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.
J Cereb Blood Flow Metab ; 36(8): 1449-63, 2016 08.
Article in English | MEDLINE | ID: mdl-26661154

ABSTRACT

Stroke is the second leading cause of death and the most frequent cause of adult disability. Neuronal Per-Arnt-Sim domain protein 4 (Npas4) is an activity-dependent transcription factor whose expression is induced in various brain insults, including cerebral ischaemia. Although previous studies have demonstrated that Npas4 plays a critical role in protecting neurons against neurodegenerative insults, the neuroprotective effect of Npas4 in response to ischaemic brain injury remains unknown. In this study, we used a loss-of-function approach to examine the neuroprotective potential of Npas4 in the context of ischaemic damage. Using oxygen and glucose deprivation, we demonstrated that the knockdown of Npas4 in mouse cortical neurons resulted in increased susceptibility to cell death. The protective effect of Npas4 was further investigated in vivo using a photochemically-induced stroke model in mice. We found a significantly larger lesion size and increased neurodegeneration in Npas4 knockout mice as compared to wild-type mice. Moreover, we also showed that ablation of Npas4 caused an increase in activated astrocytes and microglia, pro-inflammatory cytokines interleukin-6 and tumour necrosis factor alpha levels and a switch from apoptotic to necrotic cell death. Taken together, these data suggest that Npas4 plays a neuroprotective role in ischaemic stroke by limiting progressive neurodegeneration and neuroinflammation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Ischemia/metabolism , Brain/metabolism , Brain/pathology , Neurons/metabolism , Neurons/pathology , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Brain/immunology , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Survival/drug effects , Cells, Cultured , Culture Media , Gene Knockout Techniques , Glucose/metabolism , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/immunology , Oxygen/metabolism , Primary Cell Culture , Real-Time Polymerase Chain Reaction
2.
Stem Cell Res Ther ; 5(3): 64, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24887558

ABSTRACT

INTRODUCTION: Npas4 is a calcium-dependent transcription factor expressed within neurons of the brain where it regulates the expression of several genes that are important for neuronal survival and synaptic plasticity. It is known that in the adult brain Npas4 plays an important role in several key aspects of neurobiology including inhibitory synapse formation, neuroprotection and memory, yet very little is known about the role of Npas4 during neurodevelopment. The aim of this study was to examine the expression and function of Npas4 during nervous system development by using a combination of in vivo experiments in the developing mouse embryo and neural differentiation of embryonic stem cells (ESCs) as an in vitro model of the early stages of embryogenesis. METHODS: Two different neural differentiation paradigms were used to investigate Npas4 expression during neurodevelopment in vitro; adherent monolayer differentiation of mouse ESCs in N2B27 medium and Noggin-induced differentiation of human ESCs. This work was complemented by direct analysis of Npas4 expression in the mouse embryo. The function of Npas4 in the context of neurodevelopment was investigated using loss-of-function experiments in vitro. We created several mouse ESC lines in which Npas4 expression was reduced during neural differentiation through RNA interference and we then analyzed the ability of these Npas4 knockdown mouse ESCs lines to undergo neural differentiation. RESULTS: We found that while Npas4 is not expressed in undifferentiated ESCs, it becomes transiently up-regulated during neural differentiation of both mouse and human ESCs at a stage of differentiation that is characterized by proliferation of neural progenitor cells. This was corroborated by analysis of Npas4 expression in the mouse embryo where the Npas4 transcript was detected specifically in the developing forebrain beginning at embryonic day 9.5. Finally, knockdown of Npas4 expression in mouse ESCs undergoing neural differentiation affected their ability to differentiate appropriately, resulting in delayed neural differentiation. CONCLUSIONS: Here we provide the first evidence that Npas4 is expressed during embryonic development and that it may have a developmental role that is unrelated to its function in the adult brain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Neurogenesis/physiology , Animals , Cell Line , Embryo, Mammalian , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/metabolism , Polymerase Chain Reaction
3.
Stem Cells ; 31(6): 1040-3, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23390084

ABSTRACT

Stroke, a debilitating brain insult, afflicts millions of individuals globally each year. In the last decade, researchers have investigated cell-based therapy as an alternative strategy to improve neurological outcome following stroke. This concise review critically examines preclinical reports using human adult and fetal stem/progenitor cells in rodent models of ischemic stroke. As we enter the second decade of study, we should aim to optimize our collective likelihood to translational success for stroke victims worldwide. We advocate international consensus recommendations be developed for future preclinical research.


Subject(s)
Brain Ischemia/therapy , Cell- and Tissue-Based Therapy/methods , Stem Cells/cytology , Stroke/therapy , Animals , Disease Models, Animal , Humans
4.
Eur J Neurosci ; 37(11): 1875-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23431968

ABSTRACT

The neuronal Per-Arnt-Sim domain protein 4 (Npas4) is an important transcriptional regulator of synaptic plasticity and cognition. The present study characterises the in vivo neuroanatomical expression pattern of the Npas4 protein in a rat model of focal cerebral ischemia. Animals were subjected to unilateral middle cerebral artery occlusion for 2 h, after which the spatiotemporal and neuronal profiles of Npas4 protein expression were analysed by immunohistochemistry at different time points post-reperfusion. Focal cerebral ischemia induced an early, transient and robust upregulation of Npas4 in a brain region-dependent manner involving predominantly principal neurons. Interestingly, we observed a unique differential induction of Npas4 protein expression in corticolimbic regions of the rat brain that are critically linked to cognition and emotion. These findings suggest that stroke-induced Npas4 upregulation may be involved in a transcriptional regulatory program within the corticolimbic circuitry following an ischemic insult.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/metabolism , Infarction, Middle Cerebral Artery/metabolism , Limbic System/metabolism , Up-Regulation , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cerebral Cortex/pathology , Limbic System/pathology , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley
5.
Stem Cells Transl Med ; 1(3): 177-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-23197777

ABSTRACT

Human adult dental pulp stem cells (DPSCs), derived from third molar teeth, are multipotent and have the capacity to differentiate into neurons under inductive conditions both in vitro and following transplantation into the avian embryo. In this study, we demonstrate that the intracerebral transplantation of human DPSCs 24 hours following focal cerebral ischemia in a rodent model resulted in significant improvement in forelimb sensorimotor function at 4 weeks post-treatment. At this time, 2.3 ± 0.7% of engrafted cells had survived in the poststroke brain and demonstrated targeted migration toward the stroke lesion. In the peri-infarct striatum, transplanted DPSCs differentiated into astrocytes in preference to neurons. Our data suggest that the dominant mechanism of action underlying DPSC treatment that resulted in enhanced functional recovery is unlikely to be due to neural replacement. Functional improvement is more likely to be mediated through DPSC-dependent paracrine effects. This study provides preclinical evidence for the future use of human DPSCs in cell therapy to improve outcome in stroke patients.


Subject(s)
Adult Stem Cells/cytology , Astrocytes/cytology , Brain Ischemia/therapy , Cell Differentiation , Dental Pulp/cytology , Stem Cell Transplantation , Stroke/prevention & control , Adult , Adult Stem Cells/physiology , Animals , Astrocytes/physiology , Behavior, Animal , Brain Ischemia/metabolism , Brain Ischemia/pathology , Dental Pulp/physiology , Forelimb/cytology , Forelimb/physiology , Humans , Immunoenzyme Techniques , Male , Neuropsychological Tests , Rats , Rats, Sprague-Dawley , Sensory Gating
SELECTION OF CITATIONS
SEARCH DETAIL
...