cell lines provide abundant homogeneous populations of cells to produce a consistently reproducible in vitro model of endothelial morphogenesis and the ensuing NVU. Vasculature-like structures (VLS) interspersed with patches of differentiating neural cells only occurred when hNSCs were seeded onto a differentiated endothelium. These VLS emerged within 3 days of coculture and by day 6 were stabilizing. After 7 days of coculture, neuronal differentiation of hNSCs was increased 3-fold, but had no significant effect on astrocyte or oligodendrocyte differentiation. ZO1, a marker of adherens and tight junctions, was highly expressed in both undifferentiated and differentiated endothelial cells, but the adherens junction markers CD31 and VE-cadherin were significantly reduced in coculture by approximately 20%. A basement membrane, consisting of laminin, vitronectin, and collagen I and IV, separated the VLS from neural patches. This simple assay can assist in elucidating the cellular and molecular signaling involved in the formation of VLS, as well as the enhancement of neuronal differentiation through endothelial signaling.
Collapse
Affiliation(s)
- Chung-Hsing Chou
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
- Kings College London, Institute of Psychiatry, Department of Neuroscience, London, United Kingdom
- Tri-service General Hospital, Department of Neurology, National Defense Medical Centre, Taipei, Taiwan
| | | | - Pierre-Olivier Couraud
- INSERM U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Michel Modo
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh, Department of Bioengineering, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
171
|
Xu X, Kriegel AJ, Jiao X, Liu H, Bai X, Olson J, Liang M, Ding X. miR-21 in ischemia/reperfusion injury: a double-edged sword? Physiol Genomics 2014; 46:789-97. [PMID: 25159851 DOI: 10.1152/physiolgenomics.00020.2014] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are endogenous, small RNA molecules that suppress expression of targeted mRNA. miR-21, one of the most extensively studied miRNAs, is importantly involved in divergent pathophysiological processes relating to ischemia/reperfusion (I/R) injury, such as inflammation and angiogenesis. The role of miR-21 in renal I/R is complex, with both protective and pathological pathways being regulated by miR-21. Preconditioning-induced upregulation of miR-21 contributes to the protection against subsequent renal I/R injury through the targeting of genes such as the proapoptotic gene programmed cell death 4 and interactions between miR-21 and hypoxia-inducible factor. Conversely, long-term elevation of miR-21 may be detrimental to the organ by promoting the development of renal interstitial fibrosis following I/R injury. miR-21 is importantly involved in several pathophysiological processes related to I/R injury including inflammation and angiogenesis as well as the biology of stem cells that could be used to treat I/R injury; however, the effect of miR-21 on these processes in renal I/R injury remains to be studied.
Collapse
Affiliation(s)
- Xialian Xu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoyan Jiao
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Hong Liu
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jessica Olson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoqiang Ding
- Division of Nephrology, Fudan University Zhongshan Hospital, Shanghai, Peoples Republic of China; Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, Peoples Republic of China; Kidney and Dialysis Institute of Shanghai, Shanghai, Peoples Republic of China; and Kidney and Blood Purification Laboratory of Shanghai, Shanghai, Peoples Republic of China
| |
Collapse
|
172
|
Merson TD, Bourne JA. Endogenous neurogenesis following ischaemic brain injury: insights for therapeutic strategies. Int J Biochem Cell Biol 2014; 56:4-19. [PMID: 25128862 DOI: 10.1016/j.biocel.2014.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/18/2014] [Accepted: 08/04/2014] [Indexed: 01/19/2023]
Abstract
Ischaemic stroke is among the most common yet most intractable types of central nervous system (CNS) injury in the adult human population. In the acute stages of disease, neurons in the ischaemic lesion rapidly die and other neuronal populations in the ischaemic penumbra are vulnerable to secondary injury. Multiple parallel approaches are being investigated to develop neuroprotective, reparative and regenerative strategies for the treatment of stroke. Accumulating evidence indicates that cerebral ischaemia initiates an endogenous regenerative response within the adult brain that potentiates adult neurogenesis from populations of neural stem and progenitor cells. A major research focus has been to understand the cellular and molecular mechanisms that underlie the potentiation of adult neurogenesis and to appreciate how interventions designed to modulate these processes could enhance neural regeneration in the post-ischaemic brain. In this review, we highlight recent advances over the last 5 years that help unravel the cellular and molecular mechanisms that potentiate endogenous neurogenesis following cerebral ischaemia and are dissecting the functional importance of this regenerative mechanism following brain injury. This article is part of a Directed Issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
Affiliation(s)
- Tobias D Merson
- Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, 30 Royal Parade, Parkville, VIC 3010, Australia.
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, Building 75, Level 1 North STRIP 1, Clayton, VIC 3800, Australia.
| |
Collapse
|
173
|
Ishrat T, Pillai B, Soliman S, Fouda AY, Kozak A, Johnson MH, Ergul A, Fagan SC. Low-dose candesartan enhances molecular mediators of neuroplasticity and subsequent functional recovery after ischemic stroke in rats. Mol Neurobiol 2014; 51:1542-53. [PMID: 25084762 DOI: 10.1007/s12035-014-8830-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/22/2014] [Indexed: 01/19/2023]
Abstract
We have previously reported that angiotensin type 1 receptor (AT1R) blockade with candesartan exerts neurovascular protection after experimental cerebral ischemia. Here, we tested the hypothesis that a low, subhypotensive dose of candesartan enhances neuroplasticity and subsequent functional recovery through enhanced neurotrophic factor expression in rats subjected to ischemia reperfusion injury. Male Wistar rats (290-300 g) underwent 90 min of middle cerebral artery occlusion (MCAO) and received candesartan (0.3 mg/kg) or saline at reperfusion and then once every 24 h for 7 days. Functional deficits were assessed in a blinded manner at 1, 3, 7, and 14 days after MCAO. Animals were sacrificed 14-day post-stroke and the brains perfused for infarct size by cresyl violet. Western blot and immunohistochemistry were used to assess the expression of growth factors and synaptic proteins. Candesartan-treated animals showed a significant reduction in the infarct size [t (13) = -5.5, P = 0.0001] accompanied by functional recovery in Bederson [F (1, 13) = 7.9, P = 0.015], beam walk [F (1, 13) = 6.7, P = 0.023], grip strength [F (1, 13) = 15.2, P = 0.0031], and rotarod performance [F (1, 14) = 29.8, P < 0.0001]. In addition, candesartan-treated animals showed significantly higher expression of active metalloproteinase-3 (MMP-3), laminin, and angiopoietin-1 (Ang-1). The expression of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) and its receptor was significantly increased in the animals treated with candesartan. Also, we observed significant increases in neuroplasticity markers, synaptophysin, and PSD-95. These results indicate that low-dose candesartan had a large and enduring effect on measures of plasticity, and this accompanied the functional recovery after ischemic stroke.
Collapse
|
174
|
Austin MW, Ploughman M, Glynn L, Corbett D. Aerobic exercise effects on neuroprotection and brain repair following stroke: a systematic review and perspective. Neurosci Res 2014; 87:8-15. [PMID: 24997243 DOI: 10.1016/j.neures.2014.06.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/04/2014] [Accepted: 06/24/2014] [Indexed: 01/02/2023]
Abstract
Aerobic exercise (AE) enhances neuroplasticity and improves functional outcome in animal models of stroke, however the optimal parameters (days post-stroke, intensity, mode, and duration) to influence brain repair processes are not known. We searched PubMed, CINAHL, PsychInfo, the Cochrane Library, and the Central Register of Controlled Clinical Trials, using predefined criteria, including all years up to July 2013 (English language only). Clinical studies were included if participants had experienced an ischemic or hemorrhagic stroke. We included animal studies that utilized any method of global or focal ischemic stroke or intracerebral hemorrhage. Any intervention utilizing AE-based activity with the intention of improving cardiorespiratory fitness was included. Of the 4250 titles returned, 47 studies (all in animal models) met criteria and measured the effects of exercise on brain repair parameters (lesion volume, oxidative damage, inflammation and cell death, neurogenesis, angiogenesis and markers of stress). Our synthesized findings show that early-initiated (24-48h post-stroke) moderate forced exercise (10m/min, 5-7 days per week for about 30min) reduced lesion volume and protected perilesional tissue against oxidative damage and inflammation at least for the short term (4 weeks). The applicability and translation of experimental exercise paradigms to clinical trials are discussed.
Collapse
Affiliation(s)
- Mark W Austin
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Michelle Ploughman
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada.
| | - Lindsay Glynn
- Health Sciences Library, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Dale Corbett
- Canadian Partnership for Stroke Recovery and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
175
|
Qin W, Li Z, Luo S, Wu R, Pei Z, Huang R. Exogenous fractalkine enhances proliferation of endothelial cells, promotes migration of endothelial progenitor cells and improves neurological deficits in a rat model of ischemic stroke. Neurosci Lett 2014; 569:80-4. [PMID: 24704182 DOI: 10.1016/j.neulet.2014.03.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/21/2014] [Accepted: 03/22/2014] [Indexed: 11/18/2022]
Abstract
Fractalkine/CX3CL1, also called neurotactin, has been described as an angiogenic agent, and its expression is up-regulated in the penumbra after ischemia. This study was conducted to investigate the neovascular potential of fractalkine on rat models of transient middle cerebral artery occlusion (MCAO). Rats receiving intracerebroventricular injections of fractalkine were found to have improved neurological deficits, reduced cerebral infarct size and increased neuron survival for both doses (100ng and 1μg). Fractalkine exerted angiogenic effects that showed dose-dependent higher vascular densities in the peri-infarct area. Furthermore, exogenous fractalkine increased the proliferation of endothelial cells in a dose-dependent manner and enhanced the migration of endothelial progenitor cells at the higher dose (1μg) in ischemic penumbra. In conclusion, intracerebroventricular administration of fractalkine reduces ischemic damage by promoting neuroprotection and by inducing endothelial cell proliferation and endothelial progenitor cell migration, thereby enhancing neovascularization in the peri-infarct region.
Collapse
Affiliation(s)
- Wenjing Qin
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China
| | - Zhendong Li
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China.
| | - Shijian Luo
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China
| | - Rui Wu
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China
| | - Zhong Pei
- Department of Neurology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Ruxun Huang
- Department of Neurology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
176
|
Ergul A, Abdelsaid M, Fouda AY, Fagan SC. Cerebral neovascularization in diabetes: implications for stroke recovery and beyond. J Cereb Blood Flow Metab 2014; 34:553-63. [PMID: 24496174 PMCID: PMC3982092 DOI: 10.1038/jcbfm.2014.18] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 01/30/2023]
Abstract
Neovascularization is an innate physiologic response by which tissues respond to various stimuli through collateral remodeling (arteriogenesis) and new vessel formation from existing vessels (angiogenesis) or from endothelial progenitor cells (vasculogenesis). Diabetes has a major impact on the neovascularization process but the response varies between different organ systems. While excessive angiogenesis complicates diabetic retinopathy, impaired neovascularization contributes to coronary and peripheral complications of diabetes. How diabetes influences cerebral neovascularization remained unresolved until recently. Diabetes is also a major risk factor for stroke and poor recovery after stroke. In this review, we discuss the impact of diabetes, stroke, and diabetic stroke on cerebral neovascularization, explore potential mechanisms involved in diabetes-mediated neovascularization as well as the effects of the diabetic milieu on poststroke neovascularization and recovery, and finally discuss the clinical implications of these effects.
Collapse
Affiliation(s)
- Adviye Ergul
- 1] Charlie Norwood VA Medical Center, Augusta, Georgia, USA [2] Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA [3] Center for Pharmacy and Experimental Therapeutics, Medical College of Georgia and University of Georgia College of Pharmacy, Augusta, Georgia, USA
| | - Mohammed Abdelsaid
- 1] Charlie Norwood VA Medical Center, Augusta, Georgia, USA [2] Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | - Abdelrahman Y Fouda
- 1] Charlie Norwood VA Medical Center, Augusta, Georgia, USA [2] Center for Pharmacy and Experimental Therapeutics, Medical College of Georgia and University of Georgia College of Pharmacy, Augusta, Georgia, USA
| | - Susan C Fagan
- 1] Charlie Norwood VA Medical Center, Augusta, Georgia, USA [2] Center for Pharmacy and Experimental Therapeutics, Medical College of Georgia and University of Georgia College of Pharmacy, Augusta, Georgia, USA [3] Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
177
|
Venna VR, Li J, Hammond MD, Mancini NS, McCullough LD. Chronic metformin treatment improves post-stroke angiogenesis and recovery after experimental stroke. Eur J Neurosci 2014; 39:2129-38. [PMID: 24649970 DOI: 10.1111/ejn.12556] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 02/06/2023]
Abstract
Metformin is currently the first-line treatment drug for type 2 diabetes. Metformin is a well-known activator of AMP-activated protein kinase (AMPK). In experimental studies, metformin has been shown to exert direct vascular effects by increasing vascular endothelial growth factor expression and improving microvascular density. As stroke is the leading cause of long-term disability and angiogenesis is implicated as an important mechanism in functional recovery, we hypothesized that chronic metformin treatment would improve post-stroke functional recovery by enhancing functional microvascular density. For this study, C57BL/6N male mice were subjected to a 60-min middle cerebral artery occlusion, and were given 50 mg/kg/day metformin beginning 24 h post-stroke for 3 weeks. Behavioral recovery was assessed using adhesive-tape removal and the apomorphine-induced turning test. The role of angiogenesis was assessed by counting vessel branch points from fluorescein-conjugated lectin-perfused brain sections. Importantly even if metformin treatment was initiated 24 h after injury it enhanced recovery and significantly improved stroke-induced behavioral deficits. This recovery occurred in parallel with enhanced angiogenesis and with restoration of endogenous cerebral dopaminergic tone and revascularization of ischemic tissue. We assessed if the effects on recovery and angiogenesis were mediated by AMPK. When tested in AMPK α-2 knockout mice, we found that metformin treatment did not have the same beneficial effects on recovery and angiogenesis, suggesting that metformin-induced angiogenic effects are mediated by AMPK. The results from this study suggest that metformin mediates post-stroke recovery by enhancing angiogenesis, and these effects are mediated by AMPK signaling.
Collapse
Affiliation(s)
- Venugopal R Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | |
Collapse
|
178
|
Mao L, Huang M, Chen SC, Li YN, Xia YP, He QW, Wang MD, Huang Y, Zheng L, Hu B. Endogenous endothelial progenitor cells participate in neovascularization via CXCR4/SDF-1 axis and improve outcome after stroke. CNS Neurosci Ther 2014; 20:460-8. [PMID: 24581269 DOI: 10.1111/cns.12238] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/17/2014] [Accepted: 01/18/2014] [Indexed: 11/26/2022] Open
Abstract
AIM To study whether endogenous endothelial progenitor cells (EPCs) are involved in neovascularization after stroke. MATERIALS AND METHODS Animal stroke models were established by subjecting male SD rats to permanent middle cerebral artery occlusion (pMCAO). Vessels in ischemic boundary zone (IBZ) were stained with antibody against laminin at 1 to 21 days after pMCAO. EPCs and newly formed vessels were identified by staining with special markers. After inhibiting recruitment of EPCs with AMD3100, a CXCR4 antagonist, endogenous EPCs, capillary density, cerebral blood flow (CBF) in IBZ, and neurobehavioral functions were assessed by staining, FITC-dextran, laser-Doppler perfusion monitor, and neurologic severity score. RESULTS After pMCAO, vessels were found in IBZ at day 3, reaching a peak at day 14. The change in number of laminin-positive cells showed a similar pattern with that of vessels. Apart from few endothelial cells, most of laminin-positive cells were endogenous EPCs. After treatment with AMD3100, the number of endogenous EPCs, capillary density, and CBF in IBZ were significantly reduced, and neurobehavioral functions were worse as compared with the normal saline group. CONCLUSIONS Our findings suggested that endogenous EPCs participated in the neovascularization via CXCR4/SDF-1 axis after pMCAO and mobilizing endogenous EPCs could be a treatment alternative for stroke.
Collapse
Affiliation(s)
- Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Ju R, Wen Y, Gou R, Wang Y, Xu Q. The Experimental Therapy on Brain Ischemia by Improvement of Local Angiogenesis with Tissue Engineering in the Mouse. Cell Transplant 2014; 23 Suppl 1:S83-95. [DOI: 10.3727/096368914x684998] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neural restoration has proven to be difficult after brain stroke, especially in its chronic stage. This is mainly due to the generation of an unpropitious niche in the injured area, including loss of vascular support but production of numerous inhibitors against neuronal regeneration. Reconstruction of a proper niche for promoting local angiogenesis, therefore, should be a key approach for neural restoration after stroke. In the present study, a new biomaterial composite that could be implanted in the injured area of the brain was created for experimental therapy of brain ischemia in the mouse. This composite was made using a hyaluronic acid (HA)-based biodegradable hydrogel scaffold, mixed with poly(lactic- co-glycolic acid) (PLGA) microspheres containing vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang1), two factors that stimulate angiogenesis. In addition, the antibody of Nogo receptor (NgR-Ab), which can bind to multiple inhibitory myelin proteins and promote neural regeneration, was covalently attached to the hydrogel, making the hydrogel more bioactive and suitable for neural survival. This composite (HA–PLGA) was implanted into the mouse model with middle cerebral artery occlusion (MCAO) to explore a new approach for restoration of brain function after ischemia. A good survival and proliferation of human umbilical artery endothelial cells (HUAECs) and neural stem cells (NSCs) were seen on the HA hydrogel with PLGA microspheres in vitro. This new material was shown to have good compatibility with the brain tissue and inhibition to gliosis and inflammation after its implantation in the normal or ischemic brain of mice. Particularly, good angiogenesis was found around the implanted HA–PLGA hydrogel, and the mouse models clearly showed a behavioral improvement. The results in this present study indicate, therefore, that the HA–PLGA hydrogel is a promising material, which is able to induce angiogenesis in the ischemic region by releasing VEGF and Ang1, thus creating a suitable niche for neural restoration in later stages of stroke. This manuscript is published as part of the International Association of Neurorestoratology (IANR) special issue of Cell Transplantation.
Collapse
Affiliation(s)
- Rongkai Ju
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| | - Yujun Wen
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| | - Rongbin Gou
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| | - Qunyuan Xu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| |
Collapse
|
180
|
Abstract
Angiogenesis, the sprouting of new capillaries from existing blood vessels, accompanies clinical and experimental stroke and is focused particularly in the salvageable ischemic border zone. As this endogenous angiogenic response correlates positively with clinical prognosis, a more complete understanding of the underlying molecular mechanisms and timing of these events may help in the design of novel therapies for vascular regeneration after stroke. In this review we discuss endogenous protective mechanisms, including angiogenesis and vasculogenesis, and underlying molecular mechanisms. We also consider the feasibility of angiogenic therapy for stroke and its optimal timing.
Collapse
|
181
|
Liu X, Ye R, Yan T, Yu SP, Wei L, Xu G, Fan X, Jiang Y, Stetler RA, Liu G, Chen J. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol 2013; 115:92-115. [PMID: 24333397 DOI: 10.1016/j.pneurobio.2013.11.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Cell therapy is emerging as a viable therapy to restore neurological function after stroke. Many types of stem/progenitor cells from different sources have been explored for their feasibility and efficacy for the treatment of stroke. Transplanted cells not only have the potential to replace the lost circuitry, but also produce growth and trophic factors, or stimulate the release of such factors from host brain cells, thereby enhancing endogenous brain repair processes. Although stem/progenitor cells have shown a promising role in ischemic stroke in experimental studies as well as initial clinical pilot studies, cellular therapy is still at an early stage in humans. Many critical issues need to be addressed including the therapeutic time window, cell type selection, delivery route, and in vivo monitoring of their migration pattern. This review attempts to provide a comprehensive synopsis of preclinical evidence and clinical experience of various donor cell types, their restorative mechanisms, delivery routes, imaging strategies, future prospects and challenges for translating cell therapies as a neurorestorative regimen in clinical applications.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Neurology, Tianjin General Hospital, Tianjin University School of Medicine, Tianjin, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xinying Fan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
182
|
Salat DH. Imaging small vessel-associated white matter changes in aging. Neuroscience 2013; 276:174-86. [PMID: 24316059 DOI: 10.1016/j.neuroscience.2013.11.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 11/21/2013] [Accepted: 11/21/2013] [Indexed: 01/18/2023]
Abstract
Alterations in cerebrovascular structure and function may underlie the most common age-associated cognitive, psychiatric, and neurological conditions presented by older adults. Although much remains to understand, existing research suggests several age-associated detrimental conditions may be mediated through sometimes subtle small vessel-induced damage to the cerebral white matter. Here we review a selected portion of the vast work that demonstrates links between changes in vascular and neural health as a function of advancing age, and how even changes in low-to-moderate risk individuals, potentially beginning early in the adult age-span, may have an important impact on functional status in late life.
Collapse
Affiliation(s)
- D H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Department of Radiology, Charlestown, MA, USA; Neuroimaging Research for Veterans Center, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
183
|
Hamel D, Sanchez M, Duhamel F, Roy O, Honoré JC, Noueihed B, Zhou T, Nadeau-Vallée M, Hou X, Lavoie JC, Mitchell G, Mamer OA, Chemtob S. G-protein-coupled receptor 91 and succinate are key contributors in neonatal postcerebral hypoxia-ischemia recovery. Arterioscler Thromb Vasc Biol 2013; 34:285-93. [PMID: 24285580 DOI: 10.1161/atvbaha.113.302131] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Prompt post-hypoxia-ischemia (HI) revascularization has been suggested to improve outcome in adults and newborn subjects. Other than hypoxia-inducible factor, sensors of metabolic demand remain largely unknown. During HI, anaerobic respiration is arrested resulting in accumulation of carbohydrate metabolic intermediates. As such succinate readily increases, exerting its biological effects via a specific receptor, G-protein-coupled receptor (GPR) 91. We postulate that succinate/GPR91 enhances post-HI vascularization and reduces infarct size in a model of newborn HI brain injury. APPROACH AND RESULTS The Rice-Vannucci model of neonatal HI was used. Succinate was measured by mass spectrometry, and microvascular density was evaluated by quantification of lectin-stained cryosection. Gene expression was evaluated by real-time polymerase chain reaction. Succinate levels rapidly increased in the penumbral region of brain infarcts. GPR91 was foremost localized not only in neurons but also in astrocytes. Microvascular density increased at 96 hours after injury in wild-type animals; it was diminished in GPR91-null mice leading to an increased infarct size. Stimulation with succinate led to an increase in growth factors implicated in angiogenesis only in wild-type mice. To explain the mode of action of succinate/GPR91, we investigated the role of prostaglandin E2-prostaglandin E receptor 4, previously proposed in neural angiogenesis. Succinate-induced vascular endothelial growth factor expression was abrogated by a cyclooxygenase inhibitor and a selective prostaglandin E receptor 4 antagonist. This antagonist also abolished succinate-induced neovascularization. CONCLUSIONS We uncover a dominant metabolic sensor responsible for post-HI neurovascular adaptation, notably succinate/GPR91, acting via prostaglandin E2-prostaglandin E receptor 4 to govern expression of major angiogenic factors. We propose that pharmacological intervention targeting GPR91 could improve post-HI brain recovery.
Collapse
Affiliation(s)
- David Hamel
- From the Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, Quebec, Canada (D.H., O.R., J.C.H., T.Z., X.H., J.-C.L., G.A.M., S.C.); Departments of Pharmacology (D.H., F.D., S.C.) and Biomedical Sciences (O.R., M.N.-V.), Université de Montréal, Montréal, Quebec, Canada; and Department of Pharmacology and Therapeutics (M.S., B.N., S.C.), Goodman Cancer Research and Metabolomics Core Facility (O.A.M.), McGill University, Montréal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Vemuganti R. All's well that transcribes well: non-coding RNAs and post-stroke brain damage. Neurochem Int 2013; 63:438-49. [PMID: 23954844 PMCID: PMC3805745 DOI: 10.1016/j.neuint.2013.07.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/26/2013] [Accepted: 07/28/2013] [Indexed: 12/13/2022]
Abstract
The mammalian genome is replete with various classes of non-coding (nc) RNA genes. Many of them actively transcribe, and their relevance to CNS diseases is just beginning to be understood. CNS is one of the organs in the body that shows very high ncRNAs activity. Recent studies demonstrated that cerebral ischemia rapidly changes the expression profiles of different classes of ncRNAs: including microRNA, long noncoding RNA and piwi-interacting RNA. Several studies further showed that post-ischemic neuronal death and/or plasticity/regeneration can be altered by modulating specific microRNAs. These studies are of interest for therapeutic development as they may contribute to identifying new ncRNA targets that can be modulated to prevent secondary brain damage after stroke.
Collapse
Affiliation(s)
- Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
185
|
Alvarez-Sabín J, Román GC. The role of citicoline in neuroprotection and neurorepair in ischemic stroke. Brain Sci 2013; 3:1395-414. [PMID: 24961534 PMCID: PMC4061873 DOI: 10.3390/brainsci3031395] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/10/2013] [Accepted: 08/14/2013] [Indexed: 12/12/2022] Open
Abstract
Advances in acute stroke therapy resulting from thrombolytic treatment, endovascular procedures, and stroke units have improved significantly stroke survival and prognosis; however, for the large majority of patients lacking access to advanced therapies stroke mortality and residual morbidity remain high and many patients become incapacitated by motor and cognitive deficits, with loss of independence in activities of daily living. Therefore, over the past several years, research has been directed to limit the brain lesions produced by acute ischemia (neuroprotection) and to increase the recovery, plasticity and neuroregenerative processes that complement rehabilitation and enhance the possibility of recovery and return to normal functions (neurorepair). Citicoline has therapeutic effects at several stages of the ischemic cascade in acute ischemic stroke and has demonstrated efficiency in a multiplicity of animal models of acute stroke. Long-term treatment with citicoline is safe and effective, improving post-stroke cognitive decline and enhancing patients' functional recovery. Prolonged citicoline administration at optimal doses has been demonstrated to be remarkably well tolerated and to enhance endogenous mechanisms of neurogenesis and neurorepair contributing to physical therapy and rehabilitation.
Collapse
Affiliation(s)
- José Alvarez-Sabín
- Neurovascular Unit, Department of Neurology, Universitat Autónoma de Barcelona, 119-129 Passeig de la Vall d'Hebron, Barcelona 08035, Spain.
| | - Gustavo C Román
- Department of Neurology, Nantz National Alzheimer Center, Methodist Neurological Institute, Houston, TX 77030, USA.
| |
Collapse
|
186
|
Prakash R, Li W, Qu Z, Johnson MA, Fagan SC, Ergul A. Vascularization pattern after ischemic stroke is different in control versus diabetic rats: relevance to stroke recovery. Stroke 2013; 44:2875-82. [PMID: 23920018 DOI: 10.1161/strokeaha.113.001660] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND PURPOSE Pre-existing diabetes mellitus worsens brain functionality in ischemic stroke. We have previously shown that type 2 diabetic rats exhibit enhanced dysfunctional cerebral neovascularization and when these rats are subjected to cerebral ischemic reperfusion injury develop hemorrhagic transformation and greater neurological deficits. However, our knowledge of vascular and functional plasticity during the recovery phase of diabetic stroke is limited. This study tested the hypothesis that vascular repair is impaired in the poststroke period in diabetes mellitus, and this is associated with poor sensorimotor and cognitive function. We further hypothesized that glycemic control prevents impaired vascularization and improves functional outcome in diabetes mellitus. METHODS Vascularization was assessed in the ipsilateral and contralateral hemispheres in control, diabetes mellitus and diabetes mellitus plus metformin groups 14 days after ischemic reperfusion injury, as well as in respective sham controls. Three-dimensional reconstruction of the fluorescein isothiocyanate (FITC)-stained vasculature was achieved by confocal microscopy, and stereological parameters, including vascular volume and surface area, were measured. Astrogliosis was determined by glial fibrillary acidic protein staining. The relative rates of sensorimotor recovery, cognitive decline, and spontaneous activity were assessed. RESULTS Vascular density in the peri-infarct area was significantly reduced in diabetes mellitus, whereas there was reparative neovascularization in control rats. Astroglial swelling and reactivity were more pronounced in diabetic stroke compared with control stroke. Diabetes mellitus blunted sensorimotor recovery and also exacerbated anxiety-like symptoms and cognitive deficits. Glycemic control started after stroke partially prevented these changes. CONCLUSIONS Diabetes mellitus impairs poststroke reparative neovascularization and impedes the recovery. Glycemic control after stroke can improve neurovascular repair and improve functional outcome.
Collapse
Affiliation(s)
- Roshini Prakash
- From the Department of Physiology, Charlie Norwood Veterans Administration Medical Center, Augusta, GA (R.P., W.L., S.C.F., A.E.); Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA (R.P., S.C.F., A.E.); and Departments of Physiology (W.L., Z.Q., A.E.), and Biostatistics (M.A.J.), Georgia Regents University, Augusta, GA
| | | | | | | | | | | |
Collapse
|
187
|
Abstract
Angiogenesis is a key feature of central nervous system injury. A neovessel-derived signal mediated by prostacyclin triggers axonal sprouting and functional recovery in a mouse model of inflammatory spinal cord injury (pages 1658-1664). Are such angiocrine signals relevant to neurovascular remodeling and recovery in other neurological contexts?
Collapse
|
188
|
NADPH oxidase and angiogenesis following endothelin-1 induced stroke in rats: role for nox2 in brain repair. Brain Sci 2013; 3:294-317. [PMID: 24961316 PMCID: PMC4061826 DOI: 10.3390/brainsci3010294] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/07/2013] [Accepted: 02/19/2013] [Indexed: 12/29/2022] Open
Abstract
NADPH oxidases contribute to brain injury, yet they may also have a role in brain repair, particularly in vascular signaling and angiogenesis. This study determined the temporal and spatial profile of NADPH oxidase subunit expression/activity concurrently with angiogenesis in the brain following transient ischemic stroke induced by prolonged constriction of the middle cerebral artery by perivascular injection of endothelin-1 in conscious Hooded Wistar rats (n = 47). VEGF mRNA expression was increased in the ipsilateral cortex and striatum between 6 h and 28 days post-stroke concurrently with a marked increase in Nox2 mRNA expression up to 7 days, and increased Nox4 mRNA expression detected between 7 and 28 days. Point counting of blood vessels using Metamorph imaging software showed increased vascular sprouting between 3 and 7 days after stroke with new vascular networks detected in the core infarct region by 14 days. Angiogenic blood vessels 3 and 7 days post-stroke were observed to co-localise with both Nox2 antibody and dihydroethidium fluorescence suggesting a role for Nox2 generated superoxide during the phase of vascular remodeling, whilst Nox4 expression was detected once new cerebral vessels had formed. These results indicate for the first time that ROS signaling through a cerebrovascular Nox2 NADPH oxidase may be important in initiating brain angiogenesis.
Collapse
|
189
|
Cerebral neovascularization and remodeling patterns in two different models of type 2 diabetes. PLoS One 2013; 8:e56264. [PMID: 23441170 PMCID: PMC3575336 DOI: 10.1371/journal.pone.0056264] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/07/2013] [Indexed: 11/19/2022] Open
Abstract
We previously reported intense pial cerebral collateralization and arteriogenesis in a mild and lean model of type 2 diabetes (T2D), Goto-Kakizaki (GK) rats. Increased cerebral neovascularization differed regionally and was associated with poor vessel wall maturity. Building upon these findings, the goals of this study were to determine whether a) glycemic control prevents this erratic cerebral neovascularization in the GK model, and b) this pathological neovascularization pattern occurs in Lepr(db/db) model, which is the most commonly used model of T2D for studies involving cerebral complications of diabetes. Vascular volume, surface area and structural parameters including microvessel/macrovessel ratio, non-FITC (fluorescein) perfusing vessel abundance, vessel tortuosity, and branch density were measured by 3D reconstruction of FITC stained vasculature in GK rats or Lepr(db/db) mice. GK rats exhibited an increase in all of these parameters, which were prevented by glycemic control with metformin. In Lepr(db/db) mice, microvascular density was increased but there was no change in nonFITC-perfusing vessels. Increased PA branch density was associated with reduced branch diameter. These results suggest that T2D leads to cerebral neovascularization and remodeling but some structural characteristics of newly formed vessels differ between these models of T2D. The prevention of dysfunctional cerebral neovascularization by early glucose control suggests that hyperglycemia is a mediator of this response.
Collapse
|
190
|
Lei C, Lin S, Zhang C, Tao W, Dong W, Hao Z, Liu M, Wu B. Activation of cerebral recovery by matrix metalloproteinase-9 after intracerebral hemorrhage. Neuroscience 2013. [DOI: 10.1016/j.neuroscience.2012.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
191
|
Yang R, Zhang Q, Wu Y, Dunn JF. Monitoring angiogenesis using a human compatible calibration for broadband near-infrared spectroscopy. JOURNAL OF BIOMEDICAL OPTICS 2013; 18:16011. [PMID: 23314579 PMCID: PMC3595713 DOI: 10.1117/1.jbo.18.1.016011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/06/2012] [Accepted: 12/06/2012] [Indexed: 05/20/2023]
Abstract
Angiogenesis is a hallmark of many conditions, including cancer, stroke, vascular disease, diabetes, and high-altitude exposure. We have previously shown that one can study angiogenesis in animal models by using total hemoglobin (tHb) as a marker of cerebral blood volume (CBV), measured using broadband near-infrared spectroscopy (bNIRS). However, the method was not suitable for patients as global anoxia was used for the calibration. Here we determine if angiogenesis could be detected using a calibration method that could be applied to patients. CBV, as a marker of angiogenesis, is quantified in a rat cortex before and after hypoxia acclimation. Rats are acclimated at 370-mmHg pressure for three weeks, while rats in the control group are housed under the same conditions, but under normal pressure. CBV increased in each animal in the acclimation group. The mean CBV (%volume/volume) is 3.49%± 0.43% (mean ± SD) before acclimation for the experimental group, and 4.76%± 0.29% after acclimation. The CBV for the control group is 3.28%± 0.75%, and 3.09%± 0.48% for the two measurements. This demonstrates that angiogenesis can be monitored noninvasively over time using a bNIRS system with a calibration method that is compatible with human use and less stressful for studies using animals.
Collapse
Affiliation(s)
- Runze Yang
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Qiong Zhang
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Ying Wu
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Jeff F. Dunn
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
- University of Calgary, Experimental Imaging Centre, Calgary, Alberta, Canada
- Address all correspondence: Jeff F. Dunn, University of Calgary, Department of Radiology, 3330 Hospital Drive, N.W., Calgary, AB T2N 4N1, Canada. Tel: 403-210-3886; E-mail:
| |
Collapse
|