Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
Restor Neurol Neurosci ; 37(4): 347-362, 2019.
Article in English | MEDLINE | ID: mdl-31282441

ABSTRACT

BACKGROUND: Exosomes from mesenchymal stromal cells (MSCs) are endosome-derived vesicles that have been shown to enhance functional recovery in rodent models of stroke. OBJECTIVE: Building on these findings, we tested exosomes as a treatment in monkeys with cortical injury. METHODS: After being trained on a task of fine motor function of the hand, monkeys received a cortical injury to the hand representation in primary motor cortex. Twenty-four hours later and again 14 days after injury, monkeys received exosomes or vehicle control. Recovery of motor function was followed for 12 weeks. RESULTS: Compared to monkeys that received vehicle, exosome treated monkeys returned to pre-operative grasp patterns and latency to retrieve a food reward in the first three-five weeks of recovery. CONCLUSIONS: These results provide evidence that in monkeys exosomes delivered after cortical injury enhance recovery of motor function.


Subject(s)
Exosomes , Motor Cortex/drug effects , Motor Cortex/injuries , Motor Skills/drug effects , Recovery of Function/drug effects , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Macaca mulatta
2.
Article in English | MEDLINE | ID: mdl-28529979

ABSTRACT

AIM: This study was designed to determine any rebleeding after atorvastatin treatment following spontaneous intracerebral hemorrhage (ICH) in a prospective safety trial. PATIENTS: Atorvastatin (80 mg/day) therapy was initiated in 6 patients with primary ICH with admission Glasgow Coma Score (GCS) >5 within 24 hours of ictus and continued for 7 days, with the dose tapered and treatment terminated over the next 5 days. Patients were studied longitudinally by multiparametric magnetic resonance imaging (MRI) at three time points: acute (3 to 5 days), subacute (4 to 6 weeks) and chronic (3 to 4 months). Imaging sequences included T1, T2-weighted imaging (T2WI), diffusion tensor imaging (DTI) and contrast-enhanced MRI measures of cerebral perfusion, blood volume and blood-brain barrier (BBB) permeability. Susceptibility weighted imaging (SWI) was used to identify primary ICH and to check for secondary rebleeding. Final outcome was assessed using Glasgow Outcome Score (GOS) at 3-4 months. RESULTS: Mean admission GCS was 13.2±4.0 and mean GOS at 3 months was 4.5±0.6. Hemorrhagic lesions were segmented into core and rim areas. Mean lesion volumes decreased significantly between the acute and chronic study time points (p=0.008). Average ipsilateral hemispheric tissue loss at 3 to 4 months was 11.4±4.6 cm3. MRI showed acutely reduced CBF (p=0.004) and CBV (p=0.002) in the rim, followed by steady normalization. Apparent diffusion coefficient of water (ADC) in the rim demonstrated no alterations at any of the time points (p>0.2). The T2 values were significantly elevated in the rim acutely (p=0.02), but later returned to baseline. The ICH core showed sustained low CBF and CBV values concurrent with a small reduction in ADC acutely, but significant ADC elevation at the end suggestive of irreversible injury. CONCLUSION: Despite the presence of a small, probably permanent, cerebral lesion in the ICH core, no patients exhibited post-treatment rebleeding. These data suggest that larger, Phase 2 trials are warranted to establish long term clinical safety of atorvastatin in spontaneous ICH.

3.
J Neurol Sci ; 345(1-2): 61-7, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25060418

ABSTRACT

BACKGROUND: Thymosin ß4 (Tß4) is a 5K actin binding peptide. Tß4 improves neurological outcome in a rat model of embolic stroke and research is now focused on optimizing its dose for clinical trials. The purpose of this study was to perform a dose-response study of Tß4 to determine the optimal dose of neurological improvement in a rat model of embolic stroke. METHODS: Male Wistar rats were subjected to embolic middle cerebral artery occlusion (MCAo). Rats were divided into 4 groups of 10 animals/group: control, 2, 12 and 18 mg/kg. Tß4 was administered intraperitoneally 24h after MCAo and then every 3 days for 4 additional doses in a randomized controlled fashion. Neurological tests were performed after MCAo and before treatment and up to 8 weeks after treatment. The rats were sacrificed 56 days after MCAo and lesion volumes measured. Generalized estimating equation was used to compare the treatment effect on long term functional recovery at day 56. A quartic regression model was used for an optimal dose determination. RESULTS: Tß4 significantly improved neurological outcome at dose of 2 and 12 mg/kg at day 14 and extended to day 56 (p-values <0.05). The higher dose of 18 mg/kg did not show significant improvement. The estimated optimal dose of 3.75 mg/kg would provide optimal neurological improvement. CONCLUSIONS: This study shown that Tß4 significantly improved the long term neurological functional recovery at day 56 after MCAo with an optimal dose of 3.75 mg/kg. These results provide preclinical data for human clinical trials.


Subject(s)
Stroke/drug therapy , Thymosin/therapeutic use , Acute Disease , Adenomatous Polyposis Coli/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Bromodeoxyuridine/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/complications , Male , Myelin Basic Protein/metabolism , Neuroimaging , Neurologic Examination , Rats , Stroke/etiology , Time Factors , Treatment Outcome , Versicans/metabolism
4.
Neuroscience ; 271: 1-8, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24769225

ABSTRACT

The primary limitation of thrombolytic treatment of ischemic stroke with tissue plasminogen activator (tPA) is the hemorrhagic risk. We tested AcSDKP (N-acetyl-seryl-aspartyl-lysyl-proline), as an auxiliary therapeutic agent, to reduce blood-brain barrier (BBB) disruption in a combination tPA thrombolytic treatment of stroke. Wistar rats subjected to embolic stroke were randomly assigned to either the tPA monotherapy group (n=9) or combination of tPA and AcSDKP treatment group (n=9) initiated at 4 h after ischemia. Magnetic resonance imaging (MRI) measurements were performed before and after the treatments. Immunohistochemical staining and measurements were performed to confirm MRI findings. Longitudinal MRI permeability measurements with gadolinium-diethylenetriamine penta-acetic acid (Gd-DTPA) demonstrated that combination treatment of acute embolic stroke with AcSDKP and tPA significantly reduced BBB leakage, compared to tPA monotherapy, at 3 and 6 days (18.3±9.8 mm3 vs. 65.0±21.0 mm3, p<0.001) after the onset of stroke, although BBB leakage was comparable between the two groups prior to the treatments (6.8±4.4 mm3 vs. 4.3±3.3 mm3, p>0.18). The substantial reduction of BBB leakage observed in the combination treatment group was closely associated with reduced ischemic lesions measured by T2 maps (113.6±24.9 mm3 vs. 188.1±60.8 mm3, p<0.04 at 6 days). Histopathological analysis of the same population of rats showed that the combination treatment significantly reduced parenchymal fibrin deposition (0.063±0.059 mm2 vs. 0.172±0.103 mm2, p<0.03) and infarct volume (146.7±35.9 mm3 vs. 199.3±60.4 mm3, p<0.05) compared to the tPA monotherapy at 6days after stroke. MRI provides biological insight into the therapeutic benefit of combination treatment of stroke with tPA and AcSDKP 4h after onset, and demonstrates significantly improved cerebrovascular integrity with neuroprotective effects compared with tPA monotherapy.


Subject(s)
Blood-Brain Barrier/drug effects , Fibrinolytic Agents/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Tissue Plasminogen Activator/pharmacology , Acute Disease , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Contrast Media , Disease Models, Animal , Drug Therapy, Combination , Fibrin/metabolism , Gadolinium DTPA , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Rats, Wistar
5.
Neuroscience ; 257: 76-85, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24211797

ABSTRACT

INTRODUCTION: Angiogenin is a member of the ribonuclease superfamily and promotes degradation of the basement membrane and the extracellular matrix. After stroke in type one diabetes (T1DM) rats, Angiogenin is significantly increased and the Angiogenin is inversely correlated with functional outcome. Neamine, an aminoglycoside antibiotic, blocks nuclear translocation of Angiogenin, thereby abolishing the biological activity of Angiogenin. In this study, we therefore investigated the effect and underlying protective mechanisms of Neamine treatment of stroke in T1DM. METHODS: T1DM was induced in male Wistar rats by streptozotocin (60mg/kg, ip), and T1DM rats were subjected to embolic middle cerebral artery occlusion (MCAo). Neamine (10mg/kg ip) was administered at 2, 24 and 48h after the induction of embolic MCAo. A battery of functional outcome tests was performed. Blood-brain barrier (BBB) leakage, and lesion volume were evaluated and immunostaining, and Western blot were performed. RESULTS: Neamine treatment of stroke in T1DM rats significantly decreased BBB leakage and lesion volume as well as improved functional outcome compared to T1DM-control. Neamine also significantly decreased apoptosis and cleaved caspase-3 in the ischemic brain. Using immunostaining, we found that Neamine treatment significantly decreased nuclear Angiogenin, nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) activity, advanced glycation endproducts receptor (RAGE) number, the positive area of toll-like receptor 4 (TLR4) and increased Angeopoietin-1 expression compared to T1DM-MCAo control rats. Western blot results are consistent with the immunostaining. CONCLUSION: Neamine treatment of stroke is neuroprotective in T1DM rats. Inhibition of neuroinflammatory factor expression and decrease of BBB leakage may contribute to Neamine-induced neuroprotective effects after stroke in T1DM rats.


Subject(s)
Diabetes Mellitus, Experimental/complications , Framycetin/therapeutic use , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Angiopoietins/metabolism , Animals , Blood Pressure/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Body Temperature/drug effects , Caspase 3/metabolism , Disease Models, Animal , Extravasation of Diagnostic and Therapeutic Materials , Male , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Severity of Illness Index , Time Factors , Toll-Like Receptor 4/metabolism , NF-kappaB-Inducing Kinase
6.
Neuroscience ; 259: 155-63, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24316060

ABSTRACT

Sensory neurons mediate diabetic peripheral neuropathy. Using a mouse model of diabetic peripheral neuropathy (BKS.Cg-m+/+Lepr(db)/J (db/db) mice) and cultured dorsal root ganglion (DRG) neurons, the present study showed that hyperglycemia downregulated miR-146a expression and elevated interleukin-1 receptor-activated kinase (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6) levels in DRG neurons. In vitro, elevation of miR-146a by miR-146a mimics in DRG neurons increased neuronal survival under high-glucose conditions. Downregulation and elevation of miR-146a in DRG neurons, respectively, were inversely related to IRAK1 and TRAF6 levels. Treatment of diabetic peripheral neuropathy with sildenafil, a phosphodiesterase type 5 inhibitor, augmented miR-146a expression and decreased levels of IRAK1 and TRAF6 in the DRG neurons. In vitro, blockage of miR-146a in DRG neurons abolished the effect of sildenafil on DRG neuron protection and downregulation of IRAK1 and TRAF6 proteins under hyperglycemia. Our data provide the first evidence showing that miR-146a plays an important role in mediating DRG neuron apoptosis under hyperglycemic conditions.


Subject(s)
Diabetic Neuropathies/pathology , Ganglia, Spinal/pathology , MicroRNAs/metabolism , Neurons/metabolism , Animals , Cells, Cultured , Diabetic Neuropathies/genetics , Disease Models, Animal , Down-Regulation/drug effects , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Mice , Mice, Mutant Strains , Neurons/drug effects , Phosphodiesterase 5 Inhibitors , Piperazines , Purines , Receptors, Leptin/genetics , Sildenafil Citrate , Sulfones , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
7.
Neuroscience ; 227: 223-31, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23041512

ABSTRACT

Human umbilical cord blood cells (HUCBCs) have been employed as a restorative treatment for experimental stroke. In this study, we investigated whether transplantation of sub-therapeutic doses of HUCBCs and Simvastatin enhances cerebral vascular remodeling after stroke. Adult male Wistar rats (n=34) were subjected to transient middle cerebral artery occlusion (MCAo) and treated with: phosphate-buffered solution (PBS, gavaged daily for 7 days); Simvastatin (0.5mg/kg, gavaged daily for 7 days); HUCBCs (1×10(6), injected once via tail vein); and combination Simvasatin with HUCBCs, starting at 24h after MCAo. There was no significant difference between Simvastatin- or HUCBC-monotherapy and MCAo-alone group. Combination treatment 24h post-stroke significantly increased the perimeter of von Willebrand factor (vWF)-positive vessels, the diameter and density of alpha smooth muscle actin (αSMA)-positive arteries, and the percentage of 5-bromodeoxyuridine (BrdU)-positive endothelial cells (ECs) in the ischemic boundary zone (IBZ) compared with MCAo-alone or HUCBC-monotherapy 14 days after MCAo (p<0.05, n=8/group); Combination treatment significantly increased the densities of vWF-vessels and αSMA-arteries as well as the densities of BrdU-ECs and BrdU-positive smooth muscle cells (SMCs) in vascular walls in the IBZ compared with Simvastatin-monotherapy. Moreover, the increased BrdU-ECs and BrdU-SMCs were significantly correlated with neurological functional outcome 14 days after MCAo. Combination treatment also significantly increased the expression of Angiopoietin-1 (Ang1), Tie2 and Occludin in the IBZ (p<0.05, n=8/group). The in vitro experiments showed that combination treatment and Ang1 significantly increased capillary-like tube formation and arterial cell migration; anti-Ang1 significantly reduced combination treatment-induced tube-formation and artery cell migration (p<0.05, n=6/group). These findings indicated that a combination of sub-therapeutic doses of Simvastatin and HUCBCs treatment of stroke increases Ang1/Tie2 and Occludin expression in the ischemic brain, amplifies endogenous angiogenesis and arteriogenesis, and enhances vascular remodeling which in concert may contribute to functional outcome after stroke.


Subject(s)
Anticholesteremic Agents/therapeutic use , Human Umbilical Vein Endothelial Cells/transplantation , Neovascularization, Physiologic/drug effects , Simvastatin/therapeutic use , Stroke/drug therapy , Stroke/surgery , Analysis of Variance , Angiopoietins/immunology , Angiopoietins/metabolism , Animals , Antibodies/pharmacology , Arteries/cytology , Bromodeoxyuridine , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/cytology , Humans , Lectins/metabolism , Linear Models , Male , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/therapy , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar , Time Factors
8.
Neuroscience ; 222: 326-32, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22820263

ABSTRACT

BACKGROUND AND PURPOSE: Diabetes mellitus (DM) is a major stroke risk factor and is associated with poor recovery compared with nondiabetic stroke patients. In the present study, we investigated the effects of tissue plasminogen activator (tPA) treatment of stroke in diabetic and non-diabetic rats. METHODS: Type-1 diabetes (T1DM) was induced by injection of streptozotocin. Non-T1DM and T1DM rats were subjected to embolic middle cerebral artery occlusion (MCAo) and treated with or without tPA 2h after MCAo. Functional outcomes and immunostaining for advanced glycation endproducts receptor (RAGE), matrix metalloproteinase-9 (MMP-9) and toll-like receptor 4 (TLR4) and Western blotting were performed. RESULTS: tPA treatment of WT-MCAo rats significantly improved the functional outcome and reduced the lesion volume compared with non-treatment WT-MCAo rats (p<0.05). There was no significant difference between treatment with or without tPA in the WT-MCAo group in brain hemorrhage, BBB leakage and expression of inflammatory mediators, RAGE, MMP-9 and TLR4. However, tPA treatment in T1DM-MCAo rats (T1DM-MCAo+tPA) significantly enlarged brain hemorrhage, augmented BBB leakage, and failed to decrease lesion volume and improve functional outcome after stroke compared to T1DM-MCAo control. tPA treatment also significantly increased the expression of RAGE, MMP-9 and TLR4 in the ischemic brain in T1DM-MCAo rats compared with T1DM-MCAo control rats (p<0.05). Brain hemorrhage was significantly correlated with functional deficit and RAGE and TLR4 expression, respectively. CONCLUSIONS: Treatment of stroke with tPA increased brain hemorrhage, BBB leakage and failed to improve functional outcome in T1DM rats. The increased inflammatory response may contribute to the failed neuroprotective effects of tPA treatment in T1DM rats.


Subject(s)
Diabetes Mellitus, Type 1/complications , Fibrinolytic Agents/therapeutic use , Stroke/drug therapy , Stroke/etiology , Tissue Plasminogen Activator/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Blotting, Western , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Coloring Agents , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Evans Blue , Immunohistochemistry , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Matrix Metalloproteinase 9/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Recovery of Function , Toll-Like Receptor 4/metabolism , Treatment Outcome
9.
Neuroscience ; 220: 313-21, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22704966

ABSTRACT

Acute treatment of stroke with histone deacetylase (HDAC) inhibitors has been shown to reduce ischemic cell damage; however, it is unclear whether delayed treatment with HDAC inhibitors will contribute to the brain repair and plasticity. In the present study, we investigated the effects of delayed treatment of stroke with a pan HDAC inhibitor, valproic acid (VPA), on white matter injury and neurogenesis during stroke recovery. Administration of VPA at a dose of 100mg/kg for 7 days starting 24h after middle cerebral artery occlusion (MCAo) in rats significantly improved neurological outcome measured 7-28 days post-MCAo. In addition, the VPA treatment significantly increased oligodendrocyte survival and newly generated oligodendrocytes, which was associated with elevation of myelinated axonal density in the ischemic boundary 28 days after MCAo. VPA treatment also increased the expression of glutamate transporter 1 (GLT1) in the ischemic boundary after stroke, and increased acetylated histone H4 expression in neuroblasts and the number of new neurons in striatal ischemic boundary region. This study provides new evidence that the delayed VPA treatment enhances white matter repair and neurogenesis in ischemic brain, which may contribute to improved functional outcome.


Subject(s)
Brain/drug effects , Histone Deacetylase Inhibitors/pharmacology , Nerve Fibers, Myelinated/drug effects , Neurogenesis/drug effects , Stroke/pathology , Valproic Acid/pharmacology , Animals , Brain/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Rats , Recovery of Function/drug effects
10.
Neuroscience ; 193: 399-410, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21820491

ABSTRACT

Peripheral neuropathy is a common and major complication of diabetes, the underlying mechanisms of which are not fully understood. Using a mouse model of type II diabetes, the present study investigated the role of phosphodiesterase-5 (PDE5) in peripheral neuropathy. BKS.Cg-m+/+Leprdb/J (db/db) mice were treated with sildenafil, a specific inhibitor of PDE5, at doses of 2 and 10 mg/kg or saline. Levels of PDE5 and morphometric parameters in sciatic nerve tissue as well as the motor and sensory function were measured in these mice. In diabetic mice, PDE5 expression in sciatic nerve tissue was significantly upregulated, whereas the myelin sheath thickness, myelin basic protein (MBP), and subcutaneous nerve fibers were significantly reduced. Treatment with sildenafil significantly improved neurological function, assayed by motor and sensory conducting velocities and thermal and mechanical noxious stimuli, concomitantly with increases in myelin sheath thickness, MBP levels, and subcutaneous nerve fibers. In vitro, hyperglycemia upregulated PDE5 in Schwann cells and reduced Schwann cell proliferation, migration, and expression of brain-derived neurotrophic factor (BDNF). Blockage of PDE5 with sildenafil increased cyclic guanosine monophosphate (cGMP) and completely abolished the effect of hyperglycemia on Schwann cells. Sildenafil upregulated cGMP-dependent protein kinase G I (PKGI), whereas inhibition of PKGI with a PKG inhibitor, KT5823, suppressed the inhibitory effect of sildenafil on Schwann cells. These data indicate that hyperglycemia substantially upregulates PDE5 expression and that the cGMP/PKG signaling pathway activated by sildenafil mediates the beneficial effects of sildenafil on diabetic peripheral neuropathy.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Diabetes Complications/physiopathology , Diabetic Neuropathies/etiology , Diabetic Neuropathies/metabolism , Gene Expression Regulation/physiology , Sciatic Neuropathy/metabolism , Analysis of Variance , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Bromodeoxyuridine/metabolism , Cells, Cultured , Diabetic Neuropathies/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nerve Tissue Proteins/metabolism , Neural Conduction/drug effects , Pain Measurement/methods , Pain Threshold/drug effects , Phosphodiesterase 5 Inhibitors/therapeutic use , Piperazines/therapeutic use , Purines/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Leptin/deficiency , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/pathology , Sildenafil Citrate , Sulfones/therapeutic use , Time Factors , Transfection/methods
11.
Neuroscience ; 190: 339-45, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21683770

ABSTRACT

OBJECTIVE: High-mobility group box 1 (HMGB1), an active receptor for advanced glycation endproducts (RAGE), functions as a potent proinflammatory cytokine-like factor that contributes to the pathogenesis of vasculature. Diabetes mellitus (DM) is associated with accelerated development of both microvascular and macrovascular disease and increases the risk of ischemic stroke. Using a model of streptozotocin-induced type-1 diabetes (T1DM) in rats, we investigated the changes in HMGB and RAGE and tested the effects of Niaspan, a slow release form of niacin, on the expression of pro-inflammatory proteins in rats after stroke. RESEARCH DESIGN AND METHODS: T1DM rats were subjected to transient middle cerebral artery occlusion (MCAo) and treated without or with Niaspan (40 mg/kg) daily for 14 days after MCAo. Non-streptozotocin rats (WT) were also subjected to MCAo. Immunostaining for inflammatory mediators including HMGB1, RAGE, matrix metalloproteinase-9 (MMP-9) and toll-like receptor 4 (TLR4) immunostaining (n=8/group) and Western blotting (n=4/group) were performed. RESULTS: Compared to WT-MCAo rats, T1DM-MCAo rats showed an increased expression of HMGB1 (0.82±0.07 vs. 1.81±0.98, P<0.05), RAGE (1.31±0.22 vs. 3.77±0.72, P<0.05), MMP-9 (0.74±0.08 vs. 1.61±0.09, P<0.05) and TLR4 (2.85±0.22 vs. 6.72±0.44, P<0.05) after stroke. Niaspan treatment significantly attenuated the expression of HMGB1 (1.80±0.98 vs. 1.31±0.01, P<0.05), RAGE (3.77±0.71 vs. 1.78±0.45, P<0.05), MMP-9 (1.61±0.09 vs. 0.97±0.07, P<0.05) and TLR4 (6.72±0.44 vs. 2.28±0.43, P<0.05) in the ischemic brain in T1DM-MCAo rats. CONCLUSIONS: T1DM increases HMGB1/RAGE, TLR4 and MMP-9 expression after stroke. Niaspan treatment of stroke in T1DM rats inhibits HMGB1/RAGE, TLR4 and MMP-9 expression which may contribute to the reduced inflammatory response after stroke in T1DM rats.


Subject(s)
Brain/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , HMGB Proteins/metabolism , Niacin/pharmacology , Receptors, Immunologic/metabolism , Stroke/metabolism , Animals , Brain/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Male , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Stroke/complications
12.
Neuroscience ; 169(2): 674-82, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20627173

ABSTRACT

UNLABELLED: Thymosin beta4 (Tbeta4) is a developmentally expressed 43-amino acid peptide that inhibits organization of the actin-cytoskeleton by sequestration of G-actin monomers. Tbeta4 improves cardiac function after myocardial infarction in adult mice and promotes healing properties in both dermal and corneal wounds. We tested the hypothesis that Tbeta4 improves functional neurological outcome in a rat model of embolic stroke. EXPERIMENTAL PROCEDURES: Male Wistar rats (n=18) were subjected to embolic middle cerebral artery occlusion (MCAo). Tbeta4 (6 mg/kg, IP) was administered 24 h after MCAo and then every 3 days for four additional doses (n=9). Rats treated with saline were used as a control (n=9). The adhesive-removal test (ART) and modified Neurological Severity Score (mNSS) were performed to measure functional outcome. Rats were sacrificed 56 days after MCAo. Immunostaining was performed with antibodies against NG-2 (chondroitin sulfate proteoglycan), CNPase (2", 3"-cyclic nucleotide 3'-phosphodiesterase) to detect immature and mature oligodendrocytes. Neurofilament-H (NF-H) antibodies were used to detect axons while myelinated axons were identified with Bielschowsky/Luxol (B/L) Blue staining. EBA (endothelial barrier antigen) was used for detection of mature vessels. RESULTS: Ischemic rats treated with Tbeta4 demonstrated a significant overall improvement (P<0.01) in the ART and the mNSS when compared to controls. Significant improvement was observed beginning at 14 and 35 days, respectively. Lesion volumes showed no significant differences between the two groups. Treatment with Tbeta4 increased myelinated axons and increased vessel density in the ischemic boundary (P<0.05) and augmented remyelination which was associated with an increase of oligodendrocyte progenitor cells (OPCs) and myelinating oligodendrocytes (P<0.05). CONCLUSIONS: The present study suggests that Tbeta4 improves neurological functional outcome after embolic stroke in rats. Axonal remodeling from mobilization of OPCs is proposed as contributing to Tbeta4 induced functional improvement.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Thymosin/therapeutic use , Animals , Antigens/biosynthesis , Axons/metabolism , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Proliferation , Corpus Callosum/blood supply , Corpus Striatum/blood supply , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Proteoglycans/biosynthesis , Rats , Rats, Wistar , Stem Cells/pathology
13.
Glia ; 58(9): 1074-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20468049

ABSTRACT

Bone marrow stromal cells (BMSCs) facilitate functional recovery in rats after focal ischemic attack. Growing evidence suggests that the secretion of various bioactive factors underlies BMSCs' beneficial effects. This study investigates the expression of glial cell derived neurotrophic factor (GDNF) in the ischemic hemisphere with or without BMSC administration. Adult male Wistar rats were subjected to 2 h of middle cerebral artery occlusion followed by an injection of 3 x 10(6) BMSCs (n = 11) or phosphate-buffered saline (n = 10) into the tail vein 24 h later. Animals were sacrificed seven days later. Single and double immunohistochemical staining was performed to measure GDNF, Ki67, doublecortin, and glial fibrillary acidic protein expression as well as the number of apoptotic cells along the ischemic boundary zone (IBZ) and/or in the subventricular zone (SVZ). BMSC treatment significantly increased GDNF expression and decreased the number of apoptotic cells in the IBZ (P < 0.05). GDNF expression was colocalized with GFAP. Meanwhile, BMSCs increased the number of Ki-67 positive cells and the density of DCX positive migrating neuroblasts (P < 0.05). GDNF expression was significantly increased in single astrocytes collected from animals treated with BMSCs, and in astrocytes cocultured with BMSCs after OGD (P < 0.05). Our data suggest that BMSCs increase GDNF levels in the ischemic hemisphere; the major source of GDNF protein is reactive astrocytes. We propose that the increase of GDNF in response to BMSC administration creates a hospitable environment for local cellular repair as well as for migrating neuroblasts from the SVZ, and thus contributes to the functional improvement.


Subject(s)
Astrocytes/metabolism , Bone Marrow Transplantation , Brain Ischemia/therapy , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Stroke/therapy , Stromal Cells/transplantation , Aging , Animals , Apoptosis/physiology , Brain/physiopathology , Brain Ischemia/physiopathology , Doublecortin Protein , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/therapy , Male , Neurons/physiology , Random Allocation , Rats , Rats, Wistar , Stem Cell Niche/physiopathology , Stroke/physiopathology
14.
Neuroscience ; 164(4): 1887-93, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-19782721

ABSTRACT

In the present study, we hypothesized that thymosin beta 4 (Tbeta4) is a potential therapy of multiple sclerosis (MS). To test this hypothesis, SJL/J mice (n=21) were subjected to experimental autoimmune encephalomyelitis (EAE), an animal model of MS. EAE mice were treated with saline or Tbeta4 (6 mg/kg, n=10) every 3 days starting on the day of myelin proteolipid protein (PLP) immunization for total five doses. Neurological function, inflammatory infiltration, oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes were measured in the brain of EAE mice. Double immunohistochemical staining was used to detect proliferation and differentiation of OPCs. Tbeta4 was used to treat N20.1 cells (premature oligodendrocyte cell line) in vitro, and proliferation of N20.1 cells was measured by bromodeoxyuridine (BrdU) immunostaining. Tbeta4 treatment improved functional recovery after EAE. Inflammatory infiltrates were significantly reduced in the Tbeta4 treatment group compared to the saline groups (3.6+/-0.3/slide vs 5+/-0.5/slide, P<0.05). NG2(+) OPCs (447.7+/-41.9 vs 195.2+/-31/mm(2) in subventricular zone (SVZ), 75.1+/-4.7 vs 41.7+/-3.2/mm(2) in white matter), CNPase(+) mature oligodendrocytes (267.5+/-10.3 vs 141.4+/-22.9/mm(2)), BrdU(+) with NG2(+) OPCs (32.9+/-3.7 vs 17.9+/-3.6/mm(2)), BrdU(+) with CNPase(+) mature oligodendrocytes (18.2+/-1.7 vs 10.7+/-2.2/mm(2)) were significantly increased in the Tbeta4 treated mice compared to those of saline controls (P<0.05). These data indicate that Tbeta4 treatment improved functional recovery after EAE, possibly, via reducing inflammatory infiltrates, and stimulating oligodendrogenesis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Thymosin/therapeutic use , Animals , Brain/blood supply , Brain/immunology , Brain/pathology , Cell Line , Cell Proliferation/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Inflammation/immunology , Inflammation/pathology , Mice , Oligodendroglia/pathology , Stem Cells/pathology
15.
Int J Stroke ; 4(4): 250-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19689750

ABSTRACT

BACKGROUND: Attention has turned to neurorestorative therapies, including erythropoietin, for experimental ischaemic stroke and head injury. Treatments for intracerebral haemorrhage need to be developed, as this represents a particularly devastating and common form of neurological injury. Aim The aim of this study is to investigate the therapeutic potential of erythropoietin after intracerebral haemorrhage in rats and to measure its effects on mechanisms of recovery and neurogenesis. METHODS: Intracerebral haemorrhage was induced in 24 Wistar male rats by intrastriatal infusion of autologous blood. Recombinant human erythropoietin (5000 or 10,000 U/kg BW/day) or saline was administered starting 1 day after intracerebral haemorrhage and continued daily for 1 week (n=8 for each group). To label proliferating cells, 5'-bromo-2' deoxyuridine was injected daily for 13 days after intracerebral haemorrhage. All animals survived for 2 weeks after intracerebral haemorrhage. Functional outcome, area of tissue loss and immunohistochemical staining were measured at 14 days after intracerebral haemorrhage. Global test or anova was used to test the erythropoietin dose effect. RESULTS: Rats receiving recombinant human erythropoietin after intracerebral haemorrhage exhibited significant improvement in modified neurological severity score and corner test at 14 days (P<0.05). Increased expression of phenotypes of synaptogenesis and proliferating immature neurons were shown by immunohistochemical staining. Only the group receiving a lower dose of recombinant human erythropoietin had significantly less tissue loss compared with the control group (P<0.05). In rats treated with recombinant human erythropoietin, double staining for 5'-bromo-2' deoxyuridine and TUJ1 revealed a subpopulation of cells that express an immature neuronal marker while still dividing. CONCLUSIONS: Erythropoietin improves neurological outcome and increases histochemical parameters of neurogenesis when given after intracerebral haemorrhage in rats. Intriguingly, only the lower dose of recombinant human erythropoietin was effective in reducing tissue loss in the region of intracerebral haemorrhage.


Subject(s)
Cerebral Hemorrhage/drug therapy , Erythropoietin/therapeutic use , Animals , Antimetabolites , Brain/pathology , Bromodeoxyuridine , Cerebral Hemorrhage/pathology , Coloring Agents , Humans , Immunohistochemistry , Indicators and Reagents , Linear Models , Male , Nerve Tissue Proteins/metabolism , Psychomotor Performance/drug effects , Rats , Rats, Wistar , Recombinant Proteins , Recovery of Function , Tissue Fixation
16.
Neuroscience ; 159(2): 744-50, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19154781

ABSTRACT

Arteriogenesis supports restored perfusion in the ischemic brain and improves long-term functional outcome after stroke. We investigate the role of endothelial nitric oxide synthetase (eNOS) and a nitric oxide (NO) donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) amino] diazen-1-ium-1, 2-diolate (DETA-NONOate), in promoting arteriogenesis after stroke. Adult wild-type (WT, n=18) and eNOS-knockout (eNOS(-/-), n=36) mice were subjected to transient (2.5 h) right middle cerebral artery occlusion (MCAo) and were treated with or without DETA-NONOate (0.4 mg/kg) 24 h after MCAo. Functional evaluation was performed. Animals were sacrificed 3 days after MCAo for arterial cell culture studies, or 14 days for immunohistochemical analysis. Consistent with previous studies, eNOS(-/-) mice exhibited a higher mortality rate (P<0.05, n=18/group) and more severe neurological functional deficit after MCAo than WT mice (P<0.05, n=12/group). Decreased arteriogenesis, was evident in eNOS(-/-) mice compared with WT mice, as demonstrated by reduced vascular smooth muscle cell (VSMC) proliferation, arterial density and diameter in the ischemic brain. eNOS(-/-) mice treated with DETA-NONOate had a significantly decreased mortality rate and improved functional recovery, and exhibited enhanced arteriogenesis identified by increased VSMC proliferation, and upregulated arterial density and diameter compared to eNOS(-/-) mice after stroke (P<0.05, n=12/group). To elucidate the mechanisms underlying eNOS/NO mediated arteriogenesis, VSMC migration was measured in vitro. Arterial cell migration significantly decreased in the cultured common carotid artery (CCA) derived from eNOS(-/-) mice 3 days after MCAo compared to WT arterial cells. DETA-NONOate-treatment significantly attenuated eNOS(-/-)-induced decrease of arterial cell migration compared to eNOS(-/-) control artery (P<0.05; n=6/group). Using VSMC culture, DETA-NONOate significantly increased VSMC migration, while inhibition of NOS significantly decreased VSMC migration (P<0.05; n=6/group). Our data indicated that eNOS not only promotes vascular dilation but also increases VSMC proliferation and migration, and thereby enhances arteriogenesis after stroke. Therefore, increase eNOS may play an important role in regulating of arteriogenesis after stroke.


Subject(s)
Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/physiopathology , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/metabolism , Actins/metabolism , Animals , Carotid Artery, Common/cytology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/drug effects , Neurologic Examination , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase Type III/deficiency , Nitroso Compounds/therapeutic use , Recovery of Function/drug effects , Time Factors
17.
Neuroscience ; 158(4): 1356-63, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19059466

ABSTRACT

Molecular mechanisms by which stroke increases neurogenesis have not been fully investigated. Using neural progenitor cells isolated from the subventricular zone (SVZ) of the adult rat subjected to focal cerebral ischemia, we investigated the Notch pathway in regulating proliferation and differentiation of adult neural progenitor cells after stroke. During proliferation of neural progenitor cells, ischemic neural progenitor cells exhibited substantially increased levels of Notch, Notch intracellular domain (NICD), and hairy enhancer of split (Hes) 1, which was associated with a significant increase of proliferating cells. Blockage of the Notch pathway by short interfering ribonucleic acid (siRNA) against Notch or a gamma secretase inhibitor significantly reduced Notch, NICD and Hes1 expression and cell proliferation induced by stroke. During differentiation of neural progenitor cells, Notch and Hes1 expression was downregulated in ischemic neural progenitor cells, which was coincident with a significant increase of neuronal population. Inhibition of the Notch pathway with a gamma secretase inhibitor further substantially increased neurons, but did not alter astrocyte population in ischemic neural progenitor cells. These data suggest that the Notch signaling pathway mediates adult SVZ neural progenitor cell proliferation and differentiation after stroke.


Subject(s)
Adult Stem Cells/pathology , Cerebral Ventricles/pathology , Infarction, Middle Cerebral Artery/pathology , Neurons/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Gene Expression/drug effects , Leukemia Inhibitory Factor/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Receptors, Notch/genetics , Signal Transduction/drug effects , Transfection , Triglycerides , gamma-Aminobutyric Acid/analogs & derivatives
18.
Acta Neurochir Suppl ; 105: 79-83, 2008.
Article in English | MEDLINE | ID: mdl-19066087

ABSTRACT

We describe some of our studies on use of neuro-restorative agents for treatment of neural injury. We focus on cell-based therapies and select from a variety of statins. In addition, we show that cell-based and pharmacological-based therapies enhance brain plasticity and promote recovery of function after stroke and intracerebral hemorrhage (ICH). Injured brain recapitulates ontogeny. Cerebral tissue around the infarction expresses developmental genes, many of which are present only during embryonic or neonatal stages of development. Brain response to injury undergoes remodeling with induction of angiogenesis, neurogenesis, and synaptogenesis. The attempt at remodeling, although expressed as a partial improvement in patients with stroke and ICH, is clearly insufficient to promote substantial recovery in many patients. The goal of restorative therapies should be to activate and amplify this endogenous restorative brain plasticity process to potentiate functional recovery. The logic of restorative therapy is to treat intact or marginally compromised tissue and not injured or dying tissue. Thus, these treatments can be made available for all neurological injury. Once demonstrated to be effective for treatment of a large middle cerebral artery occlusion (MCAo), these restorative treatments can be applied to many types of injury, including ICH, traumatic brain injury, and neurodegenerative disease such as experimental autoimmune encephalomyelitis and multiple sclerosis.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cerebral Hemorrhage/therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Stroke/therapy , Humans , Stem Cell Transplantation
19.
Neuroscience ; 156(1): 155-64, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18691637

ABSTRACT

Neovascularization may contribute to functional recovery after neural injury. Combination treatment of stroke with a nitric oxide donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) amino] diazen-1-ium-1, 2-diolate (DETA-NONOate) and bone marrow stromal cells promotes functional recovery. However, the mechanisms underlying functional improvement have not been elucidated. In this study, we tested the hypothesis that combination treatment upregulates angiopoietin-1 and its receptor Tie2 in the ischemic brain and bone marrow stromal cells, thereby enhancing cerebral neovascularization after stroke. Adult wild type male C57BL/6 mice were i.v. administered PBS, bone marrow stromal cells 5x10(5), DETA-NONOate 0.4 mg/kg or combination DETA-NONOate with bone marrow stromal cells (n=12/group) after middle cerebral artery occlusion. Combination treatment significantly upregulated angiopoietin-1/Tie2 and tight junction protein (occludin) expression, and increased the number, diameter and perimeter of blood vessels in the ischemic brain compared with vehicle control (mean+ or -S.E., P<0.05). In vitro, DETA-NONOate significantly increased angiopoietin-1/Tie2 protein (n=6/group) and Tie2 mRNA (n=3/group) expression in bone marrow stromal cells. DETA-NONOate also significantly increased angiopoietin-1 protein (n=6/group) and mRNA (n=3/group) expression in mouse brain endothelial cells (P<0.05). Angiopoietin-1 mRNA (n=3/group) was significantly increased in mouse brain endothelial cells treated with DETA-NONOate in combination with bone marrow stromal cell-conditioned medium compared with cells treated with bone marrow stromal cell-conditioned medium or DETA-NONOate alone. Mouse brain endothelial cell capillary tube-like formation assays (n=6/group) showed that angiopoietin-1 peptide, the supernatant of bone marrow stromal cells and DETA-NONOate significantly increased capillary tube formation compared with vehicle control. Combination treatment significantly increased capillary tube formation compared with DETA-NONOate treatment alone. Inhibition of angiopoietin-1 significantly attenuated combination treatment-induced tube formation. Our data indicated that combination treatment of stroke with DETA-NONOate and bone marrow stromal cells promotes neovascularization, which is at least partially mediated by upregulation of the angiopoietin-1/Tie2 axis.


Subject(s)
Angiopoietin-1/metabolism , Bone Marrow Transplantation/methods , Neovascularization, Physiologic/drug effects , Nitroso Compounds/pharmacology , Stroke/drug therapy , Stroke/surgery , Angiopoietin-1/agonists , Angiopoietin-1/antagonists & inhibitors , Animals , Capillaries/drug effects , Capillaries/metabolism , Cells, Cultured , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Coculture Techniques , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Nitroso Compounds/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, TIE-2/genetics , Regeneration/drug effects , Regeneration/physiology , Stromal Cells/transplantation , Tight Junctions/drug effects , Tight Junctions/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Neuroscience ; 152(3): 646-55, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18313231

ABSTRACT

Transplantation of bone marrow stromal cells (BMSCs) improves animal neurological functional recovery after stroke. To obtain insight into the mechanisms underlying the therapeutic benefit, we directed our attention to the interaction of BMSCs with astrocytes. Astrocytes become reactive (astrogliosis) after a brain injury, such as stroke. Astrogliosis plays both beneficial and detrimental roles in brain recovery. Previously, we have shown that administration of BMSCs to animals with stroke significantly reduces the thickness of the scar wall formed by reactive astrocytes. We tested the influence of mouse bone marrow stromal cell (mBMSC) on astrogliosis under oxygen-glucose deprivation (OGD)/reoxygenation conditions in vitro, employing an anaerobic chamber. Our data indicate that mBMSCs down-regulate glial fibrillary acidic protein (GFAP) expression in astrocytes after 2 h of OGD and an additional 16 h reoxygenation. mBMSCs protected astrocytes from ischemia, maintaining morphological integrity and proliferation. The IL-6/IL-6R/gp130 pathway is associated with astrogliosis in response to CNS (disorders. Therefore, we examined the effects of mBMSC on the IL-6/IL-6R/gp130 pathway as an underlying mechanism of mBMSC-altered astrogliosis. Furthermore, IL-6 siRNA was used to block IL-6 expression in astrocytes to further investigate IL-6 involvement in mBMSC-altered astrogliosis. Our results indicate that the mBMSC-conferred decline of astrogliosis post-ischemia may derive from the down-regulation of the IL-6/IL-6R/gp130 pathway.


Subject(s)
Astrocytes/metabolism , Brain Ischemia/physiopathology , Gliosis/physiopathology , Stromal Cells/metabolism , Animals , Astrocytes/pathology , Bone Marrow Cells/cytology , Brain Ischemia/pathology , Cell Communication , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Cytoprotection , Down-Regulation/immunology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Gliosis/prevention & control , Graft Survival , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , RNA, Small Interfering , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Stromal Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL