1
|
Seder DB, Ryzhov S. Therapeutic opportunities for cerebral edema after resuscitation. Resuscitation 2022; 181:168-169. [PMID: 36423738 DOI: 10.1016/j.resuscitation.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Affiliation(s)
- David B Seder
- Maine Medical Center Department of Critical Care Services, Portland, ME, USA; MaineHealth Institute for Research, Scarborough, ME, USA
| | - Sergey Ryzhov
- Maine Medical Center Department of Critical Care Services, Portland, ME, USA; MaineHealth Institute for Research, Scarborough, ME, USA
| |
Collapse
|
2
|
Halder SK, Milner R. The impact of chronic mild hypoxia on cerebrovascular remodelling; uncoupling of angiogenesis and vascular breakdown. Fluids Barriers CNS 2021; 18:50. [PMID: 34789271 PMCID: PMC8597176 DOI: 10.1186/s12987-021-00284-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic mild hypoxia (CMH, 8% O2) stimulates robust vascular remodelling in the brain, but it also triggers transient vascular disruption. This raises the fundamental question: is the vascular leak an unwanted side-effect of angiogenic remodelling or is it a pathological response, unrelated to endothelial proliferation, in which declining oxygen levels trigger endothelial dysfunction? METHODS To answer this question, mice were exposed to CMH (8% O2) for periods up to 14 days, after which, brain tissue was examined by immunofluorescence (IF) to determine which type of blood vessel (arteriole, capillary or venule) was most commonly associated with endothelial proliferation and vascular leak and how this correlated with tight junction protein expression. Vascular perfusion was examined using DiI. Data were analysed using one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison post-hoc test. RESULTS The following was observed: (1) most endothelial proliferation and extravascular fibrinogen leak occurred in capillaries and to a lesser degree in venules, (2) much to our surprise, endothelial proliferation and extravascular fibrinogen leak never colocalized, (3) interestingly however, endothelial proliferation was strongly associated with an intravascular fibrinogen staining pattern not seen in stable blood vessels, (4) DiI perfusion studies revealed that angiogenic vessels were adequately perfused, suggesting that fibrinogen retention in angiogenic vessels is not due to temporary closure of the vessel, but more likely because fibrinogen is retained within the vessel wall, (5) bromodeoxyuridine (BrdU) labelling as a means to more permanently label proliferating endothelial cells, confirmed lack of any connection between endothelial proliferation and extravascular fibrinogen leak, while (6) in contrast, proliferating microglia were detected within extravascular leaks. CONCLUSIONS Taken together, our findings support the concept that in the short-term, hypoxia-induced endothelial proliferation triggers transient fibrinogen deposition within the walls of angiogenic blood vessels, but no overt vascular leak occurs in these vessels. Importantly, endothelial proliferation and extravascular fibrinogen leaks never co-localize, demonstrating that extravascular leak is not an unwanted side-effect of angiogenic endothelial proliferation, but rather a dysfunctional vascular response to hypoxia that occurs in a distinct group of non-angiogenic blood vessels.
Collapse
Affiliation(s)
- Sebok K Halder
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA, 92121, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA, 92121, USA.
| |
Collapse
|
3
|
Early Post-ischemic Brain Glucose Metabolism Is Dependent on Function of TLR2: a Study Using [ 18F]F-FDG PET-CT in a Mouse Model of Cardiac Arrest and Cardiopulmonary Resuscitation. Mol Imaging Biol 2021; 24:466-478. [PMID: 34779968 PMCID: PMC8592082 DOI: 10.1007/s11307-021-01677-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/17/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022]
Abstract
Purpose The mammalian brain glucose metabolism is tightly and sensitively regulated. An ischemic brain injury caused by cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) affects cerebral function and presumably also glucose metabolism. The majority of patients who survive CA suffer from cognitive deficits and physical disabilities. Toll-like receptor 2 (TLR2) plays a crucial role in inflammatory response in ischemia and reperfusion (I/R). Since deficiency of TLR2 was associated with increased survival after CA-CPR, in this study, glucose metabolism was measured using non-invasive [18F]F-FDG PET-CT imaging before and early after CA-CPR in a mouse model comparing wild-type (WT) and TLR2-deficient (TLR2−/−) mice. The investigation will evaluate whether FDG-PET could be useful as an additional methodology in assessing prognosis. Procedures Two PET-CT scans using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]F-FDG) tracer were carried out to measure dynamic glucose metabolism before and early after CPR. To achieve this, anesthetized and ventilated adult female WT and TLR2−/− mice were scanned in PET-CT. After recovery from the baseline scan, the same animals underwent 10-min KCL-induced CA followed by CPR. Approximately 90 min after CA, measurements of [18F]F-FDG uptake for 60 min were started. The [18F]F-FDG standardized uptake values (SUVs) were calculated using PMOD-Software on fused FDG-PET-CT images with the included 3D Mirrione-Mouse-Brain-Atlas. Results The absolute SUVmean of glucose in the whole brain of WT mice was increased about 25.6% after CA-CPR. In contrast, the absolute glucose SUV in the whole brain of TLR2−/− mice was not significantly different between baseline and measurements post CA-CPR. In comparison, baseline measurements of both mouse strains show a highly significant difference with regard to the absolute glucose SUV in the whole brain. Values of TLR2−/− mice revealed a 34.6% higher glucose uptake. Conclusions The altered mouse strains presented a different pattern in glucose uptake under normal and ischemic conditions, whereby the post-ischemic differences in glucose metabolism were associated with the function of key immune factor TLR2. There is evidence for using early FDG-PET-CT as an additional diagnostic tool after resuscitation. Further studies are needed to use PET-CT in predicting neurological outcomes.
Collapse
|
4
|
Li J, Li C, Yuan W, Wu J, Li J, Li Z, Zhao Y. Targeted Temperature Management Suppresses Hypoxia-Inducible Factor-1α and Vascular Endothelial Growth Factor Expression in a Pig Model of Cardiac Arrest. Neurocrit Care 2021; 35:379-388. [PMID: 33403582 PMCID: PMC7785329 DOI: 10.1007/s12028-020-01166-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND The hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF)/VEGF receptor subtype 2 (VEGFR-2) pathway has been implicated in ischemia/reperfusion injury. The aim of this study was to clarify whether whole-body hypothermic targeted temperature management (HTTM) inhibits the HIF-1α/VEGF/VEGFR-2 pathway in a swine model of cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). METHODS Twenty-four domestic male Beijing Landrace pigs were used in this study. CA was electrically induced with ventricular fibrillation and left untreated for 8 min. Return of spontaneous circulation (ROSC) was achieved in 16 pigs, which were randomly assigned either to normothermia at 38 °C or to HTTM at 33 °C (each group: n = 8). HTTM was intravascularly induced immediately after ROSC. The core temperature was reduced to 33 °C and maintained for 12 h after ROSC. The serum levels of HIF-1α, VEGF, VEGFR-2, and neuron-specific enolase (NSE) were measured with enzyme immunoassay kits 0.5, 6, 12, and 24 h after ROSC. The expression of HIF-1α, VEGF, and VEGFR-2 in cerebral cortical tissue was measured by RT-PCR and Western blot analysis 24 h after ROSC. Neurological deficit scores and brain cortical tissue water content were evaluated 24 h after ROSC. RESULTS The serum levels of HIF-1α, VEGF, and VEGFR-2 were significantly increased under normothermia within 24 h after ROSC. However, these increases were significantly reduced by HTTM. HTTM also decreased cerebral cortical HIF-1α, VEGF, and VEGFR-2 mRNA and protein expression 24 h after ROSC (all p < 0.05). HTTM pigs had better neurological outcomes and less brain edema than normothermic pigs. CONCLUSION The HIF-1α/VEGF/VEGFR-2 system is activated following CA and CPR. HTTM protects against cerebral injury after ROSC, which may be part of the mechanism by which it inhibits the expression of components of the HIF-1α/VEGF/VEGFR-2 signaling pathway.
Collapse
Affiliation(s)
- Jiebin Li
- Department of Emergency Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Chunsheng Li
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng, Beijing, 100050 China
| | - Wei Yuan
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020 China
| | - Junyuan Wu
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020 China
| | - Jie Li
- Department of Emergency Medicine, Beijing Fuxing Hospital, Capital Medical University, Beijing, 100038 China
| | - Zhenhua Li
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng, Beijing, 100050 China
| | - Yongzhen Zhao
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020 China
| |
Collapse
|
5
|
Azedi F, Mehrpour M, Talebi S, Zendedel A, Kazemnejad S, Mousavizadeh K, Beyer C, Zarnani AH, Joghataei MT. Melatonin regulates neuroinflammation ischemic stroke damage through interactions with microglia in reperfusion phase. Brain Res 2019; 1723:146401. [PMID: 31445031 DOI: 10.1016/j.brainres.2019.146401] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022]
Abstract
Even today, ischemic stroke is a major cause of death and disabilities because of its high incidence, limited treatments and poor understanding of the pathophysiology of ischemia/reperfusion, neuroinflammation and secondary injuries following ischemic stroke. The function of microglia as a part of the immune system of the brain following ischemic stroke can be destructive or protective. Recent surveys indicate that melatonin, a strong antioxidant agent, has receptors on microglial cells and can regulate them to protective form; yet, more findings are required for better understanding of this mechanism, particularly in the reperfusion phase. In this study, we initially aimed to evaluate the therapeutic efficacy of melatonin intra-arterially and to clarify the underlying mechanisms. After that by using an in vitro approach, we evaluated the protective effects of melatonin on microglial cells following the hypoxia condition. Our results proved that a single dose of melatonin at the beginning of reperfusion phase improved structural and behavioral outcomes. Melatonin increased NeuN and decreased GFAP, Iba1 and active caspase-3 at protein level. Furthermore, melatonin elevated BDNF, MAP2, HSPA1A and reduced VEGF at mRNA level. We also showed that melatonin receptor 1B highly expressed in microglial cells after 3 h hypoxia. Besides, melatonin increased the ratio of TREM2/iNOS as a marker of the most protective form of microglia (M2). In summary, our data suggest that melatonin has the possibility to serve as targeting microglial action for preventing secondary injury of reperfusion phase after ischemic stroke.
Collapse
Affiliation(s)
- Fereshteh Azedi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Mehrpour
- Department of Neurology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Talebi
- Department of Medical Genetics, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Somaieh Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Kazem Mousavizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Ryzhov S, May T, Dziodzio J, Emery IF, Lucas FL, Leclerc A, McCrum B, Lord C, Eldridge A, Robich MP, Ichinose F, Sawyer DB, Riker R, Seder DB. Number of Circulating CD 73-Expressing Lymphocytes Correlates With Survival After Cardiac Arrest. J Am Heart Assoc 2019; 8:e010874. [PMID: 31237169 PMCID: PMC6662342 DOI: 10.1161/jaha.118.010874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Patients resuscitated from cardiac arrest (CA) have highly variable neurological, circulatory, and systemic ischemia‐reperfusion injuries. After the initial hypoxic‐ischemic insult, a cascade of immune and inflammatory responses develops and is often fatal. The role of the immune response in pathophysiological characteristics and recovery is not well understood. We studied immune cell activity and its association with outcomes in a cohort of CA survivors. Methods and Results After informed consent, we collected blood samples at intervals over a week after resuscitation from CA. We examined the expression of CD39 and CD73 (alias 5′‐nucleotidase), production of tumor necrosis factor‐α, generation of reactive oxygen species, and secretion of vascular endothelial growth factor by circulating myeloid and lymphoid cells, in comparison to cells obtained from control subjects before coronary artery bypass grafting surgery. The number of circulating total and CD73‐expressing lymphocytes correlated with survival after CA. Incubation of immune cells, obtained from post‐CA subjects, with AMP, a substrate for CD73, resulted in inhibition of tumor necrosis factor‐α production and generation of reactive oxygen species. This effect was blocked by adenosine 5′‐(α, β‐methylene) diphosphate, a specific inhibitor of CD73 and ZM 241385, an A2 adenosine receptor antagonist. We also found that AMP‐dependent activation of CD73 induces production of vascular endothelial growth factor. Conclusions CD73‐expressing lymphocytes mediate cellular protection from inflammation after CA through inhibition of proinflammatory activation of myeloid cells and promotion of vascular endothelial growth factor secretion. The contribution of CD73 lymphocytes in the regulation of acute inflammation and tissue injury after CA warrants further study.
Collapse
Affiliation(s)
- Sergey Ryzhov
- 1 Maine Medical Center Research Institute Scarborough ME
| | - Teresa May
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| | - John Dziodzio
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Ivette F Emery
- 1 Maine Medical Center Research Institute Scarborough ME
| | - F L Lucas
- 3 Center for Outcomes Research and Evaluation Maine Medical Center Portland ME
| | - Angela Leclerc
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Barbara McCrum
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Christine Lord
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Ashley Eldridge
- 2 Department of Critical Care Services Maine Medical Center Portland ME
| | - Michel P Robich
- 1 Maine Medical Center Research Institute Scarborough ME.,4 Maine Medical Center Cardiovascular Institute Portland ME
| | - Fumito Ichinose
- 5 Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston MA
| | - Douglas B Sawyer
- 1 Maine Medical Center Research Institute Scarborough ME.,4 Maine Medical Center Cardiovascular Institute Portland ME
| | - Richard Riker
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| | - David B Seder
- 1 Maine Medical Center Research Institute Scarborough ME.,2 Department of Critical Care Services Maine Medical Center Portland ME
| |
Collapse
|
7
|
Rehni AK, Liu A, Perez-Pinzon MA, Dave KR. Diabetic aggravation of stroke and animal models. Exp Neurol 2017; 292:63-79. [PMID: 28274862 PMCID: PMC5400679 DOI: 10.1016/j.expneurol.2017.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/03/2017] [Accepted: 03/03/2017] [Indexed: 12/16/2022]
Abstract
Cerebral ischemia in diabetics results in severe brain damage. Different animal models of cerebral ischemia have been used to study the aggravation of ischemic brain damage in the diabetic condition. Since different disease conditions such as diabetes differently affect outcome following cerebral ischemia, the Stroke Therapy Academic Industry Roundtable (STAIR) guidelines recommends use of diseased animals for evaluating neuroprotective therapies targeted to reduce cerebral ischemic damage. The goal of this review is to discuss the technicalities and pros/cons of various animal models of cerebral ischemia currently being employed to study diabetes-related ischemic brain damage. The rational use of such animal systems in studying the disease condition may better help evaluate novel therapeutic approaches for diabetes related exacerbation of ischemic brain damage.
Collapse
Affiliation(s)
- Ashish K Rehni
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Allen Liu
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
8
|
Li J, Li C, Yuan W, Wu J, Li J, Li Z, Zhao Y. Mild hypothermia alleviates brain oedema and blood-brain barrier disruption by attenuating tight junction and adherens junction breakdown in a swine model of cardiopulmonary resuscitation. PLoS One 2017; 12:e0174596. [PMID: 28355299 PMCID: PMC5371345 DOI: 10.1371/journal.pone.0174596] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/11/2017] [Indexed: 12/21/2022] Open
Abstract
Mild hypothermia improves survival and neurological recovery after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). However, the mechanism underlying this phenomenon is not fully elucidated. The aim of this study was to determine whether mild hypothermia alleviates early blood-brain barrier (BBB) disruption. We investigated the effects of mild hypothermia on neurologic outcome, survival rate, brain water content, BBB permeability and changes in tight junctions (TJs) and adherens junctions (AJs) after CA and CPR. Pigs were subjected to 8 min of untreated ventricular fibrillation followed by CPR. Mild hypothermia (33°C) was intravascularly induced and maintained at this temperature for 12 h, followed by active rewarming. Mild hypothermia significantly reduced cortical water content, decreased BBB permeability and attenuated TJ ultrastructural and basement membrane breakdown in brain cortical microvessels. Mild hypothermia also attenuated the CPR-induced decreases in TJ (occludin, claudin-5, ZO-1) and AJ (VE-cadherin) protein and mRNA expression. Furthermore, mild hypothermia decreased the CA- and CPR-induced increases in matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF) expression and increased angiogenin-1 (Ang-1) expression. Our findings suggest that mild hypothermia attenuates the CA- and resuscitation-induced early brain oedema and BBB disruption, and this improvement might be at least partially associated with attenuation of the breakdown of TJ and AJ, suppression of MMP-9 and VEGF expression, and upregulation of Ang-1 expression.
Collapse
Affiliation(s)
- Jiebin Li
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chunsheng Li
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- * E-mail:
| | - Wei Yuan
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Junyuan Wu
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jie Li
- Department of Emergency Medicine, Beijing FuXing Hospital, Capital Medical University, Beijing, China
| | - Zhenhua Li
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yongzhen Zhao
- Beijing Key Laboratory of Cardiopulmonary-Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Egervari K, Potter G, Guzman-Hernandez ML, Salmon P, Soto-Ribeiro M, Kastberger B, Balla T, Wehrle-Haller B, Kiss JZ. Astrocytes spatially restrict VEGF signaling by polarized secretion and incorporation of VEGF into the actively assembling extracellular matrix. Glia 2015; 64:440-56. [PMID: 26539695 DOI: 10.1002/glia.22939] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 01/13/2023]
Abstract
The spatial organization of vascular endothelial growth factor (VEGF) signaling is a key determinant of vascular patterning during development and tissue repair. How VEGF signaling becomes spatially restricted and the role of VEGF secreting astrocytes in this process remains poorly understood. Using a VEGF-GFP fusion protein and confocal time-lapse microscopy, we observed the intracellular routing, secretion and immobilization of VEGF in scratch-activated living astrocytes. We found VEGF to be directly transported to cell-extracellular matrix attachments where it is incorporated into fibronectin fibrils. VEGF accumulated at β1 integrin containing fibrillar adhesions and was translocated along the cell surface prior to internalization and degradation. We also found that only the astrocyte-derived, matrix-bound, and not soluble VEGF decreases β1 integrin turnover in fibrillar adhesions. We suggest that polarized VEGF release and ECM remodeling by VEGF secreting cells is key to control the local concentration and signaling of VEGF. Our findings highlight the importance of astrocytes in directing VEGF functions and identify these mechanisms as promising target for angiogenic approaches.
Collapse
Affiliation(s)
| | - Gael Potter
- Department of Neurosciences, University of Geneva, Switzerland
| | - Maria Luisa Guzman-Hernandez
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Patrick Salmon
- Department of Neurosciences, University of Geneva, Switzerland
| | | | - Birgit Kastberger
- Department of Cell Physiology and Metabolism, University of Geneva, Switzerland
| | - Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
10
|
Schmidt-Kastner R. Genomic approach to selective vulnerability of the hippocampus in brain ischemia–hypoxia. Neuroscience 2015; 309:259-79. [DOI: 10.1016/j.neuroscience.2015.08.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023]
|
11
|
Geng Y, Li E, Mu Q, Zhang Y, Wei X, Li H, Cheng L, Zhang B. Hydrogen sulfide inhalation decreases early blood-brain barrier permeability and brain edema induced by cardiac arrest and resuscitation. J Cereb Blood Flow Metab 2015; 35:494-500. [PMID: 25492119 PMCID: PMC4348391 DOI: 10.1038/jcbfm.2014.223] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/12/2022]
Abstract
The effects of hydrogen sulfide (H2S) on blood-brain barrier (BBB) and brain edema after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) remain poorly understood. We investigated the effects of exogenous 80-p.p.m. H2S gas on BBB, brain water content, neurologic outcome, and survival rate after CA and CPR. Cardiopulmonary resuscitation followed CA induced in rats by ventricular fibrillation for 6 minutes. Results show that inhalation of 80-p.p.m. H2S significantly reduced the permeability of the BBB in both in the cortex and hippocampus at 24 hours after resuscitation. Hydrogen sulfide also lessened brain edema in the cortex and hippocampus, ameliorated neurologic outcome as evaluated by neurologic deficit score and tape removal test, and improved the 14-day survival rate. Hydrogen sulfide also attenuated CA and CPR-induced increases of matrix metalloproteinase-9 (MMP-9) activity and vascular endothelial growth factor (VEGF) expression, and increased the expression of angiogenin-1 (Ang-1). These results indicate that inhalation of 80-p.p.m. H2S immediately after CPR attenuated BBB permeability and brain edema, and improved neurologic outcome and 14-day survival of rats after CA. The therapeutic benefits of H2S could be associated with suppression of MMP-9 and VEGF expression and increased expression of Ang-1.
Collapse
Affiliation(s)
- Yingjie Geng
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Eerdunmutu Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qier Mu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Wei
- Department of Anesthesiology, Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hangbing Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Cheng
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Boroujerdi A, Milner R. Defining the critical hypoxic threshold that promotes vascular remodeling in the brain. Exp Neurol 2015; 263:132-40. [PMID: 25448157 PMCID: PMC4261640 DOI: 10.1016/j.expneurol.2014.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/07/2014] [Accepted: 10/13/2014] [Indexed: 11/18/2022]
Abstract
In animal models, hypoxic pre-conditioning confers protection against subsequent neurological insults, mediated in part through an extensive vascular remodeling response. In light of the therapeutic potential of this effect, the goal of this study was to establish the dose-response relationship between level of hypoxia and the extent of cerebrovascular modeling, and to define the mildest level of hypoxia that promotes remodeling. Mice were exposed to different levels of continuous hypoxia (8-21% O2) for seven days before several aspects of vascular remodeling were evaluated, including endothelial proliferation, total vascular area, arteriogenesis, and fibronectin/α5β1 integrin expression. For most events, the threshold level of hypoxia that stimulated remodeling was 12-13% O2. Interestingly, many parameters displayed a biphasic dose-response curve, with peak levels attained at 10% O2, but declined thereafter. Further analysis in the 12-13% O2 range revealed that vascular remodeling occurs by two separate mechanisms: (i) endothelial hyperplasia, triggered by a hypoxic threshold of 13% O2, which leads to increased capillary growth, and (ii) endothelial hypertrophy, triggered by a more severe hypoxic threshold of 12% O2, which leads to expansion of large vessels and arteriogenesis. Taken together, these results define the hypoxic thresholds for vascular remodeling in the brain, and point to two separate mechanisms mediating this process.
Collapse
Affiliation(s)
- Amin Boroujerdi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Richard Milner
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Rocha SF, Schiller M, Jing D, Li H, Butz S, Vestweber D, Biljes D, Drexler HC, Nieminen-Kelhä M, Vajkoczy P, Adams S, Benedito R, Adams RH. Esm1 Modulates Endothelial Tip Cell Behavior and Vascular Permeability by Enhancing VEGF Bioavailability. Circ Res 2014; 115:581-90. [DOI: 10.1161/circresaha.115.304718] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Susana F. Rocha
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Maria Schiller
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Ding Jing
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Hang Li
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Stefan Butz
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Dietmar Vestweber
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Daniel Biljes
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Hannes C.A. Drexler
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Melina Nieminen-Kelhä
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Peter Vajkoczy
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Susanne Adams
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Rui Benedito
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Ralf H. Adams
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| |
Collapse
|
14
|
The beneficial effect of melatonin in brain endothelial cells against oxygen-glucose deprivation followed by reperfusion-induced injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:639531. [PMID: 25126203 PMCID: PMC4122057 DOI: 10.1155/2014/639531] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/07/2014] [Accepted: 06/20/2014] [Indexed: 12/31/2022]
Abstract
Melatonin has a cellular protective effect in cerebrovascular and neurodegenerative diseases. Protection of brain endothelial cells against hypoxia and oxidative stress is important for treatment of central nervous system (CNS) diseases, since brain endothelial cells constitute the blood brain barrier (BBB). In the present study, we investigated the protective effect of melatonin against oxygen-glucose deprivation, followed by reperfusion- (OGD/R-) induced injury, in bEnd.3 cells. The effect of melatonin was examined by western blot analysis, cell viability assays, measurement of intracellular reactive oxygen species (ROS), and immunocytochemistry (ICC). Our results showed that treatment with melatonin prevents cell death and degradation of tight junction protein in the setting of OGD/R-induced injury. In response to OGD/R injury of bEnd.3 cells, melatonin activates Akt, which promotes cell survival, and attenuates phosphorylation of JNK, which triggers apoptosis. Thus, melatonin protects bEnd.3 cells against OGD/R-induced injury.
Collapse
|
15
|
Dave KR, Della-Morte D, Saul I, Prado R, Perez-Pinzon MA. Ventricular fibrillation-induced cardiac arrest in the rat as a model of global cerebral ischemia. Transl Stroke Res 2013; 4:571-8. [PMID: 24187598 PMCID: PMC3811953 DOI: 10.1007/s12975-013-0267-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cardiopulmonary arrest remains one of the leading causes of death and disability in Western countries. Although ventricular fibrillation (VF) models in rodents mimic the "square wave" type of insult (rapid loss of pulse and pressure) commonly observed in adult humans at the onset of cardiac arrest (CA), they are not popular because of the complicated animal procedure, poor animal survival and thermal injury. Here we present a modified, simple, reliable, ventricular fibrillation-induced rat model of CA that will be useful in studying mechanisms of CA-induced delayed neuronal death as well as the efficacy of neuroprotective drugs. CA was induced in male Sprague Dawley rats using a modified method of von Planta et al. In brief, VF was induced in anesthetized, paralyzed, mechanically ventilated rats by an alternating current delivered to the entrance of the superior vena cava into the heart. Resuscitation was initiated by administering a bolus injection of epinephrine and sodium bicarbonate followed by mechanical ventilation and manual chest compressions and countershock with a 10-J DC current. Neurologic deficit score was higher in the CA group compared to the sham group during early reperfusion periods, suggesting brain damage. Significant damage in CA1 hippocampus (21% normal neurons compared to control animals) was observed following histopathological assessment at seven days of reperfusion. We propose that this method of VF-induced CA in rat provides a tool to study the mechanism of CA-induced neuronal death without compromising heart functions.
Collapse
Affiliation(s)
- Kunjan R. Dave
- The Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, FL 33136
| | - David Della-Morte
- The Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, FL 33136
- Department of Advanced Biotechnologies and Bioimaging, IRCCS San Raffaele, Rome, Italy
| | - Isabel Saul
- The Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, FL 33136
| | - Ricardo Prado
- The Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, FL 33136
| | - Miguel A. Perez-Pinzon
- The Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, FL 33136
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136
| |
Collapse
|
16
|
Exercise therapy augments the ischemia-induced proangiogenic state and results in sustained improvement after stroke. Int J Mol Sci 2013; 14:8570-84. [PMID: 23598418 PMCID: PMC3645762 DOI: 10.3390/ijms14048570] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/11/2013] [Accepted: 04/11/2013] [Indexed: 01/19/2023] Open
Abstract
The induction of angiogenesis will stimulate endogenous recovery mechanisms, which are involved in the long-term repair and restoration process of the brain after an ischemic event. Here, we tested whether exercise influences the pro-angiogenic factors and outcomes after cerebral infarction in rats. Wistar rats were exposed to two hours of middle-cerebral artery occlusion and reperfusion. Different durations of treadmill training were performed on the rats. The expression of matrix metalloproteinase 2 (MMP2) and vascular endothelial growth factor (VEGF)-related genes and proteins were higher over time post-ischemia, and exercise enhanced their expression. Sixteen days post-ischemia, the regional cerebral blood flow in the ischemic striatum was significantly increased in the running group over the sedentary. Although no difference was seen in infarct size between the running and sedentary groups, running evidently improved the neurobehavioral score. The effects of running on MMP2 expression, regional cerebral blood flow and outcome were abolished when animals were treated with bevacizumab (BEV), a VEGF-targeting antibody. Exercise therapy improves long-term stroke outcome by MMP2-VEGF-dependent mechanisms related to improved cerebral blood flow.
Collapse
|
17
|
Seiffge DJ, Lapina NE, Tsagogiorgas C, Theisinger B, Henning RH, Schilling L. Improvement of oxygen supply by an artificial carrier in combination with normobaric oxygenation decreases the volume of tissue hypoxia and tissue damage from transient focal cerebral ischemia. Exp Neurol 2012; 237:18-25. [PMID: 22728375 DOI: 10.1016/j.expneurol.2012.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 06/04/2012] [Accepted: 06/09/2012] [Indexed: 11/29/2022]
Abstract
Tissue hypoxia may play an important role in the development of ischemic brain damage. In the present study we investigated in a rat model of transient focal brain ischemia the neuroprotective effects of increasing the blood oxygen transport capacity by applying a semifluorinated alkane (SFA)-containing emulsion together with normobaric hyperoxygenation (NBO). The spread of tissue hypoxia was studied using pimonidazole given prior to filament-induced middle cerebral artery occlusion (MCAO, 2 h). Treatment consisted of intravenous injection of saline or the SFA-containing emulsion (0.5 or 1.0 ml/100g body weight; [SFA(0.5) or SFA(1.0)]) either upon establishing MCAO (early treatment) or after filament removal (delayed treatment). After injection NBO was administered for 8 h (early treatment) or 6 h (delayed treatment). Experiments were terminated 8 or 24 h after MCAO. In serial brain sections tissue hypoxia and irreversible cell damage were quantitatively determined. Furthermore, we studied hypoxia-related gene expression (VEGF, flt-1). Early treatment significantly (p<0.05) reduced the volumes of tissue damage (8 h after MCAO: SFA(1.0), 57±34 mm³; controls, 217±70 mm³; 24 h after MCAO: SFA(1.0), 189±82 mm³; controls, 317±60 mm³) and of P-Add immunoreactivity (8 h after MCAO: SFA(1.0), 261±37 mm³; controls, 339±26 mm³; 24h after MCAO: SFA(1.0), 274±47 mm³; controls, 364±46 mm³). Delayed treatment was comparably successful. The volume of the hypoxic penumbra was not decreased by the treatment. Similarly, VEGF and flt-1 mRNA levels did not differ between the experimental groups. From these data we conclude that increasing the blood oxygen transport capacity in the plasma compartment provides a neuroprotective effect by alleviating the severity of hypoxia to a level sufficient to prevent cells from transition into irreversible damage.
Collapse
Affiliation(s)
- David J Seiffge
- Division of Neurosurgical Research, Medical Faculty Mannheim, University of Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Wuestefeld R, Chen J, Meller K, Brand-Saberi B, Theiss C. Impact of vegf on astrocytes: analysis of gap junctional intercellular communication, proliferation, and motility. Glia 2012; 60:936-47. [PMID: 22431192 DOI: 10.1002/glia.22325] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 02/16/2012] [Indexed: 12/12/2022]
Abstract
The purpose of the present study was to investigate the effects of vascular endothelial growth factor (VEGF) on gap junctional intercellular communication (GJIC), cell proliferation, and cell dynamics in primary astrocytes. VEGF is known as a dimeric polypeptide that potentially binds to two receptors, VEGFR-1 and VEGFR-2, however many effects are mediated by VEGFR-2, for example, actin polymerization, forced cell migration, angiogenesis, and cell proliferation. Recently it has been shown that in case of hypoxia, ischemia or injury VEGF is upregulated to stimulate angiogenesis and cell proliferation. Besides this, VEGF reveals a potent therapeutical target for averting tumor vascularization, emerging in bevacizumab, the first humanized anti-VEGF-A antibody for treating recurrent Glioblastoma multiforme. To expand our knowledge about VEGF effects in glial cells, we cultivated rat astrocytes in medium containing VEGF for 1 and 2 days. To investigate the effects of VEGF on GJIC, we microinjected neurobiotin into a single cell and monitored dye-spreading into adjacent cells. These experiments showed that VEGF significantly enhances astrocytic GJIC compared with controls. Cell proliferation measured by BrdU-labeling also revealed a significant increase of astrocytic mitose rates subsequent to 1 day of VEGF exposure, whereas longer VEGF treatment for 2 days did not have additive effects. To study cell-dynamics of astrocytes subsequent to VEGF treatment, we additionally transfected astrocytes with LifeAct-RFP. Live-cell imaging and quantitative analysis of these cells with aid of confocal laser scanning microscopy revealed higher process movement of VEGF-treated astrocytes. In conclusion, VEGF strongly affects cell proliferation, GJIC, and motility in astrocytes.
Collapse
Affiliation(s)
- Ricarda Wuestefeld
- Institute of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | |
Collapse
|
19
|
Dexamethasone pre-treatment protects brain against hypoxic-ischemic injury partially through up-regulation of vascular endothelial growth factor A in neonatal rats. Neuroscience 2011; 179:223-32. [PMID: 21277350 DOI: 10.1016/j.neuroscience.2011.01.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/21/2011] [Accepted: 01/22/2011] [Indexed: 10/18/2022]
Abstract
Dexamethasone (Dex) provides neuroprotection against subsequent hypoxia ischemia (HI) in newborn rats, but the mechanism of this neuroprotection is not well understood. It is known that vascular endothelial growth factor A (VEGF) has neuroprotective effects. The objective of this study was to evaluate the role of the VEGF signaling pathway in the Dex-induced neuroprotection in newborn rats. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 140 or 160 min of hypoxia (8% oxygen). Rat pups received two i.p. injections of either saline or Dex (0.25 mg/kg) at 24 and 4 h before HI exposure. To quantify the effects of a glucocorticoid receptor (GR) blocker, on postnatal day (PD) 6 and 15 min prior to Dex treatment rat pups received s.c. vehicle or RU486 (GR blocker, 60 mg/kg). After 24 h at PD 7, all rat pups had HI as described earlier. To quantify the effects of a VEGFR 2 blocker, at 24 h after Dex/Veh treatment (PD7), SU5416, a VEGFR 2 inhibitor or vehicle was injected intracerebroventricularly in the right hemisphere at 30 min before and 2 h after HI. Dex pre-treatment reduced brain injury and enhanced the HI-induced brain VEGF protein while a GR blocker inhibited these effects. Treatment with VEGFR 2 blocker decreased Dex-induced neuroprotection also. Dex pre-treatment enhanced the HI-induced increase in mRNA expression of VEGF splice variants and decreased the HI-induced reduction of Akt phosphorylation. Additionally, it also decreased HI-induced increase of caspase-3 activity and DNA fragments in neonatal rat brain. We conclude that Dex provides robust neuroprotection against subsequent HI in newborn rats via GR likely with the partial involvement of VEGF signaling pathway.
Collapse
|
20
|
Pathology and new players in the pathogenesis of brain edema. Acta Neuropathol 2009; 118:197-217. [PMID: 19404652 DOI: 10.1007/s00401-009-0541-0] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 04/16/2009] [Accepted: 04/21/2009] [Indexed: 12/20/2022]
Abstract
Brain edema continues to be a major cause of mortality after diverse types of brain pathologies such as major cerebral infarcts, hemorrhages, trauma, infections and tumors. The classification of edema into vasogenic, cytotoxic, hydrocephalic and osmotic has stood the test of time although it is recognized that in most clinical situations there is a combination of different types of edema during the course of the disease. Basic information about the types of edema is provided for better understanding of the expression pattern of some of the newer molecules implicated in the pathogenesis of brain edema. These molecules include the aquaporins, matrix metalloproteinases and growth factors such as vascular endothelial growth factors A and B and the angiopoietins. The potential of these agents in the treatment of edema is discussed. Since many molecules are involved in the pathogenesis of brain edema, effective treatment cannot be achieved by a single agent but will require the administration of a "magic bullet" containing a variety of agents released at different times during the course of edema in order to be successful.
Collapse
|
21
|
Schmid-Brunclik N, Bürgi-Taboada C, Antoniou X, Gassmann M, Ogunshola OO. Astrocyte responses to injury: VEGF simultaneously modulates cell death and proliferation. Am J Physiol Regul Integr Comp Physiol 2008; 295:R864-73. [PMID: 18614764 DOI: 10.1152/ajpregu.00536.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia is linked to changes in blood-brain barrier (BBB) permeability, and loss of BBB integrity is characteristic of many pathological brain diseases including stroke. In particular, astrocytes play a central role in brain homeostasis and BBB function. We investigated how hypoxia affects astrocyte survival and assessed whether VEGF release through hypoxia-inducible factor-1alpha (HIF-1alpha) induction plays a role in tolerance of these cells to insult. Thus primary astrocytes were subjected to normoxic (21% O(2)), hypoxic (1% O(2)), or near-anoxic (<0.1% O(2)) conditions in the presence or absence of glucose. Cell death was significantly initiated after combined oxygen glucose deprivation, and, surprisingly, astrocyte proliferation increased concomitantly. Near anoxic, but not hypoxic, conditions stabilized HIF-1alpha protein and provoked DNA binding activity, whereas oxygen and glucose deprivation accelerated HIF-1alpha accumulation. Unexpectedly, Hif-1alpha knockdown studies showed that elevated VEGF levels following increased insult was only partially due to HIF-1alpha induction, suggesting alternative mechanisms of VEGF regulation. Notably, endogenous VEGF signaling during insult was essential for cell fate since VEGF inhibition appreciably augmented cell death and reduced proliferation. These data suggest Hif-1 only partially contributes to VEGF-mediated astrocyte responses during chronic injury (as occurs in clinical hypoxic/ischemic insults) that may ultimately be responsible for disrupting BBB integrity.
Collapse
Affiliation(s)
- Nicole Schmid-Brunclik
- Institute of Veterinary Physiology, Vetsuisse Faculty, Univ. of Zurich, Winterthurerstrasse 260, Zurich CH 8057, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Krum JM, Mani N, Rosenstein JM. Roles of the endogenous VEGF receptors flt-1 and flk-1 in astroglial and vascular remodeling after brain injury. Exp Neurol 2008; 212:108-17. [PMID: 18482723 DOI: 10.1016/j.expneurol.2008.03.019] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 03/07/2008] [Accepted: 03/12/2008] [Indexed: 01/13/2023]
Abstract
Following trauma to the brain significant changes occur in both the astroglial and vascular components of the neuropil. Angiogenesis is required to re-establish metabolic support and astrocyte activation encompasses several functions including scar formation and the production of growth factors. VEGF has seminal involvement in the process of brain repair and is upregulated during many pathological events. VEGF signaling is regulated mainly through its two primary receptors: flk-1 (KDR/VEGF-R2) is expressed on vascular endothelium and some neurons and flt-1 (VEGF-R1) in the CNS, is expressed predominantly by activated astrocytes. Using an injury model of chronic minipump infusion of neutralizing antibodies (NA) to block VEGF receptor signaling, this study takes advantage of these differences in VEGF receptor distribution in order to understand the role the cytokine plays after brain injury. Infusion of NA to flk-1 caused a significant decrease in vascular proliferation and increased endothelial cell degeneration compared to control IgG infusions but had no effect on astrogliosis. By contrast infusion of NA to flt-1 significantly decreased astroglial mitogenicity and scar formation and caused some increase in endothelial degeneration. Neutralization of the flt-1 receptor function, but not flk-1, caused significant reduction in the astroglial expression of the growth factors, CNTF and FGF by 7days. These data suggest that after CNS injury, endogenous VEGF upregulation (by astrocytes) induces angiogenesis and, by autocrine signaling, increases both astrocyte proliferation and facilitates expression of growth factors. It is likely that VEGF plays an important role in aspects of astroglial scar formation.
Collapse
Affiliation(s)
- Janette M Krum
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, 2300 I Street NW, Washington, DC 20037, USA.
| | | | | |
Collapse
|
23
|
Ekerbicer N, Tarakci F, Barut T, Inan S. Immunolocalization of VEGF, VEGFR-1 and VEGFR-2 in lung tissues after acute hemorrhage in rats. Acta Histochem 2008; 110:285-93. [PMID: 18321563 DOI: 10.1016/j.acthis.2007.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/28/2007] [Accepted: 10/09/2007] [Indexed: 11/28/2022]
Abstract
In treatment of hypovolemia it is important to reestablish normal tissue hemodynamics after fluid resuscitation. Vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) have been identified as important in many physiological and pathological processes. In this study, we aimed to investigate the histo-physiological effects of VEGF, VEGFR-1 (flt-1) and VEGFR-2 (KDR/flk-1) in resuscitation with different plasma substitutes on lung tissues after acute hemorrhage in rats. Male Sprague-Dawley rats (n=25) were used in this study. The left femoral vein and artery were cannulated for the administration of volume expanders and for direct measurement of mean arterial blood pressure (MAP) (Power-Lab) and heart rate (HR). Fifteen rats were bled (5 ml/10 min) and infused (5 ml/5 min) with one of three randomly selected fluids: (a) dextran-70 (Macrodex); (b) gelatin (Gelofusine); or (c) physiological saline (PS, 0.9% isotonic saline) solutions. Five rats were bled and none were infused (hypovolemia group) and five rats were untreated as the control group. At the end of the experiment, rats were sacrificed and lung tissues were removed for routine processing and paraffin wax embedding. Sections of tissue were stained with hematoxylin and eosin (H&E) and selected blocks were then prepared for indirect immunohistochemical labeling for anti-VEGF, anti-VEGFR-1 and anti-VEGFR-2 primary antibodies. It was observed that both MAP and HR decreased parallel to blood withdrawn in this time interval. The MAP and HR were restored in the following periods. In the control rats, positive immunoreactivity of VEGF and its receptors (VEGFR-1 and VEGFR-2) were detected in respiratory epithelial cells, respiratory and vascular smooth muscle cells, alveolar cells and endothelial cells. While strong immunoreactivities of VEGF and VEGFR-1 were observed in the hypovolemia group, only moderate immunoreactivity of VEGFR-2 was seen in this group. Moderately strong immunolabeling of VEGF and VEGFR-1 were observed in the dextran-70, gelatin and PS resuscitated groups, whereas only weak immunolabeling of VEGFR-2 was observed in these groups. In summary, the vascular protecting effects of these factors were observed with fluid resuscitation, contributing to the pathophysiological changes seen in hypovolemia.
Collapse
Affiliation(s)
- Nuran Ekerbicer
- Department of Physiology, Faculty of Medicine, Celal Bayar University, Dekanlik Binasi, Uncubozkoy Mevkii, 45030 Manisa, Turkey.
| | | | | | | |
Collapse
|
24
|
Dombrowski SM, Deshpande A, Dingwall C, Leichliter A, Leibson Z, Luciano MG. Chronic hydrocephalus-induced hypoxia: increased expression of VEGFR-2+ and blood vessel density in hippocampus. Neuroscience 2007; 152:346-59. [PMID: 18262364 DOI: 10.1016/j.neuroscience.2007.11.049] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 11/09/2007] [Accepted: 11/29/2007] [Indexed: 12/11/2022]
Abstract
Chronic hydrocephalus (CH) is a neurological disease characterized by increased cerebrospinal fluid volume and pressure that is often associated with impaired cognitive function. By and large, CH is a complex and heterogeneous cerebrospinal fluid (CSF) disorder where the exact site of brain insult is uncertain. Several mechanisms including neural compression, fiber stretch, and local or global hypoxia have been implicated in the underlying pathophysiology of CH. Specifically, the hippocampus, which plays a significant role in memory processing and is in direct contact with expanding CSF ventricles, may be involved. Using our model of chronic hydrocephalus, we quantified the density of vascular endothelial growth factor receptor 2 (VEGFR-2(+)) neurons, glial, endothelial cells, and blood vessels in hippocampal regions CA1, CA2-3, dentate gyrus and hilus using immunohistochemical and stereological methods. Density and %VEGFR-2(+) cell populations were estimated for CH animals (2-3 weeks vs. 12-16 weeks) and surgical controls (SC). Overall, we found approximately six- to eightfold increase in the cellular density of VEGFR-2(+) and more than double blood vessel density (BVd) in the hippocampus of CH compared with SC. There were no significant regional differences in VEGFR-2(+) cellular and BVd expression in the CH group. VEGFR-2(+) and BVds were significantly related to changes in CSF volume (P<or=0.05), and not intracranial pressure (ICP). The %VEGFR-2(+) was significantly greater in CH than SC (P<or=0.05), and was significantly correlated with BVd (P<or=0.05). These results showed that CH elicited a profound increase in VEGFR-2(+) in hippocampus that corresponded to increased BVd. It was unclear whether increased VEGFR-2(+) and blood vessel expression was related to focal compression alone or in combination with global ischemia/hypoxia conditions as previously described. These findings suggest that VEGFR-2 may play an adaptive role in angiogenesis after CH-induced hypoxia. Modulation of vascular endothelial growth factor/VEGFR-2(+) may be important in developing treatments for hypoxic conditions including hydrocephalus and other forms of cerebral ischemia.
Collapse
Affiliation(s)
- S M Dombrowski
- Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Zhang MC, Wang Y, Yang Y. The expression of nuclear factor kappa B in inflammation-induced rat corneal neovascularization. Ocul Immunol Inflamm 2007; 14:359-65. [PMID: 17162607 DOI: 10.1080/09273940601001322] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To investigate the involvement of the nuclear-transfactor-kappa B (NF-kappa B) in the rat model of inflammation-induced corneal neovascularization (CNV). METHODS The CNV model in Sprague-Dawley rats was induced by alkaline cauterization of the central cornea. The corneas were examined by a slit lamp microscope. NF-kappa B was assayed by Western blot. Vascular endothelial growth factor (VEGF) protein was evaluated by immunohistochemistry. VEGF mRNA levels were determined by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS Morphologically, the CNV was shown on the second day after cautery. In corneas after cautery, NF-kappa B protein increased 6 hours after cautery, peaked 4 days after cautery, and decreased to near baseline by day 14. VEGF protein and mRNA increased gradually in the early stage after cautery, reached the highest level on the fourth day, and then decreased to near baseline slowly after 7 days. CONCLUSIONS The activated NF-kappa B was up-regulated in the early stage of CNV of rat induced by cauterization, which suggests it may participate in the pathogenesis of wound healing, inflammation, and neovascularization processes in the cornea.
Collapse
Affiliation(s)
- Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | |
Collapse
|
26
|
Schmidt-Kastner R, Haberkamp M, Schmitz C, Hankeln T, Burmester T. Neuroglobin mRNA expression after transient global brain ischemia and prolonged hypoxia in cell culture. Brain Res 2006; 1103:173-80. [PMID: 16796995 DOI: 10.1016/j.brainres.2006.05.047] [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] [Received: 02/20/2006] [Revised: 05/09/2006] [Accepted: 05/16/2006] [Indexed: 11/18/2022]
Abstract
Neuroglobin is a nerve-specific respiratory protein that has been proposed to play an important role in the protection of brain neurons from ischemic and hypoxic injuries. Here, we investigated the regulation of neuroglobin expression after transient global ischemia in the rat brain using mRNA in situ hybridization and under hypoxic stress in cultured neuronal cell lines (PC12, HN33) by quantitative RT-PCR. While neuroglobin mRNA expression was significantly enhanced in cell culture after severe prolonged hypoxia (0-1% O2 for 24 h), we did not find any significant increases in neuroglobin mRNA levels in the rat brain after transient global ischemia. Vegf and Glut1 mRNAs showed increases in the hippocampus as expected. Therefore, it is unlikely that neuroglobin is instrumental in the acute response of neurons to hypoxic or ischemic insults, for which the mammalian brain is not adapted.
Collapse
|
27
|
Xu K, Puchowicz MA, Lust WD, LaManna JC. Adenosine treatment delays postischemic hippocampal CA1 loss after cardiac arrest and resuscitation in rats. Brain Res 2006; 1071:208-17. [PMID: 16412392 DOI: 10.1016/j.brainres.2005.11.060] [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] [Received: 08/04/2005] [Revised: 11/09/2005] [Accepted: 11/11/2005] [Indexed: 10/25/2022]
Abstract
Resuscitation from cardiac arrest results in reperfusion injury that leads to increased postresuscitation mortality and delayed neuronal death. One of the many consequences of resuscitation from cardiac arrest is a derangement of energy metabolism and the loss of adenylates, impairing the tissue's ability to regain proper energy balance. In this study, we investigated the effects of adenosine (ADO) on the recovery of the brain from 12 min of ischemia using a rat model of cardiac arrest and resuscitation. Compared to the untreated group, treatment with adenosine (7.2 mg/kg) initiated immediately after resuscitation increased the proportion of rats surviving to 4 days and significantly delayed hippocampal CA1 neuronal loss. Brain blood flow was increased significantly in the adenosine-treated rats 1 h after cardiac arrest and resuscitation. Adenosine-treated rats exhibited less edema in cortex, brainstem and hippocampus during the first 48 h of recovery. Adenosine treatment significantly lowered brain temperature during recovery, and a part of the neuroprotective effects of adenosine treatment could be ascribed to adenosine-induced hypothermia. With this dose, adenosine may have a delayed transient effect on the restoration of the adenylate pool (AXP = ATP + ADP + AMP) 24 h after cardiac arrest and resuscitation. Our findings suggested that improved postischemic brain blood flow and ADO-induced hypothermia, rather than adenylate supplementation, may be the two major contributors to the neuroprotective effects of adenosine following cardiac arrest and resuscitation. Although adenosine did not prevent eventual CA1 neuronal loss in the long term, it did delay neuronal loss and promoted long-term survival. Thus, adenosine or specific agonists of adenosine receptors should be evaluated as adjuncts to broaden the window of opportunity in the treatment of the reperfusion injury following cardiac arrest and resuscitation.
Collapse
Affiliation(s)
- Kui Xu
- Department of Anatomy, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
28
|
Chodobski A, Chung I, Koźniewska E, Ivanenko T, Chang W, Harrington JF, Duncan JA, Szmydynger-Chodobska J. Early neutrophilic expression of vascular endothelial growth factor after traumatic brain injury. Neuroscience 2004; 122:853-67. [PMID: 14643756 DOI: 10.1016/j.neuroscience.2003.08.055] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The formation of edema after traumatic brain injury (TBI) is in part associated with the disruption of the blood-brain barrier. However, the molecular and cellular mechanisms underlying these phenomena have not been fully understood. One possible factor involved in edema formation is vascular endothelial growth factor (VEGF). This growth factor has previously been demonstrated to increase the blood-brain barrier permeability to the low molecular weight markers and macromolecules. In this study, we analyzed the temporal changes in VEGF expression after TBI in rats. In the intact brain, VEGF was expressed at relatively low levels and was found in the cells located close to the cerebrospinal fluid space. These were the astrocytes located under the ependyma and the pia-glial lining, as well as the epithelial cells of the choroid plexus. In addition, several groups of neurons, including those located in the frontoparietal cortex and in all hippocampal regions, were VEGF-positive. The pattern of VEGF-immunopositive staining of neurons and choroidal epithelium suggested that in these cells, VEGF binds to the cell membrane-associated heparan sulfate proteoglycans. Following TBI, there was an early (within 4 h post-injury) increase in VEGF expression in the traumatized parenchyma associated with neutrophilic invasion. The ipsilateral choroid plexus appeared to play a role in facilitating the migration of neutrophils from blood into the cerebrospinal fluid space, from where many of these cells infiltrated the brain parenchyma. VEGF-immunopositive staining of neutrophils resembled haloes and was found ipsilaterally within the frontoparietal cortex and around the velum interpositum, a part of the subarachnoid space. These haloes likely represent the deposition of neutrophil-derived VEGF within the extracellular matrix, from where this growth factor may be gradually released during an early post-traumatic period. The maximum number of VEGF-secreting neutrophils was observed between 8 h and 1 day after TBI. In addition, from 4 h post-TBI, there was a progressive increase in the number of VEGF-immunoreactive astrocytes in the ipsilateral frontoparietal cortex. The maximum number of astrocytes expressing VEGF was observed 4 days after TBI, and then the levels of astroglial VEGF expression declined gradually. Early invasion of brain parenchyma by VEGF-secreting neutrophils together with a delayed increase in astrocytic synthesis of this growth factor correlate with the biphasic opening of the blood-brain barrier and formation of edema previously observed after TBI. Therefore, these findings suggest that VEGF plays an important role in promoting the formation of post-traumatic brain edema.
Collapse
Affiliation(s)
- A Chodobski
- Department of Clinical Neurosciences, Brown University School of Medicine, Aldrich Building, Room 405, 593 Eddy Street, Providence, RI 02903, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Croll SD, Goodman JH, Scharfman HE. Vascular endothelial growth factor (VEGF) in seizures: a double-edged sword. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 548:57-68. [PMID: 15250585 PMCID: PMC2504497 DOI: 10.1007/978-1-4757-6376-8_4] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a vascular growth factor which induces angiogenesis (the development of new blood vessels), vascular permeability, and inflammation. In brain, receptors for VEGF have been localized to vascular endothelium, neurons, and glia. VEGF is upregulated after hypoxic injury to the brain, which can occur during cerebral ischemia or high-altitude edema, and has been implicated in the blood-brain barrier breakdown associated with these conditions. Given its recently-described role as an inflammatory mediator, VEGF could also contribute to the inflammatory responses observed in cerebral ischemia. After seizures, blood-brain barrier breakdown and inflammation is also observed in brain, albeit on a lower scale than that observed after stroke. Recent evidence has suggested a role for inflammation in seizure disorders. We have described striking increases in VEGF protein in both neurons and glia after pilocarpine-induced status epilepticus in the brain. Increases in VEGF could contribute to the blood-brain barrier breakdown and inflammation observed after seizures. However, VEGF has also been shown to be neuroprotective across several experimental paradigms, and hence could potentially protect vulnerable cells from damage associated with seizures. Therefore, the role of VEGF after seizures could be either protective or destructive. Although only further research will determine the exact nature of VEGF's role after seizures, preliminary data indicate that VEGF plays a protective role after seizures.
Collapse
Affiliation(s)
- Susan D Croll
- Department of Psychology, Queens College and the Graduate Center of the City University of New York, Flushing, USA
| | | | | |
Collapse
|
30
|
Hai J, Li ST, Lin Q, Pan QG, Gao F, Ding MX. Vascular Endothelial Growth Factor Expression and Angiogenesis Induced by Chronic Cerebral Hypoperfusion in Rat Brain. Neurosurgery 2003; 53:963-70; discussion 970-2. [PMID: 14519228 DOI: 10.1227/01.neu.0000083594.10117.7a] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2003] [Accepted: 06/04/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In a rat model, we studied the time courses of vascular endothelial growth factor (VEGF) expression and angiogenesis induced by chronic cerebral hypoperfusion in the brain, and we investigated the histological basis of normal-perfusion pressure breakthrough. METHODS Twenty-one Sprague-Dawley rats were randomly divided into a control group (n = 3) and a model group assessed at various time points after the creation of a carotid artery-jugular vein fistula (12 h, n = 3; 24 h, n = 3; 72 h, n = 3; 7 d, n = 3; 21 d, n = 3; 90 d, n = 3). The time courses of the expression of VEGF messenger ribonucleic acid (mRNA) and protein in rat brain were analyzed with semiquantitative reverse transcriptase-polymerase chain reaction and Western blot assays, respectively. Immunohistochemical techniques were used to evaluate VEGF protein localization with rabbit polyclonal anti-rat VEGF, VEGF receptor (VEGFR) expression with rabbit polyclonal antibodies to VEGFR-1 and -2, microvascular density with mouse monoclonal anti-rat CD31, and astrocytic reactivity with polyclonal anti-glial fibrillary acidic protein, in cerebral cortical tissue of the right middle cerebral artery territory. RESULTS Three alternative splicing forms, i.e., VEGF(188), VEGF(164), and VEGF(120), were observed in cerebral cortical tissue of the right middle cerebral artery territory in semiquantitative reverse transcriptase-polymerase chain reaction analyses. VEGF(164) mRNA was the predominant isoform expressed in rat brain. VEGF(188) mRNA and VEGF(120) mRNA were also detected but at very low levels (not statistically significant). Low levels of VEGF(164) mRNA were observed in the control brains. However, VEGF(164) mRNA levels were significantly increased in the model brains at 24 hours postoperatively, peaked by 7 days, decreased by 21 days, and returned to basal levels by 90 days after fistula formation. VEGF protein expression, as measured in Western blot assays, was also increased in rat brains in the model group from 24 hours to 21 days postoperatively but returned to control levels by 90 days after fistula formation. VEGF immunohistochemical analyses indicated that this increased expression was mostly associated with endothelial cells. Consistent with the VEGF protein expression findings, up-regulation of VEGFR-1 but not VEGFR-2 expression on endothelial cells in the model brains was observed. Microvascular density in the rat brains began to increase significantly 7 days after fistula formation in the model group, as assessed immunohistochemically, and the increase was maintained for 90 days. Although no prominent astrocytic reactivity was observed in the rat brains throughout the experiments, there was an absence of astrocytic foot processes surrounding some cerebral capillaries 90 days after fistula formation in the model group. CONCLUSION These results demonstrated that chronic cerebral hypoperfusion could induce sustained up-regulation of VEGF mRNA and protein expression in rat brain, which was correlated with angiogenesis. An absence of corresponding astrocytic reactivity during angiogenesis may be an important factor accounting for structural deficits of the blood-brain barrier and the occurrence of normal-perfusion pressure breakthrough.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
31
|
Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest 2003. [PMID: 12813020 DOI: 10.1172/jci200317977] [Citation(s) in RCA: 823] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic protein with therapeutic potential in ischemic disorders, including stroke. VEGF confers neuroprotection and promotes neurogenesis and cerebral angiogenesis, but the manner in which these effects may interact in the ischemic brain is poorly understood. We produced focal cerebral ischemia by middle cerebral artery occlusion for 90 minutes in the adult rat brain and measured infarct size, neurological function, BrdU labeling of neuroproliferative zones, and vWF-immunoreactive vascular profiles, without and with intracerebroventricular administration of VEGF on days 1-3 of reperfusion. VEGF reduced infarct size, improved neurological performance, enhanced the delayed survival of newborn neurons in the dentate gyrus and subventricular zone, and stimulated angiogenesis in the striatal ischemic penumbra, but not the dentate gyrus. We conclude that in the ischemic brain VEGF exerts an acute neuroprotective effect, as well as longer latency effects on survival of new neurons and on angiogenesis, and that these effects appear to operate independently. VEGF may, therefore, improve histological and functional outcome from stroke through multiple mechanisms.
Collapse
Affiliation(s)
- Yunjuan Sun
- Buck Institute for Age Research, Novato, California 94945, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest 2003; 111:1843-51. [PMID: 12813020 PMCID: PMC161428 DOI: 10.1172/jci17977] [Citation(s) in RCA: 405] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic protein with therapeutic potential in ischemic disorders, including stroke. VEGF confers neuroprotection and promotes neurogenesis and cerebral angiogenesis, but the manner in which these effects may interact in the ischemic brain is poorly understood. We produced focal cerebral ischemia by middle cerebral artery occlusion for 90 minutes in the adult rat brain and measured infarct size, neurological function, BrdU labeling of neuroproliferative zones, and vWF-immunoreactive vascular profiles, without and with intracerebroventricular administration of VEGF on days 1-3 of reperfusion. VEGF reduced infarct size, improved neurological performance, enhanced the delayed survival of newborn neurons in the dentate gyrus and subventricular zone, and stimulated angiogenesis in the striatal ischemic penumbra, but not the dentate gyrus. We conclude that in the ischemic brain VEGF exerts an acute neuroprotective effect, as well as longer latency effects on survival of new neurons and on angiogenesis, and that these effects appear to operate independently. VEGF may, therefore, improve histological and functional outcome from stroke through multiple mechanisms.
Collapse
Affiliation(s)
- Yunjuan Sun
- Buck Institute for Age Research, Novato, California 94945, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Krum JM, Khaibullina A. Inhibition of endogenous VEGF impedes revascularization and astroglial proliferation: roles for VEGF in brain repair. Exp Neurol 2003; 181:241-57. [PMID: 12781997 DOI: 10.1016/s0014-4886(03)00039-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Vascular endothelial growth factor (VEGF) is upregulated following injury to the CNS. Our previous work has shown that exogenous application of VEGF promotes angiogenesis, blood-brain barrier permeability, and astroglial mitogenicity in the traumatized brain. To develop a model that could link endogenously secreted VEGF to brain tissue repair, a specific neutralizing antibody to VEGF was infused by osmotic minipump directly into the neocortex and striatum for up to 1 week. Tissues adjacent to the infusion/wound site were analyzed for specific vascular and astroglial protein markers and proliferation, necrosis/apoptosis (via TUNEL staining), VEGF, the VEGF receptors flt-1 and flk-1, and bFGF expression using immunohistochemistry and semi-quantitative RT-PCR. Neutralization of native VEGF caused significant decreases in angiogenic activity, astroglial proliferation, and nestin immunoexpression, while vascular and astroglial degeneration was substantially increased, resulting in much larger wound cavities when compared to controls. The hindrance of brain tissue repair occurred despite an increase in bFGF expression at the wound sites. VEGF appears to be an integral factor in CNS wound healing that is essential for vascular endothelial proliferation and survival and may also be necessary for astroglial proliferation and maintenance during the repair of brain injury.
Collapse
Affiliation(s)
- Janette M Krum
- Department of Anatomy and Cell Biology, George Washington University Medical Center, Washington, DC 20037, USA.
| | | |
Collapse
|
34
|
Payen JF, Fauvage B, Falcon D, Lavagne P. [Brain oedema following blood-brain barrier disruption: mechanisms and diagnosis]. ANNALES FRANCAISES D'ANESTHESIE ET DE REANIMATION 2003; 22:220-5. [PMID: 12747990 DOI: 10.1016/s0750-7658(03)00010-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Brain oedema following blood-brain barrier (BBB) disruption, or vasogenic oedema, is present in most cases of brain oedema. According to the Starling's law, water, ions and plasma proteins cross the BBB toward the interstitium if the driving forces for transmural bulk flow are excessive (mechanical origin) and/or if the BBB permeability is enhanced (chemical origin). Both mechanisms coexist in most cases. Excessive elevation of the gradient of hydrostatic pressure with lost of cerebral autoregulation has been proved in ischaemia/reperfusion and trauma, and suggested in acute mountain sickness and eclampsia. The BBB permeability can be enhanced by immediate (chemical mediators) or delayed (cellular infiltration) inflammatory response, or by alteration of the membrane integrity. This later can be transient (hyperosmolar BBB disruption), or permanent by activation of matrix metalloproteinase or by neovascularization with BBB breakdown. The reference method for the diagnosis of vasogenic oedema is the MRI diffusion-weighted imaging.
Collapse
Affiliation(s)
- J F Payen
- Département d'anesthésie-réanimation, hôpital Michallon, BP 217, 38043 Grenoble, France.
| | | | | | | |
Collapse
|
35
|
Kimura R, Nakase H, Sakaki T, Taoka T, Tsuji T. Vasogenic edema and VEGF expression in a rat two-vein occlusion model. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 86:213-7. [PMID: 14753438 DOI: 10.1007/978-3-7091-0651-8_46] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Vasogenic edema plays an important etiologic role in the pathogenesis of cerebral venous circulation disturbances (CVCDs). Since vascular endothelial growth factor (VEGF) is a major mediator in angiogenesis and vascular permeability, including induction of vasogenic edema, the present study was undertaken to investigate whether it has any relevance to CVCDs. Male Wistar rats (n = 15) were used. Two adjacent cortical veins were occluded photochemically using rose bengal dye and fiberoptic illumination, with evaluation 24 hours thereafter by magnetic resonance imaging (MRI). Each brain was removed from the skull immediately after MRI and processed for hematoxylin-eosin staining (H&E staining) of sections for histopathology and comparison with MRI data. VEGF expression as demonstrated immunohistochemically appeared to coincide with vasogenic edema, diagnosed as high intensity areas on apparent diffusion coefficient of water (ADCw) maps. On the basis of these data, we conclude that VEGF is related to formation of vasogenic edema in the acute stage of CVCD.
Collapse
Affiliation(s)
- R Kimura
- Department of Neurosurgery, Nara Medical University, Nara, Japan.
| | | | | | | | | |
Collapse
|
36
|
Activation of hypoxia-inducible factor-1 in the rat cerebral cortex after transient global ischemia: potential role of insulin-like growth factor-1. J Neurosci 2002. [PMID: 12388599 DOI: 10.1523/jneurosci.22-20-08922.2002] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that regulates the adaptive response to hypoxia in mammalian cells. It consists of a regulatory subunit HIF-1alpha, which accumulates under hypoxic conditions, and a constitutively expressed subunit HIF-1beta. In this study we analyzed HIF-1alpha expression in the rat cerebral cortex after transient global ischemia induced by cardiac arrest and resuscitation. Our results showed that HIF-1alpha accumulates as early as 1 hr of recovery and persists for at least 7 d. In addition, the expression of HIF-1 target genes, erythropoietin and Glut-1, were induced at 12 hr to 7d of recovery. A logical explanation for HIF-1alpha accumulation might be that the brain remained hypoxic for prolonged periods after resuscitation. By using the hypoxic marker 2-(2-nitroimidazole-1[H]-y1)-N-(2,2,3,3,3-pentafluoropropyl)-acetamide (EF5), we showed that the brain is hypoxic during the first hours of recovery from cardiac arrest, but the tissue is no longer hypoxic at 2 d. Thus, the initial ischemic episode must have activated other nonhypoxic mechanisms that maintain prolonged HIF-1alpha accumulation. One such mechanism might be initiated by insulin-like growth factor-1 (IGF-1). Our results showed that IGF-1 expression was upregulated after cardiac arrest and resuscitation. In addition, we showed that IGF-1 was able to induce HIF-1alpha in pheochromocytoma cells and cultured neurons as well as in the brain of rats that received intracerebroventricular and systemic IGF-1 infusion. Moreover, infusion of a selective IGF-1 receptor antagonist abrogates HIF-1alpha accumulation after cardiac arrest and resuscitation. Our study suggest that activation of HIF-1 might be part of the mechanism by which IGF-1 promotes cell survival after cerebral ischemia.
Collapse
|
37
|
Nag S, Eskandarian MR, Davis J, Eubanks JH. Differential expression of vascular endothelial growth factor-A (VEGF-A) and VEGF-B after brain injury. J Neuropathol Exp Neurol 2002; 61:778-88. [PMID: 12230324 DOI: 10.1093/jnen/61.9.778] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our previous study demonstrated that vascular endothelial growth factor (VEGF), now referred to as VEGF-A, plays a significant role in blood-brain barrier (BBB) breakdown and angiogenesis after brain injury. In this study, VEGF-A expression was compared with that of VEGF-B in the rat cortical cold injury model over a period of 6 hours to 6 days post-injury. VEGF-A and VEGF-B mRNA were detected by in situ hybridization and their protein was detected by immunohistochemistry. The presence of VEGF-A and VEGF-B proteins in endothelium of lesion vessels was related to BBB breakdown by double labeling for either of these growth factors and fibronectin, which was used as a marker of BBB breakdown. Significant induction of both VEGF-A and VEGF-B mRNA occurred at the lesion site during the period of maximal endothelial proliferation. VEGF-A mRNA levels peaked at 3 and 4 days post-injury and returned to basal expression by day 6, while VEGF-B mRNA levels remained elevated up to day 6. VEGF-B protein was constitutively expressed in endothelium of all cerebral vessels. After brain injury, there was increased immunoreactivity for VEGF-B at the lesion site, this protein being present in the endothelium and vascular smooth muscle cells of pial vessels, in inflammatory cells, and later in proliferating endothelial cells, endothelium of neovessels, and astrocytes. Lesion vessels showing BBB breakdown to fibronectin showed endothelial VEGF-A protein but not VEGF-B protein. Constitutive expression of VEGF-B in normal endothelium suggests that it may have a role in maintenance of the BBB in steady states, while its induction at both the gene and protein level post-injury indicates that it has an essential role in angiogenesis and the repair processes after brain injury.
Collapse
Affiliation(s)
- Sukriti Nag
- Toronto Western Research Institute at University Health Network, Department of Laboratory Medicine, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
38
|
Krum JM, Mani N, Rosenstein JM. Angiogenic and astroglial responses to vascular endothelial growth factor administration in adult rat brain. Neuroscience 2002; 110:589-604. [PMID: 11934468 DOI: 10.1016/s0306-4522(01)00615-7] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effects of exogenous vascular endothelial growth factor (VEGF) on angiogenesis, blood-brain barrier permeability and astroglial proliferation in the adult rat CNS in situ were investigated. Recombinant human VEGF(165) (25 or 50 ng/ml) was delivered for up to 1 week using either intracerebral osmotic minipumps or less traumatic subdural gelatin sponge placement. By 3 days, VEGF delivery caused significantly increased cerebral angiogenesis (25 ng/ml was most effective) in both experimental models when compared to saline controls; VEGF infusion resulted in a 100% increase in an index of vascular proliferation, and gelatin sponge delivery produced a 65% increase. The blood-brain barrier hallmark endothelial glucose transporter-1 was not present in nascent vascular sprouts. Infusion of VEGF produced extensive protein leakage that persisted after saline-induced permeability was mostly resolved, while gelatin sponge administration caused milder barrier dysfunction. Administration of the angiogenic factor had unexpected proliferative effects on astroglia in both models, resulting in an 80-85% increase in mitotically active astroglia when compared to controls. Immunohistochemical results and semi-quantitative reverse transcriptase-polymerase chain reaction indicated that the VEGF receptors flk-1 and flt-1 were up-regulated in response to the infusion trauma; flt-1 was localized to reactive astroglia, while flk-1 was expressed in vascular endothelium but predominantly in neuronal somata and processes adjacent to the delivery site. mRNA for the VEGF(121), VEGF(165) and VEGF(188) isoforms was also increased after delivery of the recombinant protein. These data show that VEGF application has substantial proliferative effects on CNS endothelium and astroglia and causes up-regulation of its own message. Flt-1 and flk-1 receptor mRNAs and proteins are up-regulated in both vascular and non-vascular cell types following infusion trauma. From these results we suggest that administered VEGF has heretofore unanticipated pleiotrophic effects in the adult CNS.
Collapse
Affiliation(s)
- J M Krum
- Department of Anatomy and Cell Biology, The George Washington University Medical Center, Washington, DC 20037, USA.
| | | | | |
Collapse
|