1
|
Faber JE. Genetic determinants of insufficiency of the collateral circulation. J Cereb Blood Flow Metab 2025:271678X251317880. [PMID: 39901795 DOI: 10.1177/0271678x251317880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
It has been estimated that approximately two million neurons, sixteen billion synapses and twelve kilometers of axons are lost each minute following anterior large-vessel stroke. The level of collateral blood flow has become recognized as a primary determinant of the pace of this loss and an important factor in clinical decision-making. Many of the topics in this review cover recent developments that have not been reviewed elsewhere. These include that: the number and diameter of collaterals and collateral blood flow vary greatly in the brain and other tissues of healthy individuals; a large percentage of individuals are deficient in collaterals; the underlying mechanism arises primarily from naturally occurring polymorphisms in genes/genetic loci within the pathway that drives collateral formation during development; evidence indicates collateral abundance does not exhibit sexual dimorphism; and that collaterals-besides their function as endogenous bypass vessels-may have a physiological role in optimizing oxygen delivery. Animal and human studies in brain and other tissues, where available, are reviewed. Details of many of the studies are provided so that the strength of the findings and conclusions can be assessed without consulting the original literature. Key questions that remain unanswered and strategies to address them are also discussed.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, Curriculum in Neuroscience, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Nilsson I, Su EJ, Fredriksson L, Sahlgren BH, Bagoly Z, Moessinger C, Stefanitsch C, Ning FC, Zeitelhofer M, Muhl L, Lawrence ALE, Scotney PD, Lu L, Samén E, Ho H, Keep RF, Medcalf RL, Lawrence DA, Eriksson U. Thrombolysis exacerbates cerebrovascular injury after ischemic stroke via a VEGF-B dependent effect on adipose lipolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617532. [PMID: 39416206 PMCID: PMC11483068 DOI: 10.1101/2024.10.11.617532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cerebrovascular injuries leading to edema and hemorrhage after ischemic stroke are common. The mechanisms underlying these events and how they are connected to known risk factors for poor outcome, like obesity and diabetes, is relatively unknown. Herein we demonstrate that increased adipose tissue lipolysis is a dominating risk factor for the development of a compromised cerebrovasculature in ischemic stroke. Reducing adipose lipolysis by VEGF-B antagonism improved vascular integrity by reducing ectopic cerebrovascular lipid deposition. Thrombolytic therapy in ischemic stroke using tissue plasminogen activator (tPA) leads to increased risk of hemorrhagic complications, substantially limiting the use of thrombolytic therapy. We provide evidence that thrombolysis with tPA promotes adipose tissue lipolysis, leading to a rise in plasma fatty acids and lipid accumulation in the ischemic cerebrovasculature after stroke. VEGF-B blockade improved the efficacy and safety of thrombolysis suggesting the potential use of anti-VEGF-B therapy to extend the therapeutic window for stroke management.
Collapse
Affiliation(s)
- Ingrid Nilsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
- These authors contributed equally
- Lead contact: (I.N.)
| | - Enming J. Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- These authors contributed equally
| | - Linda Fredriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Heller Sahlgren
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Zsuzsa Bagoly
- MTA-DE Lendület “Momentum” Hemostasis and Stroke Research Group, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences, Faculty of Medicine, University of Debrecen, Hungary
| | - Christine Moessinger
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Stefanitsch
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Frank Chenfei Ning
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Zeitelhofer
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lars Muhl
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anna-Lisa E. Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Li Lu
- Karolinska Experimental Research and Imaging Centre, Karolinska University Hospital, Stockholm, Sweden
| | - Erik Samén
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Heidi Ho
- Australian Centre for Blood Diseases, Monash University, Melbourne 3004, Victoria, Australia
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Robert L. Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne 3004, Victoria, Australia
| | - Daniel A. Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
González-Rojas A, Valencia-Narbona M. Neurodevelopmental Disruptions in Children of Preeclamptic Mothers: Pathophysiological Mechanisms and Consequences. Int J Mol Sci 2024; 25:3632. [PMID: 38612445 PMCID: PMC11012011 DOI: 10.3390/ijms25073632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Preeclampsia (PE) is a multisystem disorder characterized by elevated blood pressure in the mother, typically occurring after 20 weeks of gestation and posing risks to both maternal and fetal health. PE causes placental changes that can affect the fetus, particularly neurodevelopment. Its key pathophysiological mechanisms encompass hypoxia, vascular and angiogenic dysregulation, inflammation, neuronal and glial alterations, and disruptions in neuronal signaling. Animal models indicate that PE is correlated with neurodevelopmental alterations and cognitive dysfunctions in offspring and in humans, an association between PE and conditions such as cerebral palsy, autism spectrum disorder, attention deficit hyperactivity disorder, and sexual dimorphism has been observed. Considering the relevance for mothers and children, we conducted a narrative literature review to describe the relationships between the pathophysiological mechanisms behind neurodevelopmental alterations in the offspring of PE mothers, along with their potential consequences. Furthermore, we emphasize aspects pertinent to the prevention/treatment of PE in pregnant mothers and alterations observed in their offspring. The present narrative review offers a current, complete, and exhaustive analysis of (i) the pathophysiological mechanisms that can affect neurodevelopment in the children of PE mothers, (ii) the relationship between PE and neurological alterations in offspring, and (iii) the prevention/treatment of PE.
Collapse
Affiliation(s)
- Andrea González-Rojas
- Laboratorio de Neurociencias Aplicadas, Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Brasil 2950, Valparaíso 2340025, Chile;
| | | |
Collapse
|
4
|
Wu LY, Chong JR, Chong JPC, Hilal S, Venketasubramanian N, Tan BY, Richards AM, Chen CP, Lai MKP. Serum Placental Growth Factor as a Marker of Cerebrovascular Disease Burden in Alzheimer's Disease. J Alzheimers Dis 2024; 97:1289-1298. [PMID: 38217598 DOI: 10.3233/jad-230811] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
BACKGROUND Concomitant cerebrovascular diseases (CeVD) have been identified as an important determinant of Alzheimer's disease (AD) progression. Development of robust blood-based biomarkers will provide critical tools to evaluate prognosis and potential interventional strategies for AD with CeVD. OBJECTIVE This study investigated circulating placental growth factor (PlGF), a potent pro-angiogenic factor related to endothelial dysfunction and vascular inflammation, in an Asian memory clinic cohort of non-demented individuals as well as AD, including its associations with neuroimaging markers of CeVD. METHODS 109 patients with AD, 76 cognitively impaired with no dementia (CIND), and 56 non-cognitively impaired (NCI) were included in this cross-sectional study. All subjects underwent 3T brain magnetic resonance imaging to assess white matter hyperintensities (WMH), lacunes, cortical infarcts, and cerebral microbleeds (CMBs). Serum PlGF concentrations were measured by electrochemiluminescence immunoassays. RESULTS Serum PlGF was elevated in AD, but not CIND, compared to the NCI controls. Adjusted concentrations of PlGF were associated with AD only in the presence of significant CeVD. Elevated PlGF was significantly associated with higher burden of WMH and with CMBs in AD patients. CONCLUSIONS Serum PlGF has potential utility as a biomarker for the presence of CeVD, specifically WMH and CMBs, in AD. Further studies are needed to elucidate the underlying pathophysiological mechanisms linking PlGF to CeVD, as well as to further assess PlGF's clinical utility.
Collapse
Affiliation(s)
- Liu-Yun Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Jenny P C Chong
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | | | | | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Heart Centre, Singapore
- Department of Medicine, National University Health System, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| |
Collapse
|
5
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 404] [Impact Index Per Article: 202.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
6
|
Roefs MT, Heusermann W, Brans MAD, Snijders Blok C, Lei Z, Vader P, Sluijter JPG. Evaluation and manipulation of tissue and cellular distribution of cardiac progenitor cell-derived extracellular vesicles. Front Pharmacol 2022; 13:1052091. [PMID: 36506565 PMCID: PMC9729535 DOI: 10.3389/fphar.2022.1052091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiac progenitor cell-derived extracellular vesicles (CPC-EVs) have been successfully applied via different delivery routes for treating post-myocardial infarction injury in several preclinical models. Hence, understanding the in vivo fate of CPC-EVs after systemic or local, i.e. myocardial, delivery is of utmost importance for the further therapeutic application of CPC-EVs in cardiac repair. Here, we studied the tissue- and cell distribution and retention of CPC-EVs after intramyocardial and intravenous injection in mice by employing different EV labeling and imaging techniques. In contrast to progenitor cells, CPC-EVs demonstrated no immediate flush-out from the heart upon intramyocardial injection and displayed limited distribution to other organs over time, as determined by near-infrared imaging in living animals. By employing CUBIC tissue clearing and light-sheet fluorescent microscopy, we observed CPC-EV migration in the interstitial space of the myocardium shortly after EV injection. Moreover, we demonstrated co-localization with cTnI and CD31-positive cells, suggesting their interaction with various cell types present in the heart. On the contrary, after intravenous injection, most EVs accumulated in the liver. To potentiate such a potential systemic cardiac delivery route, targeting the cardiac endothelium could provide openings for directed CPC-EV therapy. We therefore evaluated whether decorating EVs with targeting peptides (TPs) RGD-4C or CRPPR connected to Lamp2b could enhance EV delivery to endothelial cells. Expression of both TPs enhanced CPC-EV uptake under in vitro continuous flow, but did not affect uptake under static cell culture conditions. Together, these data demonstrate that the route of administration influences CPC-EV biodistribution pattern and suggest that specific TPs could be used to target CPC-EVs to the cardiac endothelium. These insights might lead to a better application of CPC-EV therapeutics in the heart.
Collapse
Affiliation(s)
- Marieke T. Roefs
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Maike A. D. Brans
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christian Snijders Blok
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Zhiyong Lei
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pieter Vader
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands,CDL Research, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands,*Correspondence: Joost P. G. Sluijter,
| |
Collapse
|
7
|
Du H, Xu Y, Zhu L. Role of Semaphorins in Ischemic Stroke. Front Mol Neurosci 2022; 15:848506. [PMID: 35350431 PMCID: PMC8957939 DOI: 10.3389/fnmol.2022.848506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is one of the major causes of neurological morbidity and mortality in the world. Although the management of ischemic stroke has been improved significantly, it still imposes a huge burden on the health and property. The integrity of the neurovascular unit (NVU) is closely related with the prognosis of ischemic stroke. Growing evidence has shown that semaphorins, a family of axon guidance cues, play a pivotal role in multiple pathophysiological processes in NVU after ischemia, such as regulating the immune system, angiogenesis, and neuroprotection. Modulating the NVU function via semaphorin signaling has a potential to develop a novel therapeutic strategy for ischemic stroke. We, therefore, review recent progresses on the role of semphorin family members in neurons, glial cells and vasculature after ischemic stroke.
Collapse
Affiliation(s)
- Huaping Du
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Yuan Xu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Li Zhu
- Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
- Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Collaborative Innovation Center of Hematology of Jiangsu Province, National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
- *Correspondence: Li Zhu,
| |
Collapse
|
8
|
Linton AE, Weekman EM, Wilcock DM. Pathologic sequelae of vascular cognitive impairment and dementia sheds light on potential targets for intervention. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100030. [PMID: 36324710 PMCID: PMC9616287 DOI: 10.1016/j.cccb.2021.100030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/11/2021] [Accepted: 10/08/2021] [Indexed: 11/30/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is one of the leading causes of dementia along with Alzheimer's disease (AD) and, importantly, VCID often manifests as a comorbidity of AD(Vemuri and Knopman 2016; Schneider and Bennett 2010)(Vemuri and Knopman 2016; Schneider and Bennett 2010). Despite its common clinical manifestation, the mechanisms underlying VCID disease progression remains elusive. In this review, existing knowledge is used to propose a novel hypothesis linking well-established risk factors of VCID with the distinct neurodegenerative cascades of neuroinflammation and chronic hypoperfusion. It is hypothesized that these two synergistic signaling cascades coalesce to initiate aberrant angiogenesis and induce blood brain barrier breakdown trough a mechanism mediated by vascular growth factors and matrix metalloproteinases respectively. Finally, this review concludes by highlighting several potential therapeutic interventions along this neurodegenerative sequalae providing diverse opportunities for future translational study.
Collapse
Affiliation(s)
- Alexandria E. Linton
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| | - Erica M. Weekman
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| | - Donna M. Wilcock
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| |
Collapse
|
9
|
Hung TH, Liu YC, Wu CH, Chen CC, Chao H, Yang FY, Chen SF. Antenatal low-intensity pulsed ultrasound reduces neurobehavioral deficits and brain injury following dexamethasone-induced intrauterine growth restriction. BRAIN PATHOLOGY (ZURICH, SWITZERLAND) 2021; 31:e12968. [PMID: 33960564 PMCID: PMC8549022 DOI: 10.1111/bpa.12968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/01/2022]
Abstract
Intrauterine growth restriction (IUGR) is a leading cause of perinatal mortality and morbidity, and IUGR survivors are at increased risk of neurodevelopmental deficits. No effective interventions are currently available to improve the structure and function of the IUGR brain before birth. This study investigated the protective effects of low‐intensity pulsed ultrasound (LIPUS) on postnatal neurodevelopmental outcomes and brain injury using a rat model of IUGR induced by maternal exposure to dexamethasone (DEX). Pregnant rats were treated with DEX (200 μg/kg, s.c.) and LIPUS daily from gestational day (GD) 14 to 19. Behavioral assessments were performed on the IUGR offspring to examine neurological function. Neuropathology, levels of neurotrophic factors, and CaMKII‐Akt‐related molecules were assessed in the IUGR brain, and expression of glucose and amino acid transporters and neurotrophic factors were examined in the placenta. Maternal LIPUS treatment increased fetal weight, fetal liver weight, and placental weight following IUGR. LIPUS treatment also increased neuronal number and myelin protein expression in the IUGR brain, and attenuated neurodevelopmental deficits at postnatal day (PND) 18. However, the number of oligodendrocytes or microglia was not affected. These changes were associated with the upregulation of brain‐derived neurotrophic factor (BDNF) and placental growth factor (PlGF) protein expression, and enhancement of neuronal CaMKII and Akt activation in the IUGR brain at PND 1. Additionally, LIPUS treatment promoted glucose transporter (GLUT) 1 production and BDNF expression in the placenta, but had no effects on GLUT3 or amino acid transporter expression. Our findings suggest that antenatal LIPUS treatment may reduce IUGR‐induced brain injury via enhancing cerebral BDNF/CaMKII/Akt signaling. These data provide new evidence that LIPUS stimulation could be considered for antenatal neuroprotective therapy in IUGR.
Collapse
Affiliation(s)
- Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Cheng Liu
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Hu Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan.,Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Hsien Chao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan.,Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
10
|
Escudero C, Acurio J, López E, Rodríguez A, Benavente A, Lara E, Korzeniewski SJ. Vascular endothelial growth factor and poor prognosis after ischaemic stroke. Eur J Neurol 2020; 28:1759-1764. [PMID: 33176035 DOI: 10.1111/ene.14641] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Systemic inflammation conveys information about ischaemic stroke prognosis. Growth factors with neurotrophic and angiogenesis-regulating properties might provide additional information about sequelae. The prognostic performance of circulating vascular endothelial growth factor (VEGF), placental growth factor, interleukin 6 and C-reactive protein measured after acute ischaemic stroke was evaluated. METHODS Blood samples were collected from n = 45 patients within 24-48 h of acute ischaemic stroke. The primary outcome was death or moderate to severe disability at 6 months (modified Rankin Scale >2). Logistic regression models were used to determine the area under the receiver operating characteristic curve (AUC). Correlation and principal component analyses were performed to examine interrelationships amongst biomarkers. RESULTS Vascular endothelial growth factor was elevated in ischaemic stroke patients who died or had moderate to severe disability at six months. Correlation analysis revealed interrelationships between VEGF and HbA1c, triglycerides, erythrocyte sedimentation rate and National Institutes of Health Stroke Scale and Rankin scores, whereas principal component analyses identified VEGF as a major loading factor that discriminated good from poor prognosis. There were no significant differences in AUC using each protein individually to identify patients who had modified Rankin Scale score >2 at 6 months (n = 15/41, AUC 0.61-0.74). However, the AUC increased significantly when combining VEGF with interleukin 6 and C-reactive protein compared to the VEGF-only model (AUC 0.92 vs. 0.67, p = 0.02). CONCLUSION Circulating VEGF was elevated 24-48 h after acute ischaemic stroke and conveyed prognostic information about moderate to severe disability at 6 months.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health, GRIVAS Health, Chillán, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health, GRIVAS Health, Chillán, Chile
| | - Eduardo López
- Neurology Department, Hospital Clínico Herminda Martin, Chillán, Chile.,Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Andrés Rodríguez
- Group of Research and Innovation in Vascular Health, GRIVAS Health, Chillán, Chile.,Cell Communication and Vascular Dynamics Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Antonia Benavente
- Cell Communication and Vascular Dynamics Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health, GRIVAS Health, Chillán, Chile
| | - Steven J Korzeniewski
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine. Detroit,, USA
| |
Collapse
|
11
|
Kay VR, Rätsep MT, Figueiró-Filho EA, Croy BA. Preeclampsia may influence offspring neuroanatomy and cognitive function: a role for placental growth factor†. Biol Reprod 2020; 101:271-283. [PMID: 31175349 DOI: 10.1093/biolre/ioz095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Abstract
Preeclampsia (PE) is a common pregnancy complication affecting 3-5% of women. Preeclampsia is diagnosed clinically as new-onset hypertension with associated end organ damage after 20 weeks of gestation. Despite being diagnosed as a maternal syndrome, fetal experience of PE is a developmental insult with lifelong cognitive consequences. These cognitive alterations are associated with distorted neuroanatomy and cerebrovasculature, including a higher risk of stroke. The pathophysiology of a PE pregnancy is complex, with many factors potentially able to affect fetal development. Deficient pro-angiogenic factor expression is one aspect that may impair fetal vascularization, alter brain structure, and affect future cognition. Of the pro-angiogenic growth factors, placental growth factor (PGF) is strongly linked to PE. Concentrations of PGF are inappropriately low in maternal blood both before and during a PE gestation. Fetal concentrations of PGF appear to mirror maternal circulating concentrations. Using Pgf-/- mice that may model effects of PE on offspring, we demonstrated altered central nervous system vascularization, neuroanatomy, and behavior. Overall, we propose that development of the fetal brain is impaired in PE, making the offspring of preeclamptic pregnancies a unique cohort with greater risk of altered cognition and cerebrovasculature. These individuals may benefit from early interventions, either pharmacological or environmental. The early neonatal period may be a promising window for intervention while the developing brain retains plasticity.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Matthew T Rätsep
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
12
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
13
|
Vaquié A, Sauvain A, Duman M, Nocera G, Egger B, Meyenhofer F, Falquet L, Bartesaghi L, Chrast R, Lamy CM, Bang S, Lee SR, Jeon NL, Ruff S, Jacob C. Injured Axons Instruct Schwann Cells to Build Constricting Actin Spheres to Accelerate Axonal Disintegration. Cell Rep 2020; 27:3152-3166.e7. [PMID: 31189102 DOI: 10.1016/j.celrep.2019.05.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 05/17/2019] [Indexed: 01/26/2023] Open
Abstract
After a peripheral nerve lesion, distal ends of injured axons disintegrate into small fragments that are subsequently cleared by Schwann cells and later by macrophages. Axonal debris clearing is an early step of the repair process that facilitates regeneration. We show here that Schwann cells promote distal cut axon disintegration for timely clearing. By combining cell-based and in vivo models of nerve lesion with mouse genetics, we show that this mechanism is induced by distal cut axons, which signal to Schwann cells through PlGF mediating the activation and upregulation of VEGFR1 in Schwann cells. In turn, VEGFR1 activates Pak1, leading to the formation of constricting actomyosin spheres along unfragmented distal cut axons to mediate their disintegration. Interestingly, oligodendrocytes can acquire a similar behavior as Schwann cells by enforced expression of VEGFR1. These results thus identify controllable molecular cues of a neuron-glia crosstalk essential for timely clearing of damaged axons.
Collapse
Affiliation(s)
- Adrien Vaquié
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Alizée Sauvain
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Mert Duman
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gianluigi Nocera
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Bioimage Light Microscopy Facility, University of Fribourg, Fribourg, Switzerland
| | - Felix Meyenhofer
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Medicine, University of Fribourg, Fribourg, Switzerland; Bioimage Light Microscopy Facility, University of Fribourg, Fribourg, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Medicine, University of Fribourg, Fribourg, Switzerland; Bioinformatics Core Facility, University of Fribourg and Swiss Institute of Bioinformatics, Fribourg, Switzerland
| | - Luca Bartesaghi
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roman Chrast
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Seokyoung Bang
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Seung-Ryeol Lee
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Sophie Ruff
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Claire Jacob
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
14
|
Cheng CY, Kao ST, Lee YC. Angelica sinensis extract protects against ischemia-reperfusion injury in the hippocampus by activating p38 MAPK-mediated p90RSK/p-Bad and p90RSK/CREB/BDNF signaling after transient global cerebral ischemia in rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112612. [PMID: 31988015 DOI: 10.1016/j.jep.2020.112612] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Angelica sinensis (Oliv.) Diels, commonly known as Dang Gui (DG), is one of the most popular traditional Chinese herbal medicines for the treatment of stroke. However, the effects of DG on transient global cerebral ischemia (GCI) and its precise mechanisms remain unclear. AIM OF THE STUDY This study aimed to investigate the effects of the DG extract on ischemia-reperfusion (I/R) injury in the hippocampus 7 d after transient GCI and to identify the potential mitogen-activated protein kinase (MAPK)-related signaling pathway in the hippocampus involved in the effects. MATERIALS AND METHODS Rats were intragastrically administered DG at doses of 0.25 g/kg (DG-0.25g), 0.5 g/kg (DG-0.5g), or 1 g/kg (DG-1g) 1, 3, and 5 d after GCI. RESULTS DG-0.5g and DG-1g treatments effectively promoted hippocampal cornu ammonis 1 (CA1) neuronal survival. DG-0.5g and DG-1g treatments markedly increased phospho-p38 MAPK (p-p38 MAPK), phospho-90-kDa ribosomal S6 kinase (p-p90RSK), cytosolic and mitochondrial phospho-Bad (p-Bad), phospho-cAMP response element-binding protein (p-CREB), brain-derived neurotrophic factor (BDNF), and p-CREB/BDNF expression; decreased 4-hydroxy-2-nonenal, cytochrome c (Cytc), and cleaved caspase-3 expression, and inhibited apoptosis in the hippocampal CA1 region. Pretreatment with a specific inhibitor of p38 MAPK, SB203580, completely blocked the effects of DG-1g on the expression of the aforementioned proteins. CONCLUSIONS DG-0.5g and DG-1g treatments exerted neuroprotective effects on I/R injury by activating p38 MAPK-mediated p90RSK/p-Bad-induced anti-apoptotic-Cytc/caspase-3-related and p90RSK/CREB/BDNF survival signaling in the hippocampus 7 d after transient GCI.
Collapse
Affiliation(s)
- Chin-Yi Cheng
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan; Department of Chinese Medicine, Hui-Sheng Hospital, 42056, Taichung, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Yu-Chen Lee
- Department of Chinese Medicine, China Medical University Hospital, 40447, Taichung, Taiwan; Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung, 40402, Taiwan; Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
15
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
16
|
Hansson O, Santillo AF, Meeter LH, Nilsson K, Landqvist Waldö M, Nilsson C, Blennow K, van Swieten JC, Janelidze S. CSF placental growth factor - a novel candidate biomarker of frontotemporal dementia. Ann Clin Transl Neurol 2019; 6:863-872. [PMID: 31139684 PMCID: PMC6529985 DOI: 10.1002/acn3.763] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Objective Diagnosis of frontotemporal dementia (FTD) is complicated by the overlap of clinical symptoms with other dementia disorders. Development of robust fluid biomarkers is critical to improve the diagnostic work‐up of FTD. Methods CSF concentrations of placental growth factor (PlGF) were measured in the discovery cohort including patients with FTD (n = 27), Alzheimer disease (AD) dementia (n = 75), DLB or PDD (n = 47), subcortical vascular dementia (VaD, n = 33), mild cognitive impairment that later converted to AD (MCI‐AD, n = 34), stable MCI (sMCI, n = 62), and 50 cognitively healthy controls from the Swedish BioFINDER study. For validation, CSF PlGF was measured in additional independent cohort of FTD patients (n = 22) and controls (n = 18) from the Netherlands. Results In the discovery cohort, MCI, MCI‐AD, AD dementia, DLB‐PDD, VaD, and FTD patients all showed increased CSF levels of PlGF compared with controls (sMCI P = 0.019; MCI‐AD P = 0.005; AD dementia, DLB‐PDD, VaD, and FTD all P < 0.001). PlGF levels were 1.8–2.1‐fold higher in FTD than in AD, DLB‐PDD and VaD (all P < 0.001). PlGF distinguished with high accuracy FTD from controls and sMCI performing better than tau/Aβ42 (AUC 0.954–0.996 versus 0.564–0.754, P < 0.001). A combination of PlGF, tau, and Aβ42 (tau/Aβ42/PlGF) was more accurate than tau/Aβ42 when differentiating FTD from a group of other dementias (AUC 0.972 vs. 0.932, P < 0.01). Increased CSF levels of PlGF in FTD compared with controls were corroborated in the validation cohort. Interpretation CSF PlGF is increased in FTD compared with other dementia disorders, MCI, and healthy controls and might be useful as a diagnostic biomarker of FTD.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Memory Clinic Skåne University Hospital Malmö Sweden
| | - Alexander F Santillo
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Memory Clinic Skåne University Hospital Malmö Sweden
| | - Lieke H Meeter
- Department of Neurology Erasmus Medical Center Rotterdam The Netherlands
| | - Karin Nilsson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden
| | - Maria Landqvist Waldö
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Clinical Sciences Helsingborg Department of Clinical Sciences Lund University Lund Sweden
| | - Christer Nilsson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Department of Neurology Skåne University Hospital Lund Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry The Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden.,Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
| | - John C van Swieten
- Department of Neurology Erasmus Medical Center Rotterdam The Netherlands.,Department of Clinical Genetics VU University Medical Center Amsterdam The Netherlands
| | - Shorena Janelidze
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden
| |
Collapse
|
17
|
Sánchez O, Ruiz-Romero A, Domínguez C, Ferrer Q, Ribera I, Rodríguez-Sureda V, Alijotas J, Arévalo S, Carreras E, Cabero L, Llurba E. Brain angiogenic gene expression in fetuses with congenital heart disease. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2018; 52:734-738. [PMID: 29205570 DOI: 10.1002/uog.18977] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/09/2017] [Accepted: 11/28/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To assess potential differences in the expression of antiangiogenic and angiogenic factors and of genes associated with chronic hypoxia in cerebral tissue of euploid fetuses with congenital heart disease (CHD) vs those without. METHODS Cerebral tissue was obtained from 15 fetuses with CHD and 12 control fetuses that had undergone termination of pregnancy. Expression profiles of the antiangiogenic factor soluble fms-like tyrosine kinase-1 (sFlt-1), the angiogenic vascular endothelial growth factor-A (VEGF-A) and placental growth factor (PlGF), and of genes associated with chronic hypoxia were determined by real-time polymerase chain reaction in tissue from the frontal cortex and the basal ganglia of the fetuses. RESULTS Expression of sFlt-1 was 48% higher in the frontal cortex (P = 0.0431) and 72% higher in the basal ganglia (P = 0.0369) of CHD fetuses compared with controls. The expression of VEGF-A was 60% higher (P = 0.0432) and that of hypoxia-inducible factor 2-alpha was 98% higher (P = 0.0456) in the basal ganglia of CHD fetuses compared with controls. No significant differences were observed between the two groups in the expression of PlGF and hypoxia-inducible factor 1-alpha. CONCLUSION An overall dysregulation of angiogenesis with a net balance towards an antiangiogenic environment was observed in the cerebral tissue of fetuses with CHD, suggesting that these fetuses may have an intrinsic angiogenic impairment that could contribute to impaired brain perfusion and abnormal neurological development later in life. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- O Sánchez
- Maternal and Child Health and Development Network (SAMID), RD16/0022/0015, Instituto de Salud Carlos III, Barcelona, Spain
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - A Ruiz-Romero
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
| | - C Domínguez
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Q Ferrer
- Pediatric Cardiology Unit, Department of Pediatrics, Vall d'Hebron University Hospital, Barcelona, Spain
| | - I Ribera
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
| | - V Rodríguez-Sureda
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - J Alijotas
- Department of Internal Medicine, Vall d'Hebron University Hospital, Barcelona, Spain
- School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S Arévalo
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
| | - E Carreras
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
| | - L Cabero
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
- School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - E Llurba
- Maternal-Fetal Medicine Unit, Department of Obstetrics, Vall d'Hebron Research Institute (VHIR), SAMID Network, Vall d'Hebron University Hospital, Barcelona, Spain
- School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
19
|
The pericyte secretome: Potential impact on regeneration. Biochimie 2018; 155:16-25. [PMID: 29698670 DOI: 10.1016/j.biochi.2018.04.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
Abstract
Personalized and regenerative medicine is an emerging therapeutic strategy that is based on cell biology and biomedical engineering used to develop biological substitutes to maintain normal function or restore damaged tissues and organs. The secretory capacities of different cell types are now explored as such possible therapeutic regenerative agents in a variety of diseases. A secretome can comprise chemokines, cytokines, growth factors, but also extracellular matrix components, microvesicles and exosomes as well as genetic material and may differ depending on the tissue and the stimulus applied to the cell. With regard to clinical applications, the secretome of mesenchymal stem cells (MSC) is currently the most widely explored. However, other cell types such as pericytes may have similar properties as MSC and the potential therapeutic possibilities of these cells are only just beginning to emerge. In this review, we will summarize the currently available data describing the secretome of pericytes and its potential implications for tissue regeneration, whereby we especially focus on brain pericytes as potential new target cell for neuroregeneration and brain repair.
Collapse
|
20
|
Gaceb A, Özen I, Padel T, Barbariga M, Paul G. Pericytes secrete pro-regenerative molecules in response to platelet-derived growth factor-BB. J Cereb Blood Flow Metab 2018; 38:45-57. [PMID: 28741407 PMCID: PMC5757443 DOI: 10.1177/0271678x17719645] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Brain pericytes not only maintain the anatomical, biochemical and immune blood-brain barrier, but display features of mesenchymal stem cells (MSCs) in vitro. MSCs have pro-regenerative properties attributed to their secretome. However, whether also brain pericytes possess such pro-regenerative capacities is largely unknown. Here we characterize the secretome and microvesicle (MV) release of human brain pericytes mediated by platelet-derived growth factor-BB (PDGF-BB)/PDGF receptor beta (PDGFRβ) signalling. Upon PDGF-BB, pericytes release not only a plethora of growth factors and a panel of cytokines, but also MVs containing BDNF, FGFb, βNGF, VEGF and PLGF, a response that is specific for PDGFRβ signalling and activation of the ERK 1/2 pathway. In contrast, lipopolysaccharide (LPS), an activator of the innate immune system, stimulates the secretion of much higher amounts of mainly inflammatory cytokines and activates the NFκB pathway. Pericytes change their morphology and undergo opposite changes in surface marker expression, respectively. Our findings provide evidence that the secretome of human brain pericytes varies greatly depending on the exogenous stimulus. The differential secretory functions of pericytes may play an important role in either regulating neuroinflammation or contributing to neurorestoration and identify a possible new target cell for neuroregeneration.
Collapse
Affiliation(s)
- Abderahim Gaceb
- 1 Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Ilknur Özen
- 1 Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Thomas Padel
- 1 Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Marco Barbariga
- 1 Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Gesine Paul
- 1 Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,2 Department of Neurology, Scania University Hospital, Lund, Sweden
| |
Collapse
|
21
|
Luna RL, Kay VR, Rätsep MT, Khalaj K, Bidarimath M, Peterson N, Carmeliet P, Jin A, Croy BA. Placental growth factor deficiency is associated with impaired cerebral vascular development in mice. Mol Hum Reprod 2016; 22:130-42. [PMID: 26646502 PMCID: PMC4733225 DOI: 10.1093/molehr/gav069] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/23/2015] [Accepted: 11/27/2015] [Indexed: 12/23/2022] Open
Abstract
STUDY HYPOTHESIS Placental growth factor (PGF) is expressed in the developing mouse brain and contributes to vascularization and vessel patterning. STUDY FINDING PGF is dynamically expressed in fetal mouse brain, particularly forebrain, and is essential for normal cerebrovascular development. WHAT IS KNOWN ALREADY PGF rises in maternal plasma over normal human and mouse pregnancy but is low in many women with the acute onset hypertensive syndrome, pre-eclampsia (PE). Little is known about the expression of PGF in the fetus during PE. Pgf (-/-) mice appear normal but recently cerebral vascular defects were documented in adult Pgf (-/-) mice. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Here, temporal-spatial expression of PGF is mapped in normal fetal mouse brains and cerebral vasculature development is compared between normal and congenic Pgf (-/-) fetuses to assess the actions of PGF during cerebrovascular development. Pgf/PGF, Vegfa/VEGF, Vegf receptor (Vegfr)1 and Vegfr2 expression were examined in the brains of embryonic day (E)12.5, 14.5, 16.5 and 18.5 C57BL/6 (B6) mice using quantitative PCR and immunohistochemistry. The cerebral vasculature was compared between Pgf (-/-) and B6 embryonic and adult brains using whole mount techniques. Vulnerability to cerebral ischemia was investigated using a left common carotid ligation assay. MAIN RESULTS AND THE ROLE OF CHANCE Pgf/PGF and Vegfr1 are highly expressed in E12.5-14.5 forebrain relative to VEGF and Vegfr2. Vegfa/VEGF is relatively more abundant in hindbrain (HB). PGF and VEGF expression were similar in midbrain. Delayed HB vascularization was seen at E10.5 and 11.5 in Pgf (-/-) brains. At E14.5, Pgf (-/-) circle of Willis showed unilateral hypoplasia and fewer collateral vessels, defects that persisted post-natally. Functionally, adult Pgf (-/-) mice experienced cerebral ischemia after left common carotid arterial occlusion while B6 mice did not. LIMITATIONS, REASONS FOR CAUTION Since Pgf (-/-) mice were used, consequences of complete absence of maternal and fetal PGF were defined. Therefore, the effects of maternal versus fetal PGF deficiency on cerebrovascular development cannot be separated. However, as PGF was strongly expressed in the developing brain at all timepoints, we suggest that local PGF has a more important role than distant maternal or placental sources. Full PGF loss is not expected in PE pregnancies, predicting that the effects of PGF deficiency identified in this model will be more severe than any effects in PE-offspring. WIDER IMPLICATIONS OF THE FINDINGS These studies provoke the question of whether PGF expression is decreased and cerebral vascular maldevelopment occurs in fetuses who experience a preeclamptic gestation. These individuals have already been reported to have elevated risk for stroke and cognitive impairments. LARGE SCALE DATA N/A. STUDY FUNDING AND COMPETING INTERESTS This work was supported by awards from the Natural Sciences and Engineering Research Council, the Canada Research Chairs Program and the Canadian Foundation for Innovation to B.A.C. and by training awards from the Universidade Federal de Pernambuco and Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brazil to R.L.L.; Queen's University to V.R.K. and the Canadian Institutes of Health Research to M.T.R. The work of P.C. is supported by the Belgian Science Policy BELSPO-IUAP7/03, Structural funding by the Flemish Government-Methusalem funding, and the Flemish Science Fund-FWO grants. There were no competing interests.
Collapse
Affiliation(s)
- Rayana Leal Luna
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada Federal University of Pernambuco - UFPE, Recife, Pernambuco 50670-901, Brazil
| | - Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Kasra Khalaj
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Mallikarjun Bidarimath
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Nichole Peterson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Albert Jin
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
22
|
Baumgarten P, Blank AE, Franz K, Hattingen E, Dunst M, Zeiner P, Hoffmann K, Bähr O, Mäder L, Goeppert B, Machein M, Seifert V, Steinbach JP, Plate KH, Harter PN, Mittelbronn M. Differential expression of vascular endothelial growth factor A, its receptors VEGFR-1, -2, and -3 and co-receptors neuropilin-1 and -2 does not predict bevacizumab response in human astrocytomas. Neuro Oncol 2015; 18:173-83. [PMID: 26627848 DOI: 10.1093/neuonc/nov288] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/27/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND A major hallmark of malignant progression in human astrocytomas is the formation of new blood vessels. Antiangiogenic therapy using the anti-vascular endothelial growth factor (VEGF)-antibody bevacizumab leads to increased progression-free survival in glioblastoma patients but does not influence their overall survival. To date, it is unclear why antiangiogenic therapy fails in many glioblastoma patients, while a small subpopulation profits considerably from this treatment. METHODS The aim of our study was to determine the expression of VEGF-A and its (co-) receptors by immunohistochemistry and to test the association with patient survival in 350 glioma patients. Additionally, VEGF-A expression was analyzed by in-situ hybridization. In 18 patients, the protein expression was compared with the bevacizumab response according to extended and modified RANO criteria. RESULTS We found a heterogeneous expression pattern of VEGF and its receptors in glioblastoma patients with significantly lower levels in WHO grade II and III tumors and normal-appearing brain tissue (P < .001). Pilocytic astrocytomas (WHO grade I) showed significantly higher VEGFR-1, -2 and neuropilin-1 levels as compared to WHO grade II and III astrocytomas (P < .01) but at lower levels than glioblastomas. The expression of neuropilin-2 was low in all tumors. There was neither a significant correlation between protein expression and patient survival nor between protein levels and bevacizumab response after modified RANO criteria. CONCLUSION Since our data indicate that beneficial response to bevacizumab treatment is independent of the expression of VEGF-A and its (co-) receptors, further investigation is needed to decipher the underlying mechanisms of antiangiogenic treatment response.
Collapse
Affiliation(s)
- Peter Baumgarten
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Anna-Eva Blank
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Kea Franz
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Elke Hattingen
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Maika Dunst
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Pia Zeiner
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Katharina Hoffmann
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Oliver Bähr
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Lisa Mäder
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Benjamin Goeppert
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Marcia Machein
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Volker Seifert
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Joachim P Steinbach
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Karl H Plate
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Patrick N Harter
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany (P.B., A.-E.B., M.D., P.Z., K.H., L.M., K.H.P., P.N.H., M.Mi.); Department of Neurosurgery, Goethe University, Frankfurt, Germany (K.F., V.S.); Dr. Senckenberg Institute of Neurooncology, University of Frankfurt am Main, Frankfurt am Main, Germany (K.F., O.B., J.P.S.); Department of Neuroradiology, University of Frankfurt am Main, Frankfurt am Main, Germany (E.H.); Department of Neurosurgery, University Hospital, Freiburg, Germany (M.Ma.); Cancer Consortium (DKTK), Heidelberg, Germany (O.B., J.P.S., K.H.P, P.N.H., M.Mi.); German Cancer Research Center (DKFZ), Heidelberg, Germany (O.B., J.P.S., K.H.P., P.N.H., M.Mi.); Department of Pathology, University of Heidelberg, Heidelberg, Germany (B.G.)
| |
Collapse
|
23
|
Ergul A, Valenzuela JP, Fouda AY, Fagan SC. Cellular connections, microenvironment and brain angiogenesis in diabetes: Lost communication signals in the post-stroke period. Brain Res 2015; 1623:81-96. [PMID: 25749094 PMCID: PMC4743654 DOI: 10.1016/j.brainres.2015.02.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/18/2015] [Accepted: 02/23/2015] [Indexed: 12/16/2022]
Abstract
Diabetes not only increases the risk but also worsens the motor and cognitive recovery after stroke, which is the leading cause of disability worldwide. Repair after stroke requires coordinated communication among various cell types in the central nervous system as well as circulating cells. Vascular restoration is critical for the enhancement of neurogenesis and neuroplasticity. Given that vascular disease is a major component of all complications associated with diabetes including stroke, this review will focus on cellular communications that are important for vascular restoration in the context of diabetes. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - John Paul Valenzuela
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 15th Street, CA 2094, Augusta, GA 30912, USA
| | - Abdelrahman Y Fouda
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Susan C Fagan
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA; Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| |
Collapse
|
24
|
Ceafalan LC, Manole E, Tanase CP, Codrici E, Mihai S, Gonzalez A, Popescu BO. Interstitial Outburst of Angiogenic Factors During Skeletal Muscle Regeneration After Acute Mechanical Trauma. Anat Rec (Hoboken) 2015; 298:1864-79. [PMID: 26260512 DOI: 10.1002/ar.23254] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/01/2015] [Accepted: 06/12/2015] [Indexed: 11/09/2022]
Abstract
Angiogenesis is a key event during tissue regeneration, but the intimate mechanisms controlling this process are still largely unclear. Therefore, the cellular and molecular interplay along normal tissue regeneration should be carefully unveiled. To this matter, we investigated by xMAP assay the dynamics of some angiogenic factors known to be involved in tissue repair, such as follistatin (FST), Placental Growth Factor-2 (PLGF-2), epidermal growth factor (EGF), betacellulin (BTC), and amphiregulin (AREG) using an animal model that mimics acute muscle contusion injuries. In situ immunofluorescence was used for the evaluation and tissue distribution of their cellular sources. Tissue levels of explored factors increased significantly during degeneration and inflammatory stage of regeneration, peaking first week postinjury. However, except for PLGF-2 and EGF, their levels remained significantly elevated after the inflammatory process started to fade. Serum levels were significantly increased only after 24 h for AREG and EGF. Though, for all factors except FST, the levels in injured samples did not correlate with serum or contralateral tissue levels, excluding the systemic influence. We found significant correlations between the levels of EGF and AREG, BTC, FST and FST and AREG in injured samples. Interstitial cells expressing these factors were highlighted by in situ immunolabeling and their number correlated with measured levels dynamics. Our study provides evidence of a dynamic level variation along the regeneration process and a potential interplay between selected angiogenic factors. They are synthesized, at least partially, by cell populations residing in skeletal muscle interstitium during regeneration after acute muscle trauma.
Collapse
Affiliation(s)
- Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, School of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Department of Molecular Medicine and Neuroscience, "Victor Babes" Institute of Pathology, Bucharest, Romania
| | - Emilia Manole
- Department of Molecular Medicine and Neuroscience, "Victor Babes" Institute of Pathology, Bucharest, Romania
| | - Cristiana Pistol Tanase
- Biochemistry/Proteomics Department, "Victor Babes" Institute of Pathology, Bucharest, Romania
| | - Elena Codrici
- Biochemistry/Proteomics Department, "Victor Babes" Institute of Pathology, Bucharest, Romania
| | - Simona Mihai
- Biochemistry/Proteomics Department, "Victor Babes" Institute of Pathology, Bucharest, Romania
| | - Aldebarani Gonzalez
- Department of Cellular and Molecular Biology and Histology, School of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan Ovidiu Popescu
- Department of Molecular Medicine and Neuroscience, "Victor Babes" Institute of Pathology, Bucharest, Romania.,Department of Neurology, Colentina Clinical Hospital-Colentina Research Center, School of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
25
|
Shaikh H, Boudes E, Khoja Z, Shevell M, Wintermark P. Angiogenesis dysregulation in term asphyxiated newborns treated with hypothermia. PLoS One 2015; 10:e0128028. [PMID: 25996847 PMCID: PMC4440713 DOI: 10.1371/journal.pone.0128028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/21/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Neonatal encephalopathy following birth asphyxia is a major predictor of long-term neurological impairment. Therapeutic hypothermia is currently the standard of care to prevent brain injury in asphyxiated newborns but is not protective in all cases. More robust and versatile treatment options are needed. Angiogenesis is a demonstrated therapeutic target in adult stroke. However, no systematic study examines the expression of angiogenesis-related markers following birth asphyxia in human newborns. OBJECTIVE This study aimed to evaluate the expression of angiogenesis-related protein markers in asphyxiated newborns developing and not developing brain injury compared to healthy control newborns. DESIGN/METHODS Twelve asphyxiated newborns treated with hypothermia were prospectively enrolled; six developed eventual brain injury and six did not. Four healthy control newborns were also included. We used Rules-Based Medicine multi-analyte profiling and protein array technologies to study the plasma concentration of 49 angiogenesis-related proteins. Mean protein concentrations were compared between each group of newborns. RESULTS Compared to healthy newborns, asphyxiated newborns not developing brain injury showed up-regulation of pro-angiogenic proteins, including fatty acid binding protein-4, glucose-6-phosphate isomerase, neuropilin-1, and receptor tyrosine-protein kinase erbB-3; this up-regulation was not evident in asphyxiated newborns eventually developing brain injury. Also, asphyxiated newborns developing brain injury showed a decreased expression of anti-angiogenic proteins, including insulin-growth factor binding proteins -1, -4, and -6, compared to healthy newborns. CONCLUSIONS These findings suggest that angiogenesis pathways are dysregulated following birth asphyxia and are putatively involved in brain injury pathology and recovery.
Collapse
Affiliation(s)
- Henna Shaikh
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Elodie Boudes
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Zehra Khoja
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Michael Shevell
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Pia Wintermark
- Division of Newborn Medicine, Department of Pediatrics, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
26
|
Hou ST, Nilchi L, Li X, Gangaraju S, Jiang SX, Aylsworth A, Monette R, Slinn J. Semaphorin3A elevates vascular permeability and contributes to cerebral ischemia-induced brain damage. Sci Rep 2015; 5:7890. [PMID: 25601765 PMCID: PMC4298747 DOI: 10.1038/srep07890] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/16/2014] [Indexed: 12/25/2022] Open
Abstract
Semaphorin 3A (Sema3A) increased significantly in mouse brain following cerebral ischemia. However, the role of Sema3A in stroke brain remains unknown. Our aim was to determine wether Sema3A functions as a vascular permeability factor and contributes to ischemic brain damage. Recombinant Sema3A injected intradermally to mouse skin, or stereotactically into the cerebral cortex, caused dose- and time-dependent increases in vascular permeability, with a degree comparable to that caused by injection of a known vascular permeability factor vascular endothelial growth factor receptors (VEGF). Application of Sema3A to cultured endothelial cells caused disorganization of F-actin stress fibre bundles and increased endothelial monolayer permeability, confirming Sema3A as a permeability factor. Sema3A-mediated F-actin changes in endothelial cells were through binding to the neuropilin2/VEGFR1 receptor complex, which in turn directly activates Mical2, a F-actin modulator. Down-regulation of Mical2, using specific siRNA, alleviated Sema3A-induced F-actin disorganization, cellular morphology changes and endothelial permeability. Importantly, ablation of Sema3A expression, cerebrovascular permeability and brain damage were significantly reduced in response to transient middle cerebral artery occlusion (tMCAO) and in a mouse model of cerebral ischemia/haemorrhagic transformation. Together, these studies demonstrated that Sema3A is a key mediator of cerebrovascular permeability and contributes to brain damage caused by cerebral ischemia.
Collapse
Affiliation(s)
- Sheng Tao Hou
- 1] Department of Biology, South University of Science and Technology of China, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, P.R. China, 518055 [2] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada [3] Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Ladan Nilchi
- 1] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada [2] Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Xuesheng Li
- 1] Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada [2] Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Sandhya Gangaraju
- Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Susan X Jiang
- Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Amy Aylsworth
- Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Robert Monette
- Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| | - Jacqueline Slinn
- Human Health Therapeutics Portfolio, National Research Council Canada, 1200 Montreal Road, Bldg M54, Ottawa, Ontario, K1A 0R6, Canada
| |
Collapse
|
27
|
Yin KJ, Hamblin M, Chen YE. Angiogenesis-regulating microRNAs and Ischemic Stroke. Curr Vasc Pharmacol 2015; 13:352-65. [PMID: 26156265 PMCID: PMC4079753 DOI: 10.2174/15701611113119990016] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/12/2012] [Accepted: 11/15/2012] [Indexed: 12/19/2022]
Abstract
Stroke is a leading cause of death and disability worldwide. Ischemic stroke is the dominant subtype of stroke and results from focal cerebral ischemia due to occlusion of major cerebral arteries. Thus, the restoration or improvement of reduced regional cerebral blood supply in a timely manner is very critical for improving stroke outcomes and poststroke functional recovery. The recovery from ischemic stroke largely relies on appropriate restoration of blood flow via angiogenesis. Newly formed vessels would allow increased cerebral blood flow, thus increasing the amount of oxygen and nutrients delivered to affected brain tissue. Angiogenesis is strictly controlled by many key angiogenic factors in the central nervous system, and these molecules have been well-documented to play an important role in the development of angiogenesis in response to various pathological conditions. Promoting angiogenesis via various approaches that target angiogenic factors appears to be a useful treatment for experimental ischemic stroke. Most recently, microRNAs (miRs) have been identified as negative regulators of gene expression in a post-transcriptional manner. Accumulating studies have demonstrated that miRs are essential determinants of vascular endothelial cell biology/angiogenesis as well as contributors to stroke pathogenesis. In this review, we summarize the knowledge of stroke-associated angiogenic modulators, as well as the role and molecular mechanisms of stroke-associated miRs with a focus on angiogenesis-regulating miRs. Moreover, we further discuss their potential impact on miR-based therapeutics in stroke through targeting and enhancing post-ischemic angiogenesis.
Collapse
Affiliation(s)
- Ke-Jie Yin
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Milton Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, Louisiana 70112, USA
| | - Y. Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
28
|
Kristen AV, Rinn J, Hegenbart U, Lindenmaier D, Merkle C, Röcken C, Hardt S, Giannitsis E, Katus HA. Improvement of risk assessment in systemic light-chain amyloidosis using human placental growth factor. Clin Res Cardiol 2014; 104:250-7. [PMID: 25331161 DOI: 10.1007/s00392-014-0779-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Vascular amyloid deposition is common in light-chain amyloidosis resulting in endothelial dysfunction. Human placental growth factor (PlGF), a member of the vascular endothelial growth factor family was found to be altered in diverse pathological conditions, e.g. endothelial dysfunction. This study evaluated the clinical role of PlGF in light-chain amyloidosis. METHODS PlGF (cobas-PlGF, Roche Diagnostics, Mannheim, Germany) was analyzed in 125 consecutive patients with AL and correlated with diverse clinical parameters including mortality. RESULTS Kidney (n = 76) and heart (n = 57) were predominantly affected by amyloid deposition. Median PlGF was 26.3 (21.1-42.1) ng/L, NT-proBNP 3649 (1124-8581) pg/mL, and hs-TnT 42 (21-107) ng/L. PlGF increased with number of organs involved and with deterioration of renal function. A significant correlation of PlGF with hs-TnT (ρ = 0.306; p = 0.0007) and NT-proBNP (ρ = 0.315; p = 0.0006) was observed, but no correlation was observed with clinical, echocardiography, and electrocardiography parameters of cardiac involvement. In this cohort 1-year all-cause mortality was 19.2 %. The best cutoff discriminating survivors and non-survivors was 28.44 ng/L (sensitivity 66.7 %; specificity 78.1 %). A three-step risk model including hs-TnT and NT-proBNP revealed a better discrimination if patients at intermediary risk were additionally stratified by PlGF. Net reclassification index was 37.2 % (p = 0.002). Multivariate analysis revealed PlGF, difference of involved and uninvolved light chain, number of organs involved and risk class according to troponin T and NT-proBNP as independent predictors of mortality. CONCLUSION Plasma PlGF values in AL are invariably associated with the number of involved organs, but not with clinical, echocardiography, and electrocardiography parameters of cardiac involvement. PlGF provide useful information for risk stratification of patients at intermediary risk according to hs-TnT and NT-proBNP.
Collapse
Affiliation(s)
- Arnt V Kristen
- Department of Cardiology, Angiology, and Respiratory Medicine, Heidelberg University, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sun FL, Wang W, Cheng H, Wang Y, Li L, Xue JL, Wang XF, Ai HX, Zhang L, Xu JD, Wang XM, Ji XM. Morroniside improves microvascular functional integrity of the neurovascular unit after cerebral ischemia. PLoS One 2014; 9:e101194. [PMID: 24979385 PMCID: PMC4076313 DOI: 10.1371/journal.pone.0101194] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/04/2014] [Indexed: 12/20/2022] Open
Abstract
Treating the vascular elements within the neurovascular unit is essential for protecting and repairing the brain after stroke. Acute injury on endothelial systems results in the disruption of blood-brain barrier (BBB), while post-ischemic angiogenesis plays an important role in delayed functional recovery. Here, we considered alterations in microvessel integrity to be targets for brain recovery, and tested the natural compound morroniside as a therapeutic approach to restore the vascular elements of injured tissue in a rat model of focal cerebral ischemia. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) model, and morroniside was then administered intragastrically once a day at doses of 30, 90, and 270 mg/kg. BBB integrity and associated factors were analyzed to identify cerebrovascular permeability 3 days after MCAO. The recruitment of endothelial progenitor cells (EPCs), the expression of angiogenic factors and the new vessel formation in the peri-infarct cortex of rats were examined 7 days after MCAO to identify the angiogenesis. We demonstrated that at 3 days post-ischemia, morroniside preserved neurovascular unit function by ameliorating BBB injury. By 7 days post-ischemia, morroniside amplified angiogenesis, in part by enhancing endothelial progenitor cell proliferation and expression of angiogenic factors. Morever, the increase in the amount of vWF+ vessels induced by ischemia could be extended to 28 days after administration of morroniside, indicating the crucial role of morroniside in angiogenesis during the chronic phase. Taken together, our findings suggested that morroniside might offer a novel therapeutic approach for promoting microvascular integrity recovery and provide a thoroughly new direction for stroke therapy.
Collapse
Affiliation(s)
- Fang-Ling Sun
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| | - Hua Cheng
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lei Li
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jin-Long Xue
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiao-feng Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hou-Xi Ai
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jing-dong Xu
- Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Min Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| | - Xun-Ming Ji
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| |
Collapse
|
30
|
Pronto-Laborinho AC, Pinto S, de Carvalho M. Roles of vascular endothelial growth factor in amyotrophic lateral sclerosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:947513. [PMID: 24987705 PMCID: PMC4022172 DOI: 10.1155/2014/947513] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/24/2014] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal devastating neurodegenerative disorder, involving progressive degeneration of motor neurons in spinal cord, brainstem, and motor cortex. Riluzole is the only drug approved in ALS but it only confers a modest improvement in survival. In spite of a high number of clinical trials no other drug has proved effectiveness. Recent studies support that vascular endothelial growth factor (VEGF), originally described as a key angiogenic factor, also plays a key role in the nervous system, including neurogenesis, neuronal survival, neuronal migration, and axon guidance. VEGF has been used in exploratory clinical studies with promising results in ALS and other neurological disorders. Although VEGF is a very promising compound, translating the basic science breakthroughs into clinical practice is the major challenge ahead. VEGF-B, presenting a single safety profile, protects motor neurons from degeneration in ALS animal models and, therefore, it will be particularly interesting to test its effects in ALS patients. In the present paper the authors make a brief description of the molecular properties of VEGF and its receptors and review its different features and therapeutic potential in the nervous system/neurodegenerative disease, particularly in ALS.
Collapse
Affiliation(s)
- Ana Catarina Pronto-Laborinho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Susana Pinto
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Mamede de Carvalho
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Instituto de Medicina Molecular (IMM), Translational Clinical Physiology Unit, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Department of Neurosciences, Hospital Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
31
|
Developmental and pathological angiogenesis in the central nervous system. Cell Mol Life Sci 2014; 71:3489-506. [PMID: 24760128 DOI: 10.1007/s00018-014-1625-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vessels, in the central nervous system (CNS) is seen both as a normal physiological response as well as a pathological step in disease progression. Formation of the blood-brain barrier (BBB) is an essential step in physiological CNS angiogenesis. The BBB is regulated by a neurovascular unit (NVU) consisting of endothelial and perivascular cells as well as vascular astrocytes. The NVU plays a critical role in preventing entry of neurotoxic substances and regulation of blood flow in the CNS. In recent years, research on numerous acquired and hereditary disorders of the CNS has increasingly emphasized the role of angiogenesis in disease pathophysiology. Here, we discuss molecular mechanisms of CNS angiogenesis during embryogenesis as well as various pathological states including brain tumor formation, ischemic stroke, arteriovenous malformations, and neurodegenerative diseases.
Collapse
|
32
|
Freitas-Andrade M, Slinn J, Charlebois C, Moreno MJ. Histological assessment of angiogenesis in the hypoxic central nervous system. Methods Mol Biol 2014; 1135:157-75. [PMID: 24510863 DOI: 10.1007/978-1-4939-0320-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Angiogenesis, the sprouting of new capillaries from preexisting vessels, is an integral part of both normal development and numerous pathological conditions such as tumor growth, inflammation, and stroke. The development of angiogenesis assays has been critical in understanding this process in both the context of disease and normal physiology. With the growing availability of antibodies against angiogenic markers as well as advances in microscopy and imaging analysis software, a more comprehensive assessment of the angiogenesis process is beginning to take form (Milner et al., Stroke 39:191-197, 2008; Freitas-Andrade et al., J Cereb Blood Flow Metab 32:663-675, 2012; Li et al., Glia 58:1157-1167, 2010; Dore-Duffy and LaManna, Antioxid Redox Signal 9:1363-1371, 2007). This chapter describes an in vivo method of inducing brain angiogenesis in mice by chronic exposure to mild hypoxia. In addition, a detailed procedure of quantifying angiogenesis using multiple immunofluorescent labeling of mouse brain tissue sections is also presented.
Collapse
Affiliation(s)
- Moises Freitas-Andrade
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
33
|
Keifer OP, O'Connor DM, Boulis NM. Gene and protein therapies utilizing VEGF for ALS. Pharmacol Ther 2013; 141:261-71. [PMID: 24177067 DOI: 10.1016/j.pharmthera.2013.10.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/04/2013] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that is usually fatal within 2-5years. Unfortunately, the only treatment currently available is riluzole, which has a limited efficacy. As a redress, there is an expanding literature focusing on other potential treatments. One such potential treatment option utilizes the vascular endothelial growth factor (VEGF) family, which includes factors that are primarily associated with angiogenesis but are now increasingly recognized to have neurotrophic effects. Reduced expression of a member of this family, VEGF-A, in mice results in neurodegeneration similar to that of ALS, while treatment of animal models of ALS with either VEGF-A gene therapy or VEGF-A protein has yielded positive therapeutic outcomes. These basic research findings raise the potential for a VEGF therapy to be translated to the clinic for the treatment of ALS. This review covers the VEGF family, its receptors and neurotrophic effects as well as VEGF therapy in animal models of ALS and advances towards clinical trials.
Collapse
Affiliation(s)
- Orion P Keifer
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States
| | - Deirdre M O'Connor
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, United States.
| |
Collapse
|
34
|
Neuropilin-1 modulates vascular endothelial growth factor-induced poly(ADP-ribose)-polymerase leading to reduced cerebrovascular apoptosis. Neurobiol Dis 2013; 59:111-25. [PMID: 23816753 DOI: 10.1016/j.nbd.2013.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 05/21/2013] [Accepted: 06/15/2013] [Indexed: 01/13/2023] Open
Abstract
Cerebral ischemia is encompassed by cerebrovascular apoptosis, yet the mechanisms behind apoptosis regulation are not fully understood. We previously demonstrated inhibition of endothelial apoptosis by vascular endothelial growth factor (VEGF) through upregulation of poly(ADP-ribose)-polymerase (PARP) expression. However, PARP overactivation through oxidative stress can lead to necrosis. This study tested the hypothesis that neuropilin-1 (NP-1), an alternative VEGF receptor, regulates the response to cerebral ischemia by modulating PARP expression and, in turn, apoptosis inhibition by VEGF. In endothelial cell culture, NP-1 colocalized with VEGF receptor-2 (VEGFR-2) and acted as its coreceptor. This significantly enhanced VEGF-induced PARP mRNA and protein expression demonstrated by receptor-specific inhibitors and VEGF-A isoforms. NP-1 augmented the inhibitory effect of VEGF/VEGFR-2 interaction on apoptosis induced by adhesion inhibition through the αV-integrin inhibitor cRGDfV. NP-1/VEGFR-2 signal transduction involved JNK and Akt. In rat models of permanent and temporary middle cerebral artery occlusion, the ischemic cerebral hemispheres displayed endothelial and neuronal apoptosis next to increased endothelial NP-1 and VEGFR-2 expression compared to non-ischemic cerebral hemispheres, sham-operated or untreated controls. Increased vascular superoxide dismutase-1 and catalase expression as well as decreased glycogen reserves indicated oxidative stress in the ischemic brain. Of note, protein levels of intact PARP remained stable despite pro-apoptotic conditions through increased PARP mRNA production during cerebral ischemia. In conclusion, NP-1 is upregulated in conditions of imminent cerebrovascular apoptosis to reinforce apoptosis inhibition and modulate VEGF-dependent PARP expression and activation. We propose that NP-1 is a key modulator of VEGF maintaining cerebrovascular integrity during ischemia. Modulating the function of NP-1 to target PARP could help to prevent cellular damage in cerebrovascular disease.
Collapse
|
35
|
Wittko-Schneider IM, Schneider FT, Plate KH. Brain homeostasis: VEGF receptor 1 and 2-two unequal brothers in mind. Cell Mol Life Sci 2013; 70:1705-25. [PMID: 23475067 PMCID: PMC3632714 DOI: 10.1007/s00018-013-1279-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factors (VEGFs), initially thought to act specifically on the vascular system, exert trophic effects on neural cells during development and adulthood. Therefore, the VEGF system serves as a promising therapeutic target for brain pathologies, but its simultaneous action on vascular cells paves the way for harmful side effects. To circumvent these deleterious effects, many studies have aimed to clarify whether VEGFs directly affect neural cells or if the effects are mediated secondarily via other cell types, like vascular cells. A great number of reports have shown the expression and function of VEGF receptors (VEGFRs), mainly VEGFR-1 and -2, in neural cells, where VEGFR-2 has been described as the major mediator of VEGF-A signals. This review aims to summarize and compare the divergent roles of VEGFR-1 and -2 during CNS development and homeostasis.
Collapse
Affiliation(s)
- Ina M Wittko-Schneider
- Neuroscience Center, Institute of Neurology (Edinger Institute), Goethe University Medical School, Heinrich-Hoffmann Strasse 7, 60528, Frankfurt, Germany.
| | | | | |
Collapse
|
36
|
Koyama Y, Maebara Y, Hayashi M, Nagae R, Tokuyama S, Michinaga S. Endothelins reciprocally regulate VEGF-A and angiopoietin-1 production in cultured rat astrocytes: implications on astrocytic proliferation. Glia 2012; 60:1954-63. [PMID: 22927341 DOI: 10.1002/glia.22411] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Revised: 07/11/2012] [Accepted: 07/31/2012] [Indexed: 11/07/2022]
Abstract
Vascular endothelial growth factors (VEGFs) and angiopoietins (ANGs) are involved in pathophysiological responses in damaged nerve tissues. Astrocytes produce VEGFs and ANGs upon brain ischemia and traumatic injury. To clarify the extracellular signals regulating VEGF and ANG production, effects of endothelins (ETs), a family of endothelium-derived peptides, were examined in cultured rat astrocytes. ET-1 (100 nM) and Ala(1,3,11,15)-ET-1 (100 nM), an ET(B) receptor agonist, increased VEGF-A mRNA levels in cultured astrocytes, while ANG-1 mRNA was decreased by ETs. ET-1 did not affect astrocytic VEGF-B, placental growth factor (PLGF), and ANG-2 mRNA levels. The effects of ET-1 on VEGF-A and ANG-1 mRNAs were inhibited by BQ788, an ET(B) antagonist. Release of VEGF-A proteins from cultured astrocytes was increased by ET-1. In contrast, ET-1 reduced release of astrocytic ANG-1. Exogenous ET-1 (100 nM) and VEGF(165) (100 ng/mL), an isopeptide of VEGF-A, stimulated bromodeoxyuridine (BrdU) incorporation into cultured astrocytes. Treatment with ET-1 and VEGF(165) increased the numbers of cyclin D1-positive astrocytes. Exogenous ANG-1 (250 ng/mL) did not stimulate the BrdU incorporation. Increases in BrdU incorporation by ET-1 and VEGF(165) were not affected by ANG-1. In 60-70% confluent cultures, SU4312 (10 μM), a VEGF receptor tyrosine kinase inhibitor, partially reduced the effects of ET-1 on BrdU incorporation and cyclin D1 expression. ET-induced BrdU incorporation and cyclin D1 expression were reduced by a neutralizing antibody against VEGF-A. Our findings suggest that ET-1 is a factor regulating astrocytic VEGF-A and ANG-1, and that increased VEGF-A production potentiates ET-induced astrocytic proliferation by an autocrine mechanism.
Collapse
Affiliation(s)
- Yutaka Koyama
- Faculty of Pharmacy, Laboratory of Pharmacology, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Dewerchin M, Carmeliet P. PlGF: a multitasking cytokine with disease-restricted activity. Cold Spring Harb Perspect Med 2012; 2:cshperspect.a011056. [PMID: 22908198 DOI: 10.1101/cshperspect.a011056] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family that also comprises VEGF-A (VEGF), VEGF-B, VEGF-C, and VEGF-D. Unlike VEGF, PlGF is dispensable for development and health but has diverse nonredundant roles in tissue ischemia, malignancy, inflammation, and multiple other diseases. Genetic and pharmacological gain-of-function and loss-of-function studies have identified molecular mechanisms of this multitasking cytokine and characterized the therapeutic potential of delivering or blocking PlGF for various disorders.
Collapse
Affiliation(s)
- Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular Link, VIB Vesalius Research Center, K.U. Leuven, Leuven, Belgium
| | | |
Collapse
|
38
|
Ma Y, Zechariah A, Qu Y, Hermann DM. Effects of vascular endothelial growth factor in ischemic stroke. J Neurosci Res 2012; 90:1873-82. [DOI: 10.1002/jnr.23088] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 04/03/2012] [Accepted: 04/20/2012] [Indexed: 12/14/2022]
|
39
|
Abstract
In this study, we have investigated the potential role of placental growth factor (PlGF) in hypoxia-induced brain angiogenesis. To this end, PlGF wild-type (PlGF(+/+)) and PlGF knockout (PlGF(-/-)) mice were exposed to whole body hypoxia (10% oxygen) for 7, 14, and 21 days. PlGF(+/+) animals exhibited a significant ~40% increase in angiogenesis after 7 days of hypoxia compared with controls, while in PlGF(-/-) this effect only occurred after 14 days of hypoxia. No differences in pericyte/smooth muscle cell (SMC) coverage between the two genotypes were observed. After 14 days of hypoxia, PlGF(-/-) microvessels had a significant increase in fibrinogen accumulation and extravasation compared with those of PlGF(+/+), which correlated with endothelial cell disruption of the tight junction protein claudin-5. These vessels displayed large lumens, were surrounded by reactive astrocytes, lacked both pericyte/SMC coverage and endothelial vascular endothelial growth factor expression, and regressed after 21 days of hypoxia. Vascular endothelial growth factor expression levels were found to be significantly lower in the frontal cortex of PlGF(-/-) compared with those in PlGF(+/+) animals during the first 5 days of hypoxia, which in combination with the lack of PlGF may have contributed to the delayed angiogenic response and the prothrombotic phenotype observed in the PlGF(-/-)animals.
Collapse
|
40
|
Sentilhes L, Marret S, Leroux P, Gonzalez BJ, Laquerrière A. Vascular-endothelial growth factor and its high affinity receptor VEGFR-2 in the normal versus destructive lesions human forebrain during development: an immuno-histochemical comparative study. Brain Res 2012; 1385:77-86. [PMID: 21303671 DOI: 10.1016/j.brainres.2011.01.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 12/28/2010] [Accepted: 01/31/2011] [Indexed: 12/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic inducer and neurotrophic factor both in adult and neonatal animal models. In the destructive lesions of the developing human brain, the role and expression of VEGF and of its mitogenic receptor VEGFR-2 have been hardly studied. The aim of the present work was to determine the immunohistochemical distribution of VEGF and VEGFR-2 in premature and full-term infants presenting with hypoxic/ischemic lesions, and to compare results with normal infant brains at similar developmental stages. Paraffin embedded brain tissue samples were assessed using anti-human VEGF and VEGFR-2 antibodies. In all undamaged forebrain areas, VEGF and VEGFR-2 displayed same expression patterns in control and pathologic brains, whatever the destructive lesion occurrence's time (before 25 weeks of gestation (WG), between 25 and 34WG, perinatal period and infancy). In the destructive lesions, VEGF was always expressed in neurons, astrocytes and in neovessel walls, contrary to VEGFR-2 which was only expressed in dispersed astrocytes. VEGF was expressed in oligodendrocytes of prenatally damaged brains, whereas VEGF was expressed in these cells in undamaged areas from birth only, similarly to control brains. These data suggest that VEGF plays specific roles and may not be mediated by VEGFR-2 in human forebrain structures exposed to ischemia.
Collapse
Affiliation(s)
- Loïc Sentilhes
- EA 4309 Neovasc, Rouen Institute for Medical Research and Innovation, School of Medicine, University of Rouen, Normandy, France
| | | | | | | | | |
Collapse
|
41
|
I.c.v administration of an endothelin ETB receptor agonist stimulates vascular endothelial growth factor-A production and activates vascular endothelial growth factor receptors in rat brain. Neuroscience 2011; 192:689-98. [DOI: 10.1016/j.neuroscience.2011.05.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 05/25/2011] [Accepted: 05/25/2011] [Indexed: 11/17/2022]
|
42
|
Grandclement C, Borg C. Neuropilins: a new target for cancer therapy. Cancers (Basel) 2011; 3:1899-928. [PMID: 24212788 PMCID: PMC3757396 DOI: 10.3390/cancers3021899] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 03/23/2011] [Accepted: 04/01/2011] [Indexed: 02/07/2023] Open
Abstract
Recent investigations highlighted strong similarities between neural crest migration during embryogenesis and metastatic processes. Indeed, some families of axon guidance molecules were also reported to participate in cancer invasion: plexins/semaphorins/neuropilins, ephrins/Eph receptors, netrin/DCC/UNC5. Neuropilins (NRPs) are transmembrane non tyrosine-kinase glycoproteins first identified as receptors for class-3 semaphorins. They are particularly involved in neural crest migration and axonal growth during development of the nervous system. Since many types of tumor and endothelial cells express NRP receptors, various soluble molecules were also found to interact with these receptors to modulate cancer progression. Among them, angiogenic factors belonging to the Vascular Endothelial Growth Factor (VEGF) family seem to be responsible for NRP-related angiogenesis. Because NRPs expression is often upregulated in cancer tissues and correlated with poor prognosis, NRPs expression might be considered as a prognostic factor. While NRP1 was intensively studied for many years and identified as an attractive angiogenesis target for cancer therapy, the NRP2 signaling pathway has just recently been studied. Although NRP genes share 44% homology, differences in their expression patterns, ligands specificities and signaling pathways were observed. Indeed, NRP2 may regulate tumor progression by several concurrent mechanisms, not only angiogenesis but lymphangiogenesis, epithelial-mesenchymal transition and metastasis. In view of their multiples functions in cancer promotion, NRPs fulfill all the criteria of a therapeutic target for innovative anti-tumor therapies. This review focuses on NRP-specific roles in tumor progression.
Collapse
Affiliation(s)
- Camille Grandclement
- INSERM UMR 645, F-25020 Besançon, France; E-Mail:
- University of Franche-Comté, IFR133, F-25020 Besançon, France
- EFS Bourgogne Franche-Comté, F-25020 Besançon, France
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-3-81-61-56-15 or +33-3-81-66-93-21; Fax: +33-3-81-61-56-17
| | - Christophe Borg
- INSERM UMR 645, F-25020 Besançon, France; E-Mail:
- University of Franche-Comté, IFR133, F-25020 Besançon, France
- EFS Bourgogne Franche-Comté, F-25020 Besançon, France
- Department of Medical Oncology, CHU Besançon, F-25000 Besançon, France
| |
Collapse
|
43
|
Johnson EC, Doser TA, Cepurna WO, Dyck JA, Jia L, Guo Y, Lambert WS, Morrison JC. Cell proliferation and interleukin-6-type cytokine signaling are implicated by gene expression responses in early optic nerve head injury in rat glaucoma. Invest Ophthalmol Vis Sci 2011; 52:504-18. [PMID: 20847120 DOI: 10.1167/iovs.10-5317] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE In glaucoma, the optic nerve head (ONH) is the principal site of initial axonal injury, and elevated intraocular pressure (IOP) is the predominant risk factor. However, the initial responses of the ONH to elevated IOP are unknown. Here the authors use a rat glaucoma model to characterize ONH gene expression changes associated with early optic nerve injury. METHODS Unilateral IOP elevation was produced in rats by episcleral vein injection of hypertonic saline. ONH mRNA was extracted, and retrobulbar optic nerve cross-sections were graded for axonal degeneration. Gene expression was determined by microarray and quantitative PCR (QPCR) analysis. Significantly altered gene expression was determined by multiclass analysis and ANOVA. DAVID gene ontology determined the functional categories of significantly affected genes. RESULTS The Early Injury group consisted of ONH from eyes with <15% axon degeneration. By array analysis, 877 genes were significantly regulated in this group. The most significant upregulated gene categories were cell cycle, cytoskeleton, and immune system process, whereas the downregulated categories included glucose and lipid metabolism. QPCR confirmed the upregulation of cell cycle-associated genes and leukemia inhibitory factor (Lif) and revealed alterations in expression of other IL-6-type cytokines and Jak-Stat signaling pathway components, including increased expression of IL-6 (1553%). In contrast, astrocytic glial fibrillary acidic protein (Gfap) message levels were unaltered, and other astrocytic markers were significantly downregulated. Microglial activation and vascular-associated gene responses were identified. CONCLUSIONS Cell proliferation and IL-6-type cytokine gene expression, rather than astrocyte hypertrophy, characterize early pressure-induced ONH injury.
Collapse
Affiliation(s)
- Elaine C Johnson
- Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon 97201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Chaballe L, Close P, Sempels M, Delstanche S, Fanielle J, Moons L, Carmeliet P, Schoenen J, Chariot A, Franzen R. Involvement of placental growth factor in Wallerian degeneration. Glia 2010; 59:379-96. [PMID: 21264946 DOI: 10.1002/glia.21108] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 10/18/2010] [Indexed: 01/13/2023]
Abstract
Wallerian degeneration (WD) is an inflammatory process of nerve degeneration, which occurs more rapidly in the peripheral nervous system compared with the central nervous system, resulting, respectively in successful and aborted axon regeneration. In the peripheral nervous system, Schwann cells (SCs) and macrophages, under the control of a network of cytokines and chemokines, represent the main cell types involved in this process. Within this network, the role of placental growth factor (PlGF) remains totally unknown. However, properties like monocyte activation/attraction, ability to increase expression of pro-inflammatory molecules, as well as neuroprotective effects, make it a candidate likely implicated in this process. Also, nothing is described about the expression and localization of this molecule in the peripheral nervous system. To address these original questions, we decided to study PlGF expression under physiological and degenerative conditions and to explore its role in WD, using a model of sciatic nerve transection in wild-type and Pgf(-/-) mice. Our data show dynamic changes of PlGF expression, from periaxonal in normal nerve to SCs 24h postinjury, in parallel with a p65/NF-κB recruitment on Pgf promoter. After injury, SC proliferation is reduced by 30% in absence of PlGF. Macrophage invasion is significantly delayed in Pgf(-/-) mice compared with wild-type mice, which results in worse functional recovery. MCP-1 and proMMP-9 exhibit a 3-fold reduction of their relative expressions in Pgf(-/-) injured nerves, as demonstrated by cytokine array. In conclusion, this work originally describes PlGF as a novel member of the cytokine network of WD.
Collapse
Affiliation(s)
- Linda Chaballe
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain unit, University of Liege, Avenue de l'Hopital, Liege, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Minor KH, Bournat JC, Toscano N, Giger RJ, Davies SJA. Decorin, erythroblastic leukaemia viral oncogene homologue B4 and signal transducer and activator of transcription 3 regulation of semaphorin 3A in central nervous system scar tissue. ACTA ACUST UNITED AC 2010; 134:1140-55. [PMID: 21115466 DOI: 10.1093/brain/awq304] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Scar tissue at sites of traumatic injury in the adult central nervous system presents a combined physical and molecular impediment to axon regeneration. Of multiple known central nervous system scar associated axon growth inhibitors, semaphorin 3A has been shown to be strongly expressed by invading leptomeningeal fibroblasts. We have previously demonstrated that infusion of the small leucine-rich proteoglycan decorin results in major suppression of several growth inhibitory chondroitin sulphate proteoglycans and growth of adult sensory axons across acute spinal cord injuries. Furthermore, decorin treatment of leptomeningeal fibroblasts significantly increases their ability to support neurite growth of co-cultured adult dorsal root ganglion neurons. In the present study we show that decorin has the ability to suppress semaphorin 3A expression within adult rat cerebral cortex scar tissue and in primary leptomeningeal fibroblasts in vitro. Infusion of decorin core protein for eight days resulted in a significant reduction of semaphorin 3A messenger RNA expression within injury sites compared with saline-treated control animals. Both in situ hybridization and immunostaining confirmed that semaphorin 3A messenger RNA expression and protein levels are significantly reduced in decorin-treated animals. Similarly, decorin treatment decreased the expression of semaphorin 3A messenger RNA in cultured rat leptomeningeal fibroblasts compared with untreated cells. Mechanistic studies revealed that decorin-mediated suppression of semaphorin 3A critically depends on erythroblastic leukaemia viral oncogene homologue B4 and signal transducer and activator of transcription 3 function. Collectively, our studies show that in addition to suppressing the levels of inhibitory chondroitin sulphate proteoglycans, decorin has the ability to suppress semaphorin 3A in the injured central nervous system. Our findings provide further evidence for the use of decorin as a potential therapy for promoting axonal growth and repair in the injured adult mammalian brain and spinal cord.
Collapse
Affiliation(s)
- Kenneth H Minor
- Department of Neurosurgery, University of Colorado at Denver, Aurora, CO 80045, USA
| | | | | | | | | |
Collapse
|
46
|
Petcu EB, Smith RA, Miroiu RI, Opris MM. Angiogenesis in old-aged subjects after ischemic stroke: a cautionary note for investigators. JOURNAL OF ANGIOGENESIS RESEARCH 2010; 2:26. [PMID: 21110846 PMCID: PMC3000373 DOI: 10.1186/2040-2384-2-26] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 11/26/2010] [Indexed: 12/12/2022]
Abstract
Angiogenesis represents a form of neovascularisation of exceptional importance in numerous pathological conditions including stroke. In this context it is directly related to neuroregeneration which is seen in close proximity. However, numerous experimental data have been drawn from studies that have ignored the age criterion. This is extremely important as angiogenesis is different in young versus old subjects. Extrapolating data obtained from studies performed in young subjects or "in vitro" to old-age patients could lead to inexact conclusions since the dynamics of angiogenesis is age-dependent. The current review covers the key features of brain senescence including morphological and functional changes related to the brain parenchyma, its vascular network and blood flow which could possibly influence the process of angiogenesis. This is followed by a description of post-stroke angiogenesis and its relationship to neuroregeneration and its modulation by vascular endothelial growth factor (VEGF) and insulin-like growth factor 1 (IGF 1), the most important factors active in old brain after ischemic injury.
Collapse
Affiliation(s)
- Eugen B Petcu
- Griffith University School of Medicine, Gold Coast Campus, Griffith University, QLD 4222, Australia.
| | | | | | | |
Collapse
|
47
|
VEGFR-1 signaling regulates the homing of bone marrow-derived cells in a mouse stroke model. J Neuropathol Exp Neurol 2010; 69:168-75. [PMID: 20084017 DOI: 10.1097/nen.0b013e3181c9c05b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular endothelial growth factor receptor 1 (VEGFR-1) is highly expressed in endothelial cells and regulates developmental angiogenesis by acting as a decoy receptor and trapping VEGF-A. Vascular endothelial growth factor receptor 1 is also expressed in monocytes and macrophages; mice lacking the VEGFR-1 tyrosine kinase (TK) domain (VEGFR-1 TK mice) display impaired macrophage function. Because macrophages are recruited to sites of cerebral ischemic infarcts, we hypothesized that lack of VEGFR-1 TK in bone marrow(BM) cells would affect the outcome in an experimental stroke model. We performed BM transplantation experiments in C57BL/6J mice using VEGFR-1 TK and VEGFR-1 TK mice as BM donors and analyzed cell infiltration after cerebral ischemia. There was reduced initial recruitment of VEGFR-1 TK myeloid cells into the infarcted tissue and reduced postischemic angiogenesis at 3days postischemia. By 10 days, the numbers of infiltrating cells and the densities of vessels in the infarct peri-infarct zone were similar for both groups. Neither infarct size at 3 and 10 days postischemia nor neurological performance at 24 hours was different between the experimental groups. These results support a role of VEGFR-1 signaling in the early regulation of BM infiltration and angiogenesis after brain ischemia.
Collapse
|
48
|
Jiang SX, Whitehead S, Aylsworth A, Slinn J, Zurakowski B, Chan K, Li J, Hou ST. Neuropilin 1 directly interacts with Fer kinase to mediate semaphorin 3A-induced death of cortical neurons. J Biol Chem 2010; 285:9908-9918. [PMID: 20133938 DOI: 10.1074/jbc.m109.080689] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Neuropilins (NRPs) are receptors for the major chemorepulsive axonal guidance cue semaphorins (Sema). The interaction of Sema3A/NRP1 during development leads to the collapse of growth cones. Here we show that Sema3A also induces death of cultured cortical neurons through NRP1. A specific NRP1 inhibitory peptide ameliorated Sema3A-evoked cortical axonal retraction and neuronal death. Moreover, Sema3A was also involved in cerebral ischemia-induced neuronal death. Expression levels of Sema3A and NRP1, but not NRP2, were significantly increased early during brain reperfusion following transient focal cerebral ischemia. NRP1 inhibitory peptide delivered to the ischemic brain was potently neuroprotective and prevented the loss of motor functions in mice. The integrity of the injected NRP1 inhibitory peptide into the brain remained unchanged, and the intact peptide permeated the ischemic hemisphere of the brain as determined using MALDI-MS-based imaging. Mechanistically, NRP1-mediated axonal collapse and neuronal death is through direct and selective interaction with the cytoplasmic tyrosine kinase Fer. Fer RNA interference effectively attenuated Sema3A-induced neurite retraction and neuronal death in cortical neurons. More importantly, down-regulation of Fer expression using Fer-specific RNA interference attenuated cerebral ischemia-induced brain damage. Together, these studies revealed a previously unknown function of NRP1 in signaling Sema3A-evoked neuronal death through Fer in cortical neurons.
Collapse
Affiliation(s)
- Susan X Jiang
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada
| | - Shawn Whitehead
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada
| | - Amy Aylsworth
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario K1H 8M5, Canada
| | - Jacqueline Slinn
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada
| | - Bogdan Zurakowski
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada
| | - Kenneth Chan
- Mass Spectrometry Glycoanalysis Laboratory, National Research Council (NRC) Institute for Biological Sciences, NRC Canada, Ottawa, Ontario K1A 0R6, Canada
| | - Jianjun Li
- Mass Spectrometry Glycoanalysis Laboratory, National Research Council (NRC) Institute for Biological Sciences, NRC Canada, Ottawa, Ontario K1A 0R6, Canada
| | - Sheng T Hou
- Experimental NeuroTherapeutics Laboratory, Ottawa, Ontario K1A 0R6, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
49
|
Hou ST, Jiang SX, Slinn J, O'Hare M, Karchewski L. Neuropilin 2 deficiency does not affect cortical neuronal viability in response to oxygen-glucose-deprivation and transient middle cerebral artery occlusion. Neurosci Res 2009; 66:396-401. [PMID: 20036291 DOI: 10.1016/j.neures.2009.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 12/01/2009] [Accepted: 12/16/2009] [Indexed: 12/01/2022]
Abstract
Neuropilin 2 (NRP2) is a type I transmembrane protein that binds to distinct members of the class III secreted Semaphorin subfamily. NRP2 plays important roles in repulsive axon guidance, angiogenesis and vasculogenesis through partnering with co-receptors such as vascular endothelial growth factor receptors (VEGFRs) during development. Emerging evidence also suggests that NRP2 contributes to injury response and environment changes in adult brains. In this study, we examined the contribution of NRP2 gene to cerebral ischemia-induced brain injury using NRP2 deficient mouse. To our surprise, the lack of NRP2 expression does not affect the outcome of brain injury induced by transient occlusion of the middle cerebral artery (MCAO) in mouse. The cerebral vasculature in terms of the middle cerebral artery anatomy and microvessel density in the cerebral cortex of NRP2 deficient homozygous (NRP2(-/-)) mice are normal and almost identical to those of the heterozygous (NRP2(+/-)) and wild type (NRP2(+/+)) littermates. MCAO (1h) and 24h reperfusion caused a brain infarction of 23% (compared to the contralateral side) in NRP2(-/-) mice, which is not different from those in NRP2(+/- and +/+) mice at 22 and 21%, respectively (n=19, p>0.05). Correspondingly, NRP2(-/-) mouse also showed a similar level of deterioration of neurological functions after stroke compared with their NRP2(+/- and +/+) littermates. Oxygen-glucose-deprivation (OGD) caused a significant neuronal death in NRP2(-/-) cortical neurons, at the level similar to that in NRP(+/+) cortical neurons (72% death in NRP(-/-) neurons vs. 75% death in NRP2(+/+) neurons; n=4; p>0.05). Together, these loss-of-function studies demonstrated that despite of its critical role in neuronal guidance and vascular formation during development, NRP2 expression dose not affect adult brain response to cerebral ischemia.
Collapse
Affiliation(s)
- Sheng T Hou
- Experimental NeuroTherapeutics Laboratory, NRC Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Road, Bldg M-54, Ottawa, ON, Canada K1A 0R6.
| | | | | | | | | |
Collapse
|
50
|
Implications of vascular endothelial growth factor for postischemic neurovascular remodeling. J Cereb Blood Flow Metab 2009; 29:1620-43. [PMID: 19654590 DOI: 10.1038/jcbfm.2009.100] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurovascular remodeling has been recently recognized as a promising target for neurologic therapies. Hopes have emerged that, by stimulating vessel growth, it may be possible to stabilize brain perfusion, and at the same time promote neuronal survival, brain plasticity, and neurologic recovery. In this review, we outline the role of vascular endothelial growth factor (VEGF) in the ischemic brain, analyzing how this growth factor contributes to brain remodeling. Studies with therapeutic VEGF administration resulted in quite variable results depending on the route and time point of delivery. Local VEGF administration consistently enhanced neurologic recovery, whereas acute intravenous delivery exacerbated brain infarcts due to enhanced brain edema. Future studies should answer the following questions: (1) whether increased vessel density translates into improvements in blood flow in the hemodynamically compromised brain; (2) how VEGF influences brain plasticity and contributes to motor and nonmotor recovery; (3) what are the actions of VEGF not only in young animals with preserved vasculature, on which previous studies have been conducted, but also in aged animals and in animals with preexisting atherosclerosis; and (4) whether the effects of VEGF can be mimicked by pharmacological compounds or by cell-based therapies. Only on the basis of such information can more definite conclusions be made with regard to whether the translation of therapeutic angiogenesis into clinics is promising.
Collapse
|