Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 34(9): 3419-28, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24573298

ABSTRACT

The neurotrophin receptor p75(NTR) has been implicated in mediating neuronal apoptosis after injury to the CNS. Despite its frequent induction in pathologic states, there is limited understanding of the mechanisms that regulate p75(NTR) expression after injury. Here, we show that after focal cerebral ischemia in vivo or oxygen-glucose deprivation in organotypic hippocampal slices or neurons, p75(NTR) is rapidly induced. A concomitant induction of proNGF, a ligand for p75(NTR), is also observed. Induction of this ligand/receptor system is pathologically relevant, as a decrease in apoptosis, after oxygen-glucose deprivation, is observed in hippocampal neurons or slices after delivery of function-blocking antibodies to p75(NTR) or proNGF and in p75(NTR) and ngf haploinsufficient slices. Furthermore, a significant decrease in infarct volume was noted in p75(NTR)-/- mice compared with the wild type. We also investigated the regulatory mechanisms that lead to post-ischemic induction of p75(NTR). We demonstrate that induction of p75(NTR) after ischemic injury is independent of transcription but requires active translation. Basal levels of p75(NTR) in neurons are maintained in part by the expression of microRNA miR-592, and an inverse correlation is seen between miR-592 and p75(NTR) levels in the adult brain. After cerebral ischemia, miR-592 levels fall, with a corresponding increase in p75(NTR) levels. Importantly, overexpression of miR-592 in neurons decreases the level of ischemic injury-induced p75(NTR) and attenuates activation of pro-apoptotic signaling and cell death. These results identify miR-592 as a key regulator of p75(NTR) expression and point to a potential therapeutic candidate to limit neuronal apoptosis after ischemic injury.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation/physiology , Infarction, Middle Cerebral Artery/pathology , MicroRNAs/metabolism , Neurons/physiology , Receptors, Nerve Growth Factor/metabolism , Age Factors , Animals , Apoptosis/drug effects , Apoptosis/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Glucose/deficiency , Hippocampus/pathology , Humans , Hypoxia , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Nerve Growth Factor/metabolism , Protein Precursors/metabolism , RNA, Small Interfering/metabolism , Receptors, Nerve Growth Factor/genetics
2.
J Neurosci ; 29(47): 14790-802, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19940174

ABSTRACT

Although mature neurotrophins are well described trophic factors that elicit retrograde survival signaling, the precursor forms of neurotrophins (i.e., proneurotrophins) can function as high-affinity apoptotic ligands for selected neural populations. An outstanding question is whether target-derived proneurotrophins might affect neuronal survival/death decisions through a retrograde transport mechanism. Since neurotrophin-3 (NT-3) is highly expressed in non-neural tissues that receive peripheral innervation, we investigated the localized actions of its precursor (proNT-3) on sympathetic neurons in the present study. Pharmacological inhibition of intracellular furin proteinase activity in 293T cells resulted in proNT-3 release instead of mature NT-3, whereas membrane depolarization in cerebellar granule neurons stimulated endogenous proNT-3 secretion, suggesting that proNT-3 is an inducible bona fide ligand in the nervous system. Our data also indicate that recombinant proNT-3 induced sympathetic neuron death that is p75(NTR)- and sortilin-dependent, with hallmark features of apoptosis including JNK (c-Jun N-terminal kinase) activation and nuclear fragmentation. Using compartmentalized culture systems that segregate neuronal cell bodies from axons, proNT-3, acting within the distal axon compartment, elicited sympathetic neuron death and overrode the survival-promoting actions of NGF. Together, these results raise the intriguing possibility that dysregulation of proneurotrophin processing/release by innervated targets can be deleterious to the neurons projecting to these sites.


Subject(s)
Apoptosis/physiology , Axonal Transport/physiology , Nerve Degeneration/metabolism , Nervous System/metabolism , Neurons/metabolism , Neurotrophin 3/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Ligands , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Nerve Degeneration/physiopathology , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Nervous System/cytology , Neurons/drug effects , PC12 Cells , Rats , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...