Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 39(3): 461-470, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30790096

ABSTRACT

Temporal lobe epilepsy (TLE) is associated with neurodegeneration, often leading to hippocampal sclerosis (HS). Type 1 HS, which is characterized by severe neuronal loss and gliosis predominantly in regions CA1 and CA4, is the most common subtype and is associated with the best prognosis according to the ILAE classification system. MiRNAs participate in the biological processes underlying many nervous system diseases, including epilepsy. However, the miRNA expression profile of HS ILAE type 1 is not completely understood. A total of 14 patients were identified as having the ILAE subtype, as determined by NeuN immunohistochemistry (ILAE type 1 = 7; no-HS = 7). Next-generation sequencing and reverse transcription polymerase chain reaction technology were used to validate the dysregulated miRNAs. Bioinformatics analysis of the predicted target genes was conducted using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. In total, 1643 mature miRNAs were detected in this study, along with 5 miRNAs that were upregulated and 2 miRNAs that were downregulated in the type 1 group. Bioinformatics analysis showed that 1545 target genes were predicted using the miRDB and Targetscan databases and that these predicted genes showed enrichment in pathways associated with nucleic acid binding, intracellular and cellular macromolecule metabolic processes, and the PI3K-Akt signaling pathway. This study is the first to report the miRNA expression profile of HS ILAE type 1 compared with those of no-HS. These results provide new insights into the neuronal loss pathology of type 1 HS.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Gene Expression Profiling , Hippocampus/pathology , MicroRNAs/genetics , Adolescent , Adult , Case-Control Studies , Child , Computational Biology , Female , Humans , Male , MicroRNAs/metabolism , Reproducibility of Results , Sclerosis , Young Adult
2.
Article in English | MEDLINE | ID: mdl-29963652

ABSTRACT

INTRODUCTION: Using proton magnetic resonance spectroscopy imaging (1H-MRSI), the effects of early life stress (ELS) on nonhuman primate striatal neuronal integrity were examined as reflected by N-acetyl aspartate (NAA) concentrations. NAA measures were interrogated through examining their relationship to previously documented ELS markers -- cerebrospinal fluid (CSF) corticotropin-releasing factor (CRF) concentrations, hippocampal volume, body mass and behavioral timidity. Rodent models of depression exhibit increases in neurotrophic effects in the nucleus accumbens (NAcc). We hypothesized that rearing under conditions of ELS [Variable Foraging Demand: (VFD)] would produce persistent elevations of NAA concentrations (in absolute or ratio form) in ventral striatum/caudate nucleus (VS/CN) with altered correlation to ELS markers. METHODS: Eleven bonnet macaque males reared under VFD conditions and seven age-matched control subjects underwent 1H-MRSI during young adulthood. Voxels were placed over ventral striatum/caudate nucleus (VS/CN) to capture NAcc. Cisternal CSF CRF concentrations, hippocampal volume, body mass and response to a human intruder had been previously determined. RESULTS: VFD-reared monkeys exhibited significantly increased NAA/Cr concentrations in right VS/CN in comparison to normally-reared controls, controlling for multiple comparisons. In comparison to controls, VFD CSF CRF concentrations were directly associated with right VS/CN absolute NAA. Left hippocampal volume was inversely associated with left VS/CN N-acetyl aspartate/creatine (NAA/Cr) in VFD-reared but not in controls. Disruption of a normative inverse correlation between left VS/CN NAA and body mass was noted in VFD. Only non-VFD subjects exhibited a direct relationship between timidity response to an intruder and right VS/CN NAA. CONCLUSION: ELS produced persistent increases in VS/CN NAA, which demonstrated specific patterns of association (or lack thereof) to ELS markers in comparison to non-VFD subjects. The data are broadly consistent with a stable nonhuman primate phenotype of anxiety and mood disorder vulnerability whereby in vivo indicators of neuronal integrity, although reduced in hippocampus, are increased in striatum. The findings may provide a catalyst for further studies in humans and other species regarding a reciprocal hippocampal/NAcc relationship in affective disorders.

3.
Epilepsy Res ; 144: 34-42, 2018 08.
Article in English | MEDLINE | ID: mdl-29751355

ABSTRACT

OBJECTIVE: Studies have shown that microRNAs play a role in the development of epilepsy by regulating downstream target messenger (m)RNA. The present study aims to determine the changes associated with microRNA-21-5p (miR-21-5p) during epileptogenesis in a kainic acid rat model, and to assess whether the PTEN-mTOR pathway is a target of miR-21-5p. METHOD: Reverse transcription polymerase chain reaction (RT-PCR) was used to examine the quantitative expressions of miR-21-5p and PTEN, and Western blotting was used to test the activity of mTOR in the acute, latent, and chronic stages of epileptogenesis. The antagomir of miR-21-5p was injected into the intracerebroventricular space using a microsyringe. Neuronal death and epilepsy discharge were assessed by Nissl staining and electroencephalography (EEG), respectively. The Morris water maze (MWM) was used to assess the cognitive impairment in rats after status epilepticus (SE). RESULTS: Both miR-21-5p and mTOR were upregulated and PTEN was downregulated in rats during acute, latent, and chronic stages of epileptogenesis when compared with those of the control. After using antagomir miR-21-5p in vivo, miR-21-5p and mTOR decreased and the expression of PTEN increased compared with that in the SE model. The silencing of miR-21-5p diminished the number of abnormal spikes on EEG and decreased the number of neuron deletions on Nissl staining. The cognitive and memory impairment caused by epilepsy could also be improved after miR-21-5p knockdown in vivo. CONCLUSION: The results of the present study demonstrate that PTEN-mTOR is the target of miR-21-5p in a kainic acid model of epilepsy. The knockout of miR-21-5p decreases the neuronal damage in stages of epileptogenesis. The miR-21-5p/PTEN/mTOR axis may be a potential target for preventing and treating seizures and epileptic damage.


Subject(s)
Anticonvulsants/therapeutic use , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Status Epilepticus/drug therapy , TOR Serine-Threonine Kinases/metabolism , Animals , Antagomirs/therapeutic use , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraventricular , Kainic Acid/toxicity , Male , Maze Learning/drug effects , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Rats , Rats, Wistar , Statistics, Nonparametric , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/pathology , TOR Serine-Threonine Kinases/genetics , Time Factors
4.
Neuro Oncol ; 20(1): 55-65, 2018 01 10.
Article in English | MEDLINE | ID: mdl-28651374

ABSTRACT

Background: Cancer immunotherapy represents a promising treatment approach for malignant gliomas but is hampered by the limited number of ubiquitously expressed tumor antigens and the profoundly immunosuppressive tumor microenvironment. We identified cluster of differentiation (CD)70 as a novel immunosuppressive ligand and glioma target. Methods: Normal tissues derived from 52 different organs and primary and recurrent low-grade gliomas (LGGs) and glioblastomas (GBMs) were thoroughly evaluated for CD70 gene and protein expression. The association between CD70 and patients' overall survival and its impact on T-cell death was also evaluated. Human and mouse CD70-specific chimeric antigen receptors (CARs) were tested respectively against human primary GBMs and murine glioma lines. The antitumor efficacies of these CARs were also examined in orthotopic xenograft and syngeneic models. Results: CD70 was not detected in peripheral and brain normal tissues but was constitutively overexpressed by isocitrate dehydrogenase (IDH) wild-type primary LGGs and GBMs in the mesenchymal subgroup and recurrent tumors. CD70 was also associated with poor survival in these subgroups, which may link to its direct involvement in glioma chemokine productions and selective induction of CD8+ T-cell death. To explore the potential for therapeutic targeting of this newly identified immunosuppressive axis in GBM tumors, we demonstrate that both human and mouse CD70-specific CAR T cells recognize primary CD70+ GBM tumors in vitro and mediate the regression of established GBM in xenograft and syngeneic models without illicit effect. Conclusion: These studies identify a previously uncharacterized and ubiquitously expressed immunosuppressive ligand CD70 in GBMs that also holds potential for serving as a novel CAR target for cancer immunotherapy in gliomas.


Subject(s)
Brain Neoplasms/therapy , CD27 Ligand/immunology , Receptors, Chimeric Antigen/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma , Humans , Immunotherapy, Adoptive/methods , Isocitrate Dehydrogenase/genetics , Mice , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
5.
Int J Cancer ; 141(7): 1434-1444, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28612394

ABSTRACT

Tumor migration/metastasis and immunosuppression are major obstacles in effective cancer therapy. Incidentally, these 2 hurdles usually coexist inside tumors, therefore making therapy significantly more complicated, as both oncogenic mechanisms must be addressed for successful therapeutic intervention. Our recent report highlights that the tumor expression of a TNF family member, CD70, is correlated with poor survival for primary gliomas. In this study, we investigated how CD70 expression by GBM affects the characteristics of tumor cells and the tumor microenvironment. We found that the ablation of CD70 in primary GBM decreased CD44 and SOX2 gene expression, and inhibited tumor migration, growth and the ability to attract monocyte-derived M2 macrophages in vitro. In the tumor microenvironment, CD70 was associated with immune cell infiltrates, such as T cells; myeloid-derived suppressor cells; and monocytes/macrophages based on the RNA-sequencing profile. The CD163+ macrophages were far more abundant than T cells were. This overwhelming level of macrophages was identified only in GBM and not in low-grade gliomas and normal brain specimens, implying their tumor association. CD70 was detected only on tumor cells, not on macrophages, and was highly correlated with CD163 gene expression in primary GBM. Additionally, the co-expression of the CD70 and CD163 genes was found to correlate with decreased survival for patients with primary GBM. Together, these data suggest that CD70 expression is involved in promoting tumor aggressiveness and immunosuppression via tumor-associated macrophage recruitment/activation. Our current efforts to target this molecule using chimeric antigen receptor T cells hold great potential for treating patients with GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CD27 Ligand/metabolism , Glioblastoma/metabolism , Glioblastoma/secondary , Immune Tolerance , Antigens, CD/analysis , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/analysis , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/cytology , Brain Neoplasms/immunology , CD27 Ligand/analysis , CD27 Ligand/genetics , Cell Line, Tumor , Cell Migration Assays, Macrophage/methods , Cell Movement , Gene Expression Regulation, Neoplastic , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunity, Cellular , Macrophages/chemistry , Macrophages/cytology , Macrophages/immunology , Neoplasm Metastasis , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
6.
Carcinogenesis ; 35(8): 1698-706, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24480809

ABSTRACT

Bone morphogenetic protein-2 (BMP-2), a member of the transforming growth factor-ß family, plays critical roles in cell differentiation, modeling and regeneration processes in several tissues. BMP-2 is also closely associated with various malignant tumors. microRNAs negatively and posttranscriptionally regulate gene expression and function as oncogenes or tumor suppressors. Herein, we report that miR-656 expression was significantly downregulated in glioma cell lines and tissues. We identified and confirmed that BMP receptor, type 1A (BMPR1A) is a direct target of miR-656. The expression of BMPR1A was negatively correlated with that of miR-656 in human glioma tissues. We further demonstrated that miR-656 suppressed glioma cell proliferation, neurosphere formation, migration and invasion with or without exogenous BMP-2. Engineered knockdown of BMPR1A diminished the antiproliferation effect of miR-656 in vitro. Moreover, the canonical BMP/Smad and non-canonical BMP/mitogen-activated protein kinase (MAPK) pathways were inhibited by miR-656 overexpression. Several cancer-related signaling molecules, including cyclin B, cyclin D1, matrix metalloproteinase-9, p21 and p27, were also involved in miR-656 function in glioma cells. The tumor-suppressing function of miR-656 was validated using an in vivo intracranial xenograft mouse model. Notably, ectopic expression of miR-656 markedly reduced tumor size and prolonged the survival of mice treated with or without BMP-2. These results elucidate the function of miR-656 in glioma progression and suggest a promising application for glioma treatment.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Brain Neoplasms/genetics , Brain/metabolism , Cell Transformation, Neoplastic/genetics , Glioma/genetics , MicroRNAs/genetics , Animals , Blotting, Western , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Cell Cycle , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Glioma/metabolism , Glioma/pathology , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wound Healing , Xenograft Model Antitumor Assays
7.
Int J Mol Med ; 30(4): 739-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22842798

ABSTRACT

Status epilepticus (SE) causes neuronal loss and apoptosis by inducing several apoptosis-regulatory genes. Two such genes, cysteinyl aspartate-specific protease-3 (caspase-3), an apoptosis activator, and B-cell leukemia-2 (Bcl-2), an apoptosis suppressor, are tightly regulated for their expression and activation. Statins, inhibitors of HMG-CoA reductase, have been recently recognized as neuroprotective drugs. However, their underlying mechanisms are still unclear. In this study, we examined the neuroprotective effects of simvastatin in a rat model of SE induced by kainic acid (KA). Feeding of simvastatin for 3 days after kainate injection rescued SE-induced neuronal apoptosis, as determined by histological examination of brain sections at the level of the dorsal hippocampus. Semi-quantitative RT-PCR showed that SE treatment markedly increased caspase-3 mRNA expression and reduced Bcl-2 mRNA expression in the hippocampus. Similarly, western blot analysis and immunohistochemical analysis of the rat hippocampus demonstrated that under SE treatment, caspase-3 protein levels significantly increased and peaked at 72 h, whereas Bcl-2 protein levels decreased from 6-24 h following SE. Interestingly, simvastatin could reverse the aforementioned SE-induced changes, suggesting that the neuroprotective effects of simvastatin against neuronal apoptosis may be achieved by inhibiting caspase-3 expression and increasing Bcl-2 expression.


Subject(s)
Caspase 3/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Seizures/drug therapy , Simvastatin/pharmacology , Status Epilepticus/drug therapy , Animals , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kainic Acid , Male , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/pathology , Status Epilepticus/chemically induced , Status Epilepticus/pathology
8.
PLoS One ; 6(9): e24966, 2011.
Article in English | MEDLINE | ID: mdl-21949812

ABSTRACT

In this study, we examined the effect of chronic administration of simvastatin immediately after status epilepticus (SE) on rat brain with temporal lobe epilepsy (TLE). First, we evaluated cytokines expression at 3 days post KA-lesion in hippocampus and found that simvastatin-treatment suppressed lesion-induced expression of interleukin (IL)-1ß and tumor necrosis factor-α (TNF-α). Further, we quantified reactive astrocytosis using glial fibrillary acidic protein (GFAP) staining and neuron loss using Nissl staining in hippocampus at 4-6 months after KA-lesion. We found that simvastatin suppressed reactive astrocytosis demonstrated by a significant decrease in GFAP-positive cells, and attenuated loss of pyramidal neurons in CA3 and interneurons in dentate hilar (DH). We next assessed aberrant mossy fiber sprouting (MFS) that is known to contribute to recurrence of spontaneous seizure in epileptic brain. In contrast to the robust MFS observed in saline-treated animals, the extent of MFS was restrained by simvastatin in epileptic rats. Attenuated MFS was related to decreased neuronal loss in CA3 and DH, which is possibly a mechanism underlying decreased hippocampal susceptibility in animal treated with simvastatin. Electronic encephalography (EEG) was recorded during 4 to 6 months after KA-lesion. The frequency of abnormal spikes in rats with simvastatin-treatment decreased significantly compared to the saline group. In summary, simvastatin treatment suppressed cytokines expression and reactive astrocytosis and decreased the frequency of discharges of epileptic brain, which might be due to the inhibition of MFS in DH. Our study suggests that simvastatin administration might be a possible intervention and promising strategy for preventing SE exacerbating to chronic epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/prevention & control , Kainic Acid/toxicity , Mossy Fibers, Hippocampal/drug effects , Neurons/drug effects , Simvastatin/therapeutic use , Status Epilepticus/chemically induced , Animals , Anticholesteremic Agents/therapeutic use , Behavior, Animal/drug effects , Chronic Disease , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Epilepsy, Temporal Lobe/complications , Epilepsy, Temporal Lobe/physiopathology , Excitatory Amino Acid Agonists/toxicity , Male , Mossy Fibers, Hippocampal/pathology , Neurons/pathology , Rats , Rats, Wistar , Status Epilepticus/pathology
9.
Neural Plast ; 2011: 1-6, 2011.
Article in English | MEDLINE | ID: mdl-21837282

ABSTRACT

Increased neurogenesis in feeding centers of the murine hypothalamus is associated with weight loss in diet-induced obese rodents (Kokoeva et al., 2005 and Matrisciano et al., 2010), but this relationship has not been examined in other species. Postmortem hippocampal neurogenesis rates and premortem metabolic parameters were statistically analyzed in 8 chow-fed colony-reared adult bonnet macaques. Dentate gyrus neurogenesis, reflected by the immature neuronal marker, doublecortin (DCX), and expression of the antiapoptotic gene factor, B-cell lymphoma 2 (BCL-2), but not the precursor proliferation mitotic marker, Ki67, was inversely correlated with body weight and crown-rump length. DCX and BCL-2 each correlated positively with blood glucose level and lipid ratio (total cholesterol/high-density lipoprotein). This study demonstrates that markers of dentate gyrus neuroplasticity correlate with metabolic parameters in primates.


Subject(s)
Energy Metabolism/physiology , Hippocampus/cytology , Hippocampus/metabolism , Neurogenesis/physiology , Neuronal Plasticity/physiology , Animals , Cell Differentiation/physiology , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Macaca radiata , Male
10.
Neurosurgery ; 68(5): 1363-71; discussion 1371, 2011 May.
Article in English | MEDLINE | ID: mdl-21307798

ABSTRACT

BACKGROUND: Our previous studies demonstrated that simvastatin reduced neuronal death, increased neurogenesis, and promoted functional recovery after traumatic brain injury (TBI). OBJECTIVE: To investigate the effect of simvastatin on angiogenesis after TBI and the related signaling pathways. METHODS: Saline or simvastatin (1 mg/kg) was administered orally to rats starting at day 1 after TBI or sham surgery and then daily for 14 days. Rats were sacrificed at 3 and 14 days after treatment. Brain sections and tissues were prepared for immunohistochemical staining, enzyme-linked immunosorbent assay, and Western blot analysis. Cultured rat brain microvascular endothelial cells were subjected to oxygen-glucose deprivation followed by immunocytochemical staining with phallotoxins and vascular endothelial growth factor receptor-2 (VEGFR-2). Western blot analysis was carried out to examine the simvastatin-induced activation of the v-akt murine thymoma viral oncogene homolog (Akt) signaling pathway. The expression of VEGFR-2 was detected by enzyme-linked immunosorbent assay. RESULTS: Simvastatin significantly increased the length of vascular perimeter, promoted the proliferation of endothelial cells, and improved the sensorimotor function after TBI. Simvastatin stimulated endothelial cell tube formation after oxygen-glucose deprivation in vitro. VEGFR-2 expression in both brain tissues and cultured rat brain microvascular endothelial cells was enhanced after simvastatin treatment, which may be modulated by activation of Akt. Akt-dependent endothelial nitric oxide synthase phosphorylation was also induced by simvastatin in vivo and in vitro. CONCLUSION: Simvastatin augments TBI-induced angiogenesis in the lesion boundary zone and hippocampus and improves functional recovery. Simvastatin also promotes angiogenesis in vitro. These beneficial effects on angiogenesis may be related to simvastatin-induced activation of the VEGFR-2/Akt/endothelial nitric oxide synthase signaling pathway.


Subject(s)
Brain Injuries/metabolism , Neovascularization, Physiologic/physiology , Simvastatin/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Angiogenic Proteins/metabolism , Angiogenic Proteins/physiology , Animals , Brain Injuries/drug therapy , Brain Injuries/physiopathology , Cell Line , Cells, Cultured , Male , Neovascularization, Physiologic/drug effects , Random Allocation , Rats , Rats, Wistar , Simvastatin/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
11.
J Neurosurg ; 113(3): 591-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19895202

ABSTRACT

OBJECT: The authors' previous studies have demonstrated that simvastatin treatment promotes neuronal survival and reduces inflammatory cytokine release from astrocytes after traumatic brain injury (TBI) in rats. Since reactive astrocytes produce inflammation mediators, in the current study the authors investigated the effect of simvastatin on astrocyte activation after TBI and its underlying signaling mechanisms. METHODS: Saline or simvastatin (1 mg/kg) was orally administered to rats starting at Day 1 after TBI and then daily for 14 days. Rats were killed at 1, 3, 7, and 14 days after treatment. Brain sections and tissues were prepared for immunohistochemical staining and Western blot analysis, respectively. Cultured astrocytes were subjected to oxygen-glucose deprivation (OGD) and followed by immunocytochemical staining with glial fibrillary acidic protein/caveolin-1 and Western blot analysis. Lipid rafts were isolated from the cell lysate and Western blotting was carried out to detect the changes in epidermal growth factor receptor (EGFR) expression and phosphorylation in the lipid rafts. RESULTS: Simvastatin significantly promoted neuronal survival after TBI and attenuated activation of astrocytes. Simvastatin modified the caveolin-1 expression in lipid rafts in astrocyte cell membrane, suppressed the phosphorylation of EGFR in lipid rafts of astrocytes after OGD, and inhibited the OGD-induced interleukin-1 production. CONCLUSIONS: These data suggest that simvastatin reduces reactive astrogliosis and rescues neuronal cells after TBI. These beneficial effects of simvastatin may be mediated by inhibiting astrocyte activation after TBI through modifying the caveolin-1 expression in lipid rafts and the subsequent modulation of EGFR phosphorylation in lipid rafts.


Subject(s)
Astrocytes/drug effects , Brain Injuries/drug therapy , Gliosis/drug therapy , Membrane Microdomains/drug effects , Neuroprotective Agents/pharmacology , Receptors, Vascular Endothelial Growth Factor/metabolism , Simvastatin/pharmacology , Animals , Astrocytes/metabolism , Brain Injuries/complications , Brain Injuries/metabolism , Caveolin 1/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Gliosis/complications , Glucose/deficiency , Glucose/metabolism , Interleukin-1/metabolism , Male , Membrane Microdomains/metabolism , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Random Allocation , Rats , Rats, Wistar , Simvastatin/administration & dosage
12.
Neurosurgery ; 65(6): 1087-96; discussion 1096-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934968

ABSTRACT

OBJECTIVE: In this study, we investigated the effects of simvastatin on proliferation, migration, and apoptosis in human U251 and U87 glioma cells and the underlying molecular mechanism. METHODS: We used colony formation assay to test the cell proliferation, in vitro scratch assay to examine the cell migration, and caspase-3 activity assay, annexin V staining, and cytochrome C release to evaluate the cell apoptosis. Lipid raft fractions were isolated from glioma cells. Total cholesterol content assay was used to test the change of cholesterol level in lipid raft fractions. Immunocytochemistry staining was performed to detect the changes of lipid rafts in cell membranes. Western blotting analysis was performed to examine the signal transduction both in cells and in lipid raft fractions. RESULTS: Simvastatin inhibited proliferation and migration of U251 and U87 cells dose dependently. Simvastatin induced an increase of caspase-3 activity and annexin V staining, and down-regulated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Simvastatin also decreased cholesterol content in lipid raft fractions, suppressed caveolin-1 expression in the lipid rafts, and induced Fas translocation into lipid rafts, suggesting that simvastatin may inhibit the prosurvival PI3K/Akt pathway and trigger caspase-3-dependent apoptotic cell death through the modulation of lipid rafts. CONCLUSION: These results suggest that modulation of lipid rafts, Fas translocation, and PI3K/Akt/caspase-3 pathway are involved in the antitumor effect of simvastatin and may have a potential role in cancer prevention and treatment.


Subject(s)
Anticholesteremic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioma/pathology , Simvastatin/pharmacology , Analysis of Variance , Annexin A5/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Colony-Forming Units Assay/methods , Cytosol/drug effects , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glioma/physiopathology , Humans , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Time Factors
13.
Neurosurgery ; 65(1): 179-85; discussion 185-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19574840

ABSTRACT

OBJECTIVE: Our previous studies demonstrated that simvastatin promotes neurological functional recovery after traumatic brain injury (TBI) in rat; however, the underlying mechanisms remain poorly understood. The purpose of this study was to investigate the anti-inflammatory effect of simvastatin by measuring the level of cytokines and activation of glial cells. METHODS: Controlled cortical impact injury was performed in adult male Wistar rats. The rats were randomly divided into 3 groups: sham, saline control group, and simvastatin treatment group. Simvastatin was administered orally starting at day 1 after TBI until animals were killed at days 1, 3, 7, 14, and 35 after treatment. Functional outcome was measured using modified neurological severity scores. Enzyme-linked immunosorbent assay and immunohistochemical staining were used to measure the expression of interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha and to identify activated microglial cells and astrocytes. RESULTS: At days 1 and 3 after simvastatin or saline treatment, cytokine levels in the lesion boundary zone were significantly higher in the simvastatin- and saline-treated rats compared with the sham group, peaking at day 3. Simvastatin only reduced the level of IL-1beta but not IL-6 and tumor necrosis factor-alpha, compared with the saline group. Also, simvastatin significantly reduced the number of activated microglial cells and astrocytes compared with the saline control animals. There was also a trend toward improvement of modified neurological severity score, reaching statistical significance (P = 0.003) toward the end of the trial. CONCLUSION: Our data demonstrate that TBI causes inflammatory reaction, including increased levels of IL-1beta, IL-6, and tumor necrosis factor-alpha, as well as activated microglial cells. Simvastatin selectively reduces IL-1beta expression and inhibits the activation of microglial cells and astrocytes after TBI, which might be one of the mechanisms underlying the therapeutic benefits of simvastatin treatment of TBI.


Subject(s)
Astrocytes/drug effects , Brain Injuries , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Interleukin-1beta/metabolism , Microglia/drug effects , Simvastatin/therapeutic use , Analysis of Variance , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , Rats , Rats, Wistar , Simvastatin/pharmacology , Time Factors
14.
Neurosurgery ; 65(1): 187-91; discussion 191-2, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19574841

ABSTRACT

OBJECTIVE: This study was designed to investigate the long-term effects of simvastatin treatment after traumatic brain injury (TBI) in rats. METHODS: Adult female Wistar rats (n = 24) were injured with controlled cortical impact and divided into 3 groups. The first 2 groups were treated with simvastatin (0.5 or 1.0 mg/kg) administered orally for 14 days starting 1 day after TBI. The third group (control) received phosphate-buffered saline orally for 14 days. Neurological functional outcome was measured with modified neurological severity scores performed 1 day before TBI; on days 1, 4, 7, 14 after TBI; and biweekly thereafter. All animals were sacrificed 3 months after TBI. Brain tissues of half of the animals were processed for preparation of paraffin-embedded sections for immunohistological studies. The remaining half were frozen for enzyme-linked immunosorbent assay studies for quantification of brain-derived neurotrophic factor (BDNF) in the hippocampus and cortex. RESULTS: The results showed that both doses of simvastatin significantly improved functional outcome compared with the control, with no difference between the 2 doses. Simvastatin treatment of 1.0 mg/kg increased the number of morphologically intact neurons in the hippocampus, but treatment of 0.5 mg/kg had no significant effect. Enzyme-linked immunosorbent assay studies showed that 0.5 mg/kg simvastatin significantly increased BDNF levels within the hippocampus, but 1.0 mg/kg had no significant effect. Neither dose had any effect on BDNF levels within the cortex. CONCLUSION: Simvastatin treatment provides long-lasting functional improvement after TBI in rats. It also enhances neuronal survival in the hippocampus and increases BDNF levels in the hippocampus secondary to simvastatin treatment.


Subject(s)
Brain Injuries/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Simvastatin/therapeutic use , Animals , Brain Injuries/pathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Female , Hippocampus/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Wistar , Simvastatin/pharmacology , Time Factors
16.
J Neurotrauma ; 25(12): 1441-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19072586

ABSTRACT

This study was designed to investigate the beneficial effects of combination therapy of simvastatin and marrow stromal cells (MSCs) in improving functional outcome after traumatic brain injury (TBI) in rats. Adult female Wistar rats (n=72 and 8, per group) were injured with controlled cortical impact and treated either with monotherapy of MSCs or simvastatin or a combination therapy of these two agents. Different combination doses were tested, and nine groups of animals were studied. Neurological function was evaluated using Modified Neurological Severity Score (MNSS), and animals were sacrificed 3 months after injury. Coronal brain sections were stained with standard hematoxylin and eosin immunohistochemistry. Our results showed that, though functional improvement was seen with monotherapies of MSCs and simvastatin, the combination therapy when used in optimal doses was significantly better in improving functional outcome. This improvement was long lasting and persisted until the end of the trial (3 months). The optimum combination dose was 0.5mg of simvastatin combined with 2 x 10(6) MSCs. Post mortem analysis showed the presence of donor MSCs within the injured cortex. Endogenous cellular proliferation induced by the neurorestorative treatments was also observed in the lesion boundary zone. Our data show that MSCs and simvastatin have a synergistic effect in improving functional outcome after TBI.


Subject(s)
Bone Marrow Transplantation , Brain Injuries/therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Simvastatin/administration & dosage , Stromal Cells/transplantation , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Combined Modality Therapy , Female , Male , Rats , Rats, Wistar , Recovery of Function , Time Factors , Treatment Outcome
17.
J Neurosurg ; 109(4): 691-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18826357

ABSTRACT

OBJECT: In their previous studies, the authors found that simvastatin treatment of traumatic brain injury (TBI) in rats had beneficial effects on spatial learning functions. In the current study they wanted to determine whether simvastatin suppressed neuronal cell apoptosis after TBI, and if so, they wanted to examine the underlying mechanisms of this process. METHODS: Saline or simvastatin (1 mg/kg) was administered orally to rats starting on Day 1 after TBI and then daily for 14 days. Modified Neurological Severity Scores were used to evaluate the sensory motor functional recovery. Rats were killed at 1, 3, 7, 14, and 35 days after treatment, and brain tissue was harvested for terminal deoxynucleotidyl nick-end labeling (TUNEL) staining, caspase-3 activity assay, and Western blot analysis. RESULTS: Simvastatin significantly decreased the modified Neurological Severity Scores from Days 7 to 35 after TBI, significantly reduced the number of TUNEL-positive cells at Day 3, suppressed the caspase-3 activity at Days 1 and 3 after TBI, and increased phosphorylation of Akt as well as Forkhead transcription factor 1, inhibitory-kappaB, and endothelial nitric oxide synthase, which are the downstream targets of the prosurvival Akt signaling protein. CONCLUSIONS: These data suggested that simvastatin reduces the apoptosis in neuronal cells and improves the sensory motor function recovery after TBI. These beneficial effects of simvastatin may be mediated through activation of Akt, Forkhead transcription factor 1 and nuclear factor-kappaB signaling pathways, which suppress the activation of caspase-3 and apoptotic cell death, and thereby, lead to neuronal function recovery after TBI.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology , Animals , Brain Injuries/pathology , Cell Survival/drug effects , Forkhead Transcription Factors/metabolism , I-kappa B Proteins/metabolism , In Situ Nick-End Labeling , Male , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Phosphorylation/drug effects , Rats , Rats, Wistar , Recovery of Function/drug effects , Trauma Severity Indices
18.
J Neurosurg ; 109(3): 510-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18759585

ABSTRACT

OBJECT: This study was designed to investigate the beneficial effects of recombinant human erythropoietin (rhEPO) treatment of traumatic brain injury (TBI) in mice. METHODS: Adult male C57BL/6 mice were divided into 3 groups: 1) the saline group (TBI and saline [13 mice]); 2) EPO group (TBI and rhEPO [12]); and 3) sham group (sham and rhEPO [8]). Traumatic brain injury was induced by controlled cortical impact. Bromodeoxyuridine (100 mg/kg) was injected daily for 10 days, starting 1 day after injury, for labeling proliferating cells. Recombinant human erythropoietin was administered intraperitoneally at 6 hours and at 3 and 7 days post-TBI (5000 U/kg body weight, total dosage 15,000 U/kg). Neurological function was assessed using the Morris water maze and footfault tests. Animals were killed 35 days after injury, and brain sections were stained for immunohistochemical evaluation. RESULTS: Traumatic brain injury caused tissue loss in the cortex and cell loss in the dentate gyrus (DG) as well as impairment of sensorimotor function (footfault testing) and spatial learning (Morris water maze). Traumatic brain injury alone stimulated cell proliferation and angiogenesis. Compared with saline treatment, rhEPO significantly reduced lesion volume in the cortex and cell loss in the DG after TBI and substantially improved recovery of sensorimotor function and spatial learning performance. It enhanced neurogenesis in the injured cortex and the DG. CONCLUSIONS: Recombinant human erythropoietin initiated 6 hours post-TBI provided neuroprotection by decreasing lesion volume and cell loss as well as neurorestoration by enhancing neurogenesis, subsequently improving sensorimotor and spatial learning function. It is a promising neuroprotective and neurorestorative agent for TBI and warrants further investigation.


Subject(s)
Brain Injuries/drug therapy , Erythropoietin/therapeutic use , Hematinics/therapeutic use , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Disease Models, Animal , Drug Administration Schedule , Epoetin Alfa , Male , Maze Learning , Mice , Mice, Inbred C57BL , Psychomotor Performance , Recombinant Proteins , Recovery of Function
19.
Brain Res ; 1230: 247-57, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18657521

ABSTRACT

Erythropoietin (EPO) and its receptor (EPOR), essential for erythropoiesis, are expressed in the nervous system. Recombinant human EPO treatment promotes functional outcome after traumatic brain injury (TBI) and stroke, suggesting that the endogenous EPO/EPOR system plays an important role in neuroprotection and neurorestoration. This study was designed to investigate effects of the EPOR on histological and functional outcomes after TBI. Experimental TBI was induced in adult EPOR-null and wild-type mice by controlled cortical impact. Neurological function was assessed using the modified Morris Water Maze and footfault tests. Animals were sacrificed 35 days after injury and brain sections stained for immunohistochemistry. As compared to the wild-type injured mice, EPOR-null mice did not exhibit higher susceptibility to TBI as exemplified by tissue loss in the cortex, cell loss in the dentate gyrus, impaired spatial learning, angiogenesis and cell proliferation. We observed that less cortical neurogenesis occurred and that sensorimotor function (i.e., footfault) was more impaired in the EPOR-null mice after TBI. Co-accumulation of amyloid precursor protein (axonal injury marker) and calcium was observed in the ipsilateral thalamus in both EPOR-null and wild-type mice after TBI with more calcium deposits present in the wild-type mice. This study demonstrates for the first time that EPOR null in the nervous system aggravates sensorimotor deficits, impairs cortical neurogenesis and reduces thalamic calcium precipitation after TBI.


Subject(s)
Brain Injuries/pathology , Central Nervous System/physiology , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Antimetabolites , Brain Injuries/metabolism , Bromodeoxyuridine , Calcium/metabolism , Cell Count , Cell Proliferation , Central Nervous System/metabolism , Cerebral Cortex/pathology , Dentate Gyrus/pathology , Female , Fluorescent Antibody Technique , Hippocampus/pathology , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/physiology , Neurons/physiology , Psychomotor Performance/physiology
20.
Brain Res ; 1208: 234-9, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18384759

ABSTRACT

This study was designed to investigate the potential beneficial effects of bone marrow stromal cell (MSC) treatment of traumatic brain injury (TBI) in mice. Twelve female C57BL/6J mice (weight, 21-26 g) were injured with controlled cortical impact and divided into 2 groups (n=6 each). The experimental group was injected with MSCs (0.3x10(6)) intravenously one day after TBI, whereas the control group was injected with saline. MSCs were harvested from male mice, and male to female transplantation was performed to identify male donor cells within female recipient animals. This was achieved by localizing Y chromosomes within the female mice. Neurological function was assessed using the Morris water maze and foot fault tests. All mice were sacrificed 35 days after TBI. Brain sections were stained using in situ hybridization and immunohistochemistry to identify MSCs as well as to analyze vascular density following MSC treatment. Both modalities of testing demonstrated significant improvement in neurological function in the MSC-treated group compared to the saline-treated control group (p<0.05). Histologically, Y chromosome labeled MSCs were easily identified in the injured brain, localized primarily around the lesion boundary zone. There was also a significant increase in vascular density in the lesion boundary zone and hippocampus of MSC-treated mice compared to control mice. This is the first study to show beneficial effects of MSC treatment after TBI in mice.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Brain Injuries/therapy , Stromal Cells/physiology , Analysis of Variance , Animals , Antigens/metabolism , Behavior, Animal/physiology , Disease Models, Animal , Female , Hindlimb/physiopathology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Psychomotor Performance , Rats , Recovery of Function , Time Factors , Y Chromosome/metabolism , von Willebrand Factor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...