Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 39(1): 58-73, 2019 01.
Article in English | MEDLINE | ID: mdl-29135354

ABSTRACT

Depression after traumatic brain injury (TBI) is common but the mechanisms by which TBI causes depression are unknown. TBI decreases glutamate transporters GLT-1 and GLAST and allows extravasation of thrombin. We examined the effects of thrombin on transporter expression in primary hippocampal astrocytes. Application of a PAR-1 agonist caused down-regulation of GLT-1, which was prevented by inhibition of Rho kinase (ROCK). To confirm these mechanisms in vivo, we subjected mice to closed-skull TBI. Thrombin activity in the hippocampus increased one day following TBI. Seven days following TBI, expression of GLT-1 and GLAST was reduced in the hippocampus, and this was prevented by administration of the PAR-1 antagonist SCH79797. Inhibition of ROCK attenuated the decrease in GLT-1, but not GLAST, after TBI. We measured changes in glutamate levels in the hippocampus seven days after TBI using an implanted biosensor. Stress-induced glutamate levels were significantly increased following TBI and this was attenuated by treatment with the ROCK inhibitor fasudil. We quantified depressive behavior following TBI and found that inhibition of PAR-1 or ROCK decreased these behaviors. These results identify a novel mechanism by which TBI results in down-regulation of astrocyte glutamate transporters and implicate astrocyte and glutamate transporter dysfunction in depression following TBI.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/genetics , Depression/etiology , Depression/genetics , Hippocampus/metabolism , Thrombin , Vesicular Glutamate Transport Proteins/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Behavior, Animal , Blood-Brain Barrier/pathology , Depression/psychology , Down-Regulation , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/metabolism , Hippocampus/cytology , Male , Mice , Receptor, PAR-1/genetics , Vesicular Glutamate Transport Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors
2.
Exp Neurol ; 273: 288-300, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26391563

ABSTRACT

Astrocyte glutamate transporters GLAST and GLT1 play a key role in regulating neuronal excitation and their levels are altered in patients with epilepsy, and after traumatic brain injury. The mechanisms which regulate their expression are not well understood. We tested the hypothesis that exposure of astrocytes to high levels of thrombin, as may occur after a compromise of the blood-brain barrier, would reduce astrocyte glutamate transporter levels. In isolated rat cortical astrocytes we examined the effects of thrombin on the expression and function of glutamate transporters, and the signaling pathways involved in these responses by using Western blotting and selective inhibitors. Thrombin induced a selective decrease in the expression of GLAST but not GLT1, with a corresponding decrease in the capacity of astrocytes to take up glutamate. Activation of the thrombin receptor PAR-1 with an activating peptide induced a similar decrease in the expression of GLAST and compromise of glutamate uptake. The downregulation of GLAST induced by thrombin was mediated by the mitogen activated protein kinases p38 MAPK, ERK and JNK, but inhibition of these kinases did not prevent the decrease in glutamate uptake induced by thrombin. In contrast, inhibition of the Rho kinase pathway using the specific inhibitor, Y27632, suppressed both the decrease in the expression of GLAST and the decrease in glutamate uptake induced by thrombin. In hippocampal astrocyte cultures, thrombin caused a decrease in both GLAST and GLT1. In tissue resected from brains of children with intractable epilepsy, we found a decrease in the integrity of the blood-brain barrier along with a reduction in immunoreactivity for both transporters which was associated with an increase in cleaved thrombin and reactive astrogliosis. The in vitro results suggest a specific mechanism by which thrombin may lead to a compromise of astrocyte function and enhanced synaptic excitability after the blood-brain barrier is compromised. The human in vivo results provide indirect support evidence linking the compromise of the blood-brain barrier to thrombin-induced reduction in glutamate transporter expression and an increase in neuronal excitation.


Subject(s)
Astrocytes/drug effects , Cerebral Cortex/cytology , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/metabolism , Signal Transduction/drug effects , Thrombin/pharmacology , rho-Associated Kinases/metabolism , Adolescent , Animals , Animals, Newborn , Astrocytes/metabolism , Blood-Brain Barrier/physiopathology , Cells, Cultured , Cerebral Cortex/metabolism , Child , Child, Preschool , Epilepsy/pathology , Excitatory Amino Acid Transporter 2 , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Thrombin/metabolism , Time Factors , Young Adult , Zonula Occludens-1 Protein/metabolism
3.
Neurobiol Dis ; 73: 189-203, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25251607

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a Notch3 dominant mutation-induced cerebral small vascular disease, is characterized by progressive degeneration of vascular smooth muscle cells (vSMCs) of small arteries in the brain, leading to recurrent ischemic stroke, vascular dementia and death. To date, no treatment can stop or delay the progression of this disease. Herein, we determined the therapeutic effects of stem cell factor (SCF) in combination with granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) in a mouse model of CADASIL carrying the human mutant Notch3 gene. SCF+G-CSF was subcutaneously administered for 5 days and repeated 4 times with 1-4 month intervals. We found through water maze testing that SCF+G-CSF treatment improved cognitive function. SCF+G-CSF also attenuated vSMC degeneration in small arteries, increased cerebral blood vascular density, and inhibited apoptosis in CADASIL mice. We also discovered that loss of cerebral capillary endothelial cells and neural stem cells/neural progenitor cells (NSCs/NPCs) occurred in CADASIL mice. SCF+G-CSF treatment inhibited the CADASIL-induced cell loss in the endothelia and NSCs/NPCs and promoted neurogenesis. In an in vitro model of apoptosis, SCF+G-CSF prevented apoptotic cell death in vSMCs through AKT signaling and by inhibiting caspase-3 activity. These data suggest that SCF+G-CSF restricts the pathological progression of CADASIL. This study offers new insights into developing therapeutic strategies for CADASIL.


Subject(s)
CADASIL/complications , CADASIL/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Stem Cell Factor/therapeutic use , Animals , Bone Marrow Transplantation , CADASIL/genetics , CADASIL/surgery , Caspase 3/metabolism , Cell Death/drug effects , Cells, Cultured , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Mutation/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Neurogenesis/drug effects , Neurogenesis/genetics , Receptor, Notch3 , Receptors, Notch/genetics , Time Factors
4.
J Econ Entomol ; 107(1): 98-104, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24665690

ABSTRACT

In Liaoning Province, China, tomato yellow leaf curl virus (TYLCV) was first detected in 2009 and in only four counties. To quantify the spread of TYLCV and to identify potential factors influencing its spread in Liaoning Province, we assayed for TYLCV within 1,055 whiteflies (Bemisia tabaci (Gennadius) complex) from 74 populations and 29 counties in 2011. The B. tabaci species of these individuals was determined based on molecular markers. TYLCV was found in 13 counties (Donggang, Liaoyang, Kazuo, Lingyuan, Heishan, Liaozhong, Kaiyuan, Taian, Dawa, Dashiqiao, Beizhen, Linghai, and Xingcheng) and was most frequently detected in the central plain. In addition, the percentage of whiteflies with TYLCV was significantly higher in B. tabaci Q than in B. tabaci B but was unrelated to the hosts (pepper, eggplant, tomato, cucumber, and kidney bean) on which the whiteflies had been collected. These results demonstrate that TYLCV has spread rapidly in Liaoning Province since its first detection and suggest that its spread is more closely associated with the introduction of B. tabaci Q than with the species of host plant. These findings also indicate that controls are now needed to reduce the further spread of TYLCV and that these controls should include the management of B. tabaci Q populations.


Subject(s)
Begomovirus/isolation & purification , Crops, Agricultural , Hemiptera/virology , Insect Vectors/virology , Introduced Species , Animals , China , Geography , Plant Diseases/virology
5.
Article in English | MEDLINE | ID: mdl-30090853

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common condition of hereditary stroke and vascular dementia. CADASIL is caused by Notch3 mutation, leading to progressive degeneration of vascular smooth muscle cells (vSMCs) of the small arteries in the brain. However, the pathogenesis of CADASIL remains largely unknown, and treatment that can stop or delay the progression of CADASIL is not yet available. Using both wild type mice and transgenic mice carrying the human mutant Notch3 gene (CADASIL mice), we have recently characterized the pathological features of CADASIL and determined the therapeutic efficacy of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) in CADASIL. Our findings have revealed novel pathological changes in the endothelium of cerebral capillaries and in the neural stem cells (NSCs). We have also observed the impairment of cognitive function in CADASIL mice. Moreover, SCF+G-CSF treatment improves cognitive function, inhibits Notch3 mutation-induced vSMC degeneration, cerebral blood bed reduction, cerebral capillary damage, and NSC loss, and increases neurogenesis and angiogenesis. Here we compile an overview of our recently published studies, which provide new insights into understanding the pathogenesis of CADASIL and developing therapeutic strategies for this devastating neurological disease.

6.
PLoS One ; 8(10): e75562, 2013.
Article in English | MEDLINE | ID: mdl-24116056

ABSTRACT

Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are initially discovered as the essential hematopoietic growth factors regulating bone marrow stem cell proliferation and differentiation, and SCF in combination with G-CSF (SCF+G-CSF) has synergistic effects on bone marrow stem cell mobilization. In this study we have determined the effect of SCF and G-CSF on neurite outgrowth in rat cortical neurons. Using molecular and cellular biology and live cell imaging approaches, we have revealed that receptors for SCF and G-CSF are expressed on the growth core of cortical neurons, and that SCF+G-CSF synergistically enhances neurite extension through PI3K/AKT and NFκB signaling pathways. Moreover, SCF+G-CSF induces much greater NFκB activation, NFκB transcriptional binding and brain-derived neurotrophic factor (BDNF) production than SCF or G-CSF alone. In addition, we have also observed that BDNF, the target gene of NFκB, is required for SCF+G-CSF-induced neurite outgrowth. These data suggest that SCF+G-CSF has synergistic effects to promote neurite growth. This study provides new insights into the contribution of hematopoietic growth factors in neuronal plasticity.


Subject(s)
Cerebral Cortex/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Neurites/drug effects , Neurons/drug effects , Stem Cell Factor/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , NF-kappa B/metabolism , Neurites/metabolism , Neurons/cytology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Neurotherapeutics ; 10(3): 520-38, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23775067

ABSTRACT

Spinal cord injury (SCI) causes not only sensorimotor and cognitive deficits, but frequently also severe chronic pain that is difficult to treat (SCI pain). We previously showed that hyperesthesia, as well as spontaneous pain induced by electrolytic lesions in the rat spinothalamic tract, is associated with increased spontaneous and sensory-evoked activity in the posterior thalamic nucleus (PO). We have also demonstrated that rodent impact SCI increases cell cycle activation (CCA) in the injury region and that post-traumatic treatment with cyclin dependent kinase inhibitors reduces lesion volume and motor dysfunction. Here we examined whether CCA contributes to neuronal hyperexcitability of PO and hyperpathia after rat contusion SCI, as well as to microglial and astroglial activation (gliopathy) that has been implicated in delayed SCI pain. Trauma caused enhanced pain sensitivity, which developed weeks after injury and was correlated with increased PO neuronal activity. Increased CCA was found at the thoracic spinal lesion site, the lumbar dorsal horn, and the PO. Increased microglial activation and cysteine-cysteine chemokine ligand 21 expression was also observed in the PO after SCI. In vitro, neurons co-cultured with activated microglia showed up-regulation of cyclin D1 and cysteine-cysteine chemokine ligand 21 expression. In vivo, post-injury treatment with a selective cyclin dependent kinase inhibitor (CR8) significantly reduced cell cycle protein induction, microglial activation, and neuronal activity in the PO nucleus, as well as limiting chronic SCI-induced hyperpathia. These results suggest a mechanistic role for CCA in the development of SCI pain, through effects mediated in part by the PO nucleus. Moreover, cell cycle modulation may provide an effective therapeutic strategy to improve reduce both hyperpathia and motor dysfunction after SCI.


Subject(s)
Cell Cycle/physiology , Gene Expression Regulation/physiology , Hyperesthesia/etiology , Hyperesthesia/pathology , Posterior Thalamic Nuclei/physiopathology , Spinal Cord Injuries/complications , Action Potentials/drug effects , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclins/pharmacology , Cyclins/therapeutic use , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Follow-Up Studies , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Gliosis/drug therapy , Gliosis/etiology , Male , Microglia/chemistry , Microglia/metabolism , Microglia/pathology , Nerve Fibers, Unmyelinated/pathology , Neurons/drug effects , Pain Threshold/drug effects , Pain Threshold/physiology , Posterior Thalamic Nuclei/drug effects , Posterior Thalamic Nuclei/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Time Factors
8.
Neurobiol Dis ; 54: 252-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23313314

ABSTRACT

Delayed secondary biochemical and cellular changes after traumatic brain injury continue for months to years, and are associated with chronic neuroinflammation and progressive neurodegeneration. Physical activity can reduce inflammation and facilitate recovery after brain injury. Here, we investigated the time-dependent effects, and underlying mechanisms of post-traumatic exercise initiation on outcome after moderate traumatic brain injury using a well-characterized mouse controlled cortical impact model. Late exercise initiation beginning at 5weeks after trauma, but not early initiation of exercise at 1week, significantly reduced working and retention memory impairment at 3months, and decreased lesion volume compared to non-exercise injury controls. Cognitive recovery was associated with attenuation of classical inflammatory pathways, activation of alternative inflammatory responses and enhancement of neurogenesis. In contrast, early initiation of exercise failed to alter behavioral recovery or lesion size, while increasing the neurotoxic pro-inflammatory responses. These data underscore the critical importance of timing of exercise initiation after trauma and its relation to neuroinflammation, and challenge the widely held view that effective neuroprotection requires early intervention.


Subject(s)
Brain Injuries/pathology , Brain Injuries/rehabilitation , Cognition Disorders/rehabilitation , Physical Conditioning, Animal/physiology , Recovery of Function/physiology , Animals , Blotting, Western , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/pathology , Disease Models, Animal , Immunohistochemistry , Inflammation/pathology , Inflammation/prevention & control , Mice , Real-Time Polymerase Chain Reaction
9.
Exp Transl Stroke Med ; 4(1): 25, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23254113

ABSTRACT

BACKGROUND: Stroke occurs more frequently in the elderly population and presents the number one leading cause of persistent disability worldwide. Lack of effective treatment to enhance brain repair and improve functional restoration in chronic stroke, the recovery phase of stroke, is a challenging medical problem to be solved in stroke research. Our early study has revealed the therapeutic effects of stem cell factor (SCF) in combination with granulocyte-colony stimulating factor (G-CSF) (SCF+G-CSF) on chronic stroke in young animals. However, whether this treatment is effective and safe to the aged population remains to be determined. METHODS: Cortical brain ischemia was produced in aged C57BL mice or aged spontaneously hypertensive rats. SCF+G-CSF or equal volume of vehicle solution was subcutaneously injected for 7 days beginning at 3-4 months after induction of cortical brain ischemia. Using the approaches of biochemistry assays, flow cytometry, pathology, and evaluation of functional outcome, several doses of SCF+G-CSF have been examined for their safety and efficiency on chronic stroke in aged animals. RESULTS: All tested doses did not show acute or chronic toxicity in the aged animals. Additionally, SCF+G-CSF treatment in chronic stroke of aged animals mobilized bone marrow stem cells and improved functional outcome in a dose-dependent manner. CONCLUSIONS: SCF+G-CSF treatment is a safe and effective approach to chronic stroke in the aged condition. This study provides important information needed for developing a new therapeutic strategy to improve the health of older adults with chronic stroke.

10.
J Neurochem ; 123(4): 542-54, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22909049

ABSTRACT

HSP70 is a member of the family of heat-shock proteins that are known to be up-regulated in neurons following injury and/or stress. HSP70 over-expression has been linked to neuroprotection in multiple models, including neurodegenerative disorders. In contrast, less is known about the neuroprotective effects of HSP70 in neuronal apoptosis and with regard to modulation of programmed cell death (PCD) mechanisms in neurons. We examined the effects of HSP70 over-expression by transfection with HSP70-expression plasmids in primary cortical neurons and the SH-SY5Y neuronal cell line using four independent models of apoptosis: etoposide, staurosporine, C2-ceramide, and ß-Amyloid. In these apoptotic models, neurons transfected with the HSP70 construct showed significantly reduced induction of nuclear apoptotic markers and/or cell death. Furthermore, we demonstrated that HSP70 binds and potentially inactivates Apoptotic protease-activating factor 1, as well as apoptosis-inducing factor, key molecules involved in development of caspase-dependent and caspase-independent PCD, respectively. Markers of caspase-dependent PCD, including active caspase-3, caspase-9, and cleaved PARP were attenuated in neurons over-expressing HSP70. These data indicate that HSP70 protects against neuronal apoptosis and suggest that these effects reflect, at least in part, to inhibition of both caspase-dependent and caspase-independent PCD pathways.


Subject(s)
Caspases/metabolism , Gene Expression Regulation/physiology , HSP70 Heat-Shock Proteins/metabolism , Neurons/metabolism , Signal Transduction/physiology , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Etoposide/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Immunoprecipitation , Neural Inhibition/drug effects , Neuroblastoma/pathology , Neurons/drug effects , Peptide Fragments/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Staurosporine/pharmacology , Transfection
11.
PLoS One ; 7(7): e42129, 2012.
Article in English | MEDLINE | ID: mdl-22848730

ABSTRACT

Apoptosis of post-mitotic neurons plays a significant role in secondary tissue damage following traumatic spinal cord injury (SCI). Activation of E2F1-dependent transcription promotes expression of pro-apoptotic factors, including CDK1; this signal transduction pathway is believed to represent an important mechanism for the physiological or pathological neuronal cell death. However, a specific role for this pathway in neuronal apoptosis induced by SCI has not yet been reported. Here we demonstrate up-regulation of the E2F1/CDK1 pathway that is associated with neuronal apoptosis following impact SCI in rats. Expression of E2F1 and CDK1 were robustly up-regulated as early as 15 min after injury and sustained until 3 days post-injury. CDK1 activity and E2F1 downstream targets bim and c-Myb were significantly increased after SCI. Activation of E2F1/CDK1 signaling also was associated with death of neurons in vitro; this was attenuated by shRNA knockdown or pharmacological inhibition of the E2F1/CDK1 pathway. CR8, a novel and potent CDK1 inhibitor, blocked apoptosis of primary cortical neurons at low-micromolar concentrations. Moreover, SCI-induced up-regulation of E2F1/CDK1 and associated neuronal apoptosis was significantly attenuated by systemic injection of CR8 (1 mg/kg, i.p.) at 5 min after injury. CR8 significantly decreased posttraumatic elevation of biochemical markers of apoptosis, such as products of caspase-3 and α-fodrin cleavage, as well as neuronal cell death, as indicated by TUNEL staining. Importantly, CR8 treatment also increased the number of surviving neurons at 5 weeks after injury. Together, these findings indicate that activation of the E2F1/CDK1 pathway contributes to the pathophysiology of SCI and that selective inhibition of this signaling cascade may represent an attractive therapeutic strategy.


Subject(s)
Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , E2F1 Transcription Factor/metabolism , Neurons/drug effects , Neurons/pathology , Signal Transduction/drug effects , Spinal Cord Injuries/pathology , Animals , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/deficiency , CDC2 Protein Kinase/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , Gene Silencing , Humans , Male , Neurons/cytology , Neurons/metabolism , Purines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism
12.
Cell Cycle ; 11(9): 1782-95, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22510563

ABSTRACT

Traumatic spinal cord injury (SCI) causes tissue loss and associated neurological dysfunction through mechanical damage and secondary biochemical and physiological responses. We have previously described the pathobiological role of cell cycle pathways following rat contusion SCI by examining the effects of early intrathecal cell cycle inhibitor treatment initiation or gene knockout on secondary injury. Here, we delineate changes in cell cycle pathway activation following SCI and examine the effects of delayed (24 h) systemic administration of flavopiridol, an inhibitor of major cyclin-dependent kinases (CDKs), on functional recovery and histopathology in a rat SCI contusion model. Immunoblot analysis demonstrated a marked upregulation of cell cycle-related proteins, including pRb, cyclin D1, CDK4, E2F1 and PCNA, at various time points following SCI, along with downregulation of the endogenous CDK inhibitor p27. Treatment with flavopiridol reduced induction of cell cycle proteins and increased p27 expression in the injured spinal cord. Functional recovery was significantly improved after SCI from day 7 through day 28. Treatment significantly reduced lesion volume and the number of Iba-1(+) microglia in the preserved tissue and increased the myelinated area of spared white matter as well as the number of CC1(+) oligodendrocytes. Furthermore, flavopiridol attenuated expression of Iba-1 and glactin-3, associated with microglial activation and astrocytic reactivity by reduction of GFAP, NG2, and CHL1 expression. Our current study supports the role of cell cycle activation in the pathophysiology of SCI and by using a clinically relevant treatment model, provides further support for the therapeutic potential of cell cycle inhibitors in the treatment of human SCI.


Subject(s)
Cell Cycle , Flavonoids/pharmacology , Piperidines/pharmacology , Spinal Cord Injuries/physiopathology , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , E2F1 Transcription Factor/metabolism , Flavonoids/administration & dosage , Immunohistochemistry , Locomotion , Male , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Piperidines/administration & dosage , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/physiopathology , Spinal Cord Injuries/drug therapy , Time Factors
13.
Neurobiol Dis ; 46(3): 745-58, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426396

ABSTRACT

Neuronal programmed cell death (PCD) contributes to delayed tissue damage after traumatic brain injury (TBI). Both caspase-dependent and caspase-independent mechanisms have been implicated, with the latter including apoptosis inducing factor (AIF). The peptidyl-proplyl isomerase Cyclophilin A (CypA) transports AIF from the cytosol to the nucleus, a key step for AIF-dependent cell death. We compared the effects of single versus combined inhibition of caspase and AIF pathways in a mouse controlled cortical impact (CCI) model, by examining the effects of CypA gene knockout (CypA(-/-)), caspase inhibition with a pan-caspase inhibitor (boc-aspartyl(OMe)-fluoromethylketone, BAF), or combined modulation. TBI caused caspase activation as well as translocation of AIF to the nucleus. Markers of caspase activation including caspase-specific fodrin cleavage fragments and number of FLIVO-positive cells were reduced in BAF-treated CypA(+/+) mice, whereas markers of AIF activation including AIF/H2AX interaction and AIF translocation to the nucleus were attenuated in CypA(-/-) mice. Each single intervention, (CypA(-/-) or BAF-treated CypA(+/+)) reduced the number of apoptotic cells (TUNEL-positive) in the cortex and improved long-term sensorimotor function; CypA(-/-) also attenuated microglial activation after injury. Importantly, BAF-treated CypA(-/-) mice, showed greater effects than either intervention alone on multiple outcomes including: reduction in TUNEL-positive cells, decrease in neuroinflammation, improved motor and cognitive recovery, and attenuation of lesion volume and neuronal loss in the hippocampus. Using two in vitro neuronal cell death models known to induce AIF-mediated PCD, we also showed that neurons from CypA(-/-) animals were protected and that effects were unrelated to caspase activation. These data indicate that AIF-mediated and caspase-dependent pathways contribute independently and in parallel to secondary injury after TBI, and suggest that combined therapeutic strategies directed at multiple PCD pathways may provide superior neuroprotection than those directed at single mechanisms.


Subject(s)
Apoptosis Inducing Factor/pharmacology , Brain Injuries/drug therapy , Caspases/pharmacology , Cell Death/physiology , Neuroprotective Agents , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Blotting, Western , Brain Injuries/pathology , Cell Survival/drug effects , Cells, Cultured , Cognition/drug effects , Cyclophilin A/genetics , Cyclophilin A/physiology , Cysteine Proteinase Inhibitors/pharmacology , Hippocampus/pathology , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Injections, Intraventricular , Magnetic Resonance Imaging , Mice , Mice, Knockout , Movement/drug effects , Neurons/pathology , Signal Transduction/drug effects
14.
Differentiation ; 83(1): 17-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22099173

ABSTRACT

Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) were originally discovered as growth factors for hematopoietic stem cells (HSCs). It has been well defined that SCF and G-CSF contribute to regulation of lineage commitment for HSCs. However, little is known about whether SCF and G-CSF play roles in the determination and differentiation of neural stem cells (NSCs). Here we demonstrate the novel function of SCF and G-CSF in controlling cell cycle and cell fate determination of NSCs. We also observe that SCF and G-CSF promote neuronal differentiation and inhibit astroglial differentiation at the early stage of differentiation. In addition, our research data reveal that SCF in combination with G-CSF has a dual function in promoting cell cycle exit and directing neuronal fate commitment at the stage of NSC dividing. This coordination effect of SCF+G-CSF on cell cycle arrest and neuronal differentiation is through enhancing neurogenin 1 (Ngn1) activity. These findings extend current knowledge regarding the role of SCF and G-CSF in the regulation of neurogenesis and provide insights into the contribution of hematopoietic growth factors to brain development and remodeling.


Subject(s)
Astrocytes/cytology , Brain/growth & development , Granulocyte Colony-Stimulating Factor/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Stem Cell Factor/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/embryology , Cell Cycle Checkpoints , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Granulocyte Colony-Stimulating Factor/genetics , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Stem Cell Factor/genetics
15.
Alzheimers Res Ther ; 3(2): 8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21406112

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is widely recognized as a serious public health problem and heavy financial burden. Currently, there is no treatment that can delay or stop the progressive brain damage in AD. Recently, we demonstrated that stem cell factor (SCF) in combination with granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) has therapeutic effects on chronic stroke. The purpose of the present study is to determine whether SCF+G-CSF can reduce the burden of ß-amyloid deposits in a mouse model of AD. METHODS: APP/PS1 transgenic mice were used as the model of AD. To track bone marrow-derived cells in the brain, the bone marrow of the APP/PS1 mice was replaced with the bone marrow from mice expressing green fluorescent protein (GFP). Six weeks after bone marrow transplantation, mice were randomly divided into a saline control group and a SCF+G-CSF-treated group. SCF in combination with G-CSF was administered subcutaneously for 12 days. Circulating bone marrow stem cells (CD117+ cells) were quantified 1 day after the final injection. Nine months after treatment, at the age of 18 months, mice were sacrificed. Brain sections were processed for immunohistochemistry to identify ß-amyloid deposits and GFP expressing bone marrow-derived microglia in the brain. RESULTS: Systemic administration of SCF+G-CSF to APP/PS1 transgenic mice leads to long-term reduction of ß-amyloid deposition in the brain. In addition, we have also observed that the SCF+G-CSF treatment increases circulating bone marrow stem cells and augments bone marrow-derived microglial cells in the brains of APP/PS1 mice. Moreover, SCF+G-CSF treatment results in enhancement of the co-localization of bone marrow-derived microglia and ß-amyloid deposits in the brain. CONCLUSIONS: These data suggest that bone marrow-derived microglia play a role in SCF+G-CSF-induced long-term effects to reduce ß-amyloid deposits. This study provides insights into the contribution of the hematopoeitic growth factors, SCF and G-CSF, to limit ß-amyloid accumulation in AD and may offer a new therapeutic approach for AD.

16.
Brain Res ; 1327: 91-102, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20171958

ABSTRACT

Convincing evidence has shown that brain ischemia causes the proliferation of neural stem cells/neural progenitor cells (NSCs/NPCs) in both the subventricular zone (SVZ) and the subgranular zone (SGZ) of adult brain. The role of brain ischemia-induced NSC/NPC proliferation, however, has remained unclear. Here we have determined whether brain ischemia-induced amplification of the NSCs/NPCs in adult brain is required for brain self-protection. The approach of intracerebroventricular (ICV) infusion of cytosine arabinoside (Ara-C), an inhibitor for cell proliferation, for the first 7days after brain ischemia was used to block ischemia-induced NSC/NPC proliferation. We observed that ICV infusion of Ara-C caused a complete blockade of NSC/NPC proliferation in the SVZ and a dramatic reduction of NSC/NPC proliferation in the SGZ. Additionally, as a result of the inhibition of ischemia-induced NSC/NPC pool amplification, the number of neurons in the hippocampal CA1 and CA3 was significantly reduced, the infarction size was significantly enlarged, and neurological deficits were significantly worsened after focal brain ischemia. We also found that an NSC/NPC-conditioned medium showed neuroprotective effects in vitro and that adult NSC/NPC-released brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) are required for NSC/NPC-conditioned medium-induced neuroprotection. These data suggest that NSC/NPC-generated trophic factors are neuroprotective and that brain ischemia-triggered NSC/NPC proliferation is crucial for brain protection. This study provides insights into the contribution of endogenous NSCs/NPCs to brain self-protection in adult brain after ischemia injury.


Subject(s)
Adult Stem Cells/physiology , Brain Infarction/prevention & control , Brain Ischemia/pathology , Lateral Ventricles/pathology , Neurons/physiology , Adult Stem Cells/drug effects , Analysis of Variance , Animals , Behavior, Animal , Brain Infarction/etiology , Brain Ischemia/complications , Bromodeoxyuridine/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/pharmacology , Cytarabine/pharmacology , Disease Models, Animal , Green Fluorescent Proteins/genetics , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurologic Examination/methods , Neurons/drug effects , SOXB1 Transcription Factors/metabolism
17.
Neurosci Res ; 64(4): 355-61, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19379782

ABSTRACT

alphaB-crystallin is a member of the small heat shock proteins, which is abundantly expressed in various vertebrate tissues including the central nervous system. In our previous report, we showed alphaB-crystallin induction in activated astrocytes in the postischemic brain and in H2O2-treated primary astrocyte cultures. To investigate the functional significance of alphaB-crystallin induction in astrocytes, we generated a stable C6 astroglioma cell line overexpressing alphaB-crystallin. In these cells, hydrogen peroxide-induced apoptosis was reduced by 60% compared to parent cells. Furthermore, the repression of alphaB-crystallin expression by alphaB-crystallin siRNA transfection suppressed this protective effect, indicating that alphaB-crystallin is responsible for the protection against H2O2-induced apoptosis in C6 astroglioma cells. Similar level of aggravation in H2O2-induced apoptosis was observed in primary astrocyte cultures when alphaB-crystallin expression was suppressed by alphaB-crystallin siRNA transfection, confirming the importance of alphaB-crystallin. In addition, the induction of caspase-3 activity after H2O2 treatment was markedly suppressed in alphaB-crystallin-overexpressing cells, and immunoprecipitation proved binding between alphaB-crystallin and partially processed caspase-3 (a p24 intermediate). These results indicate that alphaB-crystallin confers protection against hydrogen peroxide-induced astrocytes apoptosis in part by inhibiting caspase-3 activation.


Subject(s)
Apoptosis/physiology , Astrocytes/metabolism , Caspase 3/metabolism , Oxidative Stress/physiology , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Cell Line, Tumor , Cytoprotection/physiology , Down-Regulation/physiology , Hydrogen Peroxide/toxicity , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Oxidants/toxicity , Oxidative Stress/drug effects , Protein Binding/physiology , RNA Interference/physiology , Rats , Transfection
18.
J Cereb Blood Flow Metab ; 29(4): 759-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19209180

ABSTRACT

Chronic stroke is a highly important but under-investigated scientific problem in neurologic research. We have reported earlier that stem cell factor (SCF) in combination with granulocyte-colony stimulating factor (G-CSF) treatment during chronic stroke improves functional outcomes. Here we have determined the contribution of bone marrow-derived cells in angiogenesis and neurogenesis, which are enhanced by SCF+G-CSF treatment during chronic stroke. Using bone marrow tracking, flow cytometry, 2-photon live brain imaging, and immunohistochemistry, we observed that the levels of circulating bone marrow stem cells (BMSCs) (CD34+/c-kit+) were significantly increased by SCF+G-CSF treatment. In addition, live brain imaging revealed that numerous bone marrow-derived cells migrate into the brain parenchyma in the treated mice. We also found that bone marrow-derived cells, bone marrow-derived endothelial cells, vascular density, and bone marrow-derived neurons were significantly augmented by SCF+G-CSF. It is interesting that, in addition to the increase in bone marrow-derived endothelial cells, the number of bone marrow-derived pericytes was reduced after SCF+G-CSF treatment during chronic stroke. These data suggest that SCF+G-CSF treatment can enhance repair of brain damage during chronic stroke by mobilizing BMSCs, and promoting the contribution of bone marrow-derived cells to angiogenesis and neurogenesis.


Subject(s)
Brain/physiology , Granulocyte Colony-Stimulating Factor/physiology , Neurogenesis/physiology , Regeneration/physiology , Stem Cell Factor/physiology , Stroke/pathology , Animals , Bone Marrow Cells , Brain/pathology , Cerebrovascular Circulation , Chronic Disease , Mice , Neovascularization, Physiologic , Stem Cells/physiology
19.
Neurosci Lett ; 435(2): 131-6, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18343576

ABSTRACT

AlphaB-crystallin, known as a vertebrate lens protein, is a member of the small heat shock proteins (sHSP). AlphaB-crystallin is abundantly expressed in the vertebrate lens and striated muscles and it is also expressed constitutively in other tissues including the central nervous system (CNS). In our previous report, we showed alphaB-crystallin induction in activated astrocytes, which are enriched in the penumbra after transient focal cerebral ischemia. We also reported that alphaB-crystallin is significantly induced in astrocytes in the CA3 region of the hippocampus following KA-induced seizure. Here, we report that the expression of alphaB-crystallin is upregulated in H2O2-treated primary astrocyte cultures, which was prepared from newborn male Sprague-Dawley rats and that the proximal 408 bp of the alphaB-crystallin promoter harboring stress response element (STRE) is responsible for this induction. This effect of H2O2 was found to be virtually abolished by introducing mutations into STRE, and these mutations also impaired increased lens epithelial derived growth factor (LEDGF) binding to STRE after H2O2 treatment. Moreover, LEDGF was induced in primary astrocyte cultures after H2O2 treatment and alphaB-crystallin induction was significantly suppressed by transfecting small interfering RNA (siRNA) targeting LEDGF. Together these results indicate that the H2O2-induced upregulations of alphaB-crystallin in astrocytes are mediated by the LEDGF-STRE interaction on alphaB-crystallin promoter.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Oxidative Stress/physiology , alpha-Crystallin B Chain/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Male , Oxidative Stress/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Response Elements/physiology , Time Factors , alpha-Crystallin B Chain/genetics
20.
Exp Neurol ; 204(2): 569-73, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17307165

ABSTRACT

We have previously demonstrated that receptors for hematopoietic growth factors, stem cell factor (SCF) and granulocyte-colony stimulating factor (G-CSF) are expressed in the neurons and the neural progenitor cells (NPCs) of the adult rat brain, and that systemic administration of SCF and G-CSF in the first week after induction of cortical brain ischemia (3 h-7 days post-ischemia) significantly improves functional outcome, augments NPC proliferation, and reduces infarct volume in rats. The purpose of the present study is to determine whether SCF and G-CSF pass through the blood-brain barrier (BBB) in intact rats. The growth factors were labeled with iodine (I(125)), a radioactive compound. I(125)-SCF and I(125)-G-CSF were intravenously administered and the concentrations of I(125)-SCF and I(125)-G-CSF in the blood plasma and the brain were determined at 10, 30, 60, and 120 min after injection. We observed that both SCF and G-CSF were slowly and continuously transported from the blood stream to the brain in the same rate. In addition, both immunofluorescent staining and Western blots showed that receptors for SCF and G-CSF were expressed in the capillaries of the adult rat brain, suggesting that SCF and G-CSF entry to the brain may be mediated via receptor-mediated transport, one of the endogenous transports in the BBB. These data indicate that both SCF and G-CSF were able to pass through the BBB in intact animals. This observation will help in further exploring the physiological role of peripheral SCF and G-CSF in the brain and therapeutic possibility to chronic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Stem Cell Factor/metabolism , Animals , Biological Transport/physiology , Iodine/metabolism , Rats , Recombinant Proteins , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...