1
|
Forró T, Manu DR, Barbu-Tudoran L, Bălașa R. Astrocyte Dysfunction Reflected in Ischemia-Induced Astrocyte-Derived Extracellular Vesicles: A Pilot Study on Acute Ischemic Stroke Patients. Int J Mol Sci 2024; 25:12471. [PMID: 39596535 PMCID: PMC11594292 DOI: 10.3390/ijms252212471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Extracellular vesicles (EVs) secreted by astrocytes (ADEVs) mediate numerous biological processes, providing insights into damage, repair, and protection following ischemic stroke (IS). This pilot study aimed to broaden the current knowledge on the astrocyte response to ischemia by dynamically assessing the aquaporin-4 (AQP4) and glial cell line-derived neurotrophic factor (GDNF) as cargo proteins of these vesicles in eighteen acute IS patients and nine controls. EV proteins were detected by Western blotting and followed 24 h (D1), 7 days (D7), and one month (M1) after symptoms onset. The post-ischemic ADEV AQP4 and GDNF levels were higher at D1 compared to the control group (p = 0.006 and p = 0.023). Significant differences were observed in ADEV AQP4 during the three evaluated time points (n = 12, p = 0.013) and between D1 and D7 (z = 2.858, p = 0.012), but not in EV GDNF. There was a positive relationship between the severity of stroke at D1 according to the National Institutes of Health Stroke Scale, and ADEV AQP4 at D1 (r = 0.50, p = 0.031), as well as ADEV GDNF at D1 and D7 (r = 0.49, p = 0.035 and r = 0.53, p = 0.021, respectively). The release of EVs with distinct protein profiles can be an attractive platform for the development of biomarkers in IS.
Collapse
Affiliation(s)
- Timea Forró
- Doctoral School of Medicine and Pharmacy, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Doina Ramona Manu
- Center for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Lucian Barbu-Tudoran
- Electron Microscopy Laboratory, Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania;
- Electron Microscopy Integrated Laboratory, National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Rodica Bălașa
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
- 1st Neurology Clinic, County Emergency Clinical Hospital of Targu Mures, 540136 Targu Mures, Romania
| |
Collapse
|
2
|
Chiu YS, Wu KJ, Yu SJ, Wu KL, Hsieh CY, Chou YS, Chen KY, Wang YS, Bae EK, Hung TW, Lin SH, Lin CH, Hsu SC, Wang Y, Chen YH. Transplantation of Exosomes Derived From Human Wharton's Jelly Mesenchymal Stromal Cells Enhances Functional Improvement in Stroke Rats. Cell Transplant 2024; 33:9636897241296366. [PMID: 39624898 PMCID: PMC11613244 DOI: 10.1177/09636897241296366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/28/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Cerebral ischemic stroke is a major cerebrovascular disease and the leading cause of adult disability. We and others previously demonstrated that transplantation of human Wharton's jelly mesenchymal stromal cells (WJ-MSCs) attenuated neuronal damage and promoted functional improvement in stroke animals. This study aimed to investigate the protective effects of human WJ-MSC exosome (Exo) transplant in cellular and rat models of cerebral stroke. Administration of Exo significantly antagonized glutamate-mediated neuronal loss and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-X nick end labeling (TUNEL) in rat primary cortical neuronal cultures. Adult male rats underwent a 60-min middle cerebral artery occlusion (MCAo); Exo or vehicle was injected through the tail vein 5-10 min after the MCAo. Two days later, the rats underwent a series of behavioral tests. Stroke rats receiving Exo developed a significant improvement in locomotor function and forelimb strength while reductions in body asymmetry and Bederson's neurological score. After the behavioral test, brain tissues were harvested for histological and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analyses. Animals receiving Exo had less infarction volume, measured by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Transplantation of Exo increased the expression of protective neurotrophic factors (BMP7, GDNF) and anti-apoptotic factors (Bcl2, Bcl-xL) in the ischemic brain. These findings suggest that early post-treatment with WJ-MSC Exo, given non-invasively through the vein, improved functional recovery and reduced brain damage in the stroke brain.
Collapse
Affiliation(s)
- Yu-Sung Chiu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Kuo-Jen Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kun-Lieh Wu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung, Taiwan
| | | | | | - Kuan-Yu Chen
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Hsun Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Hsueh Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- Immunology Research and Development Center, China Medical University, Taichung City, Taiwan
- Department of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Hsiang Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
3
|
Rihal V, Khan H, Kaur A, Singh TG. Vitamin D as therapeutic modulator in cerebrovascular diseases: a mechanistic perspectives. Crit Rev Food Sci Nutr 2022; 63:7772-7794. [PMID: 35285752 DOI: 10.1080/10408398.2022.2050349] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vitamin D deficiency has been linked to several major chronic diseases, such as cardiovascular and neurodegenerative diseases, diabetes, and cancer, linked to oxidative stress, inflammation, and aging. Vitamin D deficiency appears to be particularly harmful to the cardiovascular system, as it can cause endothelial dysfunctioning and vascular abnormalities through the modulation of various downstream mechanisms. As a result, new research indicates that therapeutic approaches targeting vitamin D inadequacies or its significant downstream effects, such as impaired autophagy, abnormal pro-inflammatory and pro-oxidant reactions, may delay the onset and severity of major cerebrovascular disorders such as stroke and neurologic malformations. Vitamin D modulates the various molecular pathways, i.e., Nitric Oxide, PI3K-Akt Pathway, cAMP pathway, NF-kB Pathway, Sirtuin 1, Nrf2, FOXO, in cerebrovascular disorder. The current review shows evidence for vitamin D's mitigating or slowing the progression of these cerebrovascular disorders, which are significant causes of disability and death worldwide.
Collapse
Affiliation(s)
- Vivek Rihal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
4
|
Zhang Z, Sun GY, Ding S. Glial Cell Line-Derived Neurotrophic Factor and Focal Ischemic Stroke. Neurochem Res 2021; 46:2638-2650. [PMID: 33591443 PMCID: PMC8364922 DOI: 10.1007/s11064-021-03266-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/29/2022]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human debilitation and death. Following the onset of a FIS, the brain experiences a series of spatiotemporal changes which are exemplified in different pathological processes. One prominent feature of FIS is the development of reactive astrogliosis and glial scar formation in the peri-infarct region (PIR). During the subacute phase, astrocytes in PIR are activated, referred to as reactive astrocytes (RAs), exhibit changes in morphology (hypotrophy), show an increased proliferation capacity, and altered gene expression profile, a phenomenon known as reactive astrogliosis. Subsequently, the morphology of RAs remains stable, and proliferation starts to decline together with the formation of glial scars. Reactive astrogliosis and glial scar formation eventually cause substantial tissue remodeling and changes in permanent structure around the PIR. Glial cell line-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line and regarded as a potent survival neurotrophic factor. Under normal conditions, GDNF is expressed in neurons but is upregulated in RAs after FIS. This review briefly describes properties of GDNF, its receptor-mediated signaling pathways, as well as recent studies regarding the role of RAs-derived GDNF in neuronal protection and brain recovery. These results provide evidence suggesting an important role of RA-derived GDNF in intrinsic brain repair and recovery after FIS, and thus targeting GDNF in RAs may be effective for stroke therapy.
Collapse
Affiliation(s)
- Zhe Zhang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Dalton Cardiovascular Research Center, Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, 134 Research Park Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
5
|
Zhang N, Zhang Z, He R, Li H, Ding S. GLAST-CreER T2 mediated deletion of GDNF increases brain damage and exacerbates long-term stroke outcomes after focal ischemic stroke in mouse model. Glia 2020; 68:2395-2414. [PMID: 32497340 DOI: 10.1002/glia.23848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human death. Glial scar formation largely caused by reactive astrogliosis in peri-infarct region (PIR) is the hallmark of FIS. Glial cell-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line supernatant and is a potent survival neurotrophic factor. Here, using CreERT2 -LoxP recombination technology, we generated inducible and astrocyte-specific GDNF conditional knockout (cKO), that is, GLAST-GDNF-/- cKO mice to investigate the effect of reactive astrocytes (RAs)-derived GDNF on neuronal death, brain damage, oxidative stress and motor function recovery after photothrombosis (PT)-induced FIS. Under non-ischemic conditions, we found that adult GLAST-GDNF-/- cKO mice exhibited significant lower numbers of Brdu+, Ki67+ cells, and DCX+ cells in the dentate gyrus (DG) in hippocampus than GDNF floxed (GDNFf/f ) control (Ctrl) mice, indicating endogenous astrocytic GDNF can promote adult neurogenesis. Under ischemic conditions, GLAST-GDNF-/- cKO mice had a significant increase in infarct volume, hippocampal damage and FJB+ degenerating neurons after PT as compared with the Ctrl mice. GLAST-GDNF-/- cKO mice also had lower densities of Brdu+ and Ki67+ cells in the PIR and exhibited larger behavioral deficits than the Ctrl mice. Mechanistically, GDNF deficiency in astrocytes increased oxidative stress through the downregulation of glucose-6-phosphate dehydrogenase (G6PD) in RAs. In summary, our study indicates that RAs-derived endogenous GDNF plays important roles in reducing brain damage and promoting brain recovery after FIS through neural regeneration and suggests that promoting anti-oxidant mechanism in RAs is a potential strategy in stroke therapy.
Collapse
Affiliation(s)
- Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Zhe Zhang
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Rui He
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Hailong Li
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
6
|
Chiang YH, Borlongan CV, Zhou FC, Hoffer BJ, Wang Y. Transplantation of Fetal Kidney Cells: Neuroprotection and Neuroregeneration. Cell Transplant 2017; 14:1-9. [PMID: 15789657 DOI: 10.3727/000000005783983304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Various trophic factors in the transforming growth factor-β (TGF-β) superfamily have been reported to have neuroprotective and neuroregenerative effects. Intracerebral administration of glial cell line-derived neurotrophic factor (GDNF) or bone morphogenetic proteins (BMPs), both members of the TGF-β family, reduce ischemia- or 6-hydroxydopamine (6-OHDA)-induced injury in adult rat brain. Because BMPs and GDNF are highly expressed in fetal kidney cells, transplantation of fetal kidney tissue could serve as a cellular reservoir for such molecules and protect against neuronal injury induced by ischemia, neurotoxins, or reactive oxygen species. In this review, we discuss preclinical evidence for the efficacy of fetal kidney cell transplantation in neuroprotection and regeneration models.
Collapse
Affiliation(s)
- Yung-Hsiao Chiang
- Tri-Service General Hospital, National Defense Medical Center, Taiwan
| | | | | | | | | |
Collapse
|
7
|
Kuric E, Wieloch T, Ruscher K. Dopamine receptor activation increases glial cell line-derived neurotrophic factor in experimental stroke. Exp Neurol 2013; 247:202-8. [DOI: 10.1016/j.expneurol.2013.04.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/26/2013] [Accepted: 04/30/2013] [Indexed: 12/20/2022]
|
8
|
Zhang W, Sato K, Hayashi T, Omori N, Nagano I, Kato S, Horiuchi S, Abe K. Extension of ischemic therapeutic time window by a free radical scavenger, Edaravone, reperfused with tPA in rat brain. Neurol Res 2013; 26:342-8. [PMID: 15142331 DOI: 10.1179/016164104225014058] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
3-methyl-1-phenyl-2-pyrazolin-5-one (Edaravone) is a free radical scavenger. We tested the hypothesis that combination treatment of Edaravone and recombinant tissue plasminogen activator (tPA) extends the therapeutic time window. Male Wistar rats were subjected to 1.5-, 3.0- or 4.5-hour middle cerebral artery (MCA) occlusion (MCAO) by a nylon thread. Animals were randomly divided into four groups. The Sham group rats were operated without MCAO and drug injection. In the Vehicle-treated group the same volume of saline was given every 1.5 hours from just after MCAO to just before reperfusion. In the Vehicle + tPA-treated group saline injection was given as above and tPA (5 mg/kg, i.v.) was given once just after reperfusion. Edaravone+tPA-treated group: Edaravone (3 mg/kg, i.v.) was given every 1.5 hours instead of saline and tPA injection as above. Survival rate, infarct size and evidence of apoptosis and hemorrhage were examined in the animals. Combining administration of Edaravone+tPA significantly increased survival rate after 3 hours of transient MCAO, and reduced infarct volume after 1.5 hours of transient MCAO compared with the vehicle or vehicle+tPA groups. In Edaravone+tPA-treated group, the number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end labeling (TUNEL) and 4-hydroxynonenal (4-HNE) positive cells were reduced at 16 hours after 3 hours of transient MCAO, but not in advanced glycation end products (AGEs) and 8-hydroxy-2'-deoxyguanosine (8-OHdG). Hemorrhage rate and the area decreased in the Edaravone+tPA-treated group. The combination therapy of Edaravone+tPA increased survival rate, and reduced the infarct volume and hemorrhage with reduction of lipid peroxidation. Therefore, Edaravone combination is expected to extend the therapeutic time window of tPA in the clinical situation.
Collapse
Affiliation(s)
- Wenri Zhang
- Department of Neurology, Graduate School of Medicine and Dentistry, Okayama University, 2-5-1 Shikatacho, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Sumbria RK, Boado RJ, Pardridge WM. Combination stroke therapy in the mouse with blood-brain barrier penetrating IgG-GDNF and IgG-TNF decoy receptor fusion proteins. Brain Res 2013; 1507:91-6. [PMID: 23428543 DOI: 10.1016/j.brainres.2013.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/07/2013] [Accepted: 02/14/2013] [Indexed: 01/09/2023]
Abstract
Stroke therapy may be optimized by combination therapy with both a neuroprotective neurotrophin and an anti-inflammatory agent. In the present work, the model neurotrophin is glial cell line-derived neurotrophic factor (GDNF), and the model anti-inflammatory agent is the type II tumor necrosis factor receptor (TNFR) decoy receptor. Both the GDNF and the TNFR are large molecules that do not cross the blood-brain barrier (BBB), which is intact in the early hours after stroke when neural rescue is still possible. The GDNF and the TNFR decoy receptor were re-engineered for BBB transport as IgG fusion proteins, wherein the GDNF or the TNFR are fused to the heavy chain of a chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR), and these fusion proteins are designated cTfRMAb-GDNF and cTfRMAb-TNFR, respectively. Mice were treated intravenously with (a) saline, (b) GDNF alone, (c) the cTfRMAb-GDNF fusion protein alone, or (d) the combined cTfRMAb-GDNF and cTfRMAb-TNFR fusion proteins, following a 1-h reversible middle cerebral artery occlusion (MCAO). The cTfRMAb-GDNF fusion protein alone caused a significant 25% and 30% reduction in hemispheric and cortical stroke volumes. Combined treatment with the cTfRMAb-GDNF and cTfRMAb-TNFR fusion proteins caused a significant 54%, 69% and 30% reduction in hemispheric, cortical and subcortical stroke volumes. Conversely, intravenous GDNF had no therapeutic effect. In conclusion, combination treatment with BBB penetrating IgG-GDNF and IgG-TNFR fusion proteins enhances the therapeutic effect of single treatment with the IgG-GDNF fusion protein following delayed intravenous administration in acute stroke.
Collapse
|
10
|
Immune effects of optimized DNA vaccine and protective effects in a MPTP model of Parkinson’s disease. Neurol Sci 2013; 34:1559-70. [DOI: 10.1007/s10072-012-1284-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/17/2012] [Indexed: 11/25/2022]
|
11
|
|
12
|
Combination treatment with progesterone and vitamin D hormone is more effective than monotherapy in ischemic stroke: the role of BDNF/TrkB/Erk1/2 signaling in neuroprotection. Neuropharmacology 2012; 67:78-87. [PMID: 23154302 DOI: 10.1016/j.neuropharm.2012.10.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 09/04/2012] [Accepted: 10/09/2012] [Indexed: 02/03/2023]
Abstract
We investigated whether combinatorial post-injury treatment with progesterone (P4) and vitamin D hormone (VDH) would reduce ischemic injury more effectively than P4 alone in an oxygen glucose deprivation (OGD) model in primary cortical neurons and in a transient middle cerebral artery occlusion (tMCAO) model in rats. In the OGD model, P4 and VDH each showed neuroprotection individually, but combination of the "best" doses did not show substantial efficacy; instead, the lower dose of VDH in combination with P4 was the most effective. In the tMCAO model, P4 and VDH were given alone or in combination at different times post-occlusion for 7 days. In vivo data confirmed the in vitro findings and showed better infarct reduction at day 7 and functional outcomes (at 3, 5 and 7 days post-occlusion) after combinatorial treatment than when either agent was given alone. VDH, but not P4, upregulated heme oxygenase-1, suggesting a pathway for the neuroprotective effects of VDH differing from that of P4. The combination of P4 and VDH activated brain-derived neurotrophic factor and its specific receptor, tyrosine kinase receptor-B. Under specific conditions VDH potentiates P4's neuroprotective efficacy and should be considered as a potential partner of P4 in a low-cost, safe and effective combinatorial treatment for stroke.
Collapse
|
13
|
Yousuf S, Atif F, Sayeed I, Wang J, Stein DG. Post-stroke infections exacerbate ischemic brain injury in middle-aged rats: immunomodulation and neuroprotection by progesterone. Neuroscience 2012; 239:92-102. [PMID: 23079632 DOI: 10.1016/j.neuroscience.2012.10.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/26/2012] [Accepted: 10/05/2012] [Indexed: 01/22/2023]
Abstract
We investigated the effect of delayed, prolonged systemic inflammation on stroke outcomes and progesterone (P4) neuroprotection in middle-aged rats. After transient middle cerebral artery occlusion/reperfusion (MCAO) surgery, rats received P4 (8 or 16 mg/kg) or vehicle injections at 2h, 6h and every 24h until day 7 post-occlusion. At 24h post-injury systemic inflammation was induced by giving three doses of lipopolysaccharide (LPS; 50 μg/kg, at 4h intervals) to model post-stroke infections. We measured serum brain-derived neurotrophic factor (BDNF), pro-inflammatory cytokines, and behavioral parameters at multiple times. Serum BDNF levels decreased more in the vehicle+LPS group compared to vehicle-alone at 3 and 7 days post-injury (P<0.05). Vehicle-alone showed a significant increase in interleukin-1β, interleukin-6, and tumor necrosis factor alpha levels at different times following stroke and these levels were further elevated in the vehicle+LPS group. P4 at both doses produced a significant (P<0.05) decline in cytokine levels compared to vehicle and vehicle+LPS. P4 restored BDNF levels at 3 and 7 days post-stroke (P<0.05). Behavioral assessment (rotarod, grip strength, sensory neglect and locomotor activity tests) at 3, 5 and 7 days post-stroke revealed that the vehicle group had significant (P<0.05) deficits in all tests compared to intact controls, and performance was worse in the vehicle+LPS group. P4 at both doses produced significant functional improvement on all tests. Systemic inflammation did not show an additive effect on infarct volume but P4 at both doses showed significant infarct reduction. We suggest that post-stroke infection exacerbates stroke outcomes and P4 exerts neuroprotective/modulatory effects through its systemic anti-inflammatory and BDNF regulatory actions.
Collapse
Affiliation(s)
- S Yousuf
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
The glial cell line-derived neurotrophic factor (GDNF) was first identified as a survival factor for midbrain dopaminergic neurons, but additional studies provided evidences for a role as a trophic factor for other neurons of the central and peripheral nervous systems. GDNF regulates cellular activity through interaction with glycosyl-phosphatidylinositol-anchored cell surface receptors, GDNF family receptor-α1, which might signal through the transmembrane Ret tyrosine receptors or the neural cell adhesion molecule, to promote cell survival, neurite outgrowth, and synaptogenesis. The neuroprotective effect of exogenous GDNF has been shown in different experimental models of focal and global brain ischemia, by local administration of the trophic factor, using viral vectors carrying the GDNF gene and by transplantation of GDNF-expressing cells. These different strategies and the mechanisms contributing to neuroprotection by GDNF are discussed in this review. Importantly, neuroprotection by GDNF was observed even when administered after the ischemic injury.
Collapse
Affiliation(s)
- Emília P Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | | | | | | |
Collapse
|
15
|
Han Q, Li B, Feng H, Xiao Z, Chen B, Zhao Y, Huang J, Dai J. The promotion of cerebral ischemia recovery in rats by laminin-binding BDNF. Biomaterials 2011; 32:5077-85. [DOI: 10.1016/j.biomaterials.2011.03.072] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 03/29/2011] [Indexed: 10/18/2022]
|
16
|
Body cooling ameliorating spinal cord injury may be neurogenesis-, anti-inflammation- and angiogenesis-associated in rats. ACTA ACUST UNITED AC 2011; 70:885-93. [PMID: 20693909 DOI: 10.1097/ta.0b013e3181e7456d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Body cooling (BC) or mild hypothermia therapy (about 33°C) is reportedly effective for spinal cord injury (SCI). However, the mechanisms underlying the beneficial effects of BC remain unclear, so does BC ameliorating SCI via promoting neurogenesis, anti-inflammation, and angiogenesis. METHODS The standard rat compression SCI model was tested hypothetically in two groups: one receiving BC (33°C) and the other, normothermia (37°C). Afterward, the effects of BC therapy on the hind limb locomotion, spinal cord infarction and apoptosis, angiogenesis, neurogenesis, and inflammation in these two groups of SCI were assessed. The other group of sham SCI was used as controls. RESULTS Apoptosis (evidenced by higher numbers of terminal deoxynucleotidyl- transferase-mediated and duDP-biotin nick end-labeling-positive cells), infarct, activated inflammation (evidenced by higher levels of tumor necrosis factor-α, interleukin-1β, and myeloperoxidase), and hind limb locomotor dysfunction were inspected in the untreated (37°C) SCI rats 4 days after SCI. When compared with those of untreated SCI rats, SCI rats receiving BC (33°C) displayed lower levels of apoptosis, infarct volume, activated inflammation, and hind limb locomotor dysfunction. In addition, that BC promoted both angiogenesis (evidenced by increased numbers of both vascular endothelial growth factors and bromodeoxyuridine-positive endothelial cells) and neurogenesis (evidenced by increased numbers of both glial cell line-derived neurotrophic growth factors and bromodeoxyuridine-neuronal-specific nuclear protein double positive cells) in the injured spinal cord was evaluated 4 days after SCI. CONCLUSION BC (33°C) improved SCI outcomes by promoting angiogenesis, neurogenesis, and anti-inflammation in a rat SCI model.
Collapse
|
17
|
Degeorge ML, Marlowe D, Werner E, Soderstrom KE, Stock M, Mueller A, Bohn MC, Kozlowski DA. Combining glial cell line-derived neurotrophic factor gene delivery (AdGDNF) with L-arginine decreases contusion size but not behavioral deficits after traumatic brain injury. Brain Res 2011; 1403:45-56. [PMID: 21672665 DOI: 10.1016/j.brainres.2011.05.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/20/2011] [Accepted: 05/25/2011] [Indexed: 01/09/2023]
Abstract
Our laboratory has previously demonstrated that viral administration of glial cell line-derived neurotrophic factor (AdGDNF), one week prior to a controlled cortical impact (CCI) over the forelimb sensorimotor cortex of the rat (FL-SMC) is neuroprotective, but does not significantly enhance recovery of sensorimotor function. One possible explanation for this discrepancy is that although protected, neurons may not have been functional due to enduring metabolic deficiencies. Additionally, metabolic events following TBI may interfere with expression of therapeutic proteins administered to the injured brain via gene therapy. The current study focused on enhancing the metabolic function of the brain by increasing cerebral blood flow (CBF) with l-arginine in conjunction with administration of AdGDNF immediately following CCI. An adenoviral vector harboring human GDNF was injected unilaterally into FL-SMC of the rat immediately following a unilateral CCI over the FL-SMC. Within 30min of the CCI and AdGDNF injections, some animals were injected with l-arginine (i.v.). Tests of forelimb function and asymmetry were administered for 4weeks post-injury. Animals were sacrificed and contusion size and GDNF protein expression measured. This study demonstrated that rats treated with AdGDNF and l-arginine post-CCI had a significantly smaller contusion than injured rats who did not receive any treatment, or injured rats treated with either AdGDNF or l-arginine alone. Nevertheless, no amelioration of behavioral deficits was seen. These findings suggest that AdGDNF alone following a CCI was not therapeutic and although combining it with l-arginine decreased contusion size, it did not enhance behavioral recovery.
Collapse
Affiliation(s)
- M L Degeorge
- DePaul University, Department of Biological Sciences, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ahn M, Jin JK, Moon C, Matsumoto Y, Koh CS, Shin T. Glial cell line-derived neurotrophic factor is expressed by inflammatory cells in the sciatic nerves of Lewis rats with experimental autoimmune neuritis. J Peripher Nerv Syst 2010; 15:104-12. [DOI: 10.1111/j.1529-8027.2010.00258.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Effect of nigrostriatal damage induced by 6-hydroxydopamine on the expression of glial cell line–derived neurotrophic factor in the striatum of the rat. Neuroscience 2009; 162:148-54. [DOI: 10.1016/j.neuroscience.2009.04.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 04/08/2009] [Accepted: 04/10/2009] [Indexed: 01/18/2023]
|
20
|
Chu LF, Wang WT, Ghanta VK, Lin CH, Chiang YY, Hsueh CM. Ischemic brain cell-derived conditioned medium protects astrocytes against ischemia through GDNF/ERK/NF-kB signaling pathway. Brain Res 2008; 1239:24-35. [PMID: 18804095 DOI: 10.1016/j.brainres.2008.08.087] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 08/08/2008] [Accepted: 08/24/2008] [Indexed: 02/03/2023]
Abstract
Conditioned medium (CM) collected from cultures of ischemic microglia, astrocytes, and neurons were protective to astrocytes under the in vitro ischemic condition (deprivation of oxygen, glucose and serum). Molecular and signaling pathway(s) responsible for the CMs protective activity were investigated. Results showed that CMs from the ischemic microglia (MCM), astrocytes (ACM) and neurons (NCM) contained glial cell line-derived neurotrophic factor (GDNF), which protects astrocytes against the in vitro ischemia. Expression of extra cellular signal-regulated kinase (ERK1/2) and nuclear factor-kappa B (NF-kB) by GDNF led to the inhibition of apoptosis of the ischemic astrocytes in a caspase 3-independent manner. However, CMs- and GDNF-mediated protection of the ischemic astrocytes was protein kinase B (Akt) independent. These results provided mechanistic data regarding how GDNF- and CMs-mediated protection of the ischemic astrocytes is taking place. These observations provide information for the use of GDNF and GDNF containing CMs in the control of cerebral ischemia.
Collapse
Affiliation(s)
- Lan-Feng Chu
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | | | | | | | | | | |
Collapse
|
21
|
Chen CT, Foo NH, Liu WS, Chen SH. Infusion of human umbilical cord blood cells ameliorates hind limb dysfunction in experimental spinal cord injury through anti-inflammatory, vasculogenic and neurotrophic mechanisms. Pediatr Neonatol 2008; 49:77-83. [PMID: 18947003 DOI: 10.1016/s1875-9572(08)60017-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Human umbilical cord blood cells (HUCBCs) were used to investigate the mechanisms underlying the beneficial effects of cord blood cells in spinal cord injury (SCI). METHODS Rats were divided into three groups: (1) sham operation (laminectomy only); (2) Laminectomy+SCI+human adult peripheral blood mononucleocytes (PBMCs) (5 x 10(6)/0.3 mL); and (3) Laminectomy+SCi+HUCBCs (5 x 10(6)/0.3 mL). SCI was induced by compressing the spinal cord for 1 minute with an aneurysm clip calibrated to 55 g closing pressure. HUCBCs were infused immediately after SCI via the tail vein. Behavioral function tests measuring the maximal angle at which an animal could hold onto the inclined plane were conducted on days 1, 4 and 7 after SCI. Serum levels of tumor necrosis factor (TNF)-alpha and interleukin (IL)-10, were assayed. Furthermore, to determine if glial cell line-derived neurotrophic factor (GDNF) or vascular endothelial growth factor (VEGF) could be detected in the spinal cord injured area after systemic HUCBC infusion, analysis of these two molecules was conducted by immunofluorescence. RESULTS Systemic HUCBC infusion significantly attenuated SCI-induced hind limb dysfunction. The serum IL-10 levels were increased, but TNF-alpha levels were decreased after HUCBC infusion. Both VEGF and GDNF could be detected in the injured spinal cord after transplantation of HUCBC, but not PBMC, cells. CONCLUSION Our results demonstrate that HUCBC therapy may be beneficial for the recovery of SCI-induced hind limb dysfunction by increasing serum levels of IL-10, VEGF and GDNF in SCI rats.
Collapse
Affiliation(s)
- Chun-Ta Chen
- Department of Pediatrics and Stem Cell Lab, Chi Mei Medical Center, Yung Kung City, Tainan, Taiwan
| | | | | | | |
Collapse
|
22
|
Exogenous administration of glial cell line-derived neurotrophic factor improves recovery after spinal cord injury. Resuscitation 2008; 77:395-400. [DOI: 10.1016/j.resuscitation.2008.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 11/27/2007] [Accepted: 01/27/2008] [Indexed: 11/22/2022]
|
23
|
Kao CH, Chen SH, Chio CC, Lin MT. HUMAN UMBILICAL CORD BLOOD-DERIVED CD34+ CELLS MAY ATTENUATE SPINAL CORD INJURY BY STIMULATING VASCULAR ENDOTHELIAL AND NEUROTROPHIC FACTORS. Shock 2008; 29:49-55. [PMID: 17666954 DOI: 10.1097/shk.0b013e31805cddce] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human umbilical cord blood-derived CD34(+) cells were used to elucidate the mechanisms underlying the beneficial effects exerted by cord blood cells in spinal cord injury (SCI). Rats were divided into four groups: (1) sham operation (laminectomy only); (2) laminectomy + SCI + CD34(-) cells (5 x 10(5) human cord blood lymphocytes and monocytes that contained <0.2% CD34(+) cells); (3) laminectomy + SCI + CD34(+) cells (5 x 10(5) human cord blood lymphocytes and monocytes that contained approximately 95% CD34(+) cells); and (4) laminectomy + SCI + saline (0.3 mL). Spinal cord injury was induced by compressing the spinal cord for 1 min with an aneurysm clip calibrated to a closing pressure of 55 g. CD34 cells or saline was administered immediately after SCI via the tail vein. Behavioral tests of motor function measured by maximal angle an animal could hold to the inclined plane were conducted at days 1 to 7 after SCI. The triphenyltetrazolium chloride staining and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling assay were also conducted after SCI to evaluate spinal cord infarction and apoptosis, respectively. To elucidate whether glial cell line-derived neurotrophic factor (GDNF) or vascular endothelial growth factor (VEGF) can be secreted in spinal cord-injured area by the i.v. transplanted CD34(+) cells, analysis of spinal cord homogenate supernatants by specific enzyme-linked immunosorbent assay for GDNF or immunofluorescence for VEGF was conducted. It was found that systemic administration of CD34(+), but not CD34(-), cells significantly attenuated the SCI-induced hind limb dysfunction and spinal cord infarction and apoptosis. Both GDNF and VEGF could be detected in the injured spinal cord after transplantation of CD34(+), but not CD34(-), cells. The results indicate that CD34(+) cell therapy may be beneficial in reversing the SCI-induced spinal cord infarction and apoptosis and hindlimb dysfunction by stimulating the production of both VEGF and GDNF in a spinal cord compression model.
Collapse
Affiliation(s)
- Cheng-Hsing Kao
- Center for General Education, Southern Taiwan University of Technology, Taiwan
| | | | | | | |
Collapse
|
24
|
Yamagata K, Hakata K, Maeda A, Mochizuki C, Matsufuji H, Chino M, Yamori Y. Adenosine induces expression of glial cell line-derived neurotrophic factor (GDNF) in primary rat astrocytes. Neurosci Res 2007; 59:467-74. [PMID: 17920149 DOI: 10.1016/j.neures.2007.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 08/13/2007] [Accepted: 08/24/2007] [Indexed: 02/06/2023]
Abstract
Adenosine, which accumulates rapidly during ischemia due to the breakdown of ATP, has beneficial effects in many tissues. We examined whether adenosine induces the production of glial cell line-derived neurotrophic factor (GDNF) in cultured astrocytes. We evaluated GDNF mRNA expression and GDNF production in astrocytes cultured with adenosine and the adenosine selective receptor agonists 5-(N-ethylcarboxamido) adenosine (NECA), N(6)-cyclopentyladenosine (CPA) and 2-p-(2-carboxyethyl) phenethylamino-5'-N-ethylcarboxamindo-adenosine hydrochloride (CGS 21680). Moreover, we examined the possibility that the expression of GDNF is regulated differently in cultured astrocytes from the stroke-prone spontaneously hypertensive rat (SHRSP) than in those from Wistar Kyoto rats (WKY). In this study, we confirmed that adenosine and the selective A(2B) adenosine receptor agonist NECA induced the expression of GDNF in cultured astrocytes. The A(2B) receptor antagonist alloxazine was able to inhibit the increase in extracellular GDNF produced by adenosine. Furthermore, the amounts of GDNF produced were significantly reduced in astrocytes of the adenosine-treated SHRSP compared with those of WKY. These results indicate that adenosine induces the expression of GDNF, and adenosine A(2B) receptors participate in the regulation of GDNF levels in astrocytes. This expression was attenuated in astrocytes of SHRSP compared with those of WKY.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Science and Technology, College of Bioresource Sciences, Nihon University (NUBS), Kameino, Fujisawa, Kanagawa, Japan.
| | | | | | | | | | | | | |
Collapse
|
25
|
Chen SH, Chang FM, Chang HK, Chen WC, Huang KF, Lin MT. Human umbilical cord blood-derived CD34+ cells cause attenuation of multiorgan dysfunction during experimental heatstroke. Shock 2007; 27:663-71. [PMID: 17505307 DOI: 10.1097/01.shk.0000248593.71388.40] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Multiorgan dysfunction ensuing from severe heatstroke includes hypotension, hepatic and renal failure, hypercoagulable state, activated inflammation, and cerebral ischemia and injury. We attempted to assess whether human umbilical cord blood-derived CD34+ cell therapy improves survival during experimental heatstroke by attenuating multiorgan dysfunction. Anesthetized rats, immediately after the onset of heatstroke, were divided into 2 major groups and given CD34- or CD34+ cells (1 x 10(5)-5 x 10(5)/mL/kg body weight) i.v. They were exposed to ambient temperature of 43 degrees C to induce heatstroke. Another group of rats were exposed to room temperature (26 degrees C) and used as normothermic controls. Hypotension, hepatic and renal failure (evidenced by increased serum urea nitrogen, creatinine, aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase levels in plasma), hypercoagulable state (evidenced by increased prothrombin time, activated partial thromboplastin time, and D-dimer, and decreased platelet count and protein C in plasma), activated inflammation (evidence by increased TNF-alpha levels in serum), and cerebral dysfunction (evidenced by intracranial hypertension, cerebral hypoperfusion and hypoxia, and cerebral ischemia and injury) were monitored. When the CD34- cell-treated or untreated rats underwent heat stress, their survival time values were found to be 19 to 23 min. Resuscitation with CD34+ cells significantly improved survival time (duration, 63-291 min). As compared with normothermic controls, all CD34- cell-treated heatstroke animals displayed hypotension, hepatic and renal failure, hypercoagulable state, activated inflammation, and cerebral ischemia and injury. However, CD34+ cell therapy significantly caused attenuation of all the above-mentioned heatstroke reactions. In addition, the levels of IL-10 in plasma and glial cell line-derived neurotrophic factors in brain were all significantly increased after CD34+ cell therapy during heatstroke. Our data indicate that CD34+ cell therapy may resuscitate persons who had a heatstroke by reducing multiorgan dysfunction or failure.
Collapse
Affiliation(s)
- Sheng-Hsien Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
26
|
Yuan QL, Yang CX, Xu P, Gao XQ, Deng L, Chen P, Sun ZL, Chen QY. Neuroprotective effects of ginsenoside Rb1 on transient cerebral ischemia in rats. Brain Res 2007; 1167:1-12. [PMID: 17663984 DOI: 10.1016/j.brainres.2007.06.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 05/28/2007] [Accepted: 06/03/2007] [Indexed: 02/05/2023]
Abstract
Previous experiments showed that ginsenoside Rb1 (GRb1) reduced infarct and neuronal deficit in rats followed by transient cerebral ischemia. The mechanism of this neuroprotective function is unclear. Here, we tested whether the effect of GRb1 can be achieved through preventing ischemic neuronal death, modulating apoptotic-related genes and affecting glial-derived neurotrophic factor (GDNF) expression in rats subjected to occlusion of the middle cerebral artery. When GRb1(40 mg/kg, i.p.) was administered immediately after reperfusion, the apoptotic cells in the GRb1 group were decreased significantly from 12 to 72 h of reperfusion compared to the ischemia group by TdT-mediated dUTP-biotin nick-end labeling. Immunostaining and Western blotting analysis showed that the expression of GDNF from 3 to 120 h of the GRb1 group was significantly increased compared to the ischemia group, and GDNF expression peaked at 48 h after reperfusion. The enhanced GDNF mRNA in the GRb1 group was not detected by RT-PCR and in situ hybridization compared to the ischemia group, but GDNF mRNA at 48 h after reperfusion was strongly increased in both the ischemia and GRb1 group when compared to other time points. The number of bcl-2-positive cells was significantly increased from 12 to 120 h of reperfusion compared to the ischemia group. However, the number of bax-positive cells in the GRb1 group was significantly declined compared to the ischemia group. In the GRb1 group, the number of neuronal apoptosis inhibitory protein-positive cells from 12 to 120 h after reperfusion was evidently higher than that in the ischemia group. Therefore, ginsenoside Rb1 prevents ischemic neuronal death induced by transient cerebral ischemia, and this mechanism of which is related to increase the expression of the antiapoptotic genes and modulate the expression of GDNF.
Collapse
Affiliation(s)
- Q-L Yuan
- Department of Anatomy, Luzhou Medical College, Luzhou, and Laboratory of Ophthalmology Molecular Genetics, West China Hospital, Sichuan University, Chengdu, China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen SH, Huang KF, Lin MT, Chang FM. Human umbilical cord blood cells or estrogen may be beneficial in treating heatstroke. Taiwan J Obstet Gynecol 2007; 46:15-25. [PMID: 17389184 DOI: 10.1016/s1028-4559(08)60101-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This current review summarized animal models of heatstroke experimentation that promote our current knowledge of therapeutic effects on cerebrovascular dysfunction, coagulopathy, and/or systemic inflammation with human umbilical cord blood cells (HUCBCs) or estrogen in the setting of heatstroke. Accumulating evidences have demonstrated that HUCBCs provide a promising new therapeutic method against neurodegenerative diseases, such as stroke, traumatic brain injury, and spinal cord injury as well as blood disease. More recently, we have also demonstrated that post- or pretreatment by HUCBCs may resuscitate heatstroke rats with by reducing circulatory shock, and cerebral nitric oxide overload and ischemic injury. Moreover, CD34+ cells sorted from HUCBCs may improve survival by attenuating inflammatory, coagulopathy, and multiorgan dysfunction during experimental heatstroke. Many researchers indicated pro- (e.g. tumor necrosis factor-alpha [TNF-alpha]) and anti-inflammatory (e.g. interleukin-10 [IL-10]) cytokines in the peripheral blood stream correlate with severity of circulatory shock, cerebral ischemia and hypoxia, and neuronal damage occurring in heatstroke. It has been shown that intravenous administration of CD34+ cells can secrete therapeutic molecules, such as neurotrophic factors, and attenuate systemic inflammatory reactions by decreasing serum TNF-alpha but increasing IL-10 during heatstroke. Another line of evidence has suggested that estrogen influences the severity of injury associated with cerebrovascular shock. Recently, we also successfully demonstrated estrogen resuscitated heatstroke rats by ameliorating systemic inflammation. Conclusively, HUCBCs or estrogen may be employed as a beneficial therapeutic strategy in prevention and repair of cerebrovascular dysfunction, coagulopathy, and/or systemic inflammation during heatstroke.
Collapse
Affiliation(s)
- Sheng-Hsien Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan, Taiwan
| | | | | | | |
Collapse
|
28
|
Horita Y, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. Intravenous administration of glial cell line-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in the adult rat. J Neurosci Res 2007; 84:1495-504. [PMID: 16998918 PMCID: PMC2605367 DOI: 10.1002/jnr.21056] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intravenous administration of human mesenchymal stem cells (hMSCs) prepared from adult bone marrow has been reported to ameliorate functional deficits after cerebral artery occlusion in rats. Several hypotheses to account for these therapeutic effects have been suggested, and current thinking is that neuroprotection rather than neurogenesis is responsible. To enhance the therapeutic benefits of hMSCs potentially, we transfected hMSCs with the glial cell line-derived neurotrophic factor (GDNF) gene using a fiber-mutant F/RGD adenovirus vector and investigated whether GDNF gene-modified hMSCs (GDNF-hMSCs) could contribute to functional recovery in a rat permanent middle cerebral artery occlusion (MCAO) model. We induced MCAO by using intraluminal vascular occlusion, and GDNF-hMSCs were intravenously infused into the rats 3 hr later. MRI and behavioral analyses revealed that rats receiving GDNF-hMSCs or hMSCs exhibited increased recovery from ischemia compared with the control group, but the effect was greater in the GDNF-hMSC group. Thus, these results suggest that intravenous administration of hMSCs transfected with the GDNF gene using a fiber-mutant adenovirus vector may be useful in the cerebral ischemia and may represent a new strategy for the treatment of stroke.
Collapse
Affiliation(s)
- Yoshifumi Horita
- Department of Neurosurgery, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Lee YJ, Jin JK, Jeong BH, Carp RI, Kim YS. Increased expression of glial cell line-derived neurotrophic factor (GDNF) in the brains of scrapie-infected mice. Neurosci Lett 2006; 410:178-82. [PMID: 17101222 DOI: 10.1016/j.neulet.2006.09.090] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 09/07/2006] [Accepted: 09/08/2006] [Indexed: 11/27/2022]
Abstract
Prion diseases, also called transmissible spongiform encephalopathies (TSEs), are fatal neurodegenerative disorders characterized by neuronal loss, astrogliosis, and spongiform changes in the brain. It is postulated that appearance of astrogliosis may provide the neurotrophic factors to prevent or reduce neuronal cell loss in the pathogenesis of prion diseases. To investigate the role of the glial cell line-derived neurotrophic factor (GDNF), we studied the expression levels of GDNF mRNA and protein in an animal model of prion diseases. The expression levels of GDNF mRNA and protein were significantly increased in the brains of scrapie-infected mice at 100 and 160 days after inoculation with scrapie strain compared with those of control mice. In addition, we found more intensive immunoreactivity of GDNF in the brains of scrapie-infected mice, specifically in the hippocampal astrocytes, than was seen in control mice. These results suggest that GDNF participates in protection against neuronal cell loss and atrophy in neurodegenerative disorders, which may play one of the important roles in the pathogenic mechanisms of prion diseases.
Collapse
Affiliation(s)
- Yun-Jung Lee
- Ilsong Institute of Life Science, College of Medicine, Hallym University, Anyang, Kyonggi-do 431-060, South Korea
| | | | | | | | | |
Collapse
|
30
|
Kilic U, Kilic E, Dietz GPH, Bähr M. The TAT protein transduction domain enhances the neuroprotective effect of glial-cell-line-derived neurotrophic factor after optic nerve transection. NEURODEGENER DIS 2006; 1:44-9. [PMID: 16908973 DOI: 10.1159/000076669] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2003] [Accepted: 09/29/2003] [Indexed: 12/18/2022] Open
Abstract
Glial-cell-line-derived neurotrophic factor (GDNF) acts as a potent survival factor for many neuronal populations, including retinal ganglion cells (RGC), indicating a potential therapeutic role of GDNF for neurological disorders. To enhance the tissue distribution and applicability of the neurotrophin, we linked it to a protein transduction domain derived from the HIV TAT protein and tested it in a well-established model for traumatic injury in the CNS: After optic nerve axotomy, the number of surviving RGCs was significantly increased in mice injected with TAT-GDNF on days 0, 3, 7, and 10 after surgery compared with GDNF- or PBS-injected animals. Moreover, TAT-GDNF reduced the number of activated caspase-3-positive cells. These results show that the neuroprotective effect of substances like neurotrophins may be enhanced by linking them to a domain that has been shown to mediate efficient transduction across biological membranes.
Collapse
Affiliation(s)
- Ulkan Kilic
- Department of Neurology, Georg August University Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
31
|
Lin CH, Cheng FC, Lu YZ, Chu LF, Wang CH, Hsueh CM. Protection of ischemic brain cells is dependent on astrocyte-derived growth factors and their receptors. Exp Neurol 2006; 201:225-33. [PMID: 16765947 DOI: 10.1016/j.expneurol.2006.04.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 04/03/2006] [Accepted: 04/05/2006] [Indexed: 01/14/2023]
Abstract
An in vitro ischemia model (oxygen, glucose, and serum deprivation) is used to investigate the possible cellular and molecular mechanisms responsible for cerebral ischemia. We have previously demonstrated that supernatants derived from ischemic microglia can protect ischemic brain cells by releasing GDNF and TGF-beta1. In the present study, we investigate whether products of ischemic astrocytes can also protect ischemic microglia, astrocytes, and neurons in a similar manner. Supernatants from ischemic astrocytes were collected after various periods of ischemia and incubated with microglia, astrocytes, or neurons individually, under in vitro ischemic conditions. The components responsible for the protective effects of astrocyte-derived supernatants were then identified by Western blot, ELISA, trypan blue dye exclusion, and immunoblocking assays. Results showed that under conditions of in vitro ischemia the number of surviving microglia, astrocytes, and neurons was significantly increased by the incorporation of the astrocyte-derived supernatants. Astrocyte supernatant-mediated protection of ischemic microglia was dependent on TGF-beta1 and NT-3, ischemic astrocytes were protected by GDNF, and ischemic neurons were protected by NT-3. In addition, protein expression of TGF-beta1 and NT-3 receptors in microglia, GDNF receptors in astrocytes, and NT-3 receptors in neurons was increased by in vitro ischemia. These results suggest that astrocyte-derived protection of ischemic brain cells is dependent not only on factors released from the ischemic astrocytes, but also on the type of receptor present on the responding cells. Therapeutic potential of TGF-beta1, GDNF, and NT-3 in the control of cerebral ischemia is further suggested.
Collapse
Affiliation(s)
- Chi-Hsin Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Villadiego J, Méndez-Ferrer S, Valdés-Sánchez T, Silos-Santiago I, Fariñas I, López-Barneo J, Toledo-Aral JJ. Selective glial cell line-derived neurotrophic factor production in adult dopaminergic carotid body cells in situ and after intrastriatal transplantation. J Neurosci 2006; 25:4091-8. [PMID: 15843611 PMCID: PMC6724965 DOI: 10.1523/jneurosci.4312-04.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) exerts a notable protective effect on dopaminergic neurons in rodent and primate models of Parkinson's disease (PD). The clinical applicability of this therapy is, however, hampered by the need of a durable and stable GDNF source allowing the safe and continuous delivery of the trophic factor into the brain parenchyma. Intrastriatal carotid body (CB) autografting is a neuroprotective therapy potentially useful in PD. It induces long-term recovery of parkinsonian animals through a trophic effect on nigrostriatal neurons and causes amelioration of symptoms in some PD patients. Moreover, the adult rodent CB has been shown to express GDNF. Here we show, using heterozygous GDNF/lacZ knock-out mice, that unexpectedly CB dopaminergic glomus, or type I, cells are the source of CB GDNF. Among the neural or paraneural cells tested, glomus cells are those that synthesize and release the highest amount of GDNF in the adult rodent (as measured by standard and in situ ELISA). Furthermore, GDNF expression by glomus cells is maintained after intrastriatal grafting and in CB of aged and parkinsonian 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated animals. Thus, glomus cells appear to be prototypical abundant sources of GDNF, ideally suited to be used as biological pumps for the endogenous delivery of trophic factors in PD and other neurodegenerative diseases.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology
- Age Factors
- Analysis of Variance
- Animals
- Animals, Newborn
- Carotid Body/cytology
- Carotid Body/metabolism
- Carotid Body/ultrastructure
- Cell Differentiation
- Cells, Cultured
- Corpus Striatum/transplantation
- Disease Models, Animal
- Dopamine/metabolism
- Enzyme-Linked Immunosorbent Assay/methods
- Glial Cell Line-Derived Neurotrophic Factor/genetics
- Glial Cell Line-Derived Neurotrophic Factor/metabolism
- Glial Fibrillary Acidic Protein/metabolism
- Immunohistochemistry/methods
- MPTP Poisoning/metabolism
- MPTP Poisoning/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron, Transmission/methods
- Neurons/metabolism
- Neurons/transplantation
- Neurons/ultrastructure
- PC12 Cells
- Rats
- Rats, Wistar
- Time Factors
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Javier Villadiego
- Laboratorio de Investigaciones Biomédicas, Departamento de Fisiología and Hospital Universitario Virgen del Rocío, Universidad de Sevilla, 41013 Sevilla, Spain
| | | | | | | | | | | | | |
Collapse
|
33
|
Hwang IK, Yoo KY, Kim DW, Lee BH, Kang TC, Choi SY, Han BH, Kim JS, Won MH. Ischemia-related changes of glial-derived neurotrophic factor and phosphatidylinositol 3-kinase in the hippocampus: Their possible correlation in astrocytes. Brain Res 2006; 1072:215-23. [PMID: 16412399 DOI: 10.1016/j.brainres.2005.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 11/29/2005] [Accepted: 12/05/2005] [Indexed: 11/18/2022]
Abstract
In the present study, we observed the changes of endogenous expression of glial-cell-line-derived neurotrophic factor (GDNF) and phosphatidylinositol 3-kinase (PI-3 kinase) in the gerbil hippocampus after transient forebrain ischemia and investigated the correlation between GDNF and PI-3 kinase in the ischemic hippocampus. In the sham-operated group, GDNF and PI-3 kinase immunoreactivity was not found in any cells in the hippocampal CA1 region. GDNF, not PI-3 kinase, immunoreactivity was expressed in non-pyramidal cells in the CA1 region at 6 h after ischemic insult. At 12-24 h after ischemia, GDNF and PI-3 kinase immunoreactivity in the CA1 region was similar to that of the sham-operated group. From 2 days after ischemic insult, GDNF- and PI-3-kinase-immunoreactive astrocytes were detected in the CA1 region, and GDNF and PI-3 kinase immunoreactivity in astrocytes was highest in the CA1 region 4 days after ischemic insult. Moreover, at this time point, GDNF and PI-3 kinase were co-localized in some astrocytes. Western blotting showed that ischemia-related changes of GDNF and PI-3 kinase protein levels were similar to the immunohistochemical changes after ischemia. These results suggest that GDNF and PI-3 kinase may be related to delayed neuronal death and that GDNF and PI-3 kinase may be involved in activation of astrocytes.
Collapse
Affiliation(s)
- In Koo Hwang
- Department of Anatomy, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Trendelenburg G, Dirnagl U. Neuroprotective role of astrocytes in cerebral ischemia: focus on ischemic preconditioning. Glia 2005; 50:307-320. [PMID: 15846804 DOI: 10.1002/glia.20204] [Citation(s) in RCA: 204] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Following focal cerebral ischemia ("stroke") a complex and dynamic interaction of vascular cells, glial cells, and neurons determines the extent of the ensuing lesion. Traditionally, the focus has been on mechanisms of damage, while recently it has become clear that endogenous mechanisms of protection are equally important for the final outcome. Glial cells, in particular astrocytes, have always been viewed as supporters of neuronal function. Only recently a very active role for glial cells has been emerging in physiology and pathophysiology. Not surprisingly, then, specific protective pathways have been identified by which these cells can protect or even help to regenerate brain tissue after acute insults. However, as exemplified by the existence of the glial scar, which forms around lesioned brain tissue, is composed mainly of astrocytes and plays a key role in regeneration failure, it is an oversimplification to assign merely protective functions to astrocytes. The present review will discuss the role of astrocytes in ischemic brain injury with a focus on neuroprotection in general. In this context we will consider particularly the phenomenon of "ischemic tolerance," which is an experimental paradigm helpful in discriminating destructive from protective mechanisms after cerebral ischemia.
Collapse
Affiliation(s)
| | - Ulrich Dirnagl
- Department of Neurology, Charité, Humboldt University, Berlin, Germany
| |
Collapse
|
35
|
Sellner J, Lenhard T, Haas J, Einsiedel RV, Meyding-Lamadé U. Differential mRNA expression of neurotrophic factors GDNF, BDNF, and NT-3 in experimental herpes simplex virus encephalitis. ACTA ACUST UNITED AC 2005; 137:267-71. [PMID: 15950786 DOI: 10.1016/j.molbrainres.2005.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2004] [Revised: 02/26/2005] [Accepted: 03/13/2005] [Indexed: 01/26/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) mRNA levels were studied in the course of murine herpes simplex virus encephalitis. Induction of GNDF and NT-3 (both P < 0.05) was found during acute encephalitis. Despite absence of clinical impairment, both neurotrophic factors were overexpressed 2 months (NT-3) and 6 months (GDNF) following infection (both P < 0.05). Neurotrophic factors play an important role in neuronal survival and recovery after acute injury to the central nervous system (CNS) and may represent an additional therapeutic target for treatment of viral encephalitis.
Collapse
Affiliation(s)
- Johann Sellner
- Department of Neurology, Ruprecht-Karls-University Heidelberg, D-69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
36
|
Cheng H, Huang SS, Lin SM, Lin MJ, Chu YC, Chih CL, Tsai MJ, Lin HC, Huang WC, Tsai SK. The neuroprotective effect of glial cell line-derived neurotrophic factor in fibrin glue against chronic focal cerebral ischemia in conscious rats. Brain Res 2005; 1033:28-33. [PMID: 15680336 DOI: 10.1016/j.brainres.2004.10.067] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2004] [Indexed: 11/24/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a transforming growth factor-beta which has shown beneficial effects in rats after acute focal cerebral ischemia (FCI). To study the effects of GDNF on chronic FCI injury in conscious rats, we used fibrin glue (GDNF-fibrin glue) and fibrin glue free (GDNF-only)-GDNF topically applied to the ischemic brain after right middle cerebral artery (MCA) ligation. Infarct brain volume and functional motor deficits were measured before and after FCI injury. After FCI injury induced by right MCA ligation, rats were randomly assigned to one of four treatment groups: (a) sham, (b) control, (c) topically applied GDNF (1 mug)-only, and (d) topically applied GDNF (1 mug)-fibrin glue. The degree of ischemic brain injury was estimated by infarct volume of right MCA territory at 4 weeks after occlusion. The functional motor deficits were quantified with rotarod test and grasping power test once a week. Topically applied GDNF-fibrin glue at infarct brain tissue after 4 weeks FCI injury significantly reduced the total infarct volume by 44.3% and 36%, respectively, compared to that of control group and GDNF-only group. The mean latencies for rats to stay on the rotarod were 55.0%, 50.3%, and 92.2% (P < 0.05 vs. control group and GDNF-only group) of baseline, respectively, in the control, GDNF-only, and GDNF-fibrin glue groups at the end of the 1st week after FCI injury but 75.3%, 67.3%, and 106.6% (P < 0.05 vs. control group and GDNF-only group) of baseline at the end of the 4th week after FCI injury. The mean values of grasping power were 78.7%, 71.7%, and 101.2% (P < 0.05 vs. control group and GDNF-only group) of baseline, respectively, in the control, GDNF-only, and GDNF-fibrin glue groups at the end of 1st week after FCI injury but 89.6%, 97.6%, and 120.7% (P < 0.05 vs. control group) of baseline at the end of 4th week after FCI injury. These results indicate that GDNF-fibrin glue not only reduced the total infarct volume after FCI injury but can also improve motor deficits after FCI injury. We concluded GDNF-fibrin glue could facilitate delivery of GDNF to the damaged brain tissue with subsequent reduction of ischemic brain injury accompanied by enhancing functional recovery in rats with chronic FCI injury.
Collapse
Affiliation(s)
- Henrich Cheng
- Center for Neural Regeneration, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang D, Desbois A, Hou ST. A novel adenoviral vector which mediates hypoxia-inducible gene expression selectively in neurons. Gene Ther 2005; 12:1369-76. [PMID: 15843806 DOI: 10.1038/sj.gt.3302538] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Selective gene expression in neurons is still a challenge. We have developed several expression vectors using a combination of neuron restrictive silencer elements (NRSEs), hypoxia responsive elements (HREs) and CMV minimal promoter (CMVmp). These elements were packaged into replication defective adenovirus to target gene expression selectively in neurons in a hypoxia-regulated manner. Neuronal selectivity and responsiveness to hypoxia of these novel constructs were determined empirically in both neural cell lines and primary cerebellar granule neurons (CGNs). The construct p5HRE-3NRSE exhibited not only the highest level of reporter gene expression in neuronal cells but also in an oxygen concentration-dependent manner when compared with all other constructs. As expected, this construct did not elicit reporter gene expression in non-neuronal cells including human HEK293A and HT29 cells, rat NRK cells, mouse 3T6 cells and 3T3 L1 cells. This construct was packaged into a replication defective adenoviral vector (Ad/5HRE-3NRSE) to determine neuron-selective and hypoxia-inducible gene expression in cultured mouse postmitotic primary CGNs and differentiated human NT2 neurons (NT2/Ns). Remarkably, in response to hypoxia, Ad/5HRE-3NRSE showed strong hypoxia-inducible gene expression selectively in neurons (12-fold induction in CGNs and 22-fold in NT2/Ns), but not in glial cells. Taken together, this vector with restricted gene expression to neurons under the regulation of hypoxia will be a useful tool for investigations of mechanisms of neuronal damage caused by ischemic insult.
Collapse
Affiliation(s)
- D Huang
- Experimental Therapeutics Laboratory, NRC Institute for Biological Sciences, National Research Council of Canada, 1500 Montreal Road, Ottawa, Ontario, Canada K1A 0R6
| | | | | |
Collapse
|
38
|
Oo TF, Ries V, Cho J, Kholodilov N, Burke RE. Anatomical basis of glial cell line-derived neurotrophic factor expression in the striatum and related basal ganglia during postnatal development of the rat. J Comp Neurol 2005; 484:57-67. [PMID: 15717300 PMCID: PMC3092474 DOI: 10.1002/cne.20463] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
There is increasing evidence that glial cell line-derived neurotrophic factor (GDNF) plays a role as a limiting, striatal target-derived neurotrophic factor for dopamine neurons of the substantia nigra pars compacta (SNpc) by regulating the magnitude of the first phase of postnatal natural cell death which occurs in these neurons. While it has been shown that GDNF mRNA is relatively abundant in postnatal striatum, the cellular basis of its expression has been unknown. We therefore used nonradioactive in situ hybridization and immunohistochemistry to examine the cellular basis of GDNF mRNA and protein expression, respectively, in postnatal striatum and related structures. We found that GDNF mRNA is expressed within medium-sized striatal neurons. Expression in glia was not observed. At the protein level, regionally, GDNF expression in striatum was observed in striosomal patches, as previously described. At a cellular level a few neurons were observed, but they do not account for the striosomal pattern. This pattern is predominantly due to GDNF-positive neuropil. Some of this neuropil arises from tyrosine hydroxylase-positive nigro-striatal dopaminergic afferents. Astrocytic processes do not appear to contribute to the striosomal pattern. GDNF-positive fibers are identified not only within intrinsic striatal neuropil, but also in fibers within the major striatal efferent targets: the globus pallidus, the entopeduncular nucleus, and the SN pars reticulata. We conclude that during normal postnatal development, medium-sized neurons are the principal source of GDNF within the striatum.
Collapse
Affiliation(s)
- Tinmarla Frances Oo
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Vincent Ries
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Jinwhan Cho
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Nikolai Kholodilov
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Robert E. Burke
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
- Department of Pathology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| |
Collapse
|
39
|
Chou AK, Yang LC, Wu PC, Wong WT, Liu GS, Chen JT, Howng SL, Tai MH. Intrathecal gene delivery of glial cell line-derived neurotrophic factor ameliorated paraplegia in rats after spinal ischemia. ACTA ACUST UNITED AC 2005; 133:198-207. [PMID: 15710236 DOI: 10.1016/j.molbrainres.2004.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2004] [Indexed: 11/28/2022]
Abstract
Paraplegia is a catastrophic complication of thoracic aortic surgery. At present, there is no effective mean to prevent the ischemia-induced spinal cord trauma. Gene delivery of neurotrophic factors may hold promises for prevention of spinal injury. In the present study, we evaluated the effect of glial cell line-derived neurotrophic factor (GDNF) gene delivery on prevention of the pathological changes due to spinal ischemia. Recombinant adenovirus vectors encoding GDNF (Ad-GDNF) and green fluorescent protein (Ad-GFP) were used for gene transfer studies. Treatment with cobalt chloride induced dose-dependent bcl-2 and synaptophysin downregulation in spinal neuronal cells, which could be effectively reversed by GDNF gene transfer. Intrathecal injection of Ad-GDNF led to maximal GDNF expression in spinal cord within 2-7 days. Thus, after intrathecal administration of adenovirus vectors for 3 days, Sprague-Dawley rats received transient aortic occlusion to induce spinal ischemia and were monitored for behavior deficits. The Ad-GDNF-treated rats showed significantly lower paraplegia rate (40%) than that of Ad-GFP- or saline-treated groups (75-85%; P<0.01). In addition, the Ad-GDNF-treated rats exhibited significantly improved locomotor function comparing with rats of control groups (P<0.001). Histological analysis revealed that GDNF gene delivery profoundly attenuated the infiltration of leukocytes in spinal cord after ischemic insults. Furthermore, GDNF gene delivery prominently attenuated the ischemia-induced neuronal loss in dorsal horn lamina VI-VIII and reduction in synaptophysin expression in spinal cords. In conclusion, GDNF gene transfer confers protection to the neuronal cells and synapses networks, thereby alleviated the paraplegia due to spinal ischemia.
Collapse
Affiliation(s)
- An Kuo Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lu YZ, Lin CH, Cheng FC, Hsueh CM. Molecular mechanisms responsible for microglia-derived protection of Sprague-Dawley rat brain cells during in vitro ischemia. Neurosci Lett 2005; 373:159-64. [PMID: 15567573 DOI: 10.1016/j.neulet.2004.10.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2004] [Revised: 09/30/2004] [Accepted: 10/02/2004] [Indexed: 11/17/2022]
Abstract
Microglia-derived protection of brain cells (microglia, astrocytes, and neurons) during in vitro ischemic stress (deprivation of glucose, oxygen, and serum) was determined. Trypan blue exclusion assay, immunoblocking assay, Western blot analysis, and ELISA assay were used to determine the molecular mechanisms responsible for the microglia-derived protection. Results demonstrated that supernatants from the ischemic microglia protected all three cell-types from ischemia-induced damage by releasing the transforming growth factor-beta1 (TGF-beta1) and glial cell line-derived neurotrophic factor (GDNF). The protection of microglia was TGF-beta1 related, whereas astrocytes protection was GDNF-dependent. The protection of neurons was TGF-beta1 and GDNF independent, and the molecular nature responsible for their protection remains to be determined. These results indicate contribution from the surrounding cells and the types of receptors expressed on different brain cells probably also play an important role in determining their fate against ischemia.
Collapse
Affiliation(s)
- Yen-Zhen Lu
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan ROC
| | | | | | | |
Collapse
|
41
|
Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Ishii K, Kobune M, Hirai S, Uchida H, Sasaki K, Ito Y, Kato K, Honmou O, Houkin K, Date I, Hamada H. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther 2005; 11:96-104. [PMID: 15585410 DOI: 10.1016/j.ymthe.2004.09.020] [Citation(s) in RCA: 299] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2004] [Accepted: 09/28/2004] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cells (MSC) were reported to ameliorate functional deficits after stroke in rats, with some of this improvement possibly resulting from the action of cytokines secreted by these cells. To enhance such cytokine effects, we previously transfected the telomerized human MSC with the BDNF gene using a fiber-mutant adenovirus vector and reported that such treatment contributed to improved ischemic recovery in a rat transient middle cerebral artery occlusion (MCAO) model. In the present study, we investigated whether other cytokines in addition to BDNF, i.e., GDNF, CNTF, or NT3, might have a similar or greater effect in this model. Rats that received MSC-BDNF (P < 0.05) or MSC-GDNF (P < 0.05) showed significantly more functional recovery as demonstrated by improved behavioral test results and reduced ischemic damage on MRI than did control rats 7 and 14 days following MCAO. On the other hand, rats that received MSC-CNTF or MSC-NT3 showed neither functional recovery nor ischemic damage reduction compared to control rats. Thus, MSC transfected with the BDNF or GDNF gene resulted in improved function and reduced ischemic damage in a rat model of MCAO. These data suggest that gene-modified cell therapy may be a useful approach for the treatment of stroke.
Collapse
Affiliation(s)
- Kazuhiko Kurozumi
- Department of Molecular Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schmidt KM, Repine MJ, Hicks SD, DeFranco DB, Callaway CW. Regional changes in glial cell line-derived neurotrophic factor after cardiac arrest and hypothermia in rats. Neurosci Lett 2004; 368:135-9. [PMID: 15351435 DOI: 10.1016/j.neulet.2004.06.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Revised: 06/28/2004] [Accepted: 06/29/2004] [Indexed: 02/09/2023]
Abstract
Hypothermia after resuscitation from cardiac arrest reduces functional and histological brain injury. Stimulation of neurotrophic factors may contribute to the beneficial effects of hypothermia. This study examined the effects of cardiac arrest and induced hypothermia on regional levels of glial cell line-derived neurotrophic factor (GDNF) over the first 24 h after rat cardiac arrest. Hypothermia increased GDNF in hippocampus at 6 h, but did not prevent a subsequent decline in hippocampal GDNF. In contrast, hypothermia prevented early increases in cortical levels of GDNF at 3 and 6 h. Cerebellar GDNF increased slightly over 24 h in hypothermia-treated rats, but brainstem levels of GDNF did not change in response to cardiac arrest or hypothermia. These results suggest that temperature after resuscitation produces regionally specific changes of GNDF levels in brain.
Collapse
Affiliation(s)
- Katherine M Schmidt
- Department of Emergency Medicine, University of Pittsburgh, 230 McKee Place, Suite 400, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
43
|
Harvey BK, Hoffer BJ, Wang Y. Stroke and TGF-beta proteins: glial cell line-derived neurotrophic factor and bone morphogenetic protein. Pharmacol Ther 2004; 105:113-25. [PMID: 15670622 DOI: 10.1016/j.pharmthera.2004.09.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Accepted: 09/24/2004] [Indexed: 10/26/2022]
Abstract
Recent studies have indicated that proteins in the transforming growth factor-beta superfamily alter damage induced by various neuronal injuries. Of these proteins, glial cell line-derived neurotrophic factor (GDNF) and bone morphogenetic protein-7 (BMP-7) have unique protective and regenerative effects in stroke animals. Delivery of GDNF or BMP-7 to brain tissue reduced cerebral infarction and improved motor functions in stroke animals. Pretreatment with these factors reduced caspase-3 activity and DNA fragmentation in the ischemic brain region, suggesting that antiapoptotic effects are involved. Beside the protective effects, BMP-7 given after stroke improves locomotor function. These regenerative effects of BMP-7 may involve the enhancement of dendritic growth and remodeling. In this review, we illustrate the neuroprotective and neuroregenerative properties of GDNF and BMP-7 and emphasize their therapeutic potential for stroke.
Collapse
Affiliation(s)
- Brandon K Harvey
- Neural Protection and Regeneration Section, Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, NIH, Baltimore, MD 21124, USA
| | | | | |
Collapse
|
44
|
Shyu WC, Lin SZ, Chiang MF, Yang HI, Thajeb P, Li H. Neuregulin-1 reduces ischemia-induced brain damage in rats. Neurobiol Aging 2004; 25:935-44. [PMID: 15212847 DOI: 10.1016/j.neurobiolaging.2003.10.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2003] [Revised: 09/22/2003] [Accepted: 10/21/2003] [Indexed: 11/24/2022]
Abstract
Neuregulin-1 (NRG-1) is expressed throughout the immature and adult central nervous system and it has been demonstrated to influence the migration of a variety of cell types in developing brain. Elevated levels of NRG-1 transcript are found in the adult brain after injury, leading to the suggestion that NRG-1 is involved in the physiological response to neuronal injury. Here, we report our findings that rats pre-treated with NRG-1 protein, undergoing cerebral ischemia 30 min later, had increased motor performance and less cerebral infarction than untreated rats. In the cortex of NRG-1 treated rats, ischemia induced a decrease in caspase-3 immunoreactivity and a reduction in the density of cells positive for terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end-labeling. Improvement in behavioral assays was also found in animals treated with NRG-1. Pre-treatment with NRG-1 did not alter cerebral blood flow or other physiological parameters. NRG-1 reduced ischemia/reperfusion injury, indicating that it may act as an endogenous neuroprotective factor against stroke. Therefore, NRG-1 may represent a target for the development of new treatments for stroke.
Collapse
Affiliation(s)
- Woei-Cherng Shyu
- Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
45
|
Lee GA, Lin CH, Jiang HH, Chao HJ, Wu CL, Hsueh CM. Microglia-derived glial cell line-derived neurotrophic factor could protect Sprague-Dawley rat astrocyte from in vitro ischemia-induced damage. Neurosci Lett 2004; 356:111-4. [PMID: 14746876 DOI: 10.1016/j.neulet.2003.11.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The interrelationship between microglia and astrocytes in cerebral ischemia was determined in vitro by adding in vitro ischemia-induced supernatant from microglia into astrocytes under the same conditions (glucose-, oxygen- and serum-free). The involvement of glial cell line-derived neurotrophic factor (GDNF) was further investigated by immunoblocking assay and Western blot analysis. Results showed that microglia-derived supernatant protected against in vitro ischemia-induced damage of astrocytes and this protection was pre-blocked by anti-GDNF but not normal rabbit serum. In addition, in vitro ischemia appeared to induce the expression of GDNF in microglia. These results indicate that microglia-derived protection on astrocytes during in vitro ischemia is GDNF-dependent.
Collapse
Affiliation(s)
- Gilbert A Lee
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- K Abe
- Department of Neurology, Graduate School of Medicine and Dentistry, Okayama University, Okayama 700-8558, Japan
| | | |
Collapse
|
47
|
Sarabi A, Chang CF, Wang Y, Tomac AC, Hoffer BJ, Morales M. Differential expression of the cell line-derived neurotrophic factor (GDNF) receptor GFRalpha1 in heterozygous Gfralpha1 null-mutant mice after stroke. Neurosci Lett 2003; 341:241-5. [PMID: 12697293 DOI: 10.1016/s0304-3940(03)00195-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Exogenous administration of glial cell line-derived neurotrophic factor (GDNF) reduces ischemia-induced cerebral infarction. Cerebral ischemia induces gene expression of GDNF, GDNF-receptor alpha-1 (GFRalpha-1) and c-Ret, suggesting that a GDNF signaling cascade mechanism may be involved in endogenous neuroprotection during ischemia. In the present study, we examined if this endogenous neuroprotective pathway was altered in Gfralpha-1 deficient mice. Since mice homozygous for the Gfralpha-1 deletion (-/-) die within 24 h of birth, stroke-induced changes in the levels of Gfralpha-1 mRNA were studied in Gfralpha-1 heterozygous (+/-) mice and their wild-type (+/+) littermates. The right middle cerebral artery was transiently ligated for 45 min in anesthetized mice. Animals were killed at 0, 6, 12 and 24 h after the onset of reperfusion and levels of Gfralpha-1 mRNA were measured by in situ hybridization histochemistry. Previously, we showed that Gfralpha-1 (+/-) mice are more vulnerable to focal cerebral ischemia. In the present study, we found that basal levels of GFRalpha-1 mRNA were at similar low levels in cortex and striatum in adult Gfralpha-1 (+/+) and Gfralpha-1 (+/-) mice and that ischemia/reperfusion induced up-regulation of Gfralpha-1 mRNA in the lesioned and contralateral sides of cortex and striatum in both Gfralpha-1 (+/+) and GFRalpha-1 (+/-) mice. However, the ischemia/reperfusion induction of Gfralpha-1 mRNA was significantly higher in the cortex of wild type mice, as compared to Gfralpha-1 (+/-) mice. Moreover, the increased expression of Gfralpha-1 in striatum after reperfusion occurred earlier in the GFRalpha-1 (+/+) than in the Gfralpha-1 (+/-) mice. These results indicate that after ischemia, there is a differential up-regulation of Gfralpha-1 expression in Gfralpha-1 (+/+) and Gfralpha-1 (+/-) mice. Since GDNF has neuroprotective effects, the reduced up-regulation of Gfralpha-1 in Gfralpha-1 (+/-) mice at early time points after ischemia suggests that the responsiveness to GDNF and GDNF receptor mediated neuroprotection is attenuated in these genetically modified animals and may underlie their greater vulnerability.
Collapse
Affiliation(s)
- A Sarabi
- National Institute on Drug Abuse, IRP Department, Cellular Neurophysiology Section, NIH, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
BACKGROUND AND PURPOSE Delivery of therapeutic proteins into tissues and across the blood-brain barrier is severely limited by their size and biochemical properties. The 11-amino acid human immunodeficiency virus TAT protein transduction domain is able to cross cell membranes and the blood-brain barrier, even when coupled with larger peptides. The present studies were done to evaluate whether TAT-glial line-derived neurotrophic factor (GDNF) fusion protein is protective in focal cerebral ischemia. METHODS Anesthetized male C57BL/6j mice were submitted to intraluminal thread occlusion of the middle cerebral artery. Reperfusion was initiated 30 minutes later by thread retraction. Laser Doppler flow was monitored during the experiments. TAT-GDNF, TAT-GFP (0.6 nmol each), or vehicle was intravenously applied over 10 minutes immediately after reperfusion. After 3 days (30 minutes of ischemia), animals were reanesthetized and decapitated. Brain injury was evaluated by histochemical stainings. RESULTS Immunocytochemical experiments confirmed the presence of TAT-GDNF protein in the brains of fusion protein-treated nonischemic control animals 3 to 4 hours after TAT fusion protein delivery. TAT-GDNF significantly reduced the number of caspase-3-immunoreactive and DNA-fragmented cells and increased the number of viable neurons in the striatum, where disseminated tissue injury was observed, compared with TAT-GFP- or vehicle-treated animals. CONCLUSIONS Our results demonstrate that TAT fusion proteins are powerful tools for the treatment of focal ischemia when delivered both before and after an ischemic insult. This approach may be of clinical interest because such fusion proteins can be intravenously applied and reach the ischemic brain regions. This approach may therefore offer new perspectives for future strategies in stroke therapy.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blood-Brain Barrier
- Brain Ischemia/drug therapy
- Brain Ischemia/etiology
- Brain Ischemia/pathology
- Drug Administration Schedule
- Drug Evaluation, Preclinical
- Gene Products, tat/administration & dosage
- Gene Products, tat/pharmacokinetics
- Gene Products, tat/pharmacology
- Gene Products, tat/therapeutic use
- Genes, tat
- Glial Cell Line-Derived Neurotrophic Factor
- HIV-1/genetics
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/drug therapy
- Infusions, Intravenous
- Male
- Mice
- Mice, Inbred C57BL
- Nerve Growth Factors/administration & dosage
- Nerve Growth Factors/pharmacokinetics
- Nerve Growth Factors/pharmacology
- Nerve Growth Factors/therapeutic use
- Neurons/drug effects
- Neurons/pathology
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/pharmacokinetics
- Neuroprotective Agents/therapeutic use
- Protein Structure, Tertiary
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/pharmacokinetics
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- Reperfusion Injury/pathology
- Reperfusion Injury/prevention & control
- Reproducibility of Results
- Single-Blind Method
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Ulkan Kilic
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
49
|
Omori N, Maruyama K, Jin G, Li F, Wang SJ, Hamakawa Y, Sato K, Nagano I, Shoji M, Abe K. Targeting of post-ischemic cerebral endothelium in rat by liposomes bearing polyethylene glycol-coupled transferrin. Neurol Res 2003; 25:275-9. [PMID: 12739237 DOI: 10.1179/016164103101201508] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
To achieve an efficient delivery targeting to post-ischemic cerebral vascular endothelium, PEG-liposome conjugated with transferrin (Tf) (Tf-PEG-liposome) was intravenously administered to the rats after 90 min of transient middle cerebral occlusion. The expression of Tf receptor (TfR) in the cerebral endothelium increased with a peak at 1 day after the reperfusion and returned to the control level by 6 days. The Tf-PEG fluorescence was marginally detectable in sham control brain, but remarkably increased with a peak at 2 days, showing about 70% of TfR positive vascular endothelium double-labeled with Tf-PEG. These results indicate that the Tf-PEG-liposome could be utilized as an efficient drug delivery tool to the brain after stroke.
Collapse
Affiliation(s)
- N Omori
- Department of Neurology, Graduate School of Medicine and Dentistry, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ikeda T, Koo H, Xia YX, Ikenoue T, Choi BH. Bimodal upregulation of glial cell line-derived neurotrophic factor (GDNF) in the neonatal rat brain following ischemic/hypoxic injury. Int J Dev Neurosci 2002; 20:555-62. [PMID: 12485623 DOI: 10.1016/s0736-5748(02)00082-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In order to delineate the spatial and temporal patterns of glial cell line-derived neurotrophic factor (GDNF) expression following ischemic/hypoxic injury in immature and neonatal brain, GDNF protein levels and immunocytochemistry were studied in rats subjected to a modified Levine procedure. Significant upregulation of GDNF protein occurred in a bimodal fashion in the damaged left cerebral cortex and hippocampus, while the levels in the right cerebral hemisphere of both control and ischemic groups remained relatively unchanged. Immunocytochemical studies indicated that the early rise in GDNF levels was most likely to be related to enhanced neuronal release of GDNF. The second rise was probably related to progressive astrogliosis that occurred in response to injury. In contrast to the lack of GDNF expression among astrocytes in normal mature brains, reactive astrocytes in the neonate appear to possess a ready capacity to express GDNF. Spatial and temporal changes in the pattern of GDNF expression following injury, as determined in this study may provide insight into the functions of GDNF in vivo and into possible therapeutic approaches toward prevention of damage or rescue of neurons following brain injury.
Collapse
Affiliation(s)
- Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Miyazaki Medical College, 5200 Kihara, Kiyotake-Cho, Japan.
| | | | | | | | | |
Collapse
|