Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Neurosci ; 44(16)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38413232

RESUMO

Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disorder marked by progressive motor neuron degeneration and muscle denervation. A recent transcriptomic study integrating a wide range of human ALS samples revealed that the upregulation of p53, a downstream target of inflammatory stress, is commonly detected in familial and sporadic ALS cases by a mechanism linked to a transactive response DNA-binding protein 43 (TDP-43) dysfunction. In this study, we show that prolonged interferon-gamma (IFNγ) treatment of human induced pluripotent stem cell-derived spinal motor neurons results in a severe cytoplasmic aggregation of TDP-43. TDP-43 dysfunction resulting from either IFNγ exposure or an ALS-associated TDP-43 mutation was associated with the activation of the p53 pathway. This was accompanied by the hyperactivation of neuronal firing, followed by the complete loss of their electrophysiological function. Through a comparative single-cell transcriptome analysis, we have identified significant alterations in ALS-associated genes in motor neurons exposed to IFNγ, implicating their direct involvement in ALS pathology. Interestingly, IFNγ was found to induce significant levels of programmed death-ligand 1 (PD-L1) expression in motor neurons without affecting the levels of any other immune checkpoint proteins. This finding suggests a potential role of excessive PD-L1 expression in ALS development, given that PD-L1 was recently reported to impair neuronal firing ability in mice. Our findings suggest that exposing motor neurons to IFNγ could directly derive ALS pathogenesis, even without the presence of the inherent genetic mutation or functional glia component. Furthermore, this study provides a comprehensive list of potential candidate genes for future immunotherapeutic targets with which to treat sporadic forms of ALS, which account for 90% of all reported cases.


Assuntos
Esclerose Lateral Amiotrófica , Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Camundongos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Antígeno B7-H1/metabolismo , Biomarcadores , Proteínas de Ligação a DNA/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon gama/metabolismo , Interferon gama/farmacologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Proteína Supressora de Tumor p53/metabolismo
2.
Annu Rev Med ; 68: 431-443, 2017 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-27860545

RESUMO

Demyelination of central nervous system axons, associated with traumatic injury and demyelinating diseases such as multiple sclerosis, causes impaired neural transmission and ultimately axon degeneration. Consequently, extensive research has focused on signaling systems that promote myelinating activity of oligodendrocytes or promote production of new oligodendrocytes from oligodendrocyte progenitor cells. Many receptor systems, notably including growth factor receptors and G protein-coupled receptors, control myelination. A number of recent clinical trials target these receptor signaling pathways.


Assuntos
Doenças Desmielinizantes/tratamento farmacológico , Proteínas Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Remielinização/efeitos dos fármacos , Remielinização/fisiologia , Animais , Anticorpos Monoclonais/uso terapêutico , Humanos , Proteínas de Membrana/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
3.
Mol Cell Neurosci ; 70: 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26546150

RESUMO

Neurotrophins, essential regulators of many aspects of neuronal differentiation and function, signal via four receptors, p75, TrkA, TrkB and TrkC. The three Trk paralogs are members of the LIG superfamily of membrane proteins, which share extracellular domains consisting of leucine-rich repeat and C2 Ig domains. Another LIG protein, LINGO-1 has been reported to bind and influence signaling of p75 as well as TrkA, TrkB and TrkC. Here we examine the manner in which LINGO-1 influences the function of TrkA, TrkB and TrkC. We report that Trk activation promotes Trk association with LINGO-1, and that this association promotes Trk degradation by a lysosomal mechanism. This mechanism resembles the mechanism by which another LIG protein, LRIG1, promotes lysosomal degradation of receptor tyrosine kinases such as the EGF receptor. We present evidence indicating that the Trk/LINGO-1 interaction occurs, in part, within recycling endosomes. We show that a mutant form of LINGO-1, with much of the extracellular domain deleted, has the capacity to enhance TrkA signaling in PC12 cells, possibly by acting as an inhibitor of Trk down-regulation by full length LINGO-1. We propose that LINGO-1 functions as a negative feedback regulator of signaling by cognate receptor tyrosine kinases including TrkA, TrkB and TrkC.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Transdução de Sinais/genética , Animais , Citoplasma/metabolismo , Regulação para Baixo , Endossomos/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Células PC12 , Fosforilação , Ratos
4.
J Biol Chem ; 290(15): 9511-20, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25666623

RESUMO

Axon outgrowth inhibition in response to trauma is thought to be mediated via the binding of myelin-associated inhibitory factors (e.g. Nogo-66, myelin-associated glycoprotein, oligodendrocyte myelin glycoprotein, and myelin basic protein) to a putative tripartite LINGO-1·p75(NTR)·Nogo-66 receptor (NgR) complex at the cell surface. We found that endogenous LINGO-1 expression in neurons in the cortex and cerebellum is intracellular. Mutation or truncation of the highly conserved LINGO-1 C terminus altered this intracellular localization, causing poor intracellular retention and increased plasma membrane expression. p75(NTR) associated predominantly with natively expressed LINGO-1 containing immature N-glycans, characteristic of protein that has not completed trans-Golgi-mediated processing, whereas mutant forms of LINGO-1 with enhanced plasma membrane expression did not associate with p75(NTR). Co-immunoprecipitation experiments demonstrated that LINGO-1 and NgR competed for binding to p75(NTR) in a manner that is difficult to reconcile with the existence of a LINGO-1·p75(NTR)·NgR ternary complex. These findings contradict models postulating functional LINGO-1·p75(NTR)·NgR complexes in the plasma membrane.


Assuntos
Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Animais Recém-Nascidos , Ligação Competitiva , Encéfalo/citologia , Encéfalo/metabolismo , Membrana Celular/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mutação , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptor Nogo 1 , Polissacarídeos/metabolismo , Ligação Proteica , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Fator de Crescimento Neural/genética
5.
J Neurosci ; 32(40): 14000-9, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23035107

RESUMO

The chick embryo (Gallus domesticus) is one of the most important model systems in vertebrate developmental biology. The development and function of its auditory brainstem circuitry is exceptionally well studied. These circuits represent an excellent system for genetic manipulation to investigate mechanisms controlling neural circuit formation, synaptogenesis, neuronal polarity, and dendritic arborization. The present study investigates the auditory nucleus, nucleus magnocellularis (NM). The neurotrophin receptor TrkB regulates dendritic structure in CNS neurons. TrkB is expressed in NM neurons at E7-E8 when these neurons have dendritic arbors. Downregulation of TrkB occurs after E8 followed by retraction of dendrites and by E18 most NM cells are adendritic. Is cessation of TrkB expression in NM necessary for dendritic retraction? To answer this question we combined focal in ovo electroporation with transposon mediated gene transfer to obtain stable expression of Doxycycline (Dox) regulated transgenes, specifically TrkB coexpressed with EGFP in a temporally controlled manner. Electroporation was performed at E2 and Dox added onto the chorioallointoic membrane from E7.5 to E16. Expression of EGFP had no effect on development of the embryo, or cell morphology and organization of auditory brainstem nuclei. NM cells expressing EGFP and TrkB at E17-E18 had dendrites and biophysical properties uncharacteristic for normal NM cells, indicating that cessation of TrkB expression is essential for dendrite retraction and functional maturation of these neurons. These studies indicate that expression of transposon based plasmids is an effective method to genetically manipulate events in mid to late embryonic brain development in chick.


Assuntos
Vias Auditivas/embriologia , Tronco Encefálico/embriologia , Dendritos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/fisiologia , Neurônios/metabolismo , Receptor trkB/fisiologia , Animais , Embrião de Galinha , Elementos de DNA Transponíveis/genética , Regulação para Baixo , Doxiciclina/farmacologia , Eletroporação , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Neurogênese/genética , Neurônios/citologia , Receptor trkB/biossíntese , Receptor trkB/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Transgenes
6.
Artigo em Inglês | MEDLINE | ID: mdl-36504507

RESUMO

During protein synthesis, aminoacyl-tRNA synthetases covalently link amino acids with their cognate tRNAs. Amino acid mutations in glycyl-tRNA synthetase can disrupt protein synthesis and lead to a neurological disorder known as Charcot-Marie-Tooth disease type 2D (CMT-2D). Several studies employing diverse techniques have identified potential disease mechanisms at the molecular level. The majority of CMT-2D mutations in glycyl-tRNA are found within its dimer interface. However, no atomic structures bearing these mutations have been solved. Consequently, the specific disease-causing structural changes that occur in glycyl-tRNA synthetase have not been definitively established. Here we use molecular dynamics simulations to probe conformational changes in glycyl-tRNA synthetase caused by one mutation within the dimer interface: G240R. Our results show that the mutation alters the number of native interactions at the dimer interface and also leads to altered dynamics of two regions of glycyl-tRNA synthetase associated with tRNA binding. Additionally, we use our simulations to make predictions about the effects of other clinically reported CMT-2D mutations. Our results identify a region of the glycyl-tRNA synthetase structure that may be disrupted in a large number of CMT-2D mutations. Structural changes in this region may be a common molecular mechanism in glycyl-tRNA synthetase CMT-2D pathologies. Statement of significance: In this study, we use molecular dynamics simulations to elucidate structural conformations accessible to glycyl-tRNA synthetase (GlyRS), an enzyme that ligates cytosolic glycine with tRNA-Gly. This protein contains multiple flexible regions with dynamics that elude in vitro structural characterization. Our computational approach provides unparalleled atomistic details of structural changes in GlyRS that contribute to its role in protein synthesis. A number of mutations in GlyRS are associated with a peripheral nerve disorder, Charcot-Marie-Tooth disease type 2D (CMT-2D). Mutation-induced structural and dynamic changes in GlyRS have similarity that elude in vitro structural characterization. Our simulations provide insights into disease mechanisms for one such mutation: G240R. Additionally, we leverage our computational data to identify regions of GlyRS critical to its function and to predict the effects of other disease-associated mutations. These results open up new directions for research into the molecular characterization of GlyRS and into hypothesis-driven studies of CMT-2D disease mechanisms.

7.
Adv Biol (Weinh) ; 6(2): e2101308, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34958183

RESUMO

Charcot-Marie-Tooth disease type 2D (CMT2D), is a hereditary peripheral neuropathy caused by mutations in the gene encoding glycyl-tRNA synthetase (GARS1). Here, human induced pluripotent stem cell (hiPSC)-based models of CMT2D bearing mutations in GARS1 and their use for the identification of predictive biomarkers amenable to therapeutic efficacy screening is described. Cultures containing spinal cord motor neurons generated from this line exhibit network activity marked by significant deficiencies in spontaneous action potential firing and burst fire behavior. This result matches clinical data collected from a patient bearing a GARS1P724H mutation and is coupled with significant decreases in acetylated α-tubulin levels and mitochondrial movement within axons. Treatment with histone deacetylase 6 inhibitors, tubastatin A and CKD504, improves mitochondrial movement and α-tubulin acetylation in these cells. Furthermore, CKD504 treatment enhances population-level electrophysiological activity, highlighting its potential as an effective treatment for CMT2D.


Assuntos
Doença de Charcot-Marie-Tooth , Glicina-tRNA Ligase , Células-Tronco Pluripotentes Induzidas , Transporte Axonal , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Glicina-tRNA Ligase/genética , Desacetilase 6 de Histona/genética , Inibidores de Histona Desacetilases/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Tubulina (Proteína)/genética
8.
Mol Cell Neurosci ; 43(4): 403-13, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20123019

RESUMO

Activation of nascent receptor tyrosine kinases within the secretory pathway has been reported, yet the consequences of intracellular activation are largely unexplored. We report that overexpression of the Trk neurotrophin receptors causes accumulation of autoactivated receptors in the ER-Golgi intermediate compartment. Autoactivated receptors exhibit inhibited Golgi-mediated processing and they inhibit Golgi-mediated processing of other co-expressed transmembrane proteins, apparently by inducing fragmentation of the Golgi apparatus. Signaling from G protein-coupled receptors is known to induce Trk transactivation. Transactivation of nascent TrkB in hippocampal neurons resulting from exposure to the neuropeptide PACAP caused Golgi fragmentation, whereas BDNF-dependent activation of TrkB did not. TrkB-mediated Golgi fragmentation employs a MEK-dependent signaling pathway resembling that implicated in regulation of Golgi fragmentation in mitotic cells. Neuronal Golgi fragments, in the form of dendritically localized Golgi outposts, are important determinants of dendritic growth and branching. The capacity of transactivated TrkB to enhance neuronal Golgi fragmentation may represent a novel mechanism regulating neural plasticity.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Via Secretória/fisiologia , Western Blotting , Linhagem Celular , Células Cultivadas , Imunofluorescência , Humanos , Imunoprecipitação , Microscopia Confocal , Fosforilação/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transporte Proteico/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Transfecção
9.
Front Cell Dev Biol ; 9: 728707, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34660586

RESUMO

Gene editing technologies hold great potential to enhance our ability to model inheritable neurodegenerative diseases. Specifically, engineering multiple amyotrophic lateral sclerosis (ALS) mutations into isogenic cell populations facilitates determination of whether different causal mutations cause pathology via shared mechanisms, and provides the capacity to separate these mechanisms from genotype-specific effects. As gene-edited, cell-based models of human disease become more commonplace, there is an urgent need to verify that these models constitute consistent and accurate representations of native biology. Here, commercially sourced, induced pluripotent stem cell-derived motor neurons from Cellular Dynamics International, edited to express the ALS-relevant mutations TDP-43M337V and TDP-43Q331K were compared with in-house derived lines engineered to express the TDP-43Q331K mutation within the WTC11 background. Our results highlight electrophysiological and mitochondrial deficits in these edited cells that correlate with patient-derived cells, suggesting a consistent cellular phenotype arising from TDP-43 mutation. However, significant differences in the transcriptomic profiles and splicing behavior of the edited cells underscores the need for careful comparison of multiple lines when attempting to use these cells as a means to better understand the onset and progression of ALS in humans.

10.
Biomaterials ; 271: 120700, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33631652

RESUMO

Neurons derived from human induced pluripotent stem cells (hiPSCs) are powerful tools for modeling neural pathophysiology and preclinical efficacy/toxicity screening of novel therapeutic compounds. However, human neurons cultured in vitro typically do not fully recapitulate the physiology of the human nervous system, especially in terms of exhibiting morphological maturation, longevity, and electrochemical signaling ability comparable to that of adult human neurons. In this study, we investigated the potential for astrocyte-derived extracellular vesicles (EVs) to modulate survival and electrophysiological function of human neurons in vitro. Specifically, we demonstrate that EVs obtained from human astrocytes promote enhanced single cell electrophysiological function and anti-apoptotic behavior in a homogeneous population of human iPSC-derived cortical neurons. Furthermore, EV-proteomic analysis was performed to identify cargo proteins with the potential to promote the physiological enhancement observed. EV cargos were found to include neuroprotective proteins such as heat shock proteins, alpha-synuclein, and lipoprotein receptor-related protein 1 (LRP1), as well as apolipoprotein E (APOE), which negatively regulates neuronal apoptosis, and a peroxidasin homolog that supports neuronal oxidative stress management. Proteins that positively regulate neuronal excitability and synaptic development were also detected, such as potassium channel tetramerization domain containing 12 (KCTD12), glucose-6- phosphate dehydrogenase (G6PD), kinesin family member 5B (KIF5B), spectrin-alpha non-erythrocytic1 (SPTAN1). The remarkable improvements in electrophysiological function and evident inhibition of apoptotic signaling in cultured neurons exposed to these cargos may hold significance for improving preclinical in vitro screening modalities. In addition, our collected data highlight the potential for EV-based therapeutics as a potential class of future clinical treatment for tackling inveterate central and peripheral neuropathies.


Assuntos
Vesículas Extracelulares , Células-Tronco Pluripotentes Induzidas , Doenças do Sistema Nervoso Periférico , Astrócitos , Células Cultivadas , Humanos , Neurônios , Proteômica
11.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31583078

RESUMO

Complex mechanisms control the signaling of neurotrophins through p75 NTR and Trk receptors, allowing cellular responses that are highly context dependent, particularly in the nervous system and particularly with regard to the neurotrophin brain-derived neurotrophic factor (BDNF). Recent reports describe a variety of sophisticated regulatory mechanisms that contribute to such functional flexibility. Mechanisms described include regulation of trafficking of alternative BDNF transcripts, regulation of post-translational processing and secretion of BDNF, engagement of co-receptors that influence localization and signaling of p75 NTR and Trk receptors, and control of trafficking of receptors in the endocytic pathway and during anterograde and retrograde axonal transport.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Transdução de Sinais , Transporte Axonal , Endocitose , Humanos , Proteínas do Tecido Nervoso/farmacologia , Processamento de Proteína Pós-Traducional , Receptores de Fator de Crescimento Neural/fisiologia
12.
Neuron ; 33(1): 9-12, 2002 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-11779474

RESUMO

The family of neurotrophic factors known as neurotrophins has yielded a series of surprises, both with regard to the broad extent of their functional roles and the remarkable complexity of their signaling mechanisms. The recent discovery that a neurotrophin precursor protein and its proteolytically processed products may differentially activate pro- and antiapoptotic cellular responses, through preferential activation of Trk or p75 receptors, promises to unveil yet another level of regulatory complexity.


Assuntos
Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Fatores de Crescimento Neural/metabolismo , Precursores de Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/fisiologia , Animais , Sítios de Ligação/fisiologia , Doenças do Sistema Nervoso Central/metabolismo , Doenças do Sistema Nervoso Central/fisiopatologia , Humanos
13.
Nat Neurosci ; 5(12): 1302-8, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12426574

RESUMO

Myelin-associated glycoprotein (MAG), an inhibitor of axon regeneration, binds with high affinity to the Nogo-66 receptor (NgR). Here we report that the p75 neurotrophin receptor (p75(NTR)) is a co-receptor of NgR for MAG signaling. In cultured human embryonic kidney (HEK) cells expressing NgR, p75(NTR) was required for MAG-induced intracellular Ca2+ elevation. Co-immunoprecipitation showed an association of NgR with p75(NTR) that can be disrupted by an antibody against p75(NTR) (NGFR5), and extensive coexpression was observed in the developing rat nervous system. Furthermore, NGFR5 abolished MAG-induced repulsive turning of Xenopus axonal growth cones and Ca2+ elevation, both in neurons and in NgR/p75(NTR)-expressing HEK cells. Thus we conclude that p75(NTR) is a co-receptor of NgR for MAG signaling and a potential therapeutic target for promoting nerve regeneration.


Assuntos
Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Cones de Crescimento/metabolismo , Proteínas da Mielina/metabolismo , Glicoproteína Associada a Mielina/metabolismo , Regeneração Nervosa/genética , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Diferenciação Celular/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Sistema Nervoso Central/citologia , Embrião não Mamífero , Feto , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/ultraestrutura , Humanos , Imuno-Histoquímica , Proteínas da Mielina/genética , Glicoproteína Associada a Mielina/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Proteínas Nogo , Ratos , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/imunologia , Xenopus
15.
F1000Res ; 52016.
Artigo em Inglês | MEDLINE | ID: mdl-27540475

RESUMO

The nerve growth factor family of growth factors, collectively known as neurotrophins, are evolutionarily ancient regulators with an enormous range of biological functions. Reflecting this long history and functional diversity, mechanisms for cellular responses to neurotrophins are exceptionally complex. Neurotrophins signal through p75 (NTR), a member of the TNF receptor superfamily member, and through receptor tyrosine kinases (TrkA, TrkB, TrkC), often with opposite functional outcomes. The two classes of receptors are activated preferentially by proneurotrophins and mature processed neurotrophins, respectively. However, both receptor classes also possess neurotrophin-independent signaling functions. Signaling functions of p75 (NTR) and Trk receptors are each influenced by the other class of receptors. This review focuses on the mechanisms responsible for the functional interplay between the two neurotrophin receptor signaling systems.

16.
J Neurosci ; 23(13): 5425-36, 2003 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-12843241

RESUMO

The 75 kDa neurotrophin receptor (p75NTR) and two neurotrophin receptor homologs (NRH1, NRH2) constitute a subfamily of the nerve growth factor/tumor necrosis factor receptor superfamily. NRH1 coexists with p75NTR in fish, amphibians, and birds but is absent in mammals, whereas NRH2 exists only in mammals. Unlike p75NTR and NRH1, NRH2 lacks a canonical extracellular ligand binding domain. The similarity of NRH2 to the product of metalloproteinase cleavage of p75NTR prompted us to examine the cleavage of p75NTR in greater detail. p75NTR, NRH1, and NRH2 undergo multiple proteolytic cleavages that ultimately release cytoplasmic fragments. For p75NTR, cleavage in the extracellular domain by a PMA-inducible membrane metalloproteinase is followed by cleavage within or near the transmembrane domain, releasing the intracellular domain into the cytoplasm. This processing resembles the alpha- and gamma-secretase-mediated processing of beta-amyloid precursor protein and the similar processing of Notch. Although neurotrophins did not regulate p75NTR processing, the alpha- and gamma-secretase-mediated cleavage of p75 is modulated by receptor tyrosine kinases (Trks) TrkA and TrkB but not TrkC. Surprisingly, although NRH1 and NRH2 also undergo proteolytic cytoplasmic release of intracellular domains, a different protease mediates the cleavage. Furthermore, whereas the p75NTR soluble intracellular domain accumulates only in the presence of proteasome inhibitors, the equivalent fragment of NRH2 is stable and localizes in the nucleus. Because soluble intracellular domains of p75NTR and NRH2 were found to activate NF-kappaB in concert with TNF receptor associated factor 6 (TRAF6), we propose that cleavage of these proteins may serve conserved cytoplasmic and nuclear signaling functions through distinct proteases.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/fisiologia , Secretases da Proteína Precursora do Amiloide , Animais , Proteínas Reguladoras de Apoptose , Ácido Aspártico Endopeptidases , Proteínas de Transporte/genética , Linhagem Celular , Ecdisterona/análogos & derivados , Ecdisterona/farmacologia , Endopeptidases/metabolismo , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas de Membrana/genética , Dados de Sequência Molecular , NF-kappa B/metabolismo , Fragmentos de Peptídeos/biossíntese , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Proteínas/metabolismo , Ratos , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/genética , Homologia de Sequência de Aminoácidos , Fator 6 Associado a Receptor de TNF , Xenopus
17.
Artigo em Inglês | MEDLINE | ID: mdl-25758563

RESUMO

Sequential proteolytic cleavages of amyloid-ß protein precursor (AßPP) by ß-secretase and γ-secretase generate amyloid ß (Aß) peptides, which are thought to contribute to Alzheimer's disease (AD). Much of this processing occurs in endosomes following endocytosis of AßPP from the plasma membrane. However, this pathogenic mode of processing AßPP may occur in competition with lysosomal degradation of AßPP, a common fate of membrane proteins trafficking through the endosomal system. Following up on published reports that LINGO-1 binds and promotes the amyloidogenic processing of AßPP we have examined the consequences of LINGO-1/AßPP interactions. We report that LINGO-1 and its paralogs, LINGO-2 and LINGO-3, decrease processing of AßPP in the amyloidogenic pathway by promoting lysosomal degradation of AßPP. We also report that LINGO-1 levels are reduced in AD brain, representing a possible pathogenic mechanism stimulating the generation of Aß peptides in AD.

18.
J Comp Neurol ; 452(1): 51-64, 2002 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-12205709

RESUMO

Nucleus magnocellularis (NM) in the avian auditory brainstem receives auditory input from nerve the VIIIth and projects bilaterally to nucleus laminaris (NL). This projection preserves binaural segregation in that ipsilateral NM projects to dorsal dendrites of NL and contralateral NM projects to ventral dendrites of NL. We have begun to examine the molecular signals that influence segregation of inputs onto discrete regions of NL cells. We previously showed that the Eph receptor, EphA4, is expressed selectively in the dorsal NL neuropil from embryonic day (E) 9 to E11, when NM axons grow into the NL neuropil. This asymmetric distribution suggests that EphA4 acts as a guidance molecule during binaural segregation. We report here on the developmental changes in the expression of two other Eph receptors, EphB2 and EphB5, and two ligands, ephrin-B1 and ephrin-B2, in the chick auditory brainstem. These proteins are expressed in the auditory nuclei during the maturation of the NM-NL projection. EphB2, EphB5, and ephrin-B1 are expressed in dorsal and ventral NL neuropil and at the midline of the brainstem at E10-E12. At this age, ephrin-B2, a ligand for EphB receptors and for EphA4, is expressed in NL cell bodies and NM-NL axons. The expression of these proteins diminishs in the posthatch ages examined. These results suggest that several members of the Eph family are involved in maturation of the nuclei and their projections. Moreover, ephrin-B2 in growing axons may interact with the asymmetrically expressed EphA4 during the establishment of binaural segregation.


Assuntos
Embrião de Galinha/crescimento & desenvolvimento , Núcleo Coclear/embriologia , Proteínas de Membrana/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Animais , Western Blotting , Embrião de Galinha/anatomia & histologia , Efrina-B1 , Efrina-B2 , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Receptor EphB2
19.
Arch Ophthalmol ; 120(12): 1625-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12470134

RESUMO

OBJECTIVE: To characterize the immunophenotypic expression pattern of conjunctival melanomas, with the use of standard melanoma markers as well as microphthalmia transcription factor and p75 neurotrophin receptor. DESIGN: Eleven conjunctival melanomas, including 1 caruncular melanoma, were immunolabeled with a panel of antibodies that included S100, tyrosinase, melan-A, HMB-45 and HMB-50 combination, microphthalmia transcription factor, and p75 neurotrophin receptor. The results were tabulated on the basis of intensity and pervasiveness of labeling and compared with a previous study of uveal melanomas. RESULTS: Immunolabeling with S100 was at significantly higher levels in conjunctival melanomas than in uveal melanomas. Tyrosinase, HMB-45 and HMB-50 combination, melan-A, and microphthalmia transcription factor were expressed at high levels in conjunctival melanomas, whereas p75 neurotrophin receptor was not expressed. CONCLUSIONS: Melanomas of the conjunctiva, including the caruncle, expressed S100, tyrosinase, melan-A, HMB-45 and HMB-50 combination, and microphthalmia transcription factor at high levels, suggesting that these are good markers for this melanoma subtype. Expression of S100 was significantly higher in conjunctival melanomas than in uveal melanomas. The immunophenotypic pattern of conjunctival melanomas is most similar to the epithelioid subtype of cutaneous melanomas.


Assuntos
Neoplasias da Túnica Conjuntiva/patologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Neoplasias Uveais/patologia , Biomarcadores Tumorais/análise , Neoplasias da Túnica Conjuntiva/química , Humanos , Técnicas Imunoenzimáticas , Imunofenotipagem , Melanoma/química , Proteínas de Neoplasias/análise , Neoplasias Cutâneas/química , Neoplasias Uveais/química
20.
Arch Ophthalmol ; 120(4): 466-70, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11934320

RESUMO

OBJECTIVE: To determine the immunophenotypic differences between uveal and cutaneous melanomas, employing standard melanoma markers as well as p75 neurotrophin receptor (p75NTR) and microphthalmia transcription factor (MITF). DESIGN: Fifteen uveal melanomas (5 spindle, 5 epithelioid, and 5 mixed uveal subtypes) were immunolabeled with a panel of antibodies that included S100, tyrosinase, melan-A, HMB-45 and HMB-50 combination, MITF, and p75NTR. The results were tabulated on the basis of intensity and pervasiveness of the labeling and compared with a prior study on cutaneous spindle and epithelioid melanomas. RESULTS: In contrast to its strong labeling of cutaneous melanomas, S100 immunolabeling of uveal melanomas was weak and variable. p75NTR, known to differentiate spindle from epithelioid melanomas of the skin, did not immunolabel uveal melanomas. HMB-45, HMB-50, tyrosinase, melan-A, and MITF immunolabeled all uveal melanomas strongly, irrespective of the histologic subtype, but not cutaneous melanomas. Microphthalmia transcription factor was especially clear in its labeling of uveal melanomas. CONCLUSIONS: Although cutaneous and uveal melanomas share many molecular markers in common, there are differences between the 2 types of melanoma. First, the level of expression of S100 differs between cutaneous and uveal melanomas. Second, while cutaneous melanomas can be further subdivided into spindle and epithelioid types based on their immunophenotype, the uveal melanomas cannot.


Assuntos
Neoplasias da Coroide/classificação , Imunofenotipagem , Melanoma/classificação , Neoplasias Cutâneas/classificação , Fatores de Transcrição , Antígenos de Neoplasias/análise , Biomarcadores Tumorais/análise , Neoplasias da Coroide/química , Neoplasias da Coroide/patologia , Proteínas de Ligação a DNA/análise , Humanos , Antígeno MART-1 , Melanoma/química , Melanoma/patologia , Antígenos Específicos de Melanoma , Fator de Transcrição Associado à Microftalmia , Monofenol Mono-Oxigenase/análise , Proteínas de Neoplasias/análise , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/análise , Proteínas S100/análise , Neoplasias Cutâneas/química , Neoplasias Cutâneas/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa