Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Glia ; 67(1): 193-211, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30597659

RESUMO

Neurotrophins mediate neuronal growth, differentiation, and survival via tropomyosin receptor kinase (Trk) or p75 neurotrophin receptor (p75NTR ) signaling. The p75NTR is not exclusively expressed by neurons but also by certain immune cells, implying a role for neurotrophin signaling in the immune system. In this study, we investigated the effect of p75NTR on innate immune cell behavior and on neuronal morphology upon chronic Toxoplasma gondii (T. gondii) infection-induced neuroinflammation. Characterization of the immune cells in the periphery and central nervous system (CNS) revealed that innate immune cell subsets in the brain upregulated p75NTR upon infection in wild-type mice. Although cell recruitment and phagocytic capacity of p75NTRexonIV knockout (p75-/- ) mice were not impaired, the activation status of resident microglia and recruited myeloid cell subsets was altered. Importantly, recruited mononuclear cells in brains of infected p75-/- mice upregulated the production of the cytokines interleukin (IL)-10, IL-6 as well as IL-1α. Protein levels of proBDNF, known to negatively influence neuronal morphology by binding p75NTR , were highly increased upon chronic infection in the brain of wild-type and p75-/- mice. Moreover, upon infection the activated immune cells contributed to the proBDNF release. Notably, the neuroinflammation-induced changes in spine density were rescued in the p75-/- mice. In conclusion, these findings indicate that neurotrophin signaling via the p75NTR affects innate immune cell behavior, thus, influencing the structural plasticity of neurons under inflammatory conditions.


Assuntos
Leucócitos Mononucleares/fisiologia , Neurônios/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Toxoplasma , Toxoplasmose/imunologia , Animais , Feminino , Imunidade Inata/fisiologia , Inflamação/imunologia , Inflamação/patologia , Leucócitos Mononucleares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Toxoplasmose/patologia
2.
Vet Pathol ; 55(6): 786-801, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29940812

RESUMO

The low-affinity nerve growth factor receptor p75NTR is a major neurotrophin receptor involved in manifold and pleiotropic functions in the developing and adult central nervous system (CNS). Although known for decades, its entire functions are far from being fully elucidated. Depending on the complex interactions with other receptors and on the cellular context, p75NTR is capable of performing contradictory tasks such as mediating cell death as well as cell survival. In parallel, as a prototype marker for certain differentiation stages of Schwann cells and related CNS aldynoglial cells, p75NTR has recently gained increasing notice as a marker for cells with proposed regenerative potential in CNS diseases, such as demyelinating disease and traumatic CNS injury. Besides its pivotal role as a marker for transplantation candidate cells, recent studies in canine neuroinflammatory CNS conditions also highlight a spontaneous endogenous occurrence of p75NTR-positive glia, which potentially play a role in Schwann cell-mediated CNS remyelination. The aim of the present communication is to review the pleiotropic functions of p75NTR in the CNS with a special emphasis on its role as an immunohistochemical marker in neuropathology. Following a brief illustration of the expression of p75NTR in neurogenesis and in developed neuronal populations, the implications of p75NTR expression in astrocytes, oligodendrocytes, and microglia are addressed. A special focus is put on the role of p75NTR as a cell marker for specific differentiation stages of Schwann cells and a regeneration-promoting CNS population, collectively referred to as aldynoglia.


Assuntos
Doenças do Sistema Nervoso Central/veterinária , Sistema Nervoso Central/patologia , Receptor de Fator de Crescimento Neural/fisiologia , Regeneração/fisiologia , Animais , Biomarcadores/metabolismo , Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/fisiopatologia , Neuroglia/citologia , Neuroglia/patologia , Neuroglia/fisiologia , Receptor de Fator de Crescimento Neural/metabolismo , Remielinização/fisiologia , Células de Schwann/patologia , Células de Schwann/fisiologia
3.
J Physiol ; 593(7): 1581-95, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25639253

RESUMO

How the neural substrates for detection of paired stimuli are distinct from unpaired stimuli is poorly understood and a fundamental question for understanding the signalling mechanisms for coincidence detection during associative learning. To address this question, we used a neural correlate of eyeblink classical conditioning in an isolated brainstem from the turtle, in which the cranial nerves are directly stimulated in place of using a tone or airpuff. A bidirectional response is activated in <5 min of training, in which phosphorylated 3-phosphoinositide-dependent kinase-1 (p-PDK1) is increased in response to paired and decreased in response to unpaired nerve stimulation and is mediated by the opposing actions of neurotrophin receptors TrkB and p75(NTR) . Surprisingly, blockade of adenosine 2A (A2A ) receptors inhibits both of these responses. Pairing also induces substantially increased surface expression of TrkB that is inhibited by Src family tyrosine kinase and A2A receptor antagonists. Finally, the acquisition of conditioning is blocked by a PDK1 inhibitor. The unique action of A2A receptors to function directly as G proteins and in receptor transactivation to control distinct TrkB and p75(NTR) signalling pathways allows for convergent activation of PDK1 and protein kinase A during paired stimulation to initiate classical conditioning.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/fisiologia , Condicionamento Clássico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkB/fisiologia , Animais , Tronco Encefálico/fisiologia , Nervos Cranianos/fisiologia , Receptor A2A de Adenosina , Tartarugas/fisiologia
4.
J Neurosci ; 33(25): 10221-34, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23785138

RESUMO

The p75 neurotrophin receptor (p75(NTR)) is a member of the tumor necrosis factor receptor superfamily with a widespread pattern of expression in tissues such as the brain, liver, lung, and muscle. The mechanisms that regulate p75(NTR) transcription in the nervous system and its expression in other tissues remain largely unknown. Here we show that p75(NTR) is an oscillating gene regulated by the helix-loop-helix transcription factors CLOCK and BMAL1. The p75(NTR) promoter contains evolutionarily conserved noncanonical E-box enhancers. Deletion mutagenesis of the p75(NTR)-luciferase reporter identified the -1039 conserved E-box necessary for the regulation of p75(NTR) by CLOCK and BMAL1. Accordingly, gel-shift assays confirmed the binding of CLOCK and BMAL1 to the p75(NTR-)1039 E-box. Studies in mice revealed that p75(NTR) transcription oscillates during dark and light cycles not only in the suprachiasmatic nucleus (SCN), but also in peripheral tissues including the liver. Oscillation of p75(NTR) is disrupted in Clock-deficient and mutant mice, is E-box dependent, and is in phase with clock genes, such as Per1 and Per2. Intriguingly, p75(NTR) is required for circadian clock oscillation, since loss of p75(NTR) alters the circadian oscillation of clock genes in the SCN, liver, and fibroblasts. Consistent with this, Per2::Luc/p75(NTR-/-) liver explants showed reduced circadian oscillation amplitude compared with those of Per2::Luc/p75(NTR+/+). Moreover, deletion of p75(NTR) also alters the circadian oscillation of glucose and lipid homeostasis genes. Overall, our findings reveal that the transcriptional activation of p75(NTR) is under circadian regulation in the nervous system and peripheral tissues, and plays an important role in the maintenance of clock and metabolic gene oscillation.


Assuntos
Proteínas CLOCK/fisiologia , Ritmo Circadiano/fisiologia , Metabolismo/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Fatores de Transcrição ARNTL/biossíntese , Fatores de Transcrição ARNTL/genética , Animais , Glicemia/metabolismo , Proteínas CLOCK/genética , Ritmo Circadiano/genética , DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Células HEK293 , Homeostase/genética , Humanos , Fígado/metabolismo , Luciferases/genética , Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Fator de Crescimento Neural/genética , Choque Séptico/fisiopatologia , Núcleo Supraquiasmático/fisiologia , Transfecção
5.
Handb Exp Pharmacol ; 220: 193-221, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24668474

RESUMO

The neurotrophins play crucial roles regulating survival and apoptosis in the developing and injured nervous system. The four neurotrophins exert profound and crucial survival effects on developing peripheral neurons, and their expression and action is intimately tied to successful innervation of peripheral targets. In the central nervous system, they are dispensable for neuronal survival during development but support neuronal survival after lesion or other forms of injury. Neurotrophins also regulate apoptosis of both peripheral and central neurons, and we now recognize that there are regulatory advantages to having the same molecules regulate life and death decisions. This chapter examines the biological contexts in which these events take place and highlights the specific ligands, receptors, and signaling mechanisms that allow them to occur.


Assuntos
Apoptose , Sobrevivência Celular , Fatores de Crescimento Neural/fisiologia , Animais , Humanos , Fator de Crescimento Neural/fisiologia , Precursores de Proteínas/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkA/fisiologia
6.
Handb Exp Pharmacol ; 220: 121-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24668472

RESUMO

The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including programmed cell death, axonal growth and degeneration, cell proliferation, myelination, and synaptic plasticity. The multiplicity of cellular functions governed by the receptor arises from the variety of ligands and co-receptors which associate with p75(NTR) and regulate its signaling. P75(NTR) promotes survival through interactions with Trk receptors, inhibits axonal regeneration via partnerships with Nogo receptor (Nogo-R) and Lingo-1, and promotes apoptosis through association with Sortilin. Signals downstream of these interactions are further modulated through regulated intramembrane proteolysis (RIP) of p75(NTR) and by interactions with numerous cytosolic partners. In this chapter, we discuss the intricate signaling mechanisms of p75(NTR), emphasizing how these signals are differentially regulated to mediate these diverse cellular functions.


Assuntos
Receptor de Fator de Crescimento Neural/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Apoptose , Ciclo Celular , Sobrevivência Celular , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Bainha de Mielina/fisiologia , NF-kappa B/fisiologia , Plasticidade Neuronal , Precursores de Proteínas/fisiologia , Receptor de Fator de Crescimento Neural/química , Receptor trkA/fisiologia
7.
Int J Cancer ; 133(12): 2903-13, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23754772

RESUMO

4-1BB (CD137) is a costimulatory molecule transiently expressed on the T-cell surface after TCR engagement, whereas its ligand 4-1BBL can be found on professional antigen-presenting cells, but more importantly, also on tumor cells. As the role of the 4-1BB/4-1BBL pathway has emerged central to CD8(+) T-cell responses and survival, we sought to test its relevance in the context of genetically modified human T cells. To that end, T cells purified from healthy donors and from vaccinated-melanoma patients were transduced to express high levels of constitutive 4-1BB. 4-1BB-transduced T cells were cocultured with melanoma tumor lines and exhibited enhanced cytokine secretion, upregulation of activation markers as well as increased cytotoxicity in a chick-chorioallantoic membrane model of human melanoma tumors. In addition, these cells expanded and proliferated at a higher rate, expressed heightened levels of the antiapoptotic molecule Bcl(XL) and were also relatively insensitive to immunosuppression mediated by transforming growth factor-ß, compared to control cells. We also show that 4-1BBL expression on the target cell is essential to 4-1BB-mediated functional improvement. Overall, we conclude that the modification of human T cells with 4-1BB yields enhanced antitumor function which may have important applications in therapies based on the genetic modification of patient lymphocytes.


Assuntos
Citotoxicidade Imunológica , Melanoma/imunologia , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Ligante 4-1BB/análise , Proliferação de Células , Humanos , Receptor de Fator de Crescimento Neural/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/análise , Vacinação
8.
J Cell Physiol ; 227(3): 1017-25, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21503896

RESUMO

Neurotrophins (NTs) belong to a family of growth factors that play a critical role in the control of skin homeostasis. NTs act through the low-affinity receptor p75NTR and the high-affinity receptors TrkA, TrkB, and TrkC. Here we show that dermal fibroblasts (DF) and myofibroblasts (DM) synthesize and secrete all NTs and express NT receptors. NTs induce differentiation of DF into DM, as shown by the expression of α-SMA protein. The Trk inhibitor K252a, TrkA/Fc, TrkB/Fc, or TrkC/Fc chimera prevents DF and DM proliferation. In addition, p75NTR siRNA inhibits DF proliferation, indicating that both NT receptors mediate DF proliferation induced by endogenous NTs. Autocrine NTs also induce DF migration through p75NTR and Trk, as either silencing of p75NTR or Trk/Fc chimeras prevent this effect, in absence of exogenous NTs. Finally, NGF or BDNF statistically increase the tensile strength in a dose dependent manner, as measured in a collagen gel through the GlaSbox device. Taken together, these results indicate that NTs exert a critical role on fibroblast and could be involved in tissue re-modeling and wound healing.


Assuntos
Diferenciação Celular/fisiologia , Derme/citologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Fatores de Crescimento Neural/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Diferenciação Celular/genética , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Derme/metabolismo , Derme/fisiologia , Fibroblastos/metabolismo , Prepúcio do Pênis , Regeneração Tecidual Guiada/métodos , Humanos , Masculino , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Fatores de Crescimento Neural/metabolismo , Receptor de Fator de Crescimento Neural/genética , Receptor trkA/genética , Receptor trkA/fisiologia , Receptor trkB/genética , Receptor trkB/metabolismo , Receptor trkB/fisiologia , Receptor trkC/genética , Receptor trkC/metabolismo , Receptor trkC/fisiologia , Cicatrização/genética , Cicatrização/fisiologia
9.
J Cell Biol ; 177(6): 1119-32, 2007 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-17576803

RESUMO

Clearance of fibrin through proteolytic degradation is a critical step of matrix remodeling that contributes to tissue repair in a variety of pathological conditions, such as stroke, atherosclerosis, and pulmonary disease. However, the molecular mechanisms that regulate fibrin deposition are not known. Here, we report that the p75 neurotrophin receptor (p75(NTR)), a TNF receptor superfamily member up-regulated after tissue injury, blocks fibrinolysis by down-regulating the serine protease, tissue plasminogen activator (tPA), and up-regulating plasminogen activator inhibitor-1 (PAI-1). We have discovered a new mechanism in which phosphodiesterase PDE4A4/5 interacts with p75(NTR) to enhance cAMP degradation. The p75(NTR)-dependent down-regulation of cAMP results in a decrease in extracellular proteolytic activity. This mechanism is supported in vivo in p75(NTR)-deficient mice, which show increased proteolysis after sciatic nerve injury and lung fibrosis. Our results reveal a novel pathogenic mechanism by which p75(NTR) regulates degradation of cAMP and perpetuates scar formation after injury.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fibrose , Receptor de Fator de Crescimento Neural/fisiologia , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Animais , Cicatriz/etiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Fibrinólise , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/genética , Nervo Isquiático/lesões , Ferimentos e Lesões
10.
Proc Natl Acad Sci U S A ; 106(19): 7870-5, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19416837

RESUMO

Beta-amyloid (Abeta) has adverse effects on brain cells, but little is known about its effects on the peripheral nervous system in Alzheimer's disease (AD). Several lines of in vitro evidence suggest that the neurotrophin receptor p75 mediates or exacerbates Abeta-induced neurotoxicity. Here, we show that p75-deficient sympathetic neurons are more sensitive to Abeta-induced neurite growth inhibition. To investigate the role of p75 in the sympathetic nervous system of AD, p75 mutant mice were crossed with a mouse line of AD model. The majority of p75-deficient AD mice died by 3 weeks of age. The lethality is associated with severe defects in sympathetic innervation to multiple organs. When 1 copy of the BACE1 gene encoding a protein essential in Abeta production was deleted in p75-deficient AD mice, sympathetic innervation was significantly restored. These results suggest that p75 is neuroprotective for the sympathetic nervous system in a mouse model of AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptor de Fator de Crescimento Neural/fisiologia , Sistema Nervoso Simpático/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Gânglio Estrelado/metabolismo , Sudorese , Fatores de Tempo
11.
Nat Neurosci ; 11(6): 649-58, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18382462

RESUMO

The mechanisms that regulate the pruning of mammalian axons are just now being elucidated. Here, we describe a mechanism by which, during developmental sympathetic axon competition, winning axons secrete brain-derived neurotrophic factor (BDNF) in an activity-dependent fashion, which binds to the p75 neurotrophin receptor (p75NTR) on losing axons to cause their degeneration and, ultimately, axon pruning. Specifically, we found that pruning of rat and mouse sympathetic axons that project to the eye requires both activity-dependent BDNF and p75NTR. p75NTR and BDNF are also essential for activity-dependent axon pruning in culture, where they mediate pruning by directly causing axon degeneration. p75NTR, which is enriched in losing axons, causes axonal degeneration by suppressing TrkA-mediated signaling that is essential for axonal maintenance. These data provide a mechanism that explains how active axons can eliminate less-active, competing axons during developmental pruning by directly promoting p75NTR-mediated axonal degeneration.


Assuntos
Axônios/fisiologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Degeneração Neural/fisiopatologia , Receptor de Fator de Crescimento Neural/fisiologia , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axotomia/métodos , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Toxina da Cólera/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/genética , Fator de Crescimento Neural/farmacologia , Neurônios/citologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Fator de Crescimento Neural/deficiência , Estilbamidinas/metabolismo , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/crescimento & desenvolvimento , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/metabolismo
12.
Int J Cancer ; 128(8): 1804-12, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20549701

RESUMO

The classic medulloblastoma (CMB) and the desmoplastic medulloblastoma (DMB) subtypes represent the major medulloblastoma variants. In contrast to CMB, DMB display high levels of the low-affinity nerve growth factor receptor p75(NTR) . Given the reports of a better clinical course of DMB, we hypothesized that p75(NTR) might act as a tumor suppressor in medulloblastomas. In a large set of medulloblastomas, p75(NTR) was screened for mutations, and its mRNA expression and the DNA methylation status of its 5'-region were assessed. p75(NTR) immunostainings were performed in wild-type murine cerebella and medulloblastomas arising in patched heterozygous mice, and murine cerebellar granule cell precursors (GCP) were analyzed in vitro. Medulloblastoma cells engineered to express p75(NTR) were characterized flow cytometrically and morphologically. One CMB displayed a mutation of the p75(NTR) coding sequence. p75(NTR) mRNA levels clearly delineated DMB and CMB; however, CpG island hypermethylation was excluded as the cause of low p75(NTR) expression in CMB. Sonic Hedgehog-treated GCP showed elevated p75(NTR) expression, and strong expression of p75(NTR) was detected in the external granule cell layer of wild-type mice and in murine ptc(±) medulloblastomas. CMB cells overexpressing p75(NTR) displayed a significant increase in apoptosis. In summary, our data link activated Hedgehog signaling in DMB with p75(NTR) expression and characterize p75(NTR) as a biologically relevant inductor of apoptosis in MB.


Assuntos
Apoptose , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Receptor de Fator de Crescimento Neural/fisiologia , Animais , Western Blotting , Neoplasias Cerebelares/metabolismo , Ilhas de CpG , Metilação de DNA , DNA de Neoplasias/genética , Feminino , Citometria de Fluxo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Técnicas Imunoenzimáticas , Meduloblastoma/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estudos Multicêntricos como Assunto , Neurônios , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , RNA Mensageiro/genética , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas
13.
J Neurosci Res ; 89(9): 1331-41, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21674565

RESUMO

High-resolution immunohistochemistry shows that the receptor protein p75(NTR) is present in the nerve terminal, muscle cell, and glial Schwann cell at the neuromuscular junction (NMJ) of postnatal rats (P4-P6) during the synapse elimination period. Blocking the receptor with the antibody anti-p75-192-IgG (1-5 µg/ml, 1 hr) results in reduced endplate potentials (EPPs) in mono- and polyinnervated synapses ex vivo, but the mean number of functional inputs per NMJ does not change for as long as 3 hr. Incubation with exogenous brain-derived neurotrophic factor (BDNF) for 1 hr (50 nM) resulted in a significant increase in the size of the EPPs in all nerve terminals, and preincubation with anti-p75-192-IgG prevented this potentiation. Long exposure (24 hr) in vivo of the NMJs to the antibody anti-p75-192-IgG (1-2 µg/ml) results in a delay of postnatal synapse elimination and even some regrowth of previously withdrawn axons, but also in some acceleration of the morphologic maturation of the postsynaptic nicotinic acetylcholine receptor (nAChR) clusters. The results indicate that p75(NTR) is involved in both ACh release and axonal retraction during postnatal axonal competition and synapse elimination.


Assuntos
Axônios/fisiologia , Músculo Esquelético/inervação , Junção Neuromuscular/crescimento & desenvolvimento , Receptor de Fator de Crescimento Neural/fisiologia , Animais , Animais Recém-Nascidos , Anticorpos Bloqueadores/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Relação Dose-Resposta a Droga , Eletromiografia , Imuno-Histoquímica , Masculino , Músculo Esquelético/fisiologia , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor de Fator de Crescimento Neural/antagonistas & inibidores
14.
Fiziol Cheloveka ; 37(2): 104-16, 2011.
Artigo em Russo | MEDLINE | ID: mdl-21542326

RESUMO

Traditionally, nerve growth factor (NGF) is considered as chemoattractant that participates in the regulation of cell proliferation, differentiation and myelination of neurons. However, currently available data suggest that the physiological role of NGF in the organism is much wider. This review discusses the features of the influence of NGF on the functional activity of the cardiovascular system, as well as signaling pathways by which activated NGF TrkA and p75(ntr) receptors regulate the functional state of endothelial and vascular smooth muscle cells and cardiomyocytes. In addition, the review observes the theoretical perspectives of agonists and antagonists of TrkA and p75(ntr) receptors for the treatment of various diseases of the heart and blood vessels.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Sistema Cardiovascular , Fator de Crescimento Neural/fisiologia , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Sistema Cardiovascular/metabolismo , Humanos , Receptor de Fator de Crescimento Neural/química , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkA/química , Receptor trkA/fisiologia
15.
J Neurosci ; 29(17): 5411-24, 2009 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-19403809

RESUMO

Neurotrophins such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) act through the tropomyosin-related receptor tyrosine kinases (Trk) and the pan-neurotrophin receptor (p75) to regulate complex developmental and functional properties of neurons. While NGF activates both receptor types in sympathetic neurons, differential signaling through TrkA and p75 can result in widely divergent functional outputs for neuronal survival, growth, and synaptic function. Here we show that TrkA and p75 signaling pathways have opposing effects on the firing properties of sympathetic neurons, and define a mechanism whereby the relative level of signaling through these two receptors sets firing patterns via coordinate regulation of a set of ionic currents. We show that signaling through the p75 pathway causes sympathetic neurons to fire in a phasic pattern showing marked accommodation. Signaling through the NGF-specific TrkA, on the other hand, causes cells to fire tonically. Neurons switch rapidly between firing patterns, on the order of minutes to hours. We show that changes in firing patterns are caused by neurotrophin-dependent regulation of at least four voltage-gated currents: the sodium current and the M-type, delayed rectifier, and calcium-dependent potassium currents. Neurotrophin release, and thus receptor activation, varies among somatic tissues and physiological state. Thus, these data suggest that target-derived neurotrophins may be an important determinant of the characteristic electrical properties of sympathetic neurons and therefore regulate the functional output of the sympathetic nervous system.


Assuntos
Fibras Adrenérgicas/fisiologia , Canais Iônicos/fisiologia , Neurônios/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkA/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Ativação do Canal Iônico/fisiologia , Camundongos , Técnicas de Patch-Clamp , Ratos
16.
J Neurosci ; 29(34): 10627-37, 2009 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-19710315

RESUMO

Oligomeric forms of amyloid-beta (Abeta) are thought to play a causal role in Alzheimer's disease (AD), and the p75 neurotrophin receptor (p75(NTR)) has been implicated in Abeta-induced neurodegeneration. To further define the functions of p75(NTR) in AD, we examined the interaction of oligomeric Abeta(1-42) with p75(NTR), and the effects of that interaction on neurite integrity in neuron cultures and in a chronic AD mouse model. Atomic force microscopy was used to ascertain the aggregated state of Abeta, and fluorescence resonance energy transfer analysis revealed that Abeta oligomers interact with the extracellular domain of p75(NTR). In vitro studies of Abeta-induced death in neuron cultures isolated from wild-type and p75(NTR-/-) mice, in which the p75(NTR) extracellular domain is deleted, showed reduced sensitivity of mutant cells to Abeta-induced cell death. Interestingly, Abeta-induced neuritic dystrophy and activation of c-Jun, a known mediator of Abeta-induced deleterious signaling, were completely prevented in p75(NTR-/-) neuron cultures. Thy1-hAPP(Lond/Swe) x p75(NTR-/-) mice exhibited significantly diminished hippocampal neuritic dystrophy and complete reversal of basal forebrain cholinergic neurite degeneration relative to those expressing wild-type p75(NTR). Abeta levels were not affected, suggesting that removal of p75(NTR) extracellular domain reduced the ability of excess Abeta to promote neuritic degeneration. These findings indicate that although p75(NTR) likely does not mediate all Abeta effects, it does play a significant role in enabling Abeta-induced neurodegeneration in vitro and in vivo, establishing p75(NTR) as an important therapeutic target for AD.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neuritos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptor de Fator de Crescimento Neural/fisiologia , Acetilcolinesterase/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática/métodos , Éxons/genética , Transferência Ressonante de Energia de Fluorescência/métodos , Hipocampo/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neuritos/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Prosencéfalo/citologia , Receptor de Fator de Crescimento Neural/deficiência , Espectrofotometria Atômica/métodos
17.
Curr Biol ; 17(11): 911-21, 2007 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-17493809

RESUMO

BACKGROUND: Activity-dependent competition that operates on branch stability or formation plays a critical role in shaping the pattern and complexity of axonal terminal arbors. In the mammalian central nervous system (CNS), the effect of activity-dependent competition on axon arborization and on the assembly of sensory maps is well established. However, the molecular pathways that modulate axonal-branch stability or formation in competitive environments remain unknown. RESULTS: We establish an in vivo axonal-competition paradigm in the mouse olfactory system by employing a genetic strategy that permits suppression of neurosecretory activity in random subsets of olfactory sensory neurons (OSNs). Long-term follow up confirmed that this genetic manipulation triggers competition by revealing a bias toward selective stabilization of active arbors and local degeneration of synaptically silent ones. By using a battery of genetically modified mouse models, we demonstrate that a decrease either in the total levels or the levels of activity-dependent secreted BDNF (due to a val66met substitution), rescues silent arbors from withering. We show that this effect may be mediated, at least in part, by p75(NTR). CONCLUSIONS: We establish and experimentally validate a genetic in vivo axonal-competition paradigm in the mammalian CNS. By using this paradigm, we provide evidence for a specific effect of BDNF signaling on terminal-arbor pruning under competition in vivo. Our results have implications for the formation and refinement of the olfactory and other sensory maps, as well as for neuropsychiatric diseases and traits modulated by the BDNF val66met variant.


Assuntos
Axônios/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transdução de Sinais/genética , Substituição de Aminoácidos , Animais , Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/genética , Camundongos , Camundongos Transgênicos , Neurônios Aferentes/fisiologia , Receptor de Fator de Crescimento Neural/metabolismo , Receptor de Fator de Crescimento Neural/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Olfato/genética , Olfato/fisiologia
18.
Trends Neurosci ; 31(2): 99-104, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18199491

RESUMO

The p75 neurotrophin receptor (p75(NTR)) is expressed on many cell types and can influence a variety of cellular functions. This receptor can mediate cell survival or cell death, can promote or inhibit axonal growth and can facilitate or attenuate proliferation, depending on the cell context. The emerging picture regarding p75(NTR) indicates that it can partner with different coreceptors to dictate specific responses. It then signals by recruiting intracellular binding proteins to activate different signaling pathways. The function of p75(NTR) has mainly been studied in neurons; however, it is also expressed in a variety of glial populations, especially during development and after injury, where its roles have been poorly defined. In this review, we will examine the potential roles for p75(NTR) in glial function.


Assuntos
Neuroglia/metabolismo , Receptor de Fator de Crescimento Neural/fisiologia , Animais , Humanos , Fibras Nervosas Mielinizadas/metabolismo , Neuroglia/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
19.
Life Sci ; 243: 117293, 2020 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-31930971

RESUMO

Ca2+ overload in neurons has been implicated in Alzheimer's Disease (AD). Upregulation of Ca2+ through L-type Ca2+ channels was known to be involved in the neurodegeneration induced by amyloid-ß (Aß) peptides in AD. However, little is known about the mechanism by which upregulation of L-type Ca2+ channel currents is linked to Aß-induced neuronal toxicity. In the present study, we found that the L-type Ca2+ current in transgenic AD mice (Tg2576) neurons is greater than in wild-type (WT) neurons, and this Ca2+ channel current change were rescued in Tg2576/p75NTR+/- (p75 neurotrophin receptor) neurons. We further examined the changes in the gating of L-type Ca2+ channels following Aß42 treatment, and the results showed that the L-type Ca2+ channel current was significantly increased by Aß42 treatment in WT hippocampal neurons. Blocking or decreasing the expression of p75NTR eliminated the influence of Aß42 on the L-type Ca2+ channel current in WT hippocampal neurons. We also evaluated how Aß42 affected the voltage-dependent activation and inactivation of L-type Ca2+ channels in cultured WT neurons. The results indicated that the half-maximal activation voltage (V1/2) was left shifted, and the half-inactivation voltage (V1/2) displayed a right shift in neuron treated by Aß42. Decreasing the expression of p75NTR eliminated the effect of Aß42 on voltage-dependent activation and inactivation of the L-type Ca2+ channel. These results indicate that Aß42 changes L-type Ca2+ channel currents by modulating the channel's activation and inactivation dynamics, while decreasing p75NTR expression can remove this effect.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Canais de Cálcio Tipo L/metabolismo , Neurônios/metabolismo , Receptor de Fator de Crescimento Neural/fisiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Humanos , Ativação do Canal Iônico , Camundongos , Camundongos Transgênicos
20.
Drug Metab Pers Ther ; 35(4)2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-34704697

RESUMO

OBJECTIVES: Oncological diseases are an urgent medical and social problem. The chemotherapy induces not only the death of the tumor cells but also contributes to the development of their multidrug resistance and death of the healthy cells and tissues. In this regard, the search for the new pharmacological substances with anticancer activity against drug-resistant tumors is of utmost importance. In the present study we primarily investigated the correlation between the expression of TrkA and p75 receptors with the nerve growth factor (NGF) and cisplatin or temozolomide sensitivity of anaplastic astrocytoma (AA), glioblastoma (GB) and medulloblastoma (MB) cell cultures. We then evaluated the changing of copy numbers of MYCC and MYCN and its correlation with cytotoxicity index (CI) in MB cells under NGF exposition. METHODS: The primary cell cultures were obtained from the tumor biopsy samples of the patients with AA (n=5), GB (n=7) or MB (n=25) prior to radiotherapy and chemotherapy. The cytotoxicity effect of NGF and its combinations with cisplatin or temozolomide, the relative expression of TrkA and p75 receptors, its correlations with CI in AA, GB and MB primary cell cultures were studied by trypan blue cytotoxicity assay and immunofluorescence staining respectively. The effect of NGF on MYCC and MYCN copy numbers in MB cell cultures was studied by fluorescence in situ hybridization. RESULTS: We found that the expression of TrkA and p75 receptors (p=0.03) and its ratio (p=0.0004) depends on the sensitivity of AA and GB cells to treatment with NGF and its combinations with cisplatin or temozolomide. NGF reduces (p<0.05) the quantity of MB cells with six or eight copies of MYCN and three or eight copies of MYCC. Besides, NGF increases (p<0.05) the quantity of MB cells containing two copies of both oncogenes. The negative correlation (r=-0.65, p<0.0001) is established between MYCC average copy numbers and CI of NGF in MB cells. CONCLUSIONS: The relative expression of NGF receptors (TrkA/p75) and its correlation with CI of NGF and its combinations in AA and GB cells point to the mechanism involving a cell death signaling pathway. NGF downregulates (p<0.05) some increased copy numbers of MYCC and MYCN in the human MB cell cultures, and upregulates normal two copies of both oncogenes (p<0.05).


Assuntos
Neoplasias Encefálicas , Cisplatino , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Cisplatino/farmacologia , Variações do Número de Cópias de DNA , Humanos , Hibridização in Situ Fluorescente , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/farmacologia , Oncogenes , Receptor de Fator de Crescimento Neural/fisiologia , Receptor trkA/genética , Receptor trkA/metabolismo , Temozolomida/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA