1
|
Sun R, Peng Q, Zhang F, Gao H, Li T, Wang L, Zhang L. Effect of vascular endothelial growth factor 165 on dopamine level in the retinas of guinea pigs with form-deprivation myopia. PeerJ 2023; 11:e16255. [PMID: 37849827 PMCID: PMC10578302 DOI: 10.7717/peerj.16255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Background Myopia is the most common refractive error because excessive increase in the axial length of a myopic eye leads to the thinning of the posterior scleral pole and can cause serious complications resulting in blindness. Thus, myopia has become a great concern worldwide. Dopamine (DA) plays a role in the development of myopia. Moreover, in Parkinson's disease, it has been proved that vascular endothelial growth factor 165 (VEGF165) can promote the survival and recovery of DA neurons, resulting in increased DA secretion in the striatum, thereby treating neuropathy. Therefore, we speculate that VEGF165 can also promote the release of DA in the retina to inhibit the occurrence and development of myopia. We aimed to investigate the effect of VEGF165 on DA levels in the retinas of guinea pigs with form-deprivation myopia (FDM) and the effects of DA on myopia prevention and control. Methods Healthy 3-week-old pigmented guinea pigs were randomly divided into blank, FDM, phosphate buffer saline (PBS), 1, 5, and 10 ng groups. The FDM model was established by covering the right eye continuously with a translucent latex balloon pullover for 14 days. The pigs in the PBS, 1, 5, and 10 ng groups were injected with PBS buffer and 1, 5, and 10 ng of VEGF165 recombinant human protein, respectively, in the vitreous of the right eye before masking. The refractive error and axial length were measured before and after modeling. All retinas were used for biomolecular analyses after 14 days. Results We found that the intravitreal injection of VEGF165 elevated DA levels in the retina and was effective in slowing the progression of myopia, and 1 ng of VEGF165 was the most effective. Moreover, the number of vascular endothelial cell nuclei in the 1 ng group was lower than that in the other VEGF165 groups. Conclusions Our data suggest that VEGF165 has a promoting effect on DA in the retinas of guinea pigs with FDM, potentially controlling the development of myopia.
Collapse
Affiliation(s)
- Ruiting Sun
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Qingsheng Peng
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fengyi Zhang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Honglian Gao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Tong Li
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Lei Wang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Lei Zhang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| |
Collapse
|
2
|
Wang Y, Yu S, Li M. Neurovascular crosstalk and cerebrovascular alterations: an underestimated therapeutic target in autism spectrum disorders. Front Cell Neurosci 2023; 17:1226580. [PMID: 37692552 PMCID: PMC10491023 DOI: 10.3389/fncel.2023.1226580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Normal brain development, function, and aging critically depend on unique characteristics of the cerebrovascular system. Growing evidence indicated that cerebrovascular defects can have irreversible effects on the brain, and these defects have been implicated in various neurological disorders, including autism spectrum disorder (ASD). ASD is a neurodevelopmental disorder with heterogeneous clinical manifestations and anatomical changes. While extensive research has focused on the neural abnormalities underlying ASD, the role of brain vasculature in this disorder remains poorly understood. Indeed, the significance of cerebrovascular contributions to ASD has been consistently underestimated. In this work, we discuss the neurovascular crosstalk during embryonic development and highlight recent findings on cerebrovascular alterations in individuals with ASD. We also discuss the potential of vascular-based therapy for ASD. Collectively, these investigations demonstrate that ASD can be considered a neurovascular disease.
Collapse
Affiliation(s)
- Yiran Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shunyu Yu
- Department of Psychosomatic Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Mengqian Li
- Department of Psychosomatic Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
The Active Role of Pericytes During Neuroinflammation in the Adult Brain. Cell Mol Neurobiol 2023; 43:525-541. [PMID: 35195811 DOI: 10.1007/s10571-022-01208-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.
Collapse
|
4
|
Bairwa SC, Shaw CA, Kuo M, Yoo J, Tomljenovic L, Eidi H. Cytokines profile in neonatal and adult wild-type mice post-injection of U. S. pediatric vaccination schedule. Brain Behav Immun Health 2021; 15:100267. [PMID: 34589773 PMCID: PMC8474652 DOI: 10.1016/j.bbih.2021.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 11/09/2022] Open
Abstract
Introduction A recent study from our laboratory demonstrated a number of neurobehavioral abnormalities in mice colony injected with a mouse-weight equivalent dose of all vaccines that are administered to infants in their first 18 months of life according to the U. S. pediatric vaccination schedule. Cytokines have been studied extensively as blood immune and inflammatory biomarkers, and their association with neurodevelopmental disorders. Given the importance of cytokines in early neurodevelopment, we aimed to investigate the potential post-administration effects of the U. S. pediatric vaccines on circulatory cytokines in a mouse model. In the current study, cytokines have been assayed at early and late time points in mice vaccinated early in postnatal life and compared with placebo controls. Materials and methods Newborn mouse pups were divided into three groups: i) vaccine (V1), ii) vaccine × 3 (V3) and iii) placebo control. V1 group was injected with mouse weight-equivalent of the current U. S. pediatric vaccine schedule. V3 group was injected with same vaccines but at triple the dose and the placebo control was injected with saline. Pups were also divided according to the sampling age into two main groups: acute- and chronic-phase group. Blood samples were collected at postnatal day (PND) 23, two days following vaccine schedule for the acute-phase group or at 67 weeks post-vaccination for the chronic-phase groups. Fifteen cytokines were analyzed: GM-CSF, IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12p70, IL-13, IL-17A, MCP-1, TNF-α, and VEGF-A. Wilcoxon Rank Sum test or unpaired Student's t-test was performed where applicable. Results IL-5 levels in plasma were significantly elevated in the V1 and V3 group compared with the control only in the acute-phase group. The elevation of IL-5 levels in the two vaccine groups were significant irrespective of whether the sexes were combined or analyzed separately. Other cytokines (VEGF-A, TNF-α, IL-10, MCP-1, GM-CSF, IL-6, and IL-13) were also impacted, although to a lesser extent and in a sex-dependent manner. In the acute-phase group, females showed a significant increase in IL-10 and MCP-1 levels and a decrease in VEGF-A levels in both V1 and V3 group compared to controls. In the acute-phase, a significant increase in MCP-1 levels in V3 group and CM-CSF levels in V1 and V3 group and decrease in TNF-α levels in V1 group were observed in treated males as compared with controls. In chronic-phase females, levels of VEGF-A in V1 and V3 group, TNF-α in V3 group, and IL-13 in V1 group were significantly decreased in contrast with controls. In chronic-phase males, TNF-α levels were significantly increased in V1 group and IL-6 levels decreased in V3 group in comparison to controls. The changes in levels of most tested cytokines were altered between the early and the late postnatal assays. Conclusions IL-5 levels significantly increased in the acute-phase of the treatment in the plasma of both sexes that were subjected to V1 and V3 injections. These increases had diminished by the second test assayed at week 67. These results suggest that a profound, albeit transient, effect on cytokine levels may be induced by the whole vaccine administration supporting our recently published observations regarding the behavioral abnormalities in the same mice. These observations support the view that the administration of whole pediatric vaccines in a neonatal period may impact at least short-term CNS functions in mice.
Collapse
Affiliation(s)
- S C Bairwa
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - C A Shaw
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada.,Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada.,Program in Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - M Kuo
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Yoo
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Tomljenovic
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - H Eidi
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,French Agency for Veterinary Medicinal Products (ANMV) - French Agency for Food, Environmental and Occupational Health Safety (ANSES), Fougères, France
| |
Collapse
|
5
|
Saleki K, Banazadeh M, Miri NS, Azadmehr A. Triangle of cytokine storm, central nervous system involvement, and viral infection in COVID-19: the role of sFasL and neuropilin-1. Rev Neurosci 2021; 33:147-160. [PMID: 34225390 DOI: 10.1515/revneuro-2021-0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) is identified as the cause of coronavirus disease 2019 (COVID-19), and is often linked to extreme inflammatory responses by over activation of neutrophil extracellular traps (NETs), cytokine storm, and sepsis. These are robust causes for multi-organ damage. In particular, potential routes of SARS-CoV2 entry, such as angiotensin-converting enzyme 2 (ACE2), have been linked to central nervous system (CNS) involvement. CNS has been recognized as one of the most susceptible compartments to cytokine storm, which can be affected by neuropilin-1 (NRP-1). ACE2 is widely-recognized as a SARS-CoV2 entry pathway; However, NRP-1 has been recently introduced as a novel path of viral entry. Apoptosis of cells invaded by this virus involves Fas receptor-Fas ligand (FasL) signaling; moreover, Fas receptor may function as a controller of inflammation. Furthermore, NRP-1 may influence FasL and modulate cytokine profile. The neuroimmunological insult by SARS-CoV2 infection may be inhibited by therapeutic approaches targeting soluble Fas ligand (sFasL), cytokine storm elements, or related viral entry pathways. In the current review, we explain pivotal players behind the activation of cytokine storm that are associated with vast CNS injury. We also hypothesize that sFasL may affect neuroinflammatory processes and trigger the cytokine storm in COVID-19.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, 76169-13555, Iran
| | - Niloufar Sadat Miri
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| | - Abbas Azadmehr
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- Medical Immunology Department, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| |
Collapse
|
6
|
Abstract
The coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been declared a pandemic. Global research updates confirm that the infected patients manifest a range of clinical symptoms and sometimes remain entirely asymptomatic, posing a greater threat to the people coming in contact. Despite several case reports coming up every day, our knowledge about the neurotropic mechanism of the SARS-CoV-2, immunological responses, and the mode of disease progression and mechanism of cross-talk between the central nervous system (CNS),
heart, lungs, and other major organs is not complete. Report of anosmia, ataxia, dysgeusia, and altered psychological status of the infected COVID-19 patients offers some clue to the possible route of viral entry and multiplication. In this review, we have critically assessed the involvement of CNS dysregulation in COVID-19 patients. The probable mechanism of immunological responses, the impairment of the coagulation pathway, the onset of cytokine storm, its interplay with the HPA axis, and hypoxia are discussed in detail here. Based on the latest research findings and some case reports of hospitalized COVID-19 patients, it is evident that the CNS involvement in disease progression is alarming. Accurate and timely detection of viral load in CNS is necessary to allow prompt and effective treatment modalities. Possible entry sites of SARS-CoV-2 to the central nervous system of human being and the downstream manifestations.![]()
Collapse
|
7
|
Liu SH, Shi XJ, Fan FC, Cheng Y. Peripheral blood neurotrophic factor levels in children with autism spectrum disorder: a meta-analysis. Sci Rep 2021; 11:15. [PMID: 33420109 PMCID: PMC7794512 DOI: 10.1038/s41598-020-79080-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/25/2020] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that abnormal regulation of neurotrophic factors is involved in the etiology and pathogenesis of Autism Spectrum Disorder (ASD). However, clinical data on neurotrophic factor levels in children with ASD were inconsistent. Therefore, we performed a systematic review of peripheral blood neurotrophic factors levels in children with ASD, and quantitatively summarized the clinical data of peripheral blood neurotrophic factors in ASD children and healthy controls. A systematic search of PubMed and Web of Science identified 31 studies with 2627 ASD children and 4418 healthy controls to be included in the meta-analysis. The results of random effect meta-analysis showed that the peripheral blood levels of brain-derived neurotrophic factor (Hedges’ g = 0.302; 95% CI = 0.014 to 0.591; P = 0.040) , nerve growth factor (Hedges’ g = 0.395; 95% CI = 0.104 to 0.686; P = 0.008) and vascular endothelial growth factor (VEGF) (Hedges’ g = 0.097; 95% CI = 0.018 to 0.175; P = 0.016) in children with ASD were significantly higher than that of healthy controls, whereas blood neurotrophin-3 (Hedges’ g = − 0.795; 95% CI = − 1.723 to 0.134; P = 0.093) and neurotrophin-4 (Hedges’ g = 0.182; 95% CI = − 0.285 to 0.650; P = 0.445) levels did not show significant differences between cases and controls. Taken together, these results clarified circulating neurotrophic factor profile in children with ASD, strengthening clinical evidence of neurotrophic factor aberrations in children with ASD.
Collapse
Affiliation(s)
- Shu-Han Liu
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, 27 South Zhongguancun Avenue, Zhongguancun South St, Haidian District, Beijing, 100081, China
| | - Xiao-Jie Shi
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, 27 South Zhongguancun Avenue, Zhongguancun South St, Haidian District, Beijing, 100081, China
| | - Fang-Cheng Fan
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, 27 South Zhongguancun Avenue, Zhongguancun South St, Haidian District, Beijing, 100081, China
| | - Yong Cheng
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, 27 South Zhongguancun Avenue, Zhongguancun South St, Haidian District, Beijing, 100081, China.
| |
Collapse
|
8
|
Zamorano Cuervo N, Grandvaux N. ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. eLife 2020; 9:e61390. [PMID: 33164751 PMCID: PMC7652413 DOI: 10.7554/elife.61390] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus 19 disease (COVID-19) which presents a large spectrum of manifestations with fatal outcomes in vulnerable people over 70-years-old and with hypertension, diabetes, obesity, cardiovascular disease, COPD, and smoking status. Knowledge of the entry receptor is key to understand SARS-CoV-2 tropism, transmission and pathogenesis. Early evidence pointed to angiotensin-converting enzyme 2 (ACE2) as SARS-CoV-2 entry receptor. Here, we provide a critical summary of the current knowledge highlighting the limitations and remaining gaps that need to be addressed to fully characterize ACE2 function in SARS-CoV-2 infection and associated pathogenesis. We also discuss ACE2 expression and potential role in the context of comorbidities associated with poor COVID-19 outcomes. Finally, we discuss the potential co-receptors/attachment factors such as neuropilins, heparan sulfate and sialic acids and the putative alternative receptors, such as CD147 and GRP78.
Collapse
Affiliation(s)
| | - Nathalie Grandvaux
- CRCHUM - Centre Hospitalier de l’Université de MontréalQuébecCanada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de MontréalQuébecCanada
| |
Collapse
|
9
|
Mesquita-Britto MHR, Mendonça MCP, Soares ES, Sakane KK, da Cruz-Höfling MA. Inhibition of VEGF-Flk-1 binding induced profound biochemical alteration in the hippocampus of a rat model of BBB breakdown by spider venom. A preliminary assessment using FT-IR spectroscopy. Neurochem Int 2018; 120:64-74. [PMID: 30075232 DOI: 10.1016/j.neuint.2018.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/05/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022]
Abstract
Phoneutria nigriventer spider venom (PNV) contains ion channels-acting neuropeptides that in rat induces transitory blood-brain barrier breakdown (BBBb) in hippocampus in parallel with VEGF upregulation. We investigated whether VEGF has a neuroprotective role by inhibiting its binding to receptor Flk-1 by itraconazole (ITZ). FT-IR spectroscopy examined the biochemical status of hippocampus and evaluated BBBb in rats administered PNV or ITZ/PNV at periods with greatest toxicity (1-2h), recovery (5h) and visual absence of symptoms (24h), and compared to saline and ITZ controls. The antifungal treatment before venom intoxication aggravated the venom effects and increased BBB damage. FT-IR spectra of venom, hippocampi of controls, PNV and ITZ-PNV showed a 1400 cm-1 band linked to symmetric stretch of carboxylate and 1467 cm-1 band (CH2 bending: mainly lipids) that were considered biomarker and reference bands, respectively. Inhibition of VEGF/Flk-1 binding produced marked changes in lipid/protein stability at 1-2h. The largest differences were observed in spectra regions assigned to lipids, both symmetric (2852 cm-1) and asymmetric (2924 and 2968 cm-1). Quantitative analyses showed greatest increases in the 1400 cm-1/1467 cm-1 ratio also at 1h. Such changes at period of rats' severe intoxication referred to wavenumber region from 3106 cm-1 to 687 cm-1 assigning for C-H and N-H stretching of protein, Amide I, C=N cytosine, N-H adenine, Amide II, CH2 bending: mainly lipids, C-O stretch: glycogen, polysaccharides, glycolipids, z-type DNA, C-C, C-O and CH out-of-plane bending vibrations. We conclude that VEGF has a neuroprotective role and can be a therapeutic target in PNV envenomation. FT-IR spectroscopy showed to be instrumental for monitoring biochemical changes in this model of P. nigriventer venom-induced BBB disruption.
Collapse
Affiliation(s)
- Maria Helena Rodrigues Mesquita-Britto
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Monique Culturato Padilha Mendonça
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Edilene Siqueira Soares
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Kumiko Koibuchi Sakane
- Institute for Research and Development, University of Vale do Paraíba, São José dos Campos, São Paulo, Brazil
| | - Maria Alice da Cruz-Höfling
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
10
|
Navarro Quiroz E, Navarro Quiroz R, Ahmad M, Gomez Escorcia L, Villarreal JL, Fernandez Ponce C, Aroca Martinez G. Cell Signaling in Neuronal Stem Cells. Cells 2018; 7:E75. [PMID: 30011912 PMCID: PMC6070865 DOI: 10.3390/cells7070075] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/30/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
The defining characteristic of neural stem cells (NSCs) is their ability to multiply through symmetric divisions and proliferation, and differentiation by asymmetric divisions, thus giving rise to different types of cells of the central nervous system (CNS). A strict temporal space control of the NSC differentiation is necessary, because its alterations are associated with neurological dysfunctions and, in some cases, death. This work reviews the current state of the molecular mechanisms that regulate the transcription in NSCs, organized according to whether the origin of the stimulus that triggers the molecular cascade in the CNS is internal (intrinsic factors) or whether it is the result of the microenvironment that surrounds the CNS (extrinsic factors).
Collapse
Affiliation(s)
- Elkin Navarro Quiroz
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
- School of Medicine, Universidad Rafael Nuñez, Cartagena 130001, Colombia.
| | - Roberto Navarro Quiroz
- Centro de Investigación en Salud para el Trópico, Universidad Cooperativa de Colombia, Santa Marta 470002, Colombia.
| | - Mostapha Ahmad
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
| | - Lorena Gomez Escorcia
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
| | | | | | - Gustavo Aroca Martinez
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
- Clinica de la Costa, Barranquilla 080002, Colombia.
| |
Collapse
|
11
|
Cui J, Gong C, Cao B, Li L. MicroRNA-27a participates in the pathological process of depression in rats by regulating VEGFA. Exp Ther Med 2018; 15:4349-4355. [PMID: 29731825 PMCID: PMC5921192 DOI: 10.3892/etm.2018.5942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/19/2018] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to determine the expression of vascular endothelial growth factor A (VEGFA) and microRNA (miRNA/miR)-27a in hippocampal tissues, and serum from a depression model of rats. In addition, the present study aimed to understand the mechanism of regulation of miR-27a in depression. A total of 40 male rats were selected, and divided into the control and depression model groups. The rats in the model group were subjected to 14 types of stimulations to model depression. By determining the body weight, syrup consumption rate and open field test score, the extent of depression in the rats was evaluated. Quantitative-polymerase chain reaction was used to determine the expression of VEGFA mRNA and miR-27a in hippocampal tissues, and serum. ELISA was used to measure the content of VEGFA protein in serum, while western blotting was employed to determine the expression of VEGFA protein in hippocampal tissues. A dual luciferase assay was carried out to identify the interactions between VEGFA mRNA and miR-27a. The rats in the depression model group showed depression symptoms and the depression model was successfully constructed. Rats with depression had lower VEGFA mRNA and protein expression in the hippocampus, and peripheral blood compared with the control group. Rats in the depression model group had reduced levels of miR-27a in the hippocampus and peripheral blood, which may be associated with the levels of VEGFA. miR-27a was able to bind with the 3′-untranslated region of VEGFA mRNA to regulate its expression. The present study demonstrated that miR-27a expression in hippocampal tissues and blood from rats with depression is upregulated, while the expression of VEGFA mRNA and protein is downregulated. miR-27a may participate in the pathological process of depression in rats by regulating VEGFA.
Collapse
Affiliation(s)
- Jian Cui
- Department of Psychiatry, Jining Psychiatric Hospital, Jining, Shandong 272051, P.R. China
| | - Cunqi Gong
- Department of Psychiatry, Zaozhuang Mental Health Center, Zaozhuang, Shandong 277100, P.R. China
| | - Baorui Cao
- Department of Psychiatry, Zaozhuang Mental Health Center, Zaozhuang, Shandong 277100, P.R. China
| | - Longfei Li
- Department of Psychiatry, Jining Psychiatric Hospital, Jining, Shandong 272051, P.R. China
| |
Collapse
|
12
|
Axelsen TM, Woldbye DP. Gene Therapy for Parkinson's Disease, An Update. JOURNAL OF PARKINSON'S DISEASE 2018; 8:195-215. [PMID: 29710735 PMCID: PMC6027861 DOI: 10.3233/jpd-181331] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/25/2018] [Indexed: 12/19/2022]
Abstract
The current mainstay treatment of Parkinson's disease (PD) consists of dopamine replacement therapy which, in addition to causing several side effects, does not delay disease progression. The field of gene therapy offers a potential means to improve current therapy. The present review gives an update of the present status of gene therapy for PD. Both non-disease and disease modifying transgenes have been tested for PD gene therapy in animal and human studies. Non-disease modifying treatments targeting dopamine or GABA synthesis have been successful and promising at improving PD symptomatology in randomized clinical studies, but substantial testing remains before these can be implemented in the standard clinical treatment repertoire. As for disease modifying targets that theoretically offer the possibility of slowing the progression of disease, several neurotrophic factors show encouraging results in preclinical models (e.g., neurturin, GDNF, BDNF, CDNF, VEGF-A). However, so far, clinical trials have only tested neurturin, and, unfortunately, no trial has been able to meet its primary endpoint. Future clinical trials with neurotrophic factors clearly deserve to be conducted, considering the still enticing goal of actually slowing the disease process of PD. As alternative types of gene therapy, opto- and chemogenetics might also find future use in PD treatment and novel genome-editing technology could also potentially be applied as individualized gene therapy for genetic types of PD.
Collapse
Affiliation(s)
- Tobias M. Axelsen
- Department of Neurology, Herlev University Hospital, Herlev, Denmark
| | - David P.D. Woldbye
- Department of Neuroscience, Panum Institute, Mærsk Tower, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
13
|
Yasuhara T, Date I. Intracerebral Transplantation of Genetically Engineered Cells for Parkinson's Disease: Toward Clinical Application. Cell Transplant 2017. [DOI: 10.3727/000000007783464632] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Over the last decade, molecular biology has progressively developed, leading to new technology with subsequent clinical application for various cerebral diseases including Parkinson's disease (PD), one of the most investigated neurodegenerative disorders. The therapy for PD is mainly composed of medication, including drug replacement therapy, surgical treatment, and cell transplantation. Cell therapy for PD has been explored by using fetal nigral cells as an allo- or xenograft, autologous sympathetic ganglion, adrenal medulla, and carotid body in clinical settings. In addition, neurotrophic factors, including glial cell line-derived neurotrophic factor (GDNF), have a strong potency to rescue degenerating dopaminergic cells. Protein and/or gene therapy also might be a therapeutic option for PD. In this review, genetically engineered cell transplantation for animal models of PD, including catecholamine/neurotrophic factor-secreting cell transplantation with or without encapsulation, as performed in our laboratories, and their potential future as clinical applications are described with recent clinical studies in this field.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700–8558, Japan
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700–8558, Japan
| |
Collapse
|
14
|
Abstract
The brain has long been known as a dimorphic organ and as a target of sex steroids. It is also a site for their synthesis. Sex steroids in numerous ways can modify cerebral physiology, and along with many processes adult neurogenesis is also modulated by sex steroids. This review will focus on the effects of the main steroids, estrogens, androgens and progestogens, and unveil some aspects of their partly disclosed mechanisms of actions. Gonadal steroids act on different steps of neurogenesis: cell proliferation seems to be increased by estrogens only, while androgens and progestogens favor neuronal renewal by increasing cell survival; differentiation is a common target. Aging is characterized by a cognitive deficiency, paralleled by a decrease in the rate of neuronal renewal and in the levels of circulating gonadal hormones. Therefore, the effects of gonadal hormones on the aging brain are important to consider. The review will also be expanded to related molecules which are agonists to the nuclear receptors. Sex steroids can modify adult neuronal renewal and the extensive knowledge of their actions on neurogenesis is essential, as it can be a leading pathway to therapeutic perspectives.
Collapse
Affiliation(s)
- Christine Heberden
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.
| |
Collapse
|
15
|
Polylysine-modified polyethylenimine (PEI-PLL) mediated VEGF gene delivery protects dopaminergic neurons in cell culture and in rat models of Parkinson's Disease (PD). Acta Biomater 2017; 54:58-68. [PMID: 28025049 DOI: 10.1016/j.actbio.2016.12.048] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 11/21/2022]
Abstract
Parkinson's Disease (PD) is a chronic neurodegenerative disorder characterized by motor deficits which result from the progressive loss of dopaminergic neurons. Gene therapy using growth factors such as VEGF seems to be a viable approach for potential therapeutic treatment of PD. In this study, we utilized a novel non-viral gene carrier designated as PEI-PLL synthesized by our laboratory to deliver VEGF gene to study its effect by using both cell culture as well as animal models of PD. For cell culture experiments, we utilized 6-hydroxydopamine (6-OHDA) mediated cell death model of MN9D cells following transfection with either a control plasmid or VEGF expressing plasmid. As compared to control transfected cells, PEI-PLL mediated VEGF gene delivery to MN9D cells resulted in increased cell viability, increase in the number of Tyrosine hydroxylase (TH) positive cells and decreased apoptosis following 6-OHDA insult. Next, we studied the therapeutic potential of PEI-PLL mediated VEGF gene delivery in SNPc by using unilateral 6-OHDA Medial forebrain bundle (MFB) lesion model of PD in rats. VEGF administration prevented the loss of motor functions induced by 6-OHDA as determined by behavior analysis. Similarly, VEGF inhibited the 6-OHDA mediated loss of DA neurons in Substantia Nigra Pars Compacta (SNPc) as well as DA nerve fibers in striatum as determined by TH immunostaining. In addition, PEI-PLL mediated VEGF gene delivery also prevented apoptosis and microglial activation in PD rat models. Together, these results clearly demonstrated the beneficial effects of PEI-PLL mediated VEGF gene delivery on dopaminergic system in both cell culture and animal models of PD. STATEMENT OF SIGNIFICANCE In this report, we exploited the potential of PEI-PLL to deliver VEGF gene for the potential therapeutic treatment of PD by using both cell culture and animal models of PD. To the best of our knowledge, this is the first report describing the use of novel polymeric gene carriers for the delivery of VEGF gene to DA neurons with improved transfection efficiency. Finally, the study will lead to a significant advancement in the field of non-viral PD gene therapy treatment.
Collapse
|
16
|
VEGF-related polymorphisms identified by GWAS and risk for major depression. Transl Psychiatry 2017; 7:e1055. [PMID: 28267147 PMCID: PMC5416679 DOI: 10.1038/tp.2017.36] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 12/21/2022] Open
Abstract
Depression is a common, severe, disabling mental disease that affects millions of people of all ages worldwide. Various studies have shown that neurotrophic/growth factors have a key role in depression and, more specifically, vascular endothelial growth factor (VEGF) is implicated in the pathogenesis of depression. The purpose of this study was to investigate the potential links between four VEGF-related single-nucleotide polymorphisms (SNPs), previously identified through a genome-wide association study (GWAS) and depression. The direct effects and epistatic interactions of the four VEGF-related SNPs (rs10738760, rs6921438, rs6993770 and rs4416670) on depression were investigated through a case-control study including 437 individuals diagnosed with depression and 477 healthy volunteers as controls. Gender, age and body mass index influence was additionally analyzed. The SNP rs4416670 was associated with increased risk for depression (OR: 1.60, P: 0.010). This result demonstrates the existence of relationships between VEGF genetic determinants and depression. This novel association reveals new molecular mechanisms suggesting the potential role of VEGF in depression development that could help to promote a personalized prediction for this severe common disease.
Collapse
|
17
|
Hu J, Lang Y, Cao Y, Zhang T, Lu H. The Neuroprotective Effect of Tetramethylpyrazine Against Contusive Spinal Cord Injury by Activating PGC-1α in Rats. Neurochem Res 2015; 40:1393-401. [PMID: 25981953 PMCID: PMC4493940 DOI: 10.1007/s11064-015-1606-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 03/13/2015] [Accepted: 05/07/2015] [Indexed: 12/14/2022]
Abstract
Tetramethylpyrazine (TMP) has been suggested to have neuroprotective effects against spinal cord injury (SCI); however, few studies have examined these effects and the corresponding mechanism. Therefore, the present study aimed to investigate the neuroprotective effect and underlying mechanism of TMP against contusive SCI. Adult male Sprague-Dawley rats were randomly divided into Sham, normal saline (NS) and TMP groups. Each group was divided into subgroups according to the time of sacrifice: 1, 3, 7, 14, 21 and 28 days post-injury. Laminectomy was performed in all groups, followed by contusive SCI establishment in the TMP and NS groups. TMP (80 mg/kg) was injected thereafter daily from 3 to 7 days post-injury in the TMP group, which was replaced by equal volume of normal saline in the NS group. The Basso-Beattie-Bresnahan (BBB) Locomotor Rating Scale was measured at different time points post-injury to appraise locomotor functional recovery. Quantitative real-time PCR and immunofluorescence were used to assess the spatio-temporal expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), while western blot was adopted to detect the effect of TMP on PGC-1α. Neural apoptotic changes and neuronal survival were evaluated using the TUNEL method and Nissl staining, respectively. TMP treatment markedly increased PGC-1α expression, neuronal survival and BBB locomotor scores, while also reducing neural apoptosis. These results demonstrate that TMP is neuroprotective against contusive SCI, with the inhibition of neural apoptosis and increase of neuronal survival. The sustained expression of PGC-1α may partially contribute to the TMP-mediated neuroprotective effect.
Collapse
Affiliation(s)
- Jianzhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | |
Collapse
|
18
|
Daviaud N, Garbayo E, Sindji L, Martínez-Serrano A, Schiller PC, Montero-Menei CN. Survival, differentiation, and neuroprotective mechanisms of human stem cells complexed with neurotrophin-3-releasing pharmacologically active microcarriers in an ex vivo model of Parkinson's disease. Stem Cells Transl Med 2015; 4:670-84. [PMID: 25925835 DOI: 10.5966/sctm.2014-0139] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 03/05/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Stem cell-based regenerative therapies hold great potential for the treatment of degenerative disorders such as Parkinson's disease (PD). We recently reported the repair and functional recovery after treatment with human marrow-isolated adult multilineage inducible (MIAMI) cells adhered to neurotrophin-3 (NT3) releasing pharmacologically active microcarriers (PAMs) in hemiparkinsonian rats. In order to comprehend this effect, the goal of the present work was to elucidate the survival, differentiation, and neuroprotective mechanisms of MIAMI cells and human neural stem cells (NSCs), both adhering to NT3-releasing PAMs in an ex vivo organotypic model of nigrostriatal degeneration made from brain sagittal slices. It was shown that PAMs led to a marked increase in MIAMI cell survival and neuronal differentiation when releasing NT3. A significant neuroprotective effect of MIAMI cells adhering to PAMs was also demonstrated. NSCs barely had a neuroprotective effect and differentiated mostly into dopaminergic neuronal cells when adhering to PAM-NT3. Moreover, those cells were able to release dopamine in a sufficient amount to induce a return to baseline levels. Reverse transcription-quantitative polymerase chain reaction and enzyme-linked immunosorbent assay analyses identified vascular endothelial growth factor (VEGF) and stanniocalcin-1 as potential mediators of the neuroprotective effect of MIAMI cells and NSCs, respectively. It was also shown that VEGF locally stimulated tissue vascularization, which might improve graft survival, without excluding a direct neuroprotective effect of VEGF on dopaminergic neurons. These results indicate a prospective interest of human NSC/PAM and MIAMI cell/PAM complexes in tissue engineering for PD. SIGNIFICANCE Stem cell-based regenerative therapies hold great potential for the treatment of degenerative disorders such as Parkinson's disease (PD). The present work elucidates and compares the survival, differentiation, and neuroprotective mechanisms of marrow-isolated adult multilineage inducible cells and human neural stem cells both adhered to neurotrophin-3-releasing pharmacologically active microcarriers in an ex vivo organotypic model of PD made from brain sagittal slices.
Collapse
Affiliation(s)
- Nicolas Daviaud
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Elisa Garbayo
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Laurence Sindji
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alberto Martínez-Serrano
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Paul C Schiller
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Claudia N Montero-Menei
- INSERM U1066, Micro et nanomédecines biomimétiques, Angers, France; L'université Nantes, Angers, Le Mans, Angers University, Angers, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain; Department of Molecular Biology and Center of Molecular Biology "Severo Ochoa," Autonomous University of Madrid-Consejo Superior de Investigaciones Científicas, Campus Cantoblanco, Madrid, Spain; Miami Veterans Healthcare System, Department of Orthopedics, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
19
|
|
20
|
Muñoz A, Garrido-Gil P, Dominguez-Meijide A, Labandeira-Garcia JL. Angiotensin type 1 receptor blockage reduces l-dopa-induced dyskinesia in the 6-OHDA model of Parkinson's disease. Involvement of vascular endothelial growth factor and interleukin-1β. Exp Neurol 2014; 261:720-32. [DOI: 10.1016/j.expneurol.2014.08.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/01/2014] [Accepted: 08/16/2014] [Indexed: 12/17/2022]
|
21
|
Neuroprotection by rat Müller glia against high glucose-induced neurodegeneration through a mechanism involving ERK1/2 activation. Exp Eye Res 2014; 125:20-9. [DOI: 10.1016/j.exer.2014.05.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/17/2014] [Accepted: 05/15/2014] [Indexed: 12/15/2022]
|
22
|
Shinko A, Agari T, Kameda M, Yasuhara T, Kondo A, Tayra JT, Sato K, Sasaki T, Sasada S, Takeuchi H, Wakamori T, Borlongan CV, Date I. Spinal cord stimulation exerts neuroprotective effects against experimental Parkinson's disease. PLoS One 2014; 9:e101468. [PMID: 25009993 PMCID: PMC4092020 DOI: 10.1371/journal.pone.0101468] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/05/2014] [Indexed: 01/19/2023] Open
Abstract
In clinical practice, deep brain stimulation (DBS) is effective for treatment of motor symptoms in Parkinson’s disease (PD). However, the mechanisms have not been understood completely. There are some reports that electrical stimulation exerts neuroprotective effects on the central nervous system diseases including cerebral ischemia, head trauma, epilepsy and PD, although there are a few reports on neuroprotective effects of spinal cord stimulation (SCS). We investigated the neuroprotective effects of high cervical SCS on PD model of rats. Adult female Sprague-Dawley rats received hour-long SCS (2, 50 or 200 Hz) with an epidural electrode at C1–2 level for 16 consecutive days. At 2 days after initial SCS, 6-hydroxydopamine (6-OHDA) was injected into the right striatum of rats. Behavioral evaluations of PD symptoms were employed, including cylinder test and amphetamine-induced rotation test performed at 1 and 2 weeks after 6-OHDA injection. Animals were subsequently euthanized for immunohistochemical investigations. In order to explore neurotrophic and growth factor upregulation induced by SCS, another cohort of rats that received 50 Hz SCS was euthanized at 1 and 2 weeks after lesion for protein assays. Behavioral tests revealed that the number of amphetamine-induced rotations decreased in SCS groups. Immunohistochemically, tyrosine hydroxylase (TH)-positive fibers in the striatum were significantly preserved in SCS groups. TH-positive neurons in the substantia nigra pars compacta were significantly preserved in 50 Hz SCS group. The level of vascular endothelial growth factor (VEGF) was upregulated by SCS at 1 week after the lesion. These results suggest that high cervical SCS exerts neuroprotection in PD model of rats, at least partially by upregulation of VEGF. SCS is supposed to suppress or delay PD progression and might become a less invasive option for PD patients, although further preclinical and clinical investigations are needed to confirm the effectiveness and safety.
Collapse
Affiliation(s)
- Aiko Shinko
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takashi Agari
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
- * E-mail:
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Akihiko Kondo
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Judith Thomas Tayra
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kenichiro Sato
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hayato Takeuchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takaaki Wakamori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Cesario V. Borlongan
- Department of Neurosurgery, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
23
|
Neuroprotective effects of liraglutide for stroke model of rats. Int J Mol Sci 2013; 14:21513-24. [PMID: 24177570 PMCID: PMC3856019 DOI: 10.3390/ijms141121513] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 12/17/2022] Open
Abstract
The number of diabetes mellitus (DM) patients is increasing, and stroke is deeply associated with DM. Recently, neuroprotective effects of glucagon-like peptide-1 (GLP-1) are reported. In this study, we explored whether liraglutide, a GLP-1 analogue exerts therapeutic effects on a rat stroke model. Wistar rats received occlusion of the middle cerebral artery for 90 min. At one hour after reperfusion, liraglutide or saline was administered intraperitoneally. Modified Bederson's test was performed at 1 and 24 h and, subsequently, rats were euthanized for histological investigation. Peripheral blood was obtained for measurement of blood glucose level and evaluation of oxidative stress. Brain tissues were collected to evaluate the level of vascular endothelial growth factor (VEGF). The behavioral scores of liraglutide-treated rats were significantly better than those of control rats. Infarct volumes of liraglutide-treated rats at were reduced, compared with those of control rats. The level of derivatives of reactive oxygen metabolite was lower in liraglutide-treated rats. VEGF level of liraglutide-treated rats in the cortex, but not in the striatum significantly increased, compared to that of control rats. In conclusion, this is the first study to demonstrate neuroprotective effects of liraglutide on cerebral ischemia through anti-oxidative effects and VEGF upregulation.
Collapse
|
24
|
Therapeutic effects of repetitive transcranial magnetic stimulation in an animal model of Parkinson's disease. Brain Res 2013; 1537:290-302. [PMID: 23998987 DOI: 10.1016/j.brainres.2013.08.051] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 07/24/2013] [Accepted: 08/26/2013] [Indexed: 01/08/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is used to treat neurological diseases such as stroke and Parkinson's disease (PD). Although rTMS has been used clinically, its underlying therapeutic mechanism remains unclear. The objective of the present study was to clarify the neuroprotective effect and therapeutic mechanism of rTMS in an animal model of PD. Adult Sprague-Dawley rats were unilaterally injected with 6-hydroxydopamine (6-OHDA) into the right striatum. Rats with PD were then treated with rTMS (circular coil, 10 Hz, 20 min/day) daily for 4 weeks. Behavioral assessments such as amphetamine-induced rotational test and treadmill locomotion test were performed, and the dopaminergic (DA) neurons of substantia nigra pas compacta (SNc) and striatum were histologically examined. Expression of neurotrophic/growth factors was also investigated by multiplex ELISA, western blotting analysis and immunohistochemistry 4 weeks after rTMS application. Among the results, the number of amphetamine-induced rotations was significantly lower in the rTMS group than in the control group at 4 weeks post-treatment. Treadmill locomotion was also significantly improved in the rTMS-treated rats. Tyrosine hydroxylase-positive DA neurons and DA fibers in rTMS group rats were greater than those in untreated group in both ipsilateral SNc and striatum, respectively. The expression levels of brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, platelet-derived growth factor, and vascular endothelial growth factor were elevated in both the 6-OHDA-injected hemisphere and the SNc of the rTMS-treated rats. In conclusion, rTMS treatment improved motor functions and survival of DA neurons, suggesting that the neuroprotective effect of rTMS treatment might be induced by upregulation of neurotrophic/growth factors in the PD animal model.
Collapse
|
25
|
Wahl V, Vogler S, Grosche A, Pannicke T, Ueffing M, Wiedemann P, Reichenbach A, Hauck S, Bringmann A. Osteopontin inhibits osmotic swelling of retinal glial (Müller) cells by inducing release of VEGF. Neuroscience 2013; 246:59-72. [DOI: 10.1016/j.neuroscience.2013.04.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 04/15/2013] [Accepted: 04/17/2013] [Indexed: 12/19/2022]
|
26
|
Zhang Y, Chopp M, Meng Y, Zhang ZG, Doppler E, Mahmood A, Xiong Y. Improvement in functional recovery with administration of Cerebrolysin after experimental closed head injury. J Neurosurg 2013; 118:1343-55. [PMID: 23581594 DOI: 10.3171/2013.3.jns122061] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Cerebrolysin is a unique peptide preparation that mimics the action of neurotrophic factors. This study was designed to investigate the effects of acute treatment of experimental closed head injury (CHI) in rats with Cerebrolysin on neurological function. METHODS Adult male Wistar rats (n = 60) were subjected to impact acceleration-induced CHI. Closed head injured rats received intraperitoneal injection of saline (n = 30) or Cerebrolysin (2.5 ml/kg, n = 30) starting 1 hour postinjury and administered once daily until they were killed (2 or 14 days after CHI). To evaluate functional outcome, the modified neurological severity score (mNSS), foot fault, adhesive removal, and Morris water maze (MWM) tests were performed. Animals were killed on Day 14 (n = 20) after injury, and their brains were removed and processed for measurement of neuronal cells, axonal damage, apoptosis, and neuroblasts. The remaining rats (n = 40) were killed 2 days postinjury to evaluate cerebral microvascular patency by fluorescein isothiocyanate (FITC)-dextran perfusion (n = 16) and to measure the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) by using real-time reverse transcriptase-polymerase chain reaction (RT-PCR, n = 8) and by immunohistochemical analysis (n = 16). RESULTS At 14 days post-CHI, the Cerebrolysin treatment group exhibited significant improvements in functional outcomes (the adhesive removal, mNSS, foot-fault, and MWM tests), and significantly more neurons and neuroblasts were present in the dentate gyrus (DG) (p < 0.05) compared with the saline-treated group (p < 0.05). At 2 days post-CHI, the Cerebrolysin group exhibited a significantly higher percentage of phosphorylated neurofilament H (pNF-H)-positive staining area in the striatum (p < 0.05), a significant increase in the percentage of FITC-dextran perfused vessels in the brain cortex (p < 0.05), a significant increase in the number of VEGF-positive cells (p < 0.05), and a significant reduction in the MMP-9 staining area (p < 0.05) compared with the saline-treated group. There was no significant difference in mRNA levels of MMP-9 and VEGF in the hippocampus and cortex 48 hours postinjury between Cerebrolysin- and saline-treated rats that sustained CHI. CONCLUSIONS Acute Cerebrolysin treatment improves functional recovery in rats after CHI. Cerebrolysin is neuroprotective for CHI (increased neurons in the dentate gyrus and the CA3 regions of the hippocampus and increased neuroblasts in the dentate gyrus) and may preserve axonal integrity in the striatum (significantly increased percentage of pNF-H-positive tissue in the striatum). Reduction of MMP-9 and elevation of VEGF likely contribute to enhancement of vascular patency and integrity as well as neuronal survival induced by Cerebrolysin. These promising results suggest that Cerebrolysin may be a useful treatment in improving the recovery of patients with CHI.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Faigle R, Song H. Signaling mechanisms regulating adult neural stem cells and neurogenesis. Biochim Biophys Acta Gen Subj 2012; 1830:2435-48. [PMID: 22982587 DOI: 10.1016/j.bbagen.2012.09.002] [Citation(s) in RCA: 247] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/11/2012] [Accepted: 09/05/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Adult neurogenesis occurs throughout life in discrete regions of the mammalian brain and is tightly regulated via both extrinsic environmental influences and intrinsic genetic factors. In recent years, several crucial signaling pathways have been identified in regulating self-renewal, proliferation, and differentiation of neural stem cells, as well as migration and functional integration of developing neurons in the adult brain. SCOPE OF REVIEW Here we review our current understanding of signaling mechanisms, including Wnt, notch, sonic hedgehog, growth and neurotrophic factors, bone morphogenetic proteins, neurotransmitters, transcription factors, and epigenetic modulators, and crosstalk between these signaling pathways in the regulation of adult neurogenesis. We also highlight emerging principles in the vastly growing field of adult neural stem cell biology and neural plasticity. MAJOR CONCLUSIONS Recent methodological advances have enabled the field to identify signaling mechanisms that fine-tune and coordinate neurogenesis in the adult brain, leading to a better characterization of both cell-intrinsic and environmental cues defining the neurogenic niche. Significant questions related to niche cell identity and underlying regulatory mechanisms remain to be fully addressed and will be the focus of future studies. GENERAL SIGNIFICANCE A full understanding of the role and function of individual signaling pathways in regulating neural stem cells and generation and integration of newborn neurons in the adult brain may lead to targeted new therapies for neurological diseases in humans. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Roland Faigle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
28
|
Schwartz CM, Tavakoli T, Jamias C, Park SS, Maudsley S, Martin B, Phillips TM, Yao PJ, Itoh K, Ma W, Rao MS, Arenas E, Mattson MP. Stromal factors SDF1α, sFRP1, and VEGFD induce dopaminergic neuron differentiation of human pluripotent stem cells. J Neurosci Res 2012; 90:1367-81. [PMID: 22535492 PMCID: PMC3350575 DOI: 10.1002/jnr.23064] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 03/16/2012] [Indexed: 12/21/2022]
Abstract
Human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons hold potential for treating Parkinson's disease (PD) through cell replacement therapy. Generation of DA neurons from hESCs has been achieved by coculture with the stromal cell line PA6, a source of stromal cell-derived inducing activity (SDIA). However, the factors produced by stromal cells that result in SDIA are largely undefined. We previously reported that medium conditioned by PA6 cells can generate functional DA neurons from NTera2 human embryonal carcinoma stem cells. Here we show that PA6-conditioned medium can induce DA neuronal differentiation in both NTera2 cells and the hESC I6 cell line. To identify the factor(s) responsible for SDIA, we used large-scale microarray analysis of gene expression combined with mass spectrometric analysis of PA6-conditioned medium (CM). The candidate factors, hepatocyte growth factor (HGF), stromal cell-derived factor-1 α (SDF1α), secreted frizzled-related protein 1 (sFRP1), and vascular endothelial growth factor D (VEGFD) were identified, and their concentrations in PA6 CM were established by immunoaffinity capillary electrophoresis. Upon addition of SDF1α, sFRP1, and VEGFD to the culture medium, we observed an increase in the number of cells expressing tyrosine hydroxylase (a marker for DA neurons) and βIII-tubulin (a marker for immature neurons) in both the NTera2 and I6 cell lines. These results indicate that SDF1α, sFRP1, and VEGFD are major components of SDIA and suggest the potential use of these defined factors to elicit DA differentiation of pluripotent human stem cells for therapeutic intervention in PD.
Collapse
Affiliation(s)
- Catherine M. Schwartz
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
- Department of Medical Biochemistry and Biophysics, Laboratory of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Tahereh Tavakoli
- Stem Cell Center, Developmental Biology, American Type Culture Collection, Manassas, VA
| | - Charmaine Jamias
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
| | - Sung-Soo Park
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
| | - Stuart Maudsley
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
| | - Bronwen Martin
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
| | - Terry M. Phillips
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Laboratory of Bioengineering and Physical Science, Bethesda, MD
| | - Pamela J. Yao
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
| | - Katsuhiko Itoh
- Department of Clinical Molecular Biology, Kyoto University, Kyoto, Japan
| | - Wu Ma
- Stem Cell Center, Developmental Biology, American Type Culture Collection, Manassas, VA
| | | | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Laboratory of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Mark P. Mattson
- National Institute on Aging Intramural Research Program, National Institutes of Health, Laboratory of Neurosciences, Baltimore, MD
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Yang J, Guo L, Liu R, Liu H. Neuroprotective effects of VEGF administration after focal cerebral ischemia/reperfusion: Dose response and time window. Neurochem Int 2012; 60:592-6. [DOI: 10.1016/j.neuint.2012.02.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 01/25/2012] [Accepted: 02/20/2012] [Indexed: 01/17/2023]
|
30
|
Fournier NM, Duman RS. Role of vascular endothelial growth factor in adult hippocampal neurogenesis: implications for the pathophysiology and treatment of depression. Behav Brain Res 2012; 227:440-9. [PMID: 21536078 PMCID: PMC3176958 DOI: 10.1016/j.bbr.2011.04.022] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/21/2011] [Accepted: 04/15/2011] [Indexed: 01/15/2023]
Abstract
It is now well established that the adult brain has the capacity to generate new neurons throughout life. Although the functional significance of adult neurogenesis still remains to be established, increasing evidence has implicated compromised hippocampal neurogenesis as a possible contributor in the development of major depressive disorder. Antidepressants increase hippocampal neurogenesis and there is evidence in rodent models that the therapeutic efficacy of these agents is attributable, in part, to this neurogenic effect. As such, considerable interest has been directed at identifying molecular signals, including neurotrophic factors and related signaling pathways that are associated with antidepressant action and could operate as key modulators in the regulation of neurogenesis in the adult hippocampus. One interesting candidate is vascular endothelial growth factor (VEGF), which is known to possess strong neurogenic effects. In this review, we will discuss the involvement of VEGF signaling in the etiology and treatment of depression.
Collapse
Affiliation(s)
- Neil M. Fournier
- Laboratory of Molecular Psychiatry, Department of Psychiatry, Yale University School of Medicine, New Haven, CT., 06508
| | - Ronald S. Duman
- Laboratory of Molecular Psychiatry, Department of Psychiatry, Yale University School of Medicine, New Haven, CT., 06508
| |
Collapse
|
31
|
Effects of intravitreal bevacizumab (Avastin) on the porcine retina. Graefes Arch Clin Exp Ophthalmol 2011; 249:1821-9. [DOI: 10.1007/s00417-011-1773-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/21/2011] [Accepted: 07/28/2011] [Indexed: 11/26/2022] Open
|
32
|
VEGF-expressing human umbilical cord mesenchymal stem cells, an improved therapy strategy for Parkinson's disease. Gene Ther 2010; 18:394-402. [PMID: 21107440 DOI: 10.1038/gt.2010.152] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The umbilical cord provides a rich source of primitive mesenchymal stem cells (human umbilical cord mesenchymal stem cells (HUMSCs)), which have the potential for transplantation-based treatments of Parkinson's Disease (PD). Our pervious study indicated that adenovirus-associated virus-mediated intrastriatal delivery of human vascular endothelial growth factor 165 (VEGF 165) conferred molecular protection to the dopaminergic system. As both VEGF and HUMSCs displayed limited neuroprotection, in this study we investigated whether HUMSCs combined with VEGF expression could offer enhanced neuroprotection. HUMSCs were modified by adenovirus-mediated VEGF gene transfer, and subsequently transplanted into rotenone-lesioned striatum of hemiparkinsonian rats. As a result, HUMSCs differentiated into dopaminergic neuron-like cells on the basis of neuron-specific enolase (NSE) (neuronal marker), glial fibrillary acidic protein (GFAP) (astrocyte marker), nestin (neural stem cell marker) and tyrosine hydroxylase (TH) (dopaminergic marker) expression. Further, VEGF expression significantly enhanced the dopaminergic differentiation of HUMSCs in vivo. HUMSC transplantation ameliorated apomorphine-evoked rotations and reduced the loss of dopaminergic neurons in the lesioned substantia nigra (SNc), which was enhanced significantly by VEGF expression in HUMSCs. These findings present the suitability of HUMSC as a vector for gene therapy and suggest that stem cell engineering with VEGF may improve the transplantation strategy for the treatment of PD.
Collapse
|
33
|
Voss MJ, Entschladen F. Tumor interactions with soluble factors and the nervous system. Cell Commun Signal 2010; 8:21. [PMID: 20822525 PMCID: PMC2942890 DOI: 10.1186/1478-811x-8-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 09/07/2010] [Indexed: 12/16/2022] Open
Abstract
In the genomic era of cancer research, the development of metastases has been attributed to mutations in the tumor that enable the cells to migrate. However, gene analyses revealed that primary tumors and metastases were in some cases genetically identical and the question was raised whether metastasis formation might be an inherent feature of certain tumor cells. In contradiction to this view, the last decade of cancer research has brought to light, that tumor cell migration, similar to leukocyte and fibroblast migration, is a highly regulated process. The nervous system plays an important role in this regulation, at least in two respects: firstly, neurotransmitters are known to regulate the migratory activity of tumor cells, and secondly, nerve fibers are used as routes for perineural invasion. We also summarize here the current knowledge on the innervation of tumors. Such a process might establish a neuro-neoplastic synapse, with the close interaction of tumor cells and nerve cells supporting metastasis formation.
Collapse
Affiliation(s)
- Melanie J Voss
- Institute of Immunology, ZBAF, Witten/Herdecke University, Stockumer Str, 10, 58448 Witten, Germany.
| | | |
Collapse
|
34
|
Krügel K, Wurm A, Linnertz R, Pannicke T, Wiedemann P, Reichenbach A, Bringmann A. Erythropoietin inhibits osmotic swelling of retinal glial cells by Janus kinase- and extracellular signal-regulated kinases1/2-mediated release of vascular endothelial growth factor. Neuroscience 2010; 165:1147-58. [DOI: 10.1016/j.neuroscience.2009.11.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 11/11/2009] [Accepted: 11/12/2009] [Indexed: 11/24/2022]
|
35
|
Enhanced expression of vascular endothelial growth factor receptor-3 in the subventricular zone of stroke-lesioned rats. Neurosci Lett 2010; 469:194-8. [DOI: 10.1016/j.neulet.2009.11.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 11/18/2009] [Accepted: 11/27/2009] [Indexed: 11/17/2022]
|
36
|
Dubynina EV, Inozemtseva LS, Markov DD, Yatsenko KA, Dolotov OV, Grivennikov IA. Alpha-melanocyte-stimulating hormone increases the expression of vascular endothelial growth factor in rat hippocampal astrocytes in vitro. NEUROCHEM J+ 2009. [DOI: 10.1134/s1819712409040059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Emanuele E, Orsi P, Barale F, di Nemi SU, Bertona M, Politi P. Serum levels of vascular endothelial growth factor and its receptors in patients with severe autism. Clin Biochem 2009; 43:317-9. [PMID: 19850021 DOI: 10.1016/j.clinbiochem.2009.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 10/09/2009] [Accepted: 10/13/2009] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To study vascular endothelial growth factor (VEGF) and its soluble receptors sVEGFR-1 and -2 in autism. DESIGN AND METHODS We measured serum levels of angiogenic molecules in 22 patients with severe autism and 28 controls. RESULTS Patients and controls had similar sVEGFR-2 levels, but VEGF levels were lower and sVEGFR-1 higher in patients with autism. CONCLUSION The imbalance between VEGF and its receptor sVEGFR-1 may be involved in the pathophysiology of autism.
Collapse
Affiliation(s)
- Enzo Emanuele
- Department of Health Sciences, Section of Psychiatry, University of Pavia, Via Bassi, 21, I-27100, Pavia, Italy.
| | | | | | | | | | | |
Collapse
|
38
|
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: 7.8] [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
|
39
|
Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, Osborne NN, Reichenbach A. Cellular signaling and factors involved in Müller cell gliosis: neuroprotective and detrimental effects. Prog Retin Eye Res 2009; 28:423-51. [PMID: 19660572 DOI: 10.1016/j.preteyeres.2009.07.001] [Citation(s) in RCA: 522] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Müller cells are active players in normal retinal function and in virtually all forms of retinal injury and disease. Reactive Müller cells protect the tissue from further damage and preserve tissue function by the release of antioxidants and neurotrophic factors, and may contribute to retinal regeneration by the generation of neural progenitor/stem cells. However, Müller cell gliosis can also contribute to neurodegeneration and impedes regenerative processes in the retinal tissue by the formation of glial scars. This article provides an overview of the neuroprotective and detrimental effects of Müller cell gliosis, with accounts on the cellular signal transduction mechanisms and factors which are implicated in Müller cell-mediated neuroprotection, immunomodulation, regulation of Müller cell proliferation, upregulation of intermediate filaments, glial scar formation, and the generation of neural progenitor/stem cells. A proper understanding of the signaling mechanisms implicated in gliotic alterations of Müller cells is essential for the development of efficient therapeutic strategies that increase the supportive/protective and decrease the destructive roles of gliosis.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Bringmann A, Wiedemann P. Involvement of Müller glial cells in epiretinal membrane formation. Graefes Arch Clin Exp Ophthalmol 2009; 247:865-83. [PMID: 19415318 DOI: 10.1007/s00417-009-1082-x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 02/10/2009] [Accepted: 04/06/2009] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Proliferative retinopathies are considered to represent maladapted retinal wound repair processes driven by growth factor- and cytokine-induced overstimulation of proliferation, migration, extracellular matrix production and contraction of retinal cells. The formation of neovascular membranes represents an attempt to reoxygenize non-perfused retinal areas. Müller glial cells play a crucial role in the pathogenesis of proliferative retinopathies. This review summarizes the present knowledge regarding the role of Müller cells in periretinal membrane formation, especially in the early steps of epiretinal membrane formation, which involve an interaction of inflammatory and glial cells, and gives a survey of the factors which are suggested to be implicated in the induction of Müller cell gliosis and proliferation. CONCLUSIONS Alterations in the membrane conductance of Müller cells suggest that Müller cells may alter their phenotype into progenitor-like cells in the course of proliferative retinopathies; transdifferentiated Müller cells may have great impact for the development of new cell-based therapies.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Eye Hospital, Leipzig, Germany.
| | | |
Collapse
|
41
|
Li WY, Choi YJ, Lee PH, Huh K, Kang YM, Kim HS, Ahn YH, Lee G, Bang OY. Mesenchymal stem cells for ischemic stroke: changes in effects after ex vivo culturing. Cell Transplant 2009; 17:1045-59. [PMID: 19177841 DOI: 10.3727/096368908786991551] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Although ex vivo culture expansion is necessary to use autologous mesenchymal stem cells (MSCs) in treating stroke patients, and several researchers have utilized culture-expanded cells in their studies, the effects of culture expansion on neurogenesis and trophic support are unknown. Thus, we evaluated the impact of the passage of MSCs on their effects in a rat stroke model. The i.v. application of ex vivo-cultured human MSCs, earlier (passage 2) or later passage (passage 6), was performed in a rat stroke model. Behavioral tests, immunohistochemical studies, and quantitative analysis using the CAST-grid system were performed to evaluate the degree of neurogenesis. We also evaluated the levels of trophic factors in both control and MSC-treated rat brain extract. Compared to rats that received later-passage human MSCs, behavioral recovery and neurogenesis as revealed by bromodeoxyuridine staining were more pronounced in rats that received earlier-passage human MSCs (p < 0.01 in both cases). Double staining showed that most of the endogenous neuronal progenitor cells, but few human MSCs, expressed neuronal and glial phenotypes. Tissue levels of trophic factors, including glial cell line-derived neurotrophic factor, nerve growth factor, vascular endothelial growth factor, and hepatocyte growth factor, were higher in earlier-passage MSC-treated brains than in control or later-passage MSC-treated brains (p < 0.01 in all cases). Our results indicate that ischemia-induced neurogenesis was enhanced by the i.v. administration of human MSCs. The effects were more pronounced with earlier-passage than with later-passage human MSCs, which may be related to the differential capacity in trophic support, depending on their passage.
Collapse
Affiliation(s)
- Wen Yu Li
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Enhanced adult neurogenesis and angiogenesis and altered affective behaviors in mice overexpressing vascular endothelial growth factor 120. J Neurosci 2009; 28:14522-36. [PMID: 19118187 DOI: 10.1523/jneurosci.3673-08.2008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is implicated as a molecular mediator for adult neurogenesis and behavioral effects of antidepressant drugs. However, these potential roles of VEGF in the CNS have not been clarified in model animals. Here we have created transgenic mice overexpressing a short active variant of VEGF-A (VEGF120) in forebrain. Expression of VEGF120 significantly enhanced cell proliferation and angiogenesis, as exemplified by the formation of an enlarged reddish brain. Adult neurogenesis in hippocampus was markedly stimulated without affecting cell differentiation of neural progenitor cells. Hippocampal neurogenesis was particularly robust in young adult animals, but it declined with age and reduced to control levels by 20 weeks under continuous expression of VEGF120. Thus, VEGF alone is not sufficient to support the long-term enhancement of adult neurogenesis, and VEGF-induced vascularization per se does not necessarily predict increased neurogenesis. In transgenic mice, we observed significant changes in affective behaviors. VEGF was found to have not only antidepressant effects but also anxiolytic effects. In addition, we found that VEGF significantly reduced fear and aggression. In contrast, basal activities under natural conditions were not affected much. Unexpectedly, these characteristic behaviors were maintained in older transgenic mice undergoing a reduced level of cell proliferation in hippocampus, suggesting that there is potential dissociation between adult neurogenesis and mood regulation. Our data indicate that VEGF exerts strong neurogenic and angiogenic effects in postnatal brain and influences different forms of affective behaviors.
Collapse
|
43
|
Yasuhara T, Date I. Gene therapy for Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2009:301-309. [PMID: 20411788 DOI: 10.1007/978-3-211-92660-4_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Parkinson's disease is characterized by the degeneration of the nigrostriatal dopaminergic neurons with the manifestation of tremor, rigidity, akinesia, and disturbances of postural reflexes. Medication using L-DOPA and surgeries including deep brain stimulation are the established therapies for Parkinson's disease. Cell therapies are also effective and have rapidly developed with the recent advancement in molecular biological technology including gene transfer. In this review, ex vivo gene therapy using genetically engineered cell transplantation for Parkinson's disease model of animals is described, including catecholamine/neurotrophic factor-secreting cell transplantation with or without encapsulation, as well as in vivo gene therapy using direct injection of viral vector to increase dopamine-production, ameliorate the survival of dopaminergic neurons, correct the deteriorated microenvironment, or normalize genetic abnormality. Furthermore, the future directions for clinical application are described together with recent clinical trials of gene therapy.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Okayama, 700-8558, Japan.
| | | |
Collapse
|
44
|
Downie LE, Pianta MJ, Vingrys AJ, Wilkinson-Berka JL, Fletcher EL. AT1 receptor inhibition prevents astrocyte degeneration and restores vascular growth in oxygen-induced retinopathy. Glia 2008; 56:1076-90. [PMID: 18442090 DOI: 10.1002/glia.20680] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We investigated the effect of receptor blockade induced by an angiotensin II type-1 receptor antagonist (AT(1)-RB) on glial and vascular changes in oxygen-induced retinopathy (OIR), a model of retinopathy of prematurity (ROP). OIR was induced in Sprague-Dawley rats by exposure to 80% oxygen from postnatal (P) days 0-11, followed by 7 days in room air. Control animals were in room air for the entire duration. One cohort of OIR and control pups received the AT(1)-RB valsartan (40 mg/kg/day intraperitoneal) from P11 to P18. The vascular response was examined immunocytochemically using retinal wholemounts and vertical sections labeled with endothelial (Isolectin-B4) and pericyte (NG2, desmin) markers. Glial cell changes were assessed by measuring cell numbers and immunoreactivity (S100beta, connexin-26, and glial fibrillary acidic protein). OIR resulted in extensive intravitreal neovascularization and under-development of the outer vascular plexus. Pericyte numbers were not significantly affected in OIR, although pericyte-endothelial (desmin-IB4) interactions were impaired. Peripheral astrocyte degeneration occurred between P11 and P13 with prominent Müller cell reactivity at P18. Valsartan imparted a protective effect on glia and blood vessels in OIR. At P18, valsartan-treated OIR retinae showed significantly greater astrocyte survival, improved revascularization of the retina, and reduced preretinal neovascularization and Müller cell reactivity. This study identifies a glio-vascular protective effect with AT(1)-RB in OIR.
Collapse
Affiliation(s)
- Laura E Downie
- Department of Anatomy and Cell Biology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
45
|
Shin YJ, Choi JS, Lee JY, Choi JY, Cha JH, Chun MH, Lee MY. Differential regulation of vascular endothelial growth factor-C and its receptor in the rat hippocampus following transient forebrain ischemia. Acta Neuropathol 2008; 116:517-27. [PMID: 18704465 DOI: 10.1007/s00401-008-0423-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 07/22/2008] [Accepted: 08/06/2008] [Indexed: 12/22/2022]
Abstract
We investigated the changes in the expression of vascular endothelial growth factor-C (VEGF-C) and its receptor, VEGFR-3, in the rat hippocampus following transient forebrain ischemia. The expression profiles of VEGF-C and VEGFR-3 were very similar in the control hippocampi, where both genes were constitutively expressed in neurons in the pyramidal cell and granule cell layers. The spatiotemporal expression pattern of VEGF-C was similar to that of VEGFR-3 in the ischemic hippocampus, and in the CA1 and dentate hilar regions both VEGF-C and VEGFR-3 were strongly expressed in activated glial cells rather than in neurons. Most of the activated glial cells expressing both genes were reactive astrocytes, although some were a subpopulation of brain macrophages. In the dentate gyrus, however, VEGFR-3 expression was transiently increased in the innermost layer of granule cells on days 7-10 after reperfusion, coinciding with an increase in polysialylated neural cell adhesion molecule staining--a marker for immature neurons. These data suggest that VEGF-C may be involved in glial reaction via paracrine or autocrine mechanisms in the ischemic brain and may carry out specific roles in adult hippocampal neurogenesis during ischemic insults.
Collapse
Affiliation(s)
- Yoo-Jin Shin
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul, 137-701, South Korea
| | | | | | | | | | | | | |
Collapse
|
46
|
Flanagan SR, Cantor JB, Ashman TA. Traumatic brain injury: future assessment tools and treatment prospects. Neuropsychiatr Dis Treat 2008; 4:877-92. [PMID: 19183780 PMCID: PMC2626927 DOI: 10.2147/ndt.s1985] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is widespread and leads to death and disability in millions of individuals around the world each year. Overall incidence and prevalence of TBI are likely to increase in absolute terms in the future. Tackling the problem of treating TBI successfully will require improvements in the understanding of normal cerebral anatomy, physiology, and function throughout the lifespan, as well as the pathological and recuperative responses that result from trauma. New treatment approaches and combinations will need to be targeted to the heterogeneous needs of TBI populations. This article explores and evaluates the research evidence in areas that will likely lead to a reduction in TBI-related morbidity and improved outcomes. These include emerging assessment instruments and techniques in areas of structural/chemical and functional neuroimaging and neuropsychology, advances in the realms of cell-based therapies and genetics, promising cognitive rehabilitation techniques including cognitive remediation and the use of electronic technologies including assistive devices and virtual reality, and the emerging field of complementary and alternative medicine.
Collapse
Affiliation(s)
- Steven R Flanagan
- New York University School of Medicine, The Rusk Institute of Rehabilitation, New York, NY, USA.
| | | | | |
Collapse
|
47
|
Kuramoto S, Yasuhara T, Agari T, Kondo A, Matsui T, Miyoshi Y, Shingo T, Date I. Injection of muscimol, a GABAa agonist into the anterior thalamic nucleus, suppresses hippocampal neurogenesis in amygdala-kindled rats. Neurol Res 2008; 31:407-13. [PMID: 18826756 DOI: 10.1179/174313208x346125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The relationship between neurogenesis and epilepsy remains to be solved so far, although aberrant electric circuit recognized in epilepsy might be involved in neurogenesis. In this study, neurogenesis and the proliferation of astrocytes in the subgranular zone of the hippocampus were explored using unilateral amygdala-kindled rats with or without muscimol, a gamma-aminobutyric acid a (GABAa) agonist injection into the bilateral anterior thalamic nuclei (AN). Muscimol injection significantly ameliorated the behavioral scores of epilepsy without any significant alteration on the electroencephalography recorded at the stimulated basolateral amygdala, thus suggesting that muscimol injection might affect the secondary generalization, but not the initial discharge itself. The number of bromodeoxyuridine (BrdU), BrdU/doublecortin and BrdU/glial fibrillary acidic protein-positive cells in the subgranular zone of kindled animals increased markedly. Muscimol injection significantly suppressed neurogenesis, but not the proliferation of astrocyte, in the subgranular zone of the non-stimulated side, probably through the suppression of secondary generalization via AN. The results might indicate the underlying relationships between neurogenesis and epilepsy, that epileptic propagation in unilateral amygdala-kindled rats might go through AN into the contralateral side with subsequent neurogenesis, although further studies need to clarify the hypothesis.
Collapse
Affiliation(s)
- Satoshi Kuramoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Yuan WJ, Yasuhara T, Shingo T, Muraoka K, Agari T, Kameda M, Uozumi T, Tajiri N, Morimoto T, Jing M, Baba T, Wang F, Leung H, Matsui T, Miyoshi Y, Date I. Neuroprotective effects of edaravone-administration on 6-OHDA-treated dopaminergic neurons. BMC Neurosci 2008; 9:75. [PMID: 18671880 PMCID: PMC2533664 DOI: 10.1186/1471-2202-9-75] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Accepted: 08/01/2008] [Indexed: 11/10/2022] Open
Abstract
Background Parkinson's disease (PD) is a neurological disorder characterized by the degeneration of nigrostriatal dopaminergic systems. Free radicals induced by oxidative stress are involved in the mechanisms of cell death in PD. This study clarifies the neuroprotective effects of edaravone (MCI-186, 3-methyl-1-phenyl-2-pyrazolin-5-one), which has already been used for the treatment of cerebral ischemia in Japan, on TH-positive dopaminergic neurons using PD model both in vitro and in vivo. 6-hydroxydopamine (6-OHDA), a neurotoxin for dopaminergic neurons, was added to cultured dopaminergic neurons derived from murine embryonal ventral mesencephalon with subsequet administration of edaravone or saline. The number of surviving TH-positive neurons and the degree of cell damage induced by free radicals were analyzed. In parallel, edaravone or saline was intravenously administered for PD model of rats receiving intrastriatal 6-OHDA lesion with subsequent behavioral and histological analyses. Results In vitro study showed that edaravone significantly ameliorated the survival of TH-positive neurons in a dose-responsive manner. The number of apoptotic cells and HEt-positive cells significantly decreased, thus indicating that the neuroprotective effects of edaravone might be mediated by anti-apoptotic effects through the suppression of free radicals by edaravone. In vivo study demonstrated that edaravone-administration at 30 minutes after 6-OHDA lesion reduced the number of amphetamine-induced rotations significantly than edaravone-administration at 24 hours. Tyrosine hydroxylase (TH) staining of the striatum and substantia nigra pars compacta revealed that edaravone might exert neuroprotective effects on nigrostriatal dopaminergic systems. The neuroprotective effects were prominent when edaravone was administered early and in high concentration. TUNEL, HEt and Iba-1 staining in vivo might demonstrate the involvement of anti-apoptotic, anti-oxidative and anti-inflammatory effects of edaravone-administration. Conclusion Edaravone exerts neuroprotective effects on PD model both in vitro and in vivo. The underlying mechanisms might be involved in the anti-apoptotic effects, anti-oxidative effects, and/or anti-inflammatory effects of edaravone. Edaravone might be a hopeful therapeutic option for PD, although the high therapeutic dosage remains to be solved for the clinical application.
Collapse
Affiliation(s)
- Wen Ji Yuan
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schmid-Brunclik N, Bürgi-Taboada C, Antoniou X, Gassmann M, Ogunshola OO. Astrocyte responses to injury: VEGF simultaneously modulates cell death and proliferation. Am J Physiol Regul Integr Comp Physiol 2008; 295:R864-73. [PMID: 18614764 DOI: 10.1152/ajpregu.00536.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia is linked to changes in blood-brain barrier (BBB) permeability, and loss of BBB integrity is characteristic of many pathological brain diseases including stroke. In particular, astrocytes play a central role in brain homeostasis and BBB function. We investigated how hypoxia affects astrocyte survival and assessed whether VEGF release through hypoxia-inducible factor-1alpha (HIF-1alpha) induction plays a role in tolerance of these cells to insult. Thus primary astrocytes were subjected to normoxic (21% O(2)), hypoxic (1% O(2)), or near-anoxic (<0.1% O(2)) conditions in the presence or absence of glucose. Cell death was significantly initiated after combined oxygen glucose deprivation, and, surprisingly, astrocyte proliferation increased concomitantly. Near anoxic, but not hypoxic, conditions stabilized HIF-1alpha protein and provoked DNA binding activity, whereas oxygen and glucose deprivation accelerated HIF-1alpha accumulation. Unexpectedly, Hif-1alpha knockdown studies showed that elevated VEGF levels following increased insult was only partially due to HIF-1alpha induction, suggesting alternative mechanisms of VEGF regulation. Notably, endogenous VEGF signaling during insult was essential for cell fate since VEGF inhibition appreciably augmented cell death and reduced proliferation. These data suggest Hif-1 only partially contributes to VEGF-mediated astrocyte responses during chronic injury (as occurs in clinical hypoxic/ischemic insults) that may ultimately be responsible for disrupting BBB integrity.
Collapse
Affiliation(s)
- Nicole Schmid-Brunclik
- Institute of Veterinary Physiology, Vetsuisse Faculty, Univ. of Zurich, Winterthurerstrasse 260, Zurich CH 8057, Switzerland
| | | | | | | | | |
Collapse
|
50
|
Abstract
The sympathetic nervous system, via postganglionic innervation of blood vessels and the heart, is an important determinant of cardiovascular function. The mechanisms underlying sympathetic innervation of targets are not fully understood. This study tests the hypothesis that target-derived vascular endothelial growth factor (VEGF) promotes sympathetic innervation of blood vessels. Western blot and immunohistochemical analyses indicate that VEGF is produced by vascular cells in arteries and that VEGF receptors are expressed on sympathetic nerve fibers innervating arteries. In vitro, exogenously added VEGF and VEGF produced by vascular smooth muscle cells (VSMCs) in sympathetic neurovascular cocultures inhibited semaphorin 3A (Sema3A)-induced collapse of sympathetic growth cones. In the absence of Sema3A, VEGF and VSMCs also increased growth cone area. These effects were mediated via VEGF receptor 1. In vivo, the neutralization of VEGF inhibited the reinnervation of denervated femoral arteries. These data demonstrate that target-derived VEGF plays a previously unrecognized role in promoting the growth of sympathetic axons.
Collapse
|