Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Transplant ; 24(4): 721-35, 2015.
Article in English | MEDLINE | ID: mdl-25413246

ABSTRACT

Human umbilical cord blood (HUCB) cell therapies have shown promising results in reducing brain infarct volume and most importantly in improving neurobehavioral function in rat permanent middle cerebral artery occlusion, a model of stroke. In this study, we examined the gene expression profile in neurons subjected to oxygen-glucose deprivation (OGD) with or without HUCB treatment and identified signaling pathways (Akt/MAPK) important in eliciting HUCB-mediated neuroprotective responses. Gene chip microarray analysis was performed using RNA samples extracted from the neuronal cell cultures from four experimental groups: normoxia, normoxia+HUCB, OGD, and OGD+HUCB. Both quantitative RT-PCR and immunohistochemistry were carried out to verify the microarray results. Using the Genomatix software program, promoter regions of selected genes were compared to reveal common transcription factor-binding sites and, subsequently, signal transduction pathways. Under OGD condition, HUCB cells significantly reduced neuronal loss from 68% to 44% [one-way ANOVA, F(3, 16)=11, p=0.0003]. Microarray analysis identified mRNA expression of Prdx5, Vcam1, CCL20, Alcam, and Pax6 as being significantly altered by HUCB cell treatment. Inhibition of the Akt pathway significantly abolished the neuroprotective effect of HUCB cells [one-way ANOVA, F(3, 11)=8.663, p=0.0031]. Our observations show that HUCB neuroprotection is dependent on the activation of the Akt signaling pathway that increases transcription of the Prdx5 gene. We concluded that HUCB cell therapy would be a promising treatment for stroke and other forms of brain injury by modifying acute gene expression to promote neural cell protection.


Subject(s)
Fetal Blood/metabolism , Gene Expression Regulation , Infarction, Middle Cerebral Artery/pathology , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcriptome , Animals , Binding Sites , Cells, Cultured , Coculture Techniques , Fetal Blood/cytology , Fetal Blood/transplantation , Humans , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/therapy , Neurons/cytology , Neuroprotective Agents , Oligonucleotide Array Sequence Analysis , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/metabolism
2.
Eur J Neurosci ; 40(7): 3111-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25041106

ABSTRACT

Human umbilical cord blood (HUCB) cells have shown efficacy in rodent models of focal ischemia and in vitro systems that recapitulate stroke conditions. One potential mechanism of protection is through secretion of soluble factors that protect neurons and oligodendrocytes (OLs) from oxidative stress. To overcome practical issues with cellular therapies, identification of soluble factors released by HUCB and other stem cells may pave the way for treatment modalities that are safer for a larger percentage of stroke patients. Among these soluble factors is leukemia inhibitory factor (LIF), a cytokine that exerts pleiotropic effects on cell survival. Here, data show that LIF effectively reduced infarct volume, reduced white matter injury and improved functional outcomes when administered to rats following permanent middle cerebral artery occlusion. To further explore downstream signaling, primary oligodendrocyte cultures were exposed to oxygen-glucose deprivation to mimic stroke conditions. LIF significantly reduced lactate dehydrogenase release from OLs, reduced superoxide dismutase activity and induced peroxiredoxin 4 (Prdx4) transcript. Additionally, the protective and antioxidant capacity of LIF was negated by both Akt inhibition and co-incubation with Prdx4-neutralising antibodies, establishing a role for the Akt signaling pathway and Prdx4-mediated antioxidation in LIF protection.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Leukemia Inhibitory Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Recovery of Function/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Leukemia Inhibitory Factor/pharmacology , Neuroprotective Agents/pharmacology , Oncogene Protein v-akt/metabolism , Peroxiredoxins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stroke/drug therapy , White Matter/drug effects
3.
Metab Brain Dis ; 27(2): 131-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22354752

ABSTRACT

Delayed neuronal death associated with stroke has been increasingly linked to the immune response to the injury. Splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective and significantly reduces neuroinflammation. The present study investigated whether splenic signaling occurs through interferon gamma (IFNγ). IFNγ was elevated early in spleens but later in the brains of rats following MCAO. Splenectomy decreased the amount of IFNγ in the infarct post-MCAO. Systemic administration of recombinant IFNγ abolished the protective effects of splenectomy with a concurrent increase in INFγ expression in the brain. These results suggest a role for spleen-derived IFNγ in stroke pathology.


Subject(s)
Interferon-gamma/physiology , Nerve Degeneration/physiopathology , Spleen/physiopathology , Stroke/physiopathology , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Hypoxia , Cells, Cultured , Female , Fluoresceins , Fluorescent Dyes , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Interferon-gamma/pharmacology , Laser-Doppler Flowmetry , Ligation , Male , Middle Cerebral Artery/physiology , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/metabolism , Organic Chemicals , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Spleen/metabolism , Splenectomy
4.
Transl Stroke Res ; 3(3): 357-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-24323811

ABSTRACT

Traumatic brain injury (TBI) is complex and involves multiple processes that contribute to functional decline. Progressive neuropathies result from delayed cellular death following the initial impact. Although the precise mechanisms responsible for delayed injury are unknown, numerous data implicate a role for the peripheral immune system in perpetuating neuroinflammation after TBI. A previous report demonstrated that splenic CCL20 chemokine expression was upregulated 24 h after lateral fluid percussive impact (LFPI), prior to neuronal expression but consistent with neurodegeneration. Here, we expand on those data to report increased CCL20 protein expression in white matter 48 h after LFPI and demonstrate that CCL20 is directly toxic to primary neurons and oligodendrocytes subjected to oxygen glucose deprivation. The temporal expression profile of CCL20, coupled with in vitro toxicity to primary cells, suggests that this chemokine exerts deleterious effects on cell viability following TBI. These findings warrant further investigations into the use of CCL20 as a potential biomarker and/or therapeutic target.

5.
J Biol Chem ; 287(6): 4177-87, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22158864

ABSTRACT

Human umbilical cord blood (HUCB) cells protect the brain against ischemic injury, yet the mechanism of protection remains unclear. Using both in vitro and in vivo paradigms, this study examined the role of Akt signaling and peroxiredoxin 4 expression in human umbilical cord blood cell-mediated protection of oligodendrocytes from ischemic conditions. As previously reported, the addition of HUCB cells to oligodendrocyte cultures prior to oxygen glucose deprivation significantly enhanced oligodendrocyte survival. The presence of human umbilical cord blood cells also increased Akt phosphorylation and elevated peroxiredoxin 4 expression in oligodendrocytes. Blocking either Akt or peroxiredoxin 4 activity with Akt Inhibitor IV or a peroxiredoxin 4-neutralizing antibody, respectively, negated the protective effects of human umbilical cord blood cells. In vivo, systemic administration of human umbilical cord blood cells 48 h after middle cerebral artery occlusion increased Akt phosphorylation and peroxiredoxin 4 protein expression while reducing proteolytic cleavage of caspase 3 in oligodendrocytes residing in the ipsilateral external capsule. Moreover, human umbilical cord blood cells protected striatal white matter bundles from degeneration following middle cerebral artery occlusion. These results suggest that the soluble factors released from human umbilical cord blood cells converge on Akt to elevate peroxiredoxin 4 levels, and these effects contribute to oligodendrocyte survival.


Subject(s)
Brain Ischemia/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Umbilical Cord/cytology , Animals , Brain Ischemia/pathology , Brain Ischemia/therapy , Caspase 3/biosynthesis , Cell Survival , Humans , Oligodendroglia/pathology , Peroxiredoxins/biosynthesis , Phosphorylation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...