Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 10(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37558465

RESUMO

Traumatic brain injury (TBI) elicits neuronal loss at the site of injury and progressive neuronal loss in the penumbra. However, the consequences of TBI on afferent neurons projecting to the injured tissue from distal locations is unknown. Basal forebrain cholinergic neurons (BFCNs) extend long projections to multiple brain regions including the cortex, regulate many cognitive functions, and are compromised in numerous neurodegenerative disorders. To determine the consequence of cortical injury on these afferent neurons, we used the fluid percussion injury model of traumatic brain injury and assessed the effects on BFCN survival and axon integrity in male and female mice. Survival or death of BF neurons can be regulated by neurotrophins or proneurotrophins, respectively. The injury elicited an induction of proNGF and proBDNF in the cortex and a loss of BFCNs ipsilateral to the injury compared with sham uninjured mice. The p75NTR knock-out mice did not show loss of BFCN neurons, indicating a retrograde degenerative effect of the cortical injury on the afferent BFCNs mediated through p75NTR. In contrast, locus ceruleus neurons, which also project throughout the cortex, were unaffected by the injury, suggesting specificity in retrograde degeneration after cortical TBI. Proneurotrophins (proNTs) provided directly to basal forebrain axons in microfluidic cultures triggered retrograde axonal degeneration and cell death, which did not occur in the absence of p75NTR. This study shows that after traumatic brain injury, proNTs induced in the injured cortex promote BFCN axonal degeneration and retrograde neuron loss through p75NTR.


Assuntos
Prosencéfalo Basal , Lesões Encefálicas Traumáticas , Receptores de Fator de Crescimento Neural , Animais , Feminino , Masculino , Camundongos , Lesões Encefálicas Traumáticas/metabolismo , Neurônios Colinérgicos/metabolismo , Neurônios Aferentes , Degeneração Retrógrada/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo
2.
Glia ; 71(10): 2383-2400, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37334743

RESUMO

The precise timing of neural progenitor development and the correct balance between proliferation and differentiation are crucial to generating a functional brain. The number, survival, and differentiation of neural progenitors during postnatal neurogenesis and gliogenesis is a highly regulated process. Postnatally, the majority of brain oligodendrocytes are generated from progenitors residing in the subventricular zone (SVZ), the germinal niche surrounding the lateral ventricles. In this study, we demonstrate that the p75 neurotrophin receptor (p75NTR) is highly expressed by OPCs in the postnatal male and female rat SVZ. Whereas the p75NTR is known to initiate apoptotic signaling after brain injury, it is highly expressed by proliferating progenitors in the SVZ, suggesting that it may have a different function during development. Lack of p75NTR reduced progenitor proliferation and caused premature oligodendrocyte differentiation and maturation both in vitro and in vivo, leading to aberrant early myelin formation. Our data reveal a novel role for p75NTR as a rheostat for oligodendrocyte production and maturation during myelin formation in the postnatal rat brain.


Assuntos
Ventrículos Laterais , Células Precursoras de Oligodendrócitos , Animais , Feminino , Masculino , Ratos , Encéfalo , Diferenciação Celular , Neurogênese/fisiologia , Oligodendroglia/fisiologia , Receptor de Fator de Crescimento Neural
3.
Elife ; 112022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36040414

RESUMO

Neuronal migration is one of the fundamental processes during brain development. Several neurodevelopmental disorders can be traced back to dysregulated migration. Although substantial efforts have been placed in identifying molecular signals that stimulate migration, little is known about potential mechanisms that restrict migration. These restrictive mechanisms are essential for proper development since it helps coordinate the timing for each neuronal population to arrive and establish proper connections. Moreover, preventing migration away from a proliferative niche is necessary in maintaining a pool of proliferating cells until the proper number of neuronal progenitors is attained. Here, using mice and rats, we identify an anti-migratory role for the p75 neurotrophin receptor (p75NTR) in cerebellar development. Our results show that granule cell precursors (GCPs) robustly express p75NTR in the external granule layer (EGL) when they are proliferating during postnatal development, however, they do not express p75NTR when they migrate either from the rhombic lip during embryonic development or from the EGL during postnatal development. We show that p75NTR prevented GCP migration by maintaining elevated levels of active RhoA. The expression of p75NTR was sufficient to prevent the migration of the granule cells even in the presence of BDNF (brain-derived neurotrophic factor), a well-established chemotactic signal for this cell population. Our findings suggest that the expression of p75NTR might be a critical signal that stops and maintains the GCPs in the proliferative niche of the EGL, by promoting the clonal expansion of cerebellar granule neurons.


The human brain contains billions of neurons that form vast networks to relay information around the brain and to the rest of the body. The numbers and locations of neurons, and the connections between them, affect how the brain works, so the body carefully controls how, where and when neurons form. Most of the neurons in the brain arise before we are born from groups of supporting cells known as neuronal precursors. Often, these cells must migrate from one place to another to make neurons in the correction location. For example, neuronal precursors in an area of the embryo brain, called the rhombic lip, produce granule cells ­ a type of neuron found in the cerebellum, a region of the adult brain that controls our ability to move around. Before making the neurons, the precursor cells first have to migrate out of the rhombic lip into a neighboring area. Previous studies indicate that a protein known as p75NTR may help to control the ability of brain cells to migrate, but its precise role remained unclear. To address this question, Zanin and Friedman investigated the role of p75NTR in the migration of granule cell precursors in mice and rats. The experiments found that in animals lacking this protein, the granule cell precursors began to migrate out of the rhombic lip earlier than in normal animals, resulting in excessive numbers of granule cells in the adult cerebellum, which can affect the normal development of an animal. The p75NTR protein appeared to prevent the cells from migrating by activating another protein called RhoA. Understanding how the body controls when neuronal precursors and other brain cells migrate helps us to understand how the brain develops in healthy individuals and certain neurological disorders, including autism. The next step is to find out whether p75NTR also plays a similar role in the human brain.


Assuntos
Cerebelo , Receptor de Fator de Crescimento Neural , Animais , Encéfalo/metabolismo , Movimento Celular/fisiologia , Cerebelo/fisiologia , Camundongos , Neurônios/fisiologia , Ratos , Receptor de Fator de Crescimento Neural/metabolismo
4.
ASN Neuro ; 12: 1759091420930865, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32493127

RESUMO

The p75 neurotrophin receptor (p75NTR) can regulate multiple cellular functions including proliferation, survival, and apoptotic cell death. The p75NTR is widely expressed in the developing brain and is downregulated as the nervous system matures, with only a few neuronal subpopulations retaining expression into adulthood. However, p75NTR expression is induced following damage to the adult brain, including after traumatic brain injury, which is a leading cause of mortality and disability worldwide. A major consequence of traumatic brain injury is the progressive neuronal loss that continues secondary to the initial trauma, which ultimately contributes to cognitive decline. Understanding mechanisms governing this progressive neuronal death is key to developing targeted therapeutic strategies to provide neuroprotection and salvage cognitive function. In this study, we demonstrate that a cortical impact injury to the sensorimotor cortex elicits p75NTR expression in apoptotic neurons in the injury penumbra, confirming previous studies. To establish whether preventing p75NTR induction or blocking the ligands would reduce the extent of secondary neuronal cell death, we used a noninvasive intranasal strategy to deliver either siRNA to block the induction of p75NTR, or function-blocking antibodies to the ligands pro-nerve growth factor and pro-brain-derived neurotrophic factor. We demonstrate that either preventing the induction of p75NTR or blocking the proneurotrophin ligands provides neuroprotection and preserves sensorimotor function.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas Traumáticas/metabolismo , Neurônios/metabolismo , Receptores de Fator de Crescimento Neural/biossíntese , Administração Intranasal/métodos , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/psicologia , Morte Celular/fisiologia , Técnicas de Silenciamento de Genes/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , RNA Interferente Pequeno/administração & dosagem , Receptores de Fator de Crescimento Neural/antagonistas & inibidores
5.
Front Cell Neurosci ; 13: 485, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31736712

RESUMO

Neurotrophins activate Trk receptor signaling to support neuronal survival and many aspects of neuronal function. Early studies demonstrated that TrkA formed a complex with the p75 neurotrophin receptor (p75 NTR ), which increased the affinity and selectivity of NGF binding, however, whether interaction of p75 NTR with other Trk receptors performs a similar function to enhance ligand binding has not been demonstrated. We investigated the interaction of TrkB with full length p75 NTR in hippocampal neurons in response to BDNF and found that the association of these receptors occurs after ligand binding and requires phosphorylation of TrkB, indicating that formation of this receptor complex was not necessary for ligand binding. Moreover, the interaction of these receptors required internalization and localization to early endosomes. We found that association of TrkB with p75 NTR was necessary for optimal downstream signaling of the PI3K-Akt pathway, but not the Erk pathway, in hippocampal neurons. The absence of p75 NTR impaired the ability of BDNF to rescue hippocampal neurons in a trophic deprivation model, suggesting that p75 NTR facilitates the ability of TrkB to activate specific pathways to promote neuronal survival.

6.
J Neurosci ; 39(46): 9119-9129, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31582529

RESUMO

Development of brain circuitry requires precise regulation and timing of proliferation and differentiation of neural progenitor cells. The p75 neurotrophin receptor (p75NTR) is highly expressed in the proliferating granule cell precursors (GCPs) during development of the cerebellum. In a previous paper, we showed that proNT3 promoted GCP cell cycle exit via p75NTR. Here we used genetically modified rats and mice of both sexes to show that p75NTR regulates the duration of the GCP cell cycle, requiring activation of RhoA. Rats and mice lacking p75NTR have dysregulated GCP proliferation, with deleterious effects on cerebellar circuit development and behavioral consequences persisting into adulthood. In the absence of p75NTR, the GCP cell cycle is accelerated, leading to delayed cell cycle exit, prolonged GCP proliferation, increased glutamatergic input to Purkinje cells, and a deficit in delay eyeblink conditioning, a cerebellum-dependent form of learning. These results demonstrate the necessity of appropriate developmental timing of the cell cycle for establishment of proper connectivity and associated behavior.SIGNIFICANCE STATEMENT The cerebellum has been shown to be involved in numerous behaviors in addition to its classic association with motor function. Cerebellar function is disrupted in a variety of psychiatric disorders, including those on the autism spectrum. Here we show that the p75 neurotrophin receptor, which is abundantly expressed in the proliferating cerebellar granule cell progenitors, regulates the cell cycle of these progenitors. In the absence of this receptor, the cell cycle is dysregulated, leading to excessive progenitor proliferation, which alters the balance of inputs to Purkinje cells, disrupting the circuitry and leading to functional deficits that persist into adulthood.


Assuntos
Ciclo Celular/fisiologia , Cerebelo/crescimento & desenvolvimento , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Animais , Proliferação de Células , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Potenciais Pós-Sinápticos Excitadores , Feminino , Masculino , Camundongos Transgênicos , Proteínas do Tecido Nervoso , Células de Purkinje/fisiologia , Células de Purkinje/ultraestrutura , Ratos Transgênicos , Receptores de Fatores de Crescimento
7.
Eur J Cell Biol ; 92(8-9): 264-79, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24252516

RESUMO

Chemotactic cell migration is triggered by extracellular concentration gradients of molecules segregated by target fields. Neural crest cells (NCCs), paradigmatic as an accurately moving cell population, undergo wide dispersion along multiple pathways, invading with precision defined sites of the embryo to differentiate into many derivatives. This report addresses the involvement of NT-3 in early colonization by cephalic NCCs invading the optic vesicle region. The results of in vitro and in vivo approaches showed that NCCs migrate directionally up an NT-3 concentration gradient. We also demonstrated the expression of NT-3 in the ocular region as well as their functional TrkB, TrkC and p75 receptors on cephalic NCCs. On whole-mount embryo, a perturbed distribution of NCCs colonizing the optic vesicle target field was shown after morpholino cancelation of cephalic NT-3 or TrkC receptor on NCCs, as well as in situ blocking of TrkC receptor of mesencephalic NCCs by specific antibody released from inserted microbeads. The present results strongly suggest that, among other complementary cell guidance factor(s), the chemotactic response of NCCs toward the ocular region NT-3 gradient is essential for spatiotemporal cell orientation, amplifying the functional scope of this neurotrophic factor as a molecular guide for the embryo cells, besides its well-known canonical functions.


Assuntos
Quimiotaxia , Mesencéfalo/citologia , Crista Neural/citologia , Neurotrofina 3/metabolismo , Animais , Proliferação de Células , Embrião de Galinha , Galinhas , Crista Neural/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...