101
|
Tixier E, Leconte C, Touzani O, Roussel S, Petit E, Bernaudin M. Adrenomedullin protects neurons against oxygen glucose deprivation stress in an autocrine and paracrine manner. J Neurochem 2008; 106:1388-403. [DOI: 10.1111/j.1471-4159.2008.05494.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
102
|
Gonzalez FF, Ferriero DM. Therapeutics for neonatal brain injury. Pharmacol Ther 2008; 120:43-53. [PMID: 18718848 DOI: 10.1016/j.pharmthera.2008.07.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Accepted: 07/08/2008] [Indexed: 01/19/2023]
Abstract
Neonatal brain injury is an important cause of death and neurodevelopmental delay. Multiple pathways of oxidant stress, inflammation, and excitotoxicity lead to both early and late phases of cell damage and death. Therapies targeting these different pathways have shown potential in protecting the brain from ongoing injury. More recent therapies, such as growth factors, have demonstrated an ability to increase cell proliferation and repair over longer periods of time. Even though hypothermia, which decreases cerebral metabolism and possibly affects other mechanisms, may show some benefit in particular cases, no widely effective therapeutic interventions for human neonates exist. In this review, we summarize recent findings in neuroprotection and neurogenesis for the immature brain, including combination therapy to optimize repair.
Collapse
Affiliation(s)
- Fernando F Gonzalez
- Department of Pediatrics, University of California-San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143, USA
| | | |
Collapse
|
103
|
Kim GH, Hahn DK, Kellner CP, Hickman ZL, Komotar RJ, Starke RM, Mack WJ, Mocco J, Solomon RA, Connolly ES. Plasma levels of vascular endothelial growth factor after treatment for cerebral arteriovenous malformations. Stroke 2008; 39:2274-9. [PMID: 18535271 DOI: 10.1161/strokeaha.107.512442] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The role of abnormal angiogenesis in the formation and progression of cerebral arteriovenous malformations (AVMs) is unclear. Previous studies have demonstrated increased local expression of vascular endothelial growth factor (VEGF) in AVM tissue and increased circulating levels of VEGF in AVM patients. We sought to further investigate the role of VEGF in AVM pathophysiology by examining changes in plasma VEGF levels in patients undergoing treatment for AVMs. METHODS Three serial blood samples were obtained from 13 AVM patients undergoing treatment: (1) before any treatment, (2) 24 hours postresection, and (3) 30 days postresection. Plasma VEGF concentrations were measured via commercially available enzyme-linked immunosorbent assay (ELISA). For controls, blood samples were obtained from 29 lumbar laminectomy patients. RESULTS The mean plasma VEGF level in AVM patients at baseline was 36.08+/-13.02 pg/mL, significantly lower than that of the control group (80.52+/-14.02 pg/mL, P=0.028). Twenty-four hours postresection, plasma VEGF levels dropped to 20.09+/-4.54 pg/mL, then increased to 66.81+/-26.45 pg/mL 30 days later (P=0.048). The mean plasma VEGF concentration 30 days after resection was no longer significantly different from the control group (P=0.33). CONCLUSIONS Plasma VEGF levels in 13 AVM patients were unexpectedly lower than controls, dropped early after AVM resection, then significantly increased 30 days later. These results support the key role of abnormal angiogenesis in AVM pathophysiology and suggest that a disruption in systemic VEGF expression may contribute to the natural history of these lesions.
Collapse
Affiliation(s)
- Grace H Kim
- Department of Neurological Surgery, Columbia College of Physicians and Surgeons, Neurological Institute of New York, 630 W 168th St, Room 5-454, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Andres RH, Guzman R, Ducray AD, Mordasini P, Gera A, Barth A, Widmer HR, Steinberg GK. Cell replacement therapy for intracerebral hemorrhage. Neurosurg Focus 2008; 24:E16. [DOI: 10.3171/foc/2008/24/3-4/e15] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
✓ Intracerebral hemorrhage (ICH), for which no effective treatment strategy is currently available, constitutes one of the most devastating forms of stroke. As a result, developing therapeutic options for ICH is of great interest to the medical community. The 3 potential therapies that have the most promise are cell replacement therapy, enhancing endogenous repair mechanisms, and utilizing various neuroprotective drugs. Replacement of damaged cells and restoration of function can be accomplished by transplantation of cells derived from different sources, such as embryonic or somatic stem cells, umbilical cord blood, and genetically modified cell lines. Early experimental data showing the benefits of cell transplantation on functional recovery after ICH have been promising. Nevertheless, several studies have focused on another therapeutic avenue, investigating novel ways to activate and direct endogenous repair mechanisms in the central nervous system, through exposure to specific neuronal growth factors or by inactivating inhibitory molecules. Lastly, neuroprotective drugs may offer an additional tool for improving neuronal survival in the perihematomal area. However, a number of scientific issues must be addressed before these experimental techniques can be translated into clinical therapy. In this review, the authors outline the recent advances in the basic science of treatment strategies for ICH.
Collapse
Affiliation(s)
- Robert H. Andres
- 1Department of Neurosurgery, Stanford University Medical Center, Stanford, California
- 2Departments of Neurosurgery and
| | - Raphael Guzman
- 1Department of Neurosurgery, Stanford University Medical Center, Stanford, California
| | | | - Pasquale Mordasini
- 2Departments of Neurosurgery and
- 3Neuroradiology, University of Berne, Inselspital, CH-3010 Berne, Switzerland; and
| | - Atul Gera
- 1Department of Neurosurgery, Stanford University Medical Center, Stanford, California
| | - Alain Barth
- 4Department of Neurosurgery, Medical University of Graz, A-8036 Graz, Austria
| | | | - Gary K. Steinberg
- 1Department of Neurosurgery, Stanford University Medical Center, Stanford, California
| |
Collapse
|
105
|
Kuluz J, Huang T, Watson B, Vannucci S. Stroke in the immature brain: review of pathophysiology and animal models of pediatric stroke. FUTURE NEUROLOGY 2008. [DOI: 10.2217/14796708.3.2.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pediatric stroke research presents many challenges. Relatively low incidence, need for age stratification, diverse etiologies, delays in diagnosis, lack of an established age-based stroke severity scale and outcome measures are only some of the issues that have prevented the implementation of clinical trials in infants and children with stroke. Experimental animal models of pediatric stroke, therefore, are critical to understanding the pathophysiology and management of ischemic brain damage in the immature brain, and provide the necessary platform for future clinical trials. In this review we discuss the pertinent clinical aspects of pediatric stroke, the pathophysiology of stroke in the developing brain and the animal models established to study basic mechanisms as well as translational issues in pediatric stroke.
Collapse
Affiliation(s)
- John Kuluz
- Associate Professor of Pediatrics, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960, Miami, FL 33101, USA
| | - Tingting Huang
- Post-Doctoral Research Associate, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960 Miami, FL 33101, USA
| | - Brant Watson
- Professor of Neurology, University of Miami, Department of Neurology (D4–5), Miller School of Medicine, PO Box 016960, Miami, FL 33136, USA
| | - Susan Vannucci
- Research Professor of Neuroscience in Pediatrics/Newborn Medicine, Weill Cornell Medical College, 525 East 68th Street, N-506, NY 10065, USA
| |
Collapse
|
106
|
|
107
|
Hayashi T, Wang XQ, Zhang HZ, Deguchi K, Nagotani S, Sehara Y, Tsuchiya A, Nagai M, Shoji M, Abe K. Induction of platelet derived-endothelial cell growth factor in the brain after ischemia. Neurol Res 2007; 29:463-8. [PMID: 17535565 DOI: 10.1179/016164107x164139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Platelet derived-endothelial cell growth factor (PD-ECGF) is a highly potent angiogenic factor. Although angiogenesis plays an active role in pathophysiology of stroke, the expression pattern of this molecule in ischemic brain has not been investigated. In the present study, therefore, we investigated the change of PD-ECGF expression in the brain after ischemia. METHODS Using male Wistar rats, the right middle cerebral artery was occluded by a nylon thread for 90 minutes. The animals were decapitated 3 hours, 1, 4 and 10 days after the reperfusion, and frozen sections were prepared. We then performed immunohistochemistry for PD-ECGF and identified the cell phenotype which strongly expressed it by fluorescent double staining. RESULTS In the sham-operated brain, only small numbers of cells slightly expressed PD-ECGF. The number of positively stained cells increased at the peri-ischemic area from hour 3 of reperfusion. Not only small-sized cells but also large-sized cells became stained. The number of stained cells further increased, and peaked at day 4 for large-sized cells and at day 10 as to small-sized cells. Fluorescent double staining revealed that both large-sized and small-sized cells were neurons, indicating that neurons are the main source of PD-ECGF production in the ischemic brain. DISCUSSION PD-ECGF has a strong angiogenic property without vascular permeability increasing effect. This molecule may have a therapeutic potential for ischemic stroke treatment.
Collapse
Affiliation(s)
- T Hayashi
- Department of Neurology, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikata-cho, Okayama 700-8558, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Heinzer S, Kuhn G, Krucker T, Meyer E, Ulmann-Schuler A, Stampanoni M, Gassmann M, Marti HH, Müller R, Vogel J. Novel three-dimensional analysis tool for vascular trees indicates complete micro-networks, not single capillaries, as the angiogenic endpoint in mice overexpressing human VEGF(165) in the brain. Neuroimage 2007; 39:1549-58. [PMID: 18077185 DOI: 10.1016/j.neuroimage.2007.10.054] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 10/20/2007] [Accepted: 10/31/2007] [Indexed: 11/29/2022] Open
Abstract
To adequately supply tissues with oxygen and nutrients, the formation of functional vascular networks requires generation of normal, healthy vessels and their arrangement into an effective network architecture. While our knowledge about the development of single vessels significantly increased during the last years, mechanisms responsible for network formation are still poorly understood. This is probably due to the lack of suitable methods for quantification of structural properties of microvascular networks. Previously we showed that cerebral blood flow is not increased in mice exhibiting a 2- to 3-fold higher density of normal and perfused capillaries as a result of transgenic overexpression of the human vascular endothelial growth factor (VEGF(165)). Here we used vascular corrosion casting and hierarchical micro-computed tomography combined with a new network analysis tool to characterize the vascular architecture in gray and white matter of these mice. Our results indicate that VEGF overexpression leads to formation of additional micro-networks connected to higher order vessels rather than insertion of individual capillaries into the existing vessel structure. This implies that the smallest "angiogenic quantum", i.e. the final, stable result of angiogenesis and subsequent remodeling, is not a single microvessel, but a complete micro-network. In conclusion, high-resolution 3D imaging combined with network analysis can substantially improve our understanding of vascular architecture, beneficial for the development of therapeutic angiogenesis as a clinical tool for applications such as wound healing or treatment of ischemic diseases.
Collapse
Affiliation(s)
- Stefan Heinzer
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Nicoletti JN, Shah SK, McCloskey DP, Goodman JH, Elkady A, Atassi H, Hylton D, Rudge JS, Scharfman HE, Croll SD. Vascular endothelial growth factor is up-regulated after status epilepticus and protects against seizure-induced neuronal loss in hippocampus. Neuroscience 2007; 151:232-41. [PMID: 18065154 DOI: 10.1016/j.neuroscience.2007.09.083] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 09/23/2007] [Accepted: 10/04/2007] [Indexed: 12/20/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a protein factor which has been found to play a significant role in both normal and pathological states. Its role as an angiogenic factor is well-established. More recently, VEGF has been shown to protect neurons from cell death both in vivo and in vitro. While VEGF's potential as a protective factor has been demonstrated in hypoxia-ischemia, in vitro excitotoxicity, and motor neuron degeneration, its role in seizure-induced cell loss has received little attention. A potential role in seizures is suggested by Newton et al.'s [Newton SS, Collier EF, Hunsberger J, Adams D, Terwilliger R, Selvanayagam E, Duman RS (2003) Gene profile of electroconvulsive seizures: Induction of neurotrophic and angiogenic factors. J Neurosci 23:10841-10851] finding that VEGF mRNA increases in areas of the brain that are susceptible to cell loss after electroconvulsive-shock induced seizures. Because a linear relationship does not always exist between expression of mRNA and protein, we investigated whether VEGF protein expression increased after pilocarpine-induced status epilepticus. In addition, we administered exogenous VEGF in one experiment and blocked endogenous VEGF in another to determine whether VEGF exerts a neuroprotective effect against status epilepticus-induced cell loss in one vulnerable brain region, the rat hippocampus. Our data revealed that VEGF is dramatically up-regulated in neurons and glia in hippocampus, thalamus, amygdala, and neocortex 24 h after status epilepticus. VEGF induced significant preservation of hippocampal neurons, suggesting that VEGF may play a neuroprotective role following status epilepticus.
Collapse
Affiliation(s)
- J N Nicoletti
- City University of New York, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Nangaku M, Izuhara Y, Takizawa S, Yamashita T, Fujii-Kuriyama Y, Ohneda O, Yamamoto M, van Ypersele de Strihou C, Hirayama N, Miyata T. A novel class of prolyl hydroxylase inhibitors induces angiogenesis and exerts organ protection against ischemia. Arterioscler Thromb Vasc Biol 2007; 27:2548-54. [PMID: 17932321 DOI: 10.1161/atvbaha.107.148551] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Hypoxia inducible factor (HIF) plays a pivotal role in the adaptation to ischemic conditions. Its activity is modulated by an oxygen-dependent hydroxylation of proline residues by prolyl hydroxylases (PHD). METHODS AND RESULTS We discovered 2 unique compounds (TM6008 and TM6089), which inhibited PHD and stabilized HIF activity in vitro. Our docking simulation studies based on the 3-dimensional structure of human PHD2 disclosed that they preferentially bind to the active site of PHD. Whereas PHD inhibitors previously reported inhibit PHD activity via iron chelation, TM6089 does not share an iron chelating motif and is devoid of iron chelating activity. In vitro Matrigel assays and in vivo sponge assays demonstrated enhancement of angiogenesis by local administration of TM6008 and TM6089. Their oral administration stimulated HIF activity in various organs of transgenic rats expressing a hypoxia-responsive reporter vector. No acute toxicity was observed up to 2 weeks after a single oral dose of 2000 mg/kg for TM6008. Oral administration of TM6008 protected neurons in a model of cerebrovascular disease. The protection was associated with amelioration of apoptosis but independent of enhanced angiogenesis. CONCLUSIONS The present study uncovered beneficial effects of novel PHD inhibitors preferentially binding to the active site of PHD.
Collapse
Affiliation(s)
- Masaomi Nangaku
- Institute of Medical Sciences and Division of Nephrology, Hypertension and Metabolism, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Morgan R, Kreipke CW, Roberts G, Bagchi M, Rafols JA. Neovascularization following traumatic brain injury: possible evidence for both angiogenesis and vasculogenesis. Neurol Res 2007; 29:375-81. [PMID: 17626733 DOI: 10.1179/016164107x204693] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Our goal was to characterize the angiogenic response following traumatic brain injury (TBI). METHODS Western analysis for vascular endothelial growth factor (VEGF) expression, double immunofluorescence labeling of endothelium and vascular endothelial growth factor receptor 2 (VEGFR2), bromodioxyuridine (BrdU) incorporation and measurement of capillary density, were all used to determine the temporal angiogenic response following TBI. RESULTS The angiogenic factors, VEGF and VEGFR2, increase following trauma. Capillary density increases and BrdU incorporation confirm the presence of newly formed vessels up to 48 hours post-injury. DISCUSSION Our results indicated that following TBI, there is a substantial increase in angiogenesis and based on morphologic characterization of BrdU-positive nuclei within the endothelium, we provide evidence for vasculogenesis following injury.
Collapse
Affiliation(s)
- Randy Morgan
- Department of Anatomy and Cell Biology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
112
|
Kumai Y, Ooboshi H, Ibayashi S, Ishikawa E, Sugimori H, Kamouchi M, Kitazono T, Egashira K, Iida M. Postischemic gene transfer of soluble Flt-1 protects against brain ischemia with marked attenuation of blood-brain barrier permeability. J Cereb Blood Flow Metab 2007; 27:1152-60. [PMID: 17077813 DOI: 10.1038/sj.jcbfm.9600420] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Brain edema is a major and often mortal complication of brain ischemia. Vascular endothelial growth factor (VEGF) is also known as a potent vascular permeability factor and may play detrimental roles at the acute stage of brain infarction. Our goal in this study was to explore protective effects of gene transfer of soluble flt-1 (sFlt-1), a natural inhibitor of VEGF, on focal brain ischemia. Adenoviral vector encoding sFlt-1 or beta-galactosidase as control was injected into the lateral ventricle 90 mins after photochemical distal middle cerebral artery occlusion in male spontaneously hypertensive rats. The transduced sFlt-1 was released to the cerebrospinal fluid from the ventricular wall and significantly increased 6 h, 1 and 7 days after sFlt-1 transfection. One day after brain ischemia, sFlt-1 gene transfer significantly reduced infarct volume (by 35%), brain edema (by 35%), and blood-brain barrier permeability (Evans blue extravasation; by 69%) with diminished phosphorylation of focal adhesion kinase (FAKtyr397 and FAKtyr861) in the ischemic vessels. Seven days after ischemia, sFlt-1 gene transfer also significantly attenuated infarct volume (by 29%) and monocyte/macrophage infiltration (by 27%), although there were no reductions in angiogenesis by sFlt-1 overexpression. These results suggest that sFlt-1 gene therapy targeting brain edema in acute stage of brain ischemia may be useful for brain infarction.
Collapse
Affiliation(s)
- Yasuhiro Kumai
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Rite I, Machado A, Cano J, Venero JL. Blood-brain barrier disruption induces in vivo degeneration of nigral dopaminergic neurons. J Neurochem 2007; 101:1567-82. [PMID: 17437543 DOI: 10.1111/j.1471-4159.2007.04567.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have evaluated the possibility that changes in the vascular system may constitute a contributing factor for the death of nigral dopaminergic neurons in Parkinson's disease. Thus, we have employed intranigral injections of vascular endothelial growth factor (VEGF), the most potent inducer of blood-brain barrier (BBB) permeability. A single dose of 1 mug of VEGF, chosen from a dose-response study, highly disrupted the BBB in the ventral mesencephalon in a time-dependent manner. A strong regional correlation between BBB disruption and loss of tyrosine hydroxylase-positive neurons was evident. Moreover, Fluoro-Jade B labelling showed the presence of dying neurons in the substantia nigra in response to VEGF injection. High number of TUNEL-positive nuclei was observed in this area along with activation of caspase 3 within nigral dopaminergic neurons. Analysis of the glial population demonstrated a strong inflammatory response and activation of astroglia in response to BBB disruption. We conclude that disruption of the BBB may be a causative factor for degeneration of nigral dopaminergic neurons.
Collapse
Affiliation(s)
- Inmaculada Rite
- Departamento de Bioquímica, Bromatología, Toxicología y Medicina Legal. Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González, Sevilla, Spain
| | | | | | | |
Collapse
|
114
|
Yu SW, Friedman B, Cheng Q, Lyden PD. Stroke-evoked angiogenesis results in a transient population of microvessels. J Cereb Blood Flow Metab 2007; 27:755-63. [PMID: 16883352 DOI: 10.1038/sj.jcbfm.9600378] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The role of angiogenesis after stroke is unclear; if angiogenesis supports long-term recovery of blood flow, then microvessel hyperdensity consequent to angiogenesis should persist in infarcted cortex. Here, we assess the long-term stability of ischemia-induced microvessels after 2-h transient rat middle cerebral artery occlusion (tMCAo) followed by 30, 90, or 165 days of reperfusion. Stereological measures of microvessel density were taken adjacent to and within cortical cysts. Vascular permeability was documented by extravasation of immunoglobulin (IgG) and of fluorescein-dextran. After 30 days reperfusion, a significantly increased microvessel volume density (V(V)) was restricted to the inner margin of cystic infarcts as compared with the region external to the infarct or contralateral control cortex (F=42.675, P<0.001). The hyperdense ischemic vasculature was abnormally leaky to IgG and fluorescein-dextran. Between 30 and 90 days of reperfusion, this vessel hyperdensity regressed significantly and then regressed further but less drastically between 90 and 165 days. Phagocytic macrophages were restricted to the infarct and dynamic changes in their number correlated with microvessel regression. Additional ED-1 labeled inflammatory cells were widely distributed inside and external to the infarct, even after 165 days of reperfusion. These data show that ischemia evoked angiogenesis results, at least in part, in transient populations of leaky microvessels and phagocytic macrophages. This suggests that a major role of this angiogenesis is for the removal of necrotic brain tissue.
Collapse
Affiliation(s)
- Sung Wook Yu
- Department of Neurosciences, UCSD School of Medicine, Veterans Administration Medical Center, San Diego, California 92161, USA
| | | | | | | |
Collapse
|
115
|
Suzuki S, Brown CM, Dela Cruz CD, Yang E, Bridwell DA, Wise PM. Timing of estrogen therapy after ovariectomy dictates the efficacy of its neuroprotective and antiinflammatory actions. Proc Natl Acad Sci U S A 2007; 104:6013-8. [PMID: 17389368 PMCID: PMC1851608 DOI: 10.1073/pnas.0610394104] [Citation(s) in RCA: 197] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Recent studies describing the seemingly contradictory actions of estrogens in ischemic stroke injury have led us to reevaluate the circumstances under which estrogen therapy (ET) provides benefits against cerebral stroke and decipher its mechanisms of action. One prominent feature that follows stroke injury is massive central and peripheral inflammatory responses. Evidence now suggests that postischemic inflammatory responses strongly contribute to the extent of brain injury, and 17beta-estradiol (E(2)) may protect the ischemic brain by exerting antiinflammatory actions. In an attempt to explain recently reported dichotomous effects of E(2) in stroke injury, we tested the hypothesis that an extended period of hypoestrogenicity both prevents E(2) from protecting the brain against ischemia and simultaneously suppresses its antiinflammatory actions. We report that E(2) exerts profound neuroprotective action when administered immediately upon ovariectomy, but not when administered after 10 weeks of hypoestrogenicity. Consistently, E(2) treatment given immediately at the time of ovariectomy attenuated central and peripheral production of proinflammatory cytokines after ischemic stroke. In contrast, E(2) did not suppress production of proinflammatory molecules when it was administered after 10 weeks postovariectomy. These results demonstrate that a prolonged period of hypoestrogenicity disrupts both neuroprotective and antiinflammatory actions of E(2). Our findings may help to explain the results of the Women's Health initiative that reported no beneficial effect of ET against stroke because the majority of the subjects initiated ET after an extended period of hypoestrogenicity.
Collapse
Affiliation(s)
- Shotaro Suzuki
- Departments of *Physiology and Biophysics and
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616
| | - Candice M. Brown
- Departments of *Physiology and Biophysics and
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616
| | | | - Enhua Yang
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616
| | - David A. Bridwell
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616
| | - Phyllis M. Wise
- Departments of *Physiology and Biophysics and
- Biology, University of Washington, Seattle, WA 98195; and
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
116
|
Hayashi T, Kamada H, Jin G, Deguchi K, Nagotani S, Sehara Y, Zhang H, Nagano I, Shoji M, Abe K. Different expression of low density lipoprotein receptor and ApoE between young adult and old rat brains after ischemia. Neurol Res 2007; 28:822-5. [PMID: 17288738 DOI: 10.1179/016164105x40002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Reduction of brain plasticity underlies the poor outcome of aged stroke patients. The molecular mechanism of plasticity reduction by aging is uncertain, but disturbed lipid metabolism may be implicated. METHODS We investigated the expression of low density lipoprotein receptors (LDL-R) and apolipoprotein E (ApoE), both of which play active roles in lipid metabolism in young adult and old rat brains after ischemia. RESULTS LDL-R, trivially expressed in the sham-operated brain neurons, was increased from day 1 and became prominent at days 7 and 21 at the peri-ischemic cortex. The magnitude was smaller in the old than in the young adult rats. ApoE was increased in the astrocytes and neurons of the peri-ischemic cortex at day 1, which became further pronounced in the neurons but not in the astrocytes at days 7 and 21. ApoE expression was again less prominent in the old animals at days 7 and 21. DISCUSSION As ApoE-containing lipoprotein is recruited via LDL-R, the present results suggest that old brains had less capability to induce LDL-R, which resulted in impaired recruitment of lipoprotein after the ischemic injury. Impaired lipid recruitment causes disturbance of synaptogenesis and thus brain plasticity reduction. This molecular mechanism may result in poor functional recovery of aged stroke patients.
Collapse
Affiliation(s)
- Takeshi Hayashi
- Department of Neurology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Sato N, Shimamura M, Takeuchi D, Kurinami H, Ogihara T, Morishita R. Gene therapy for ischemic brain disease with special reference to vascular dementia. Geriatr Gerontol Int 2007. [DOI: 10.1111/j.1447-0594.2007.00373.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
118
|
Sisco M, Liu WR, Kryger ZB, Mustoe TA. Reduced up-regulation of cytoprotective genes in rat cutaneous tissue during the second cycle of ischemia?reperfusion. Wound Repair Regen 2007; 15:203-12. [PMID: 17352752 DOI: 10.1111/j.1524-475x.2007.00206.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic wounds are major health problems that affect millions of people in the United States every year. Management of these wounds costs billions of dollars annually in the United States. Despite their clinical importance, the molecular mechanisms underlying these clinical conditions remain elusive. Repetitive ischemia-reperfusion (I-R) may play a pivotal role in chronic wound formation. The development of therapies for these wounds is hindered by the lack of animal models that allow identification of the molecular mechanisms underlying chronic wound formation. In the first study of its kind, we adapted our rat pressure sore model by imposing two cycles of ischemia (2 hours) and two cycles of reperfusion (24 hours), and we examined gene expression to better understand the molecular events that occur at the very early stages of cutaneous I-R injury with a goal of devising preventing strategies. We successfully tested our hypothesis and demonstrated that while cytoprotective genes, such as heat shock protein 70, heat shock protein 90, hypoxia-inducible factor-1alpha, vascular endothelial growth factor, and heme oxygenase-1, were initially up-regulated during the first cycle of I-R, their up-regulation was subsequently reduced or completely abolished during the second cycle of I-R. These findings raise the possibility that reduced up-regulation of these cytoprotective genes may be causally linked to cutaneous I-R injury.
Collapse
Affiliation(s)
- Mark Sisco
- Wound Healing Research Laboratory, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
119
|
Lee HJ, Kim KS, Park IH, Kim SU. Human neural stem cells over-expressing VEGF provide neuroprotection, angiogenesis and functional recovery in mouse stroke model. PLoS One 2007; 2:e156. [PMID: 17225860 PMCID: PMC1764718 DOI: 10.1371/journal.pone.0000156] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 11/27/2006] [Indexed: 12/27/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is a lethal stroke type. As mortality approaches 50%, and current medical therapy against ICH shows only limited effectiveness, an alternative approach is required, such as stem cell-based cell therapy. Previously we have shown that intravenously transplanted human neural stem cells (NSCs) selectively migrate to the brain and induce behavioral recovery in rat ICH model, and that combined administration of NSCs and vascular endothelial growth factor (VEGF) results in improved structural and functional outcome from cerebral ischemia. Methods and Findings We postulated that human NSCs overexpressing VEGF transplanted into cerebral cortex overlying ICH lesion could provide improved survival of grafted NSCs, increased angiogenesis and behavioral recovery in mouse ICH model. ICH was induced in adult mice by unilateral injection of bacterial collagenase into striatum. HB1.F3.VEGF human NSC line produced an amount of VEGF four times higher than parental F3 cell line in vitro, and induced behavioral improvement and 2–3 fold increase in cell survival at two weeks and eight weeks post-transplantation. Conclusions Brain transplantation of F3 human NSCs over-expressing VEGF near ICH lesion sites provided differentiation and survival of grafted human NSCs and renewed angiogenesis of host brain and functional recovery of ICH animals. These results suggest a possible application of the human neural stem cell line, which is genetically modified to over-express VEGF, as a therapeutic agent for ICH-stroke.
Collapse
Affiliation(s)
- Hong J. Lee
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- College of Bioscience and Biotechnology, Korea University, Seoul, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
| | - Kwang S. Kim
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
| | - In H. Park
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Seung U. Kim
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
120
|
Wang YQ, Guo X, Qiu MH, Feng XY, Sun FY. VEGF overexpression enhances striatal neurogenesis in brain of adult rat after a transient middle cerebral artery occlusion. J Neurosci Res 2007; 85:73-82. [PMID: 17061257 DOI: 10.1002/jnr.21091] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To elucidate whether vascular endothelial growth factor (VEGF) improves stroke-induced striatal neurogenesis, we intraventricularly injected human VEGF(165)-expressive plasmid (phVEGF) mixed with liposome into adult rats after a transient middle cerebral artery occlusion (MCAO). The results showed that EGFP, a reporter protein, positive cells appeared at 2 hr, further enhanced at 4 hr, reached the maximum at 3 days and still remained at 14 days after a single injection. Treatment with phVEGF increased angiogenesis, as indicated by double staining of vWF, a marker of endothelial cells, and 5'-bromodeoxyuridine (BrdU), a marker of cell proliferation. The phVEGF treatment dose-dependently reduced infarct volume of brain at 2 weeks after MCAO. The neuroprotection by VEGF could be obtained when the plasmid was injected within 2 hr after stroke. Moreover, VEGF overexpression significantly increased cell proliferation in the ipsilateral SVZ and the numbers of BrdU(+)-CRMP-4(+) and BrdU(+)-Tuj1(+), two markers of immature newborn neurons, and BrdU(+)-MAP-2(+), a marker of mature newborn neurons, cells in the ipsilateral striatum to MCAO. Present results show that VEGF plasmid treatment after stroke can significantly reduce infarct volume and enhance striatal neurogenesis in adult rat brain. This suggests that VEGF overexpression acquires significant functions of neuronal protection and repair in the injured brain, which provides a possibility to develop a novel therapeutic strategy for the patients with stroke.
Collapse
Affiliation(s)
- Yong-Quan Wang
- State Key Laboratory of Medical Neurobiology and Institute for Biomedical Science, Shanghai Medical College of Fudan University, Shanghai, Peoples Republic of China
| | | | | | | | | |
Collapse
|
121
|
Tham E, Gielen AW, Khademi M, Martin C, Piehl F. Decreased Expression of VEGF-A in Rat Experimental Autoimmune Encephalomyelitis and in Cerebrospinal Fluid Mononuclear Cells from Patients with Multiple Sclerosis. Scand J Immunol 2006; 64:609-22. [PMID: 17083617 DOI: 10.1111/j.1365-3083.2006.01851.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor A (VEGF-A) stimulates angiogenesis, but is also pro-inflammatory and plays an important role in the development of neurological disease, where it can have both attenuating and exacerbating effects. VEGF-B, a related molecule, is highly expressed in the central nervous system and seems to be important in neurological injury. A few studies have indicated that VEGF-A may play a role in the pathogenesis of multiple sclerosis (MS), but the role of VEGF-B has not been studied. We have studied the expression of VEGF-A, -B and their receptors by mRNA in situ hybridization, immunohistochemistry and real-time PCR in spinal cord from LEW rats with experimental autoimmune encephalomyelitis (EAE) and in cerebrospinal fluid (CSF) and blood samples from MS patients. Whereas VEGF-A is downregulated in glia in EAE, the infiltrating inflammatory cells are positive for VEGF-A. Expression of VEGF-B and the VEGF receptors is unaltered. In addition, the levels of VEGF-A mRNA in mononuclear cells [corrected] in CSF are lower in MS patients compared with controls. These results demonstrate a complex regulation of VEGF-A during neuroinflammation and suggest that VEGF-B is not involved in the pathogenesis of MS.
Collapse
MESH Headings
- Adolescent
- Adult
- Alternative Splicing
- Animals
- Cerebrospinal Fluid/cytology
- Down-Regulation
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Humans
- Leukocytes, Mononuclear/chemistry
- Leukocytes, Mononuclear/metabolism
- Male
- Middle Aged
- Multiple Sclerosis/genetics
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Neuroglia/chemistry
- Neuroglia/metabolism
- Neuroglia/pathology
- Neurons/chemistry
- Neurons/metabolism
- Neurons/pathology
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred Lew
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Vascular Endothelial Growth Factor A/analysis
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor B/analysis
- Vascular Endothelial Growth Factor B/genetics
- Vascular Endothelial Growth Factor B/metabolism
- Vascular Endothelial Growth Factor Receptor-1/analysis
- Vascular Endothelial Growth Factor Receptor-1/genetics
- Vascular Endothelial Growth Factor Receptor-1/metabolism
Collapse
Affiliation(s)
- E Tham
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
122
|
Bogaert E, Van Damme P, Van Den Bosch L, Robberecht W. Vascular endothelial growth factor in amyotrophic lateral sclerosis and other neurodegenerative diseases. Muscle Nerve 2006; 34:391-405. [PMID: 16856151 DOI: 10.1002/mus.20609] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The angiogenic activity of vascular endothelial growth factor (VEGF) is well known. Recently, it has become evident that VEGF is involved in central nervous system physiology and may play a role in the pathogenesis of neurological diseases. In particular, it may be involved in the mechanism of motor neuron degeneration in amyotrophic lateral sclerosis (ALS), and has been hypothesized to be implicated in the pathogenesis of peripheral neuropathies such as occur in the so-called POEMS syndrome and diabetes. VEGF is also being studied as a possible treatment option in some of these disorders. In this review we critically analyze the data supporting the notion that VEGF is a factor involved in motor neuron degeneration and review the studies linking VEGF to other diseases of the peripheral and central nervous systems.
Collapse
Affiliation(s)
- Elke Bogaert
- Laboratory of Neurobiology, University of Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium
| | | | | | | |
Collapse
|
123
|
Wang Y, Galvan V, Gorostiza O, Ataie M, Jin K, Greenberg DA. Vascular endothelial growth factor improves recovery of sensorimotor and cognitive deficits after focal cerebral ischemia in the rat. Brain Res 2006; 1115:186-93. [PMID: 16928361 DOI: 10.1016/j.brainres.2006.07.060] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 07/18/2006] [Accepted: 07/20/2006] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenesis factor with neurotrophic, neuroprotective and neuroproliferative effects. Depending on the dose, route and time of administration in relation to focal cerebral ischemia, VEGF can improve histological outcome and sensorimotor function in rodents. However, VEGF also increases vascular permeability, which can lead to brain edema and exacerbate ischemic brain injury. Thus, although VEGF is a candidate therapeutic for stroke and other ischemic disorders, its benefit relative to risk is uncertain. Considering that functional rather than histological measures of outcome are probably most relevant to therapeutic prospects for human stroke, we investigated the effects of VEGF after middle cerebral artery occlusion in rats using a series of behavioral tests. We report that VEGF improves functional outcome in ischemic rats, including both sensorimotor and cognitive deficiencies.
Collapse
Affiliation(s)
- Yaoming Wang
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | |
Collapse
|
124
|
Lai AY, Todd KG. Microglia in cerebral ischemia: molecular actions and interactions. Can J Physiol Pharmacol 2006; 84:49-59. [PMID: 16845890 DOI: 10.1139/y05-143] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The precise role of microglia in stroke and cerebral ischemia has been the subject of debate for a number of years. Microglia are capable of synthesizing numerous soluble and membrane-bound biomolecules, some known to be neuroprotective, some neurotoxic, whereas others have less definitive bioactivities. The molecular mechanisms through which microglia activate these molecules have thus become an important area of ischemia research. Here we provide a survey review that summarizes the key actions of microglial factors in cerebral ischemia including complement proteins, chemokines, pro-inflammatory cytokines, neurotrophic factors, hormones, and proteinases, as well several important messenger molecules that play a part in how these factors respond to extracellular signals during ischemic injuries. We also provide some new perspectives on how microglial intracellular signaling may contribute to the seemingly contradictory roles of several microglial effector molecules.
Collapse
Affiliation(s)
- Aaron Y Lai
- Neurochemical Research Unit, Department of Psychiatry and Centre for Neuroscience, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
125
|
Hossmann KA, Buschmann IR. Granulocyte-macrophage colony-stimulating factor as an arteriogenic factor in the treatment of ischaemic stroke. Expert Opin Biol Ther 2006; 5:1547-56. [PMID: 16318419 DOI: 10.1517/14712598.5.12.1547] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces arteriogenic growth of collateral vessels after occlusion of cardiac or peripheral arteries. Recently, evidence has been provided that arteriogenesis also occurs in the brain under conditions of reduced arterial blood supply. Hemispheric hypoperfusion induced by unilateral carotid and bilateral vertebral artery occlusion (three-vessel occlusion, [3-VO]) led to the growth of the anterior and posterior segments of the circle of Willis, which is the main collateral pathway between the origins of the anterior, middle and posterior cerebral arteries. GM-CSF applied subcutaneously at daily doses of 40 microg.kg(-1) resulted in the marked acceleration of this process. Within one week after the onset of treatment, the diameter of the posterior segment of the circle of Willis enlarged to 170% of control, blood flow and the haemodynamic reserve capacity of the brain returned to normal, and haemodynamic stroke, induced after 3-VO by systemic hypotension, was greatly alleviated. GM-CSF-induced stimulation of arteriogenesis in the hypoperfused brain thus provides powerful protection against ischaemic stroke.
Collapse
|
126
|
Abstract
Focal permanent or transient cerebral artery occlusion produces massive cell death in the central core of the infarction, whereas in the peripheral zone (penumbra) nerve cells are subjected to various determining survival and death signals. Cell death in the core of the infarction and in the adult brain is usually considered a passive phenomenon, although events largely depend on the partial or complete disruption of crucial metabolic pathways. Cell death in the penumbra is currently considered an active process largely dependent on the activation of cell death programs leading to apoptosis. Yet cell death in the penumbra includes apoptosis, necrosis, intermediate and other forms of cell death. A rather simplistic view implies poor prospects regarding cell survival in the core of the infarction and therapeutic expectations in the control of cell death and cell survival in the penumbra. However, the capacity for neuroprotection depends on multiple factors, primarily the use of the appropriate agent, at the appropriate time and during the appropriate interval. Understanding the mechanisms commanding cell death and survival area is as important as delimiting the therapeutic time window and the facility of a drug to effectively impact on specific targets. Moreover, the detrimental effects of homeostasis and the activation of multiple pathways with opposing signals following ischemic stroke indicate that better outcome probably does not depend on a single compound but on several drugs acting in combination at the optimal time in a particular patient.
Collapse
Affiliation(s)
- Isidro Ferrer
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat, Spain.
| |
Collapse
|
127
|
Shimamura M, Sato N, Waguri S, Uchiyama Y, Hayashi T, Iida H, Nakamura T, Ogihara T, Kaneda Y, Morishita R. Gene Transfer of Hepatocyte Growth Factor Gene Improves Learning and Memory in the Chronic Stage of Cerebral Infarction. Hypertension 2006; 47:742-51. [PMID: 16505200 DOI: 10.1161/01.hyp.0000208598.57687.3e] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is no specific treatment to improve the functional recovery in the chronic stage of ischemic stroke. To provide the new therapeutic options, we examined the effect of overexpression of hepatocyte growth factor (HGF) in the chronic stage of cerebral infarction by transferring the HGF gene into the brain using hemagglutinating virus of Japan envelope vector. Sixty rats were exposed to permanent middle cerebral artery occlusion (day 1). Based on the sensorimotor deficits at day 7, the rats were divided equally into control vector or HGF-treated rats. At day 56, rats transfected with the HGF gene showed a significant recovery of learning and memory in Morris water maze tests (control vector 50±4 s; HGF 33±5 s;
P
<0.05) and passive avoidance task (control vector 132.4±37.5 s; HGF 214.8±26.5 s;
P
<0.05). Although the total volume of cerebral infarction was not related to the outcome, immunohistochemical analysis for Cdc42 and synaptophysin in the peri-infarct region revealed that HGF enhanced the neurite extension and increased synapses. Immunohistochemistry for glial fibriary acidic protein revealed that the formation of glial scar was also prevented by HGF gene treatment. Additionally, the number of the arteries was increased in the HGF group at day 56. These data demonstrated that HGF has a pivotal role for the functional recovery after cerebral infarction through neuritogenesis, improved microcirculation, and the prevention of gliosis. Our results also provide evidence for the feasibility of gene therapy in the chronic stage of cerebral infarction.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Division of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
McCloskey DP, Croll SD, Scharfman HE. Depression of synaptic transmission by vascular endothelial growth factor in adult rat hippocampus and evidence for increased efficacy after chronic seizures. J Neurosci 2006; 25:8889-97. [PMID: 16192378 PMCID: PMC1415170 DOI: 10.1523/jneurosci.2577-05.2005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In addition to its potent effects on vasculature, it has become clear that vascular endothelial growth factor (VEGF) has effects on both neurons and glia, and recent studies suggest that it can be neuroprotective. To determine potential mechanisms underlying this neuroprotection, recombinant human VEGF was bath applied to adult rat hippocampal slices, and both extracellular and intracellular recordings were used to examine intrinsic properties and synaptic responses of hippocampal principal neurons. Initial studies in area CA1 showed that VEGF significantly reduced the amplitude of responses elicited by Schaffer collateral stimulation, without influencing membrane properties. Similar effects occurred in CA3 pyramidal cells and dentate gyrus granule cells when their major glutamatergic afferents were stimulated. Because VEGF expression is increased after seizures, effects of VEGF were also examined in rats with recurrent spontaneous seizures. VEGF reduced spontaneous discharges in slices from these rats but had surprisingly little effect on epileptiform discharges produced by disinhibition of slices from control rats. These results demonstrate a previously unknown effect of VEGF on neuronal activity and also demonstrate a remarkable potency in the epileptic brain. Based on this, we suggest that VEGF or VEGF-related targets could provide useful endpoints to direct novel therapeutic strategies for epilepsy.
Collapse
Affiliation(s)
- Daniel P McCloskey
- Center for Neural Recovery and Rehabilitation Research, Helen Hayes Hospital, West Haverstraw, New York 10993, USA.
| | | | | |
Collapse
|
129
|
Abstract
Angiogenesis--the growth of new blood vessels--is a crucial force for shaping the nervous system and protecting it from disease. Recent advances have improved our understanding of how the brain and other tissues grow new blood vessels under normal and pathological conditions. Angiogenesis factors, especially vascular endothelial growth factor, are now known to have roles in the birth of new neurons (neurogenesis), the prevention or mitigation of neuronal injury (neuroprotection), and the pathogenesis of stroke, Alzheimer's disease and motor neuron disease. As our understanding of pathophysiology grows, these developments may point the way towards new molecular and cell-based therapies.
Collapse
Affiliation(s)
- David A Greenberg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, California 94945, USA.
| | | |
Collapse
|
130
|
Zachary I. Neuroprotective role of vascular endothelial growth factor: signalling mechanisms, biological function, and therapeutic potential. Neurosignals 2006; 14:207-21. [PMID: 16301836 DOI: 10.1159/000088637] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Accepted: 05/05/2005] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGF or VEGF-A) and its receptors play essential roles in the formation of blood vessels during embryogenesis and in disease. Most biological effects of VEGF are mediated via two receptor tyrosine kinases, VEGFR1 and VEGFR2, but specific VEGF isoforms also bind neuropilins (NP) 1 and 2, non-tyrosine kinase receptors originally identified as receptors for semaphorins, polypeptides with essential roles in neuronal patterning. There is abundant evidence that VEGF-A has neurotrophic and neuroprotective effects on neuronal and glial cells in culture and in vivo, and can stimulate the proliferation and survival of neural stem cells. VEGFR2 and NP1 are the major VEGF receptors expressed on neuronal cells and, while the mechanisms mediating neuroprotective effects of VEGF are not fully understood, VEGF stimulates several signalling events in neuronal cell types, including activation of phospholipase C-gamma, Akt and ERK. Findings in diverse models of nerve damage and disease suggest that VEGF has therapeutic potential as a neuroprotective factor. VEGF is a key mediator of the angiogenic response to cerebral and peripheral ischaemia, and promotes nerve repair following traumatic spinal injury. Recent work has revealed a role for reduced VEGF expression in the pathogenesis of amyotrophic lateral sclerosis, a rare neurodegenerative disease caused by selective loss of motor neurons. In many instances, the neuroprotective effects of VEGF appear to result from a combination of the indirect consequences of increased angiogenesis, and the direct stimulation of neuronal function. However, more work is required to determine the specific functional role of direct neuronal effects of VEGF.
Collapse
Affiliation(s)
- Ian Zachary
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Department of Medicine, The Rayne Institute, University College London, London, UK.
| |
Collapse
|
131
|
Sun Y, Jin K, Childs JT, Xie L, Mao XO, Greenberg DA. Vascular endothelial growth factor-B (VEGFB) stimulates neurogenesis: Evidence from knockout mice and growth factor administration. Dev Biol 2006; 289:329-35. [PMID: 16337622 DOI: 10.1016/j.ydbio.2005.10.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2005] [Revised: 10/07/2005] [Accepted: 10/14/2005] [Indexed: 12/27/2022]
Abstract
Vascular endothelial growth factor-B (VEGFB) is an angiogenic and neuroprotective protein that reduces hypoxic and ischemic neuronal injury. To determine if VEGFB also regulates neurogenesis in the adult brain, we studied the effects of VEGFB administration in vitro and in vivo, as well as the effect of VEGFB gene knockout (KO) in mice, on bromodeoxyuridine (BrdU) incorporation and expression of immature neuronal markers in the subgranular zone (SGZ) of the hippocampal dentate gyrus and the forebrain subventricular zone (SVZ). Intracerebroventricular VEGFB administration increased BrdU incorporation into cells of neuronal lineage both in vitro and in vivo, and VEGFB-KO mice showed impaired neurogenesis, consistent with a neurogenesis-promoting effect of VEGFB. In addition, intraventricular administration of VEGFB restored neurogenesis to wild-type levels in VEGFB-KO mice. These results suggest a role for VEGFB in the regulation of adult neurogenesis, which could have therapeutic implications for diseases associated with central neuronal loss.
Collapse
Affiliation(s)
- Yunjuan Sun
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | |
Collapse
|
132
|
Sköld MK, Risling M, Holmin S. Inhibition of vascular endothelial growth factor receptor 2 activity in experimental brain contusions aggravates injury outcome and leads to early increased neuronal and glial degeneration. Eur J Neurosci 2006; 23:21-34. [PMID: 16420412 DOI: 10.1111/j.1460-9568.2005.04527.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Angiogenesis following traumatic brain injuries (TBIs) may be of importance for post-traumatic reparative processes and the development of secondary injuries. We have previously shown expression of vascular endothelial growth factor (VEGF), a major regulator of endothelial cell proliferation, angiogenesis and vascular permeability, and VEGF receptors (VEGFR1 and 2) after TBI in rat. In the present work we tried to further elucidate the role of VEGF after TBI by performing specific VEGFR2 activity inhibition. In rats subjected to VEGFR2 blockage we report an increased haemorrhagic area (P < 0.05), early increase in serum levels of neural injury marker neuron-specific enolase (P < 0.05) and glial injury marker S100beta (P < 0.05), and increased numbers of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labelling- (TUNEL-) and FluoroJade B- (P < 0.05) positive cells, all increases preceding the known VEGF/VEGFR vascular response in brain trauma. An increase in lesion area, as measured by decreased microtubuli-associated protein 2 expression (P < 0.05) and increased glial fibrillary acidic protein reactivity (P < 0.05), could also be demonstrated. In addition, vascular density, as measured by von Willebrandt factor-positive cells, was decreased (P < 0.05). No differences in post-traumatic inflammatory response, as measured by stainings for macrophages, granulocytes and intracellular adhesion molecules, were shown between the groups. Taken together, our findings point towards VEGF/VEGFR2 up-regulation after TBI as being an important endogenous cytoprotective mechanism in TBI. The possible importance of VEGF on the vascular, neuronal and glial compartments of the neurovascular unit after TBI is discussed.
Collapse
Affiliation(s)
- Mattias K Sköld
- Department of Neuroscience, Karolinska Institutet, S-17177 Stockholm, Sweden.
| | | | | |
Collapse
|
133
|
Kusaka N, Sugiu K, Tokunaga K, Katsumata A, Nishida A, Namba K, Hamada H, Nakashima H, Date I. Enhanced brain angiogenesis in chronic cerebral hypoperfusion after administration of plasmid human vascular endothelial growth factor in combination with indirect vasoreconstructive surgery. J Neurosurg 2005; 103:882-90. [PMID: 16304993 DOI: 10.3171/jns.2005.103.5.0882] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Vascular endothelial growth factor (VEGF) is a secreted mitogen associated with angiogenesis. The conceptual basis for therapeutic angiogenesis after plasmid human VEGF gene (phVEGF) transfer has been established in patients presenting with limb ischemia and myocardial infarction. The authors hypothesized that overexpression of VEGF using a gene transfer method combined with indirect vasoreconstruction might induce effective brain angiogenesis in chronic cerebral hypoperfusion, leading to prevention of ischemic attacks. METHODS A chronic cerebral hypoperfusion model induced by permanent ligation of both common carotid arteries in rats was used in this investigation. Seven days after induction of cerebral hypoperfusion, encephalomyosynangiosis (EMS) and phVEGF administration in the temporal muscle were performed. Fourteen days after treatment, the VEGF gene therapy group displayed numbers and areas of capillary vessels in temporal muscles that were 2.2 and 2.5 times greater, respectively, in comparison with the control group. In the brain, the number and area of capillary vessels in the group treated with the VEGF gene were 1.5 and 1.8 times greater, respectively, relative to the control group. CONCLUSIONS In rat models of chronic cerebral hypoperfusion, administration of phVEGF combined with indirect vasoreconstructive surgery significantly increased capillary density in the brain. The authors' results indicate that administration of phVEGF may be an effective therapy in patients with chronic cerebral hypoperfusion, such as those with moyamoya disease.
Collapse
Affiliation(s)
- Noboru Kusaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Shao G, Gao CY, Lu GW. Alterations of Hypoxia-Inducible Factor-1 Alpha in the Hippocampus of Mice Acutely and Repeatedly Exposed to Hypoxia. Neurosignals 2005; 14:255-61. [PMID: 16301840 DOI: 10.1159/000088641] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 07/08/2005] [Indexed: 11/19/2022] Open
Abstract
This work aims at investigating the effects of hypoxic preconditioning on hypoxia-inducible factor-1 alpha (HIF-1alpha) expression in the hippocampus of mice during acute and repeated hypoxic exposures. The mice were randomly divided into three groups and exposed, respectively, to hypoxia for 4 runs (group H4), 1 run (group H1), and 0 run (group H0). Reverse transcription-polymerase chain reaction (RT-PCR), Western blot, electrophoretic mobility shift assay (EMSA), and chromatin immunoprecipitation were used to examine the HIF-1alpha responses in the mouse hippocampus following exposure to hypoxia. The tolerance of mice to hypoxia increased significantly following acute and repetitive exposure to autoprogressive hypoxia. Total mRNA, total protein, and nuclear protein were extracted from the hippocampus for RT-PCR, Western blot, and EMSA, respectively. The HIF-1alpha mRNA levels were found to be increased in group H1 and decreased in group H4. The HIF-1alpha protein levels and HIF-1 DNA-binding activities were increased in group H1 and markedly increased in group H4. One of the HIF-1 target genes, vascular endothelial growth factor, increased in group H4. HIF-1 activation is thought to be involved in the protection of the brain of hypoxic preconditioned mice.
Collapse
Affiliation(s)
- Guo Shao
- Department of Neurobiology, Capital University of Medical Sciences, Beijing, China
| | | | | |
Collapse
|
135
|
Merrill MJ, Oldfield EH. A reassessment of vascular endothelial growth factor in central nervous system pathology. J Neurosurg 2005; 103:853-68. [PMID: 16304990 DOI: 10.3171/jns.2005.103.5.0853] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
✓ Overexpression of vascular endothelial growth factor (VEGF) is associated with several central nervous system (CNS) diseases and abnormalities, and is often postulated as a causative factor and promising therapeutic target in these settings. The authors' goal was to reassess the contribution of VEGF to the biology and pathology of the CNS.
The authors review the literature relating to the following aspects of VEGF: 1) the biology of VEGF in normal brain; 2) the involvement of VEGF in CNS disorders other than tumors (traumatic and ischemic injuries, arteriovenous malformations, inflammation); and 3) the role of VEGF in brain tumor biology (gliomas and the associated vasogenic edema, and hemangioblastomas).
The authors conclude the following: first, that VEGF overexpression contributes to the phenotype associated with many CNS disorders, but VEGF is a reactive rather than a causative factor in many cases; and second, that use of VEGF as a therapeutic agent or target is complicated by the effects of VEGF not only on the cerebral vasculature, but also on astrocytes, neurons, and inflammatory cells. In many cases, therapeutic interventions targeting the VEGF/VEGF receptor axis are likely to be ineffective or even detrimental. Clinical manipulation of VEGF levels in the CNS must be approached with caution.
Collapse
Affiliation(s)
- Marsha J Merrill
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-1414, USA.
| | | |
Collapse
|
136
|
Chu K, Park KI, Lee ST, Jung KH, Ko SY, Kang L, Sinn DI, Lee YS, Kim SU, Kim M, Roh JK. Combined treatment of vascular endothelial growth factor and human neural stem cells in experimental focal cerebral ischemia. Neurosci Res 2005; 53:384-90. [PMID: 16198014 DOI: 10.1016/j.neures.2005.08.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Revised: 08/04/2005] [Accepted: 08/25/2005] [Indexed: 01/12/2023]
Abstract
Recent studies have indicated the beneficial effects of vascular endothelial growth factor (VEGF), and transplanted neural stem cells (NSCs) in cerebral ischemia. We investigated the effects of the combined administration of NSCs and VEGF on focal cerebral ischemia in adult rats. Four groups (n = 12, respectively)--group 1 (ischemia-only), group 2 (ischemia + VEGF), group 3 (ischemia + NSCs) and group 4 (ischemia + NSCs + VEGF)--were compared. Human NSCs (HB1.F3), labeled with Lac Z+ or PKH26, were given intravenously 24h after surgery (5 x 10(6) cells). At 48 h after surgery, recombinant human VEGF (50 microg/kg) was infused intravenously (1 microg/(kg min)). Behavioral tests using the modified limb placing and rotarod tests were performed every week following ischemia. Immunohistochemistry for endothelial barrier antigen (EBA), VEGF and Nissl staining were performed at day 35 after ischemia. Group 4 showed better behavioral recovery at 7, 14 and 28 days than group 3 (p = 0.020, 0.005 and 0.043, respectively). These functional recoveries were correlated with enhanced EBA immunoreactivities at day 35 after ischemia, especially in the ipsilesional striatum. Group 4 showed lesser degree of brain atrophy in cortex and striatum, when compared with other groups. The distribution of VEGF was not co-localized with NSCs. Our results suggest that VEGF may act synergistically on NSC-transplanted, ischemic brain via a pro-angiogenic effect.
Collapse
Affiliation(s)
- Kon Chu
- Stroke & Neural Stem Cell Laboratory in Clinical Research Institute, Department of Neurology, Seoul National University Hospital, 28, Yongon-dong, Chongro-gu, Seoul 110-744, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Kaya D, Gürsoy-Ozdemir Y, Yemisci M, Tuncer N, Aktan S, Dalkara T. VEGF protects brain against focal ischemia without increasing blood--brain permeability when administered intracerebroventricularly. J Cereb Blood Flow Metab 2005; 25:1111-8. [PMID: 15829918 DOI: 10.1038/sj.jcbfm.9600109] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Delayed administration of vascular endothelial growth factor (VEGF) promotes functional recovery after focal cerebral ischemia. However, early intravenous injection of VEGF increases blood-brain barrier (BBB) leakage, hemorrhagic transformation and infarct volume whereas its application to cortical surface is neuroprotective. We have investigated whether or not early intracerebroventricular administration of VEGF could replicate the neuroprotective effect observed with topical application and the mechanism of action of this protection. Mice were subjected to 90 mins middle cerebral artery (MCA) occlusion and 24 h of reperfusion. Vascular endothelial growth factor (8 ng, intracerebroventricular) was administered 1 or 3 h after reperfusion. Compared with the vehicle-treated (intracerebroventricular) group, VEGF decreased the infarct volume along with BBB leakage in both treatment groups. Neurologic disability scores improved in parallel to the changes in infarct volume. Independently of the decrease in infarct size, VEGF also reduced the number of TUNEL-positive apoptotic neurons. Phospo-Akt levels were significantly higher in ischemic hemispheres of the VEGF-treated mice. Contrary to intracerebroventricular route, intravenous administration of VEGF (15 microg/kg) enhanced the infarct volume as previously reported for the rat. In conclusion, single intracerebroventricular injection of VEGF protects brain against ischemia without adversely affecting BBB permeability, and has a relatively long therapeutic time window. This early neuroprotective action, observed well before recovery-promoting actions such as angiogenesis, possibly involves activation of the PI-3-Akt pathway.
Collapse
Affiliation(s)
- Dilaver Kaya
- Department of Neurology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
138
|
Chattopadhyay M, Krisky D, Wolfe D, Glorioso JC, Mata M, Fink DJ. HSV-mediated gene transfer of vascular endothelial growth factor to dorsal root ganglia prevents diabetic neuropathy. Gene Ther 2005; 12:1377-1384. [PMID: 15843809 PMCID: PMC1242112 DOI: 10.1038/sj.gt.3302533] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Accepted: 03/03/2005] [Indexed: 12/20/2022]
Abstract
We examined the utility of herpes simplex virus (HSV) vector-mediated gene transfer of vascular endothelial growth factor (VEGF) in a mouse model of diabetic neuropathy. A replication-incompetent HSV vector with VEGF under the control of the HSV ICP0 promoter (vector T0VEGF) was constructed. T0VEGF expressed and released VEGF from primary dorsal root ganglion (DRG) neurons in vitro, and following subcutaneous inoculation in the foot, expressed VEGF in DRG and nerve in vivo. At 2 weeks after induction of diabetes, subcutaneous inoculation of T0VEGF prevented the reduction in sensory nerve amplitude characteristic of diabetic neuropathy measured 4 weeks later, preserved autonomic function measured by pilocarpine-induced sweating, and prevented the loss of nerve fibers in the skin and reduction of neuropeptide calcitonin gene-related peptide and substance P in DRG neurons of the diabetic mice. HSV-mediated transfer of VEGF to DRG may prove useful in treatment of diabetic neuropathy.
Collapse
Affiliation(s)
- M Chattopadhyay
- Department of Neurology, University of Michigan Health System, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0316, USA
| | | | | | | | | | | |
Collapse
|
139
|
Tunçkiran A, Cayan S, Bozlu M, Yilmaz N, Acar D, Akbay E. Protective effect of vascular endothelial growth factor on histologic changes in testicular ischemia-reperfusion injury. Fertil Steril 2005; 84:468-73. [PMID: 16084892 DOI: 10.1016/j.fertnstert.2005.01.144] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 01/14/2005] [Accepted: 01/14/2005] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate the efficacy of vascular endothelial growth factor (VEGF) on long-term histologic damage in testicular ischemia-reperfusion injury. DESIGN Controlled experimental study using rats. SETTING University of Mersin School of Medicine, Mersin, Turkey. ANIMAL(S) Sixteen adult male Wistar rats. INTERVENTION(S) Five rats underwent 2 hours of testicular torsion. Six rats received VEGF injection into the testis before detorsion. Five rats underwent sham operation. MAIN OUTCOME MEASURE(S) Mean seminiferous tubular diameter (MSTD), germinal epithelial cell thickness (GECT), mean testicular biopsy score (MTBS), and apoptosis (caspase-3-positive cells). RESULT(S) Testicular torsion-detorsion caused a significant decrease in MSTD, GECT, and MTBS and a significant increase in the mean value of caspase-3-positive cells in ipsilateral testes but not in the contralateral testes. The MSTD, GECT, and MTBS of the ipsilateral testes were significantly higher in the group treated with VEGF than in the torsion-detorsion group. The animals treated with VEGF had a significant decrease in the mean value of ipsilateral testicular caspase-3-positive cells compared with the torsion-detorsion group. CONCLUSION(S) Vascular endothelial growth factor might have a role in testicular damage caused by ischemia-reperfusion. Administering VEGF before reperfusion might have the potential to decrease the long-term histologic damage after testicular torsion.
Collapse
Affiliation(s)
- Ahmet Tunçkiran
- Department of Urology, University of Mersin School of Medicine, Mersin, Turkey
| | | | | | | | | | | |
Collapse
|
140
|
Rouet V, Hamma-Kourbali Y, Petit E, Panagopoulou P, Katsoris P, Barritault D, Caruelle JP, Courty J. A synthetic glycosaminoglycan mimetic binds vascular endothelial growth factor and modulates angiogenesis. J Biol Chem 2005; 280:32792-800. [PMID: 16014624 DOI: 10.1074/jbc.m504492200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In a previous study, we showed that in situ injection of glycosaminoglycan mimetics called RGTAs (ReGeneraTing Agents) enhanced neovascularization after skeletal muscular ischemia (Desgranges, P., Barbaud, C., Caruelle, J. P., Barritault, D., and Gautron, J. (1999) FASEB J. 13, 761-766). In the present study, we showed that the RGTA OTR4120 modulated angiogenesis in the chicken embryo chorioallantoic membrane assay, in a dose-dependent manner. We therefore investigated the effect of OTR4120 on one of the most specific angiogenesis-regulating heparin-binding growth factors, vascular endothelial growth factor 165 (VEGF165). OTR4120 showed high affinity binding to VEGF165 (Kd = 2.2 nm), as compared with heparin (Kd = 15 nm), and potentiated the affinity of VEGF165 for VEGF receptor-1 and -2 and for neuropilin-1. In vitro, OTR4120 potentiated VEGF165-induced proliferation and migration of human umbilical vein endothelial cells. In the in vivo Matrigel plug angiogenesis assay, OTR4120 in a concentration as low as 3 ng/ml caused a 6-fold increase in VEGF165-induced angiogenesis. Immunohistochemical staining showed a larger number of well differentiated VEGFR-2-expressing-cells in Matrigel sections of OTR4120-treated plug than in control sections. These findings indicate that OTR4120 enhances the VEGF165-induced angiogenesis and therefore may hold promise for treating disorders characterized by deficient angiogenesis.
Collapse
Affiliation(s)
- Vincent Rouet
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires UMR CNRS 7149, Université Paris XII-Val de Marne, Avenue du Général de Gaulle, 94010 Créteil CEDEX, France
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Yano A, Shingo T, Takeuchi A, Yasuhara T, Kobayashi K, Takahashi K, Muraoka K, Matsui T, Miyoshi Y, Hamada H, Date I. Encapsulated vascular endothelial growth factor—secreting cell grafts have neuroprotective and angiogenic effects on focal cerebral ischemia. J Neurosurg 2005; 103:104-14. [PMID: 16121981 DOI: 10.3171/jns.2005.103.1.0104] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Object
The authors evaluated the neuroprotective and angiogenic effects of a continuous and low-dose infusion of vascular endothelial growth factor (VEGF)-165 on cerebral ischemia in rats.
Methods
The authors introduced VEGF complementary (c)DNA into baby hamster kidney (BHK) cells and established a cell line that produces human VEGF165 (BHK-VEGF). The BHK-VEGF cells and BHK cells that had been transfected with an expression vector that did not contain human VEGF165 cDNA (BHK-control) were encapsulated. Both capsules were implanted into rat striata. Six days after capsule implantation, the right middle cerebral artery (MCA) was occluded. Some animals were killed 24 hours after occlusion to measure the volume of the resulting infarct and to perform immunohistochemical studies. Other animals were used for subsequent behavioral studies 1, 7, and 14 days after MCA occlusion.
The encapsulated BHK-VEGF cell grafts significantly reduced the volume of the infarct and the number of apoptotic cells in the penumbral area when compared with the effect of the BHK-control cell capsule. In addition, angiogenesis and gliogenesis significantly increased in the region around the capsule in animals that received BHK-VEGF cell capsules without an increase in focal cerebral blood flow; this did not occur in animals that received the BHK-control cell capsule. In behavioral studies rats that received the BHK-VEGF cell capsule displayed significant recovery while participating in the accelerating rotarod test after stroke.
Conclusions
Continuous intracerebral administration of low-dose VEGF165 through encapsulated grafts of VEGF-producing cells produces neuroprotective and angiogenic effects. These effects improve subsequent motor function.
Collapse
Affiliation(s)
- Akimasa Yano
- Department of Neurological Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Abumiya T, Yokota C, Kuge Y, Minematsu K. Aggravation of hemorrhagic transformation by early intraarterial infusion of low-dose vascular endothelial growth factor after transient focal cerebral ischemia in rats. Brain Res 2005; 1049:95-103. [PMID: 15935998 DOI: 10.1016/j.brainres.2005.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2005] [Revised: 04/28/2005] [Accepted: 05/01/2005] [Indexed: 11/20/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a unique growth factor associated with angiogenesis, vascular permeability, and neuroprotection. The aim of this study was to observe the effects of early intraarterial infusion of low-dose VEGF on ischemia/reperfusion injury after transient focal cerebral ischemia in rats. Male Sprague-Dawley rats were subjected to 2 h of focal ischemia by middle cerebral artery occlusion. After the 2 h ischemia, the rats were infused with 0.3 microg/kg of VEGF (n = 15), or the vehicle as a control (n = 15), via the reperfused internal carotid artery. The brains were collected after a 1 h, 6 h, or 72 h reperfused period. Severity of ischemic cellular injury, serum extravasation, hemorrhagic transformation, and matrix metalloproteinase (MMP)-2 and -9 expressions were compared between the VEGF-treated and control groups. No significant difference in the extent of ischemic cellular injury and serum extravasation was observed between the two groups. However, vessel numbers with hemorrhagic transformation were significantly greater in the VEGF-treated group than in the control group after the 72 h reperfusion (9.4 +/- 1.6 versus 2.6 +/- 1.5; P = 0.028). The severity of hemorrhagic transformation was not correlated with the extent of ischemic cellular injury or serum extravasation. MMP-2 and -9 expressions were not enhanced in the VEGF-treated group compared with the control group. These results suggest that exogenous VEGF administered intravascularly at a very early point in reperfusion aggravates hemorrhagic transformation. The aggravated hemorrhagic transformation does not seem to depend on the enlargement of ischemic cellular injury, serum extravasation, or overexpressions of MMP-2 and -9.
Collapse
Affiliation(s)
- Takeo Abumiya
- Research Institute, National Cardiovascular Center, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|
143
|
Sköld MK, von Gertten C, Sandberg-Nordqvist AC, Mathiesen T, Holmin S. VEGF and VEGF receptor expression after experimental brain contusion in rat. J Neurotrauma 2005; 22:353-67. [PMID: 15785231 DOI: 10.1089/neu.2005.22.353] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Angiogenesis following traumatic brain injury (TBI) may be of importance not only for post-traumatic reparative processes but also for the development of secondary injuries. Vascular endothelial growth factor (VEGF) is a major regulator of endothelial cell proliferation, angiogenesis, and vascular permeability, though its possible involvement in secondary injuries after TBI is largely unknown. This study was undertaken to analyze the expression of VEGF and the VEGF receptors in experimental brain contusion in rat. Twenty-three adult female Sprague-Dawley rats were subjected to a focal cerebral contusion injury by use of a weight-drop model. Four additional rats underwent craniotomy only. The animals were sacrificed 6 h, or 1, 2, 4, 6, 8, or 16 days post-injury. Expression of VEGF and the VEGF receptors VEGFR1 (Flt-1) and VEGFR2 (Flk-1) were studied by in situ hybridization and immunohistochemistry. VEGF messenger (m)RNA and protein expression were detected in astrocytes, neutrophils, and macrophages in or adjacent to the injury from 1 day after injury, with a peak expression after 4-6 days. Flt-1 and Flk-1 mRNA and protein were detected in vessels adjacent to the lesion from 1 day after injury throughout day 6 after injury. It was also noted that Flt-1/Flk-1 and VEGF-positive vessels often were negative for SMI-71, a marker for vessels in areas with blood-brain barrier (BBB). In conclusion, we have demonstrated that TBI leads to an upregulation of VEGF, Flt-1, and Flk-1 mRNA and protein in and around the lesion. The data provide a foundation for future pharmacological intervention studies focusing on posttraumatic angiogenesis and possible injury repair effects of the VEGF system in TBI.
Collapse
Affiliation(s)
- Mattias K Sköld
- Department of Neuroscience, Retzius Laboratory, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
144
|
Heine VM, Zareno J, Maslam S, Joëls M, Lucassen PJ. Chronic stress in the adult dentate gyrus reduces cell proliferation near the vasculature and VEGF and Flk-1 protein expression. Eur J Neurosci 2005; 21:1304-14. [PMID: 15813940 DOI: 10.1111/j.1460-9568.2005.03951.x] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent evidence has shown that cell proliferation in the adult hippocampal dentate gyrus occurs in tight clusters located near the vasculature. Also, changes in neurogenesis often appear parallel to changes in angiogenesis. Moreover, both these processes share similar modulating factors, like vascular endothelial growth factor (VEGF) and its receptor Flk-1. In an earlier study we found that chronic stress decreased new cell proliferation in the adult dentate gyrus. We here questioned whether these effects of chronic stress are mediated through the vasculature and whether they involve an angiogenic-signaling pathway. We therefore measured the surface area covered by the vasculature, the proportion of vascular-associated newborn cells, and analysed VEGF and Flk-1 protein expression in the hippocampus of a control, chronically stressed and recovery group of rats. Our results show that 32% of the proliferating cells in the rat hippocampus is vascular associated. Chronic stress affected this population of newborn cells to a significantly larger extent than the non-associated cells. Interestingly, after 3 weeks of recovery, the decreased proliferation not associated with the vasculature was more effectively restored than vascular-associated proportion of proliferating cells. VEGF protein was expressed in high densities in GFAP-positive astrocytes located in the hilus, with VEGF-positive end feet extending into and often contacting the granule cells. After chronic stress, both VEGF and Flk-1 protein levels were significantly decreased in the granular cell layer, and again recovered after 3 weeks. This demonstrates that changes in angiogenic factors are implicated in the decreased adult proliferation found after chronic stress.
Collapse
Affiliation(s)
- Vivi M Heine
- Institute Neurobiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Kruislaan 320, 1098 SM Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
145
|
Lennmyr F, Terént A, Syvänen AC, Barbany G. Vascular endothelial growth factor gene expression in middle cerebral artery occlusion in the rat. Acta Anaesthesiol Scand 2005; 49:488-93. [PMID: 15777296 DOI: 10.1111/j.1399-6576.2005.00646.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Focal cerebral ischemia induces up-regulation of angiogenic growth factors such as vascular endothelial growth factor (VEGF), which may have both beneficial and harmful effects to the ischemic brain. Vascular endothelial growth factor is up-regulated in models of brain ischemia, but the underlying mechanisms in vivo remain unclear. In the present report we have investigated the concomitant changes in VEGF and glyceraldehyde dehydrogenase (GAPDH) mRNA expression in a model of permanent and transient cerebral ischemia. METHODS Male Sprague-Dawley rats were exposed to permanent or transient (2 h) middle cerebral artery occlusion (PMCAO, TMCAO). Brain samples were collected at survival times ranging from 6 h to 1 week, and the levels of VEGF164 and GAPDH mRNA were determined using reverse-transcriptase real-time polymerase chain reaction (RT-PCR). RESULTS The VEGF mRNA levels decreased gradually over the observation period in a similar manner in both PMCAO and TMCAO. Maximum levels, seen at early observation time points, did not significantly deviate from sham controls. No statistically significant changes in GAPDH mRNA levels were observed, but there was a tendency towards a postischemic decrease with subsequent return to control levels over time. The VEGF/GAPDH ratio followed a pattern of decrease similar to VEGF mRNA alone. CONCLUSION The VEGF mRNA levels at 6 h after MCAO remain near baseline and thereafter decline, regardless of whether the occlusion is permanent or transient (2 h). The findings raise the question of other than transcriptional regulation of VEGF in cerebral ischemia.
Collapse
Affiliation(s)
- F Lennmyr
- Department of Surgical Sciences, Section of Anesthesiology and Intensive Care, University Hospital, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
146
|
Wang WZ, Fang XH, Stepheson LL, Khiabani KT, Zamboni WA. Acute Microvascular Action of Vascular Endothelial Growth Factor in Skeletal Muscle Ischemia/Reperfusion Injury. Plast Reconstr Surg 2005; 115:1355-65. [PMID: 15809599 DOI: 10.1097/01.prs.0000156980.38387.8d] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The purpose of this study was to investigate the acute action of vascular endothelial growth factor (VEGF) in the microcirculation of skeletal muscle subject to ischemia/reperfusion in vivo and to determine the role of nitric oxide synthase in VEGF-induced microvascular protection. METHODS A vascular pedicle isolated rat cremaster muscle model coupled with local intraarterial infusion technique was used. Each muscle underwent 4 hours of zero-flow warm ischemia followed by 2 hours of reperfusion. Femoral artery cannulation was performed before reperfusion. The infusate was administered by continuous infusion into the arterial tree of the muscle beginning at 1 minute before reperfusion and at the rate of 0.1 ml/hour throughout the entire reperfusion period. Three groups were designed: (1) the ischemia/reperfusion group, with infusion normal saline; (2) the VEGF plus ische-mia/reperfusion group, with infusion of recombinant human VEGF165 protein; and (3) the L-NA plus VEGF plus ischemia/reperfusion group, with infusion of N-nitro-L-arginine (L-NA; a nonselective nitric oxide synthase antagonist) mixed with VEGF165 protein. After 2 hours of reperfusion, microcirculation measurements including arteriole diameter, capillary perfusion, and endothelium-dependent and endothelium-independent vasodilatation were performed. The muscle was harvested and processed for reverse-transcriptase polymerase chain reaction for measuring eNOS and endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) gene expression. RESULTS Reperfusion caused significant microvascular alterations including vasoconstriction, poor capillary perfusion, and endothelial dysfunction in the skeletal muscle. These alterations were significantly attenuated by intraarterial infusion of VEGF during reperfusion, but the beneficial effect of VEGF was reduced significantly by coadministration of L-NA. Reverse-transcriptase polymerase chain reaction study revealed that ischemia/reperfusion depressed eNOS mRNA expression but enhanced iNOS mRNA expression. Intraarterial infusion of VEGF during reperfusion amplified mRNA expression of eNOS but not of iNOS. CONCLUSIONS Local intraarterial infusion of VEGF produced significant microvascular protection from skeletal muscle ischemia/reperfusion injury. The VEGF-induced enhancement of eNOS may play an important mechanistic role.
Collapse
Affiliation(s)
- Wei Z Wang
- Division of Plastic Surgery, Department of Surgery, University of Nevada School Medicine, Las Vegas, Nev 89102, USA
| | | | | | | | | |
Collapse
|
147
|
Sun FY, Guo X. Molecular and cellular mechanisms of neuroprotection by vascular endothelial growth factor. J Neurosci Res 2005; 79:180-4. [PMID: 15573409 DOI: 10.1002/jnr.20321] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The present view of the neuroprotective functions and mechanisms of action of vascular endothelial growth factor (VEGF) is based on studies of neuronal ischemic/hypoxic models in vivo and in vitro. Endogenous neuronal VEGF increases in the ischemic brain and plays a neuroprotective role in the pathophysiologic processes that follow stroke. Exogenous VEGF, directly administered or overexpressed by gene delivery into rat brains, reduces ischemic brain infarct and decreases hypoxic neuronal death. The main neuroprotective mechanisms of VEGF include: (1) modulation of the phosphatidylinositol 3'-kinase (PI3K)/Akt/nuclear factor-kappaB signaling pathway, inhibition of caspase-3 activity, and reduction of ischemic neuronal apoptosis; (2) inhibition of outward delayed rectifier potassium channel currents and increase of ischemia-induced tyrosine phosphorylation of Kv1.2 potassium channel proteins via activation of the PI3K pathway; and (3) enhancement of proliferation and migration of neural progenitors in the subventricular zone and improvement of striatal neurogenesis and maturation of newborn neurons in adult rat brains after stroke.
Collapse
Affiliation(s)
- Feng-Yan Sun
- National Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, 138 Xue-Yuan Road, Shanghai 200 032, P.R. China.
| | | |
Collapse
|
148
|
Hayashi T, Hamakawa K, Nagotani S, Jin G, Li F, Deguchi K, Sehara Y, Zhang H, Nagano I, Shoji M, Abe K. HMG CoA reductase inhibitors reduce ischemic brain injury of Wistar rats through decreasing oxidative stress on neurons. Brain Res 2005; 1037:52-8. [PMID: 15777752 DOI: 10.1016/j.brainres.2004.12.051] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Revised: 12/15/2004] [Accepted: 12/17/2004] [Indexed: 11/22/2022]
Abstract
Statins possess neuroprotective effect against ischemic damage, but how they protect neurons is not exactly made clear. We speculated that anti-oxidative property of statins is implicated, and investigated statins' influences on the oxidative neuronal damage in the brain after ischemia. After 14 days of atorvastatin, pitavastatin, simvastatin, or vehicle administration, 90 min of middle cerebral artery occlusion was imposed on Wistar rats. The production of 4-hydroxynonenal (HNE) and 8-hydroxy-2'-deoxyguanosine (8-OHdG), both of which are oxidative stress markers, as well as infarction formation were investigated at 1 day after the reperfusion. In the vehicle group, massive infarction was confirmed and HNE and 8-OHdG are robustly produced. In the statins-treated group, the infarction was smaller and the HNE and 8-OHdG production was less prominent than the vehicle group. Among the statins investigated, simvastatin was most effective for reducing oxidative stress and infarction volume, which may be brought by its highly lipophilic property. Reduction of oxidative stress by statins may be one main reason in ameliorating ischemic brain damage in rats.
Collapse
Affiliation(s)
- Takeshi Hayashi
- Department of Neurology, Okayama University Graduate School of Medicine and Dentistry, 2-5-1 Shikata-cho, Okayama 700-8558, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Zhu W, Mao Y, Zhou LF. Reduction of neural and vascular damage by transplantation of VEGF-secreting neural stem cells after cerebral ischemia. ACTA NEUROCHIRURGICA. SUPPLEMENT 2005; 95:393-7. [PMID: 16463888 DOI: 10.1007/3-211-32318-x_80] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We determined the role of VEGF-transfected neural stem cells (NSCs) transplantation in rat brain subjected to ischemia. Fetal NSCs were cultured from E14 days SD rats and transfected with VEGF121 gene by using lipofectamine technique. Temporary middle cerebral artery occlusion (tMCAO) models were established and randomly divided into 1: control group, 2: PBS transplantation group, 3: NSCs transplantation group and 4: VEGF-secreting NSCs transplantation group. Grafts were transplanted into the penumbra zones 3 days after tMCAO model established. Neurological Severity Score (NSS) was checked in all groups 2-12 weeks after transplantation. By using immunofluorescent staining, VEGF expression of transplanted cells, differentiation and migration of transplanted NSCs after transplantation were detected. VEGF gene-transfected neural stem cells expressed gene products during the first 2 weeks. NSS in this group was significantly lower compared with that in other 3 groups 12 weeks after transplantation. VEGF gene-transfected NSCs migrated and expressed VEGF in hosts' brains, some of them differentiated to neurons 12 weeks after transplantation. VEGF-transfected NSCs expressed gene products during the early time after transplantation, which reduce brain injury through protecting the vascular system against ischemic attack.
Collapse
Affiliation(s)
- W Zhu
- Department of Neurosurgery, Shanghai Neurosurgical Center, Huashan Hospital, Fudan University, Shanghai, China.
| | | | | |
Collapse
|
150
|
Shimizu T, Uehara T, Nomura Y. Possible involvement of pyruvate kinase in acquisition of tolerance to hypoxic stress in glial cells. J Neurochem 2004; 91:167-75. [PMID: 15379897 DOI: 10.1111/j.1471-4159.2004.02702.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neurons are highly vulnerable to ischemic/hypoxic stress, while glial cells show tolerance to such stress. However, the mechanisms for tolerance acquisition in glial cells have yet to be established. We attempted to isolate and identify a stress protein that is upregulated in response to hypoxia in human astrocytoma CCF-STTG1 cells. In particular, pyruvate kinase (PK) was upregulated by hypoxia in CCF-STTG1 cells. Hypoxia-inducible factor 1 (HIF-1), the primary transcription factor that is responsible for multiple gene activation under hypoxia, plays a critical role in PK expression during hypoxic challenge. To determine whether newly synthesized PK is involved in tolerance to hypoxic stress, we established the PK-overexpressing neuronal cells. Overexpression of the wild-type, but not the kinase-negative mutant, resulted in attenuation of the loss of cell viability and the typical apoptotic features by hypoxia or oxidative stress in SK-N-MC cells. These findings suggest that upregulation of PK may result in acquisition of tolerance against hypoxic stress, and that the antioxidant effect may be involved in the protective effect of PK.
Collapse
MESH Headings
- Antimutagenic Agents/pharmacology
- Apoptosis/physiology
- Astrocytoma
- Blotting, Western/methods
- Cell Hypoxia/physiology
- Cell Line, Tumor
- Cell Survival/physiology
- Cobalt/pharmacology
- Deferoxamine/pharmacology
- Dose-Response Relationship, Drug
- Drug Interactions
- Electrophoresis, Gel, Two-Dimensional/methods
- Electrophoretic Mobility Shift Assay/methods
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hydrogen Peroxide/pharmacology
- Iron Chelating Agents/pharmacology
- Mutagenesis/drug effects
- Mutagenesis/physiology
- Neuroglia/drug effects
- Neuroglia/metabolism
- Oxidative Stress
- Pyruvate Kinase/physiology
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Sequence Analysis, Protein/methods
- Stress, Physiological/metabolism
- Stress, Physiological/pathology
- Time Factors
- Transcriptional Activation
- Transfection/methods
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | |
Collapse
|