Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Structure ; 32(2): 217-227.e3, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38052206

RESUMO

E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8-mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site-directed mutagenesis, and single-molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped ß-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.


Assuntos
Caderinas , Simulação de Dinâmica Molecular , Caderinas/metabolismo , Ligação Proteica , Adesão Celular
2.
bioRxiv ; 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37461464

RESUMO

E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8 mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site directed mutagenesis and single molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped ß-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.

3.
PNAS Nexus ; 1(4): pgac163, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36157596

RESUMO

E-cadherin adhesion is regulated at the cell surface, a process that can be replicated by activating antibodies. We use cryo-electron microscopy (EM) and X-ray crystallography to examine functional states of the cadherin adhesive dimer. This dimer is mediated by N-terminal beta strand-swapping involving Trp2, and forms via a different transient X-dimer intermediate. X-dimers are observed in cryo-EM along with monomers and strand-swap dimers, indicating that X-dimers form stable interactions. A novel EC4-mediated dimer was also observed. Activating Fab binding caused no gross structural changes in E-cadherin monomers, but can facilitate strand swapping. Moreover, activating Fab binding is incompatible with the formation of the X-dimer. Both cryo-EM and X-ray crystallography reveal a distinctive twisted strand-swap dimer conformation caused by an outward shift in the N-terminal beta strand that may represent a strengthened state. Thus, regulation of adhesion involves changes in cadherin dimer configurations.

4.
Tissue Barriers ; 9(4): 1940741, 2021 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-34402758

RESUMO

Deficits in gastrointestinal (GI) paracellular permeability has been implicated in etiology of Inflammatory Bowel Disease (IBD), and E-cadherin, a key component of the epithelial junctional complex, has been implicated in both barrier function and IBD. We have previously described antibodies against E-cadherin that activate cell adhesion, and in this study, we show that they increase transepithelial electrical resistance in epithelial cell monolayers in vitro. We therefore tested the hypothesis that adhesion activating E-cadherin mAbs will enhance epithelial barrier function in vivo and limit progression of inflammation in IBD. Activating mAbs to mouse E-cadherin were tested in different mouse models of IBD including the IL10-/- and adoptive T cell transfer models of colitis. Previously established histological and biomarker measures of inflammation were evaluated to monitor disease progression. Mouse E-cadherin activating mAb treatment reduced total colitis score, individual histological measures of inflammation, and other hallmarks of inflammation compared to control treatment. Activating mAbs also reduced the fecal accumulation lipocalin2 and albumin content, consistent with enhanced barrier function. Therefore, E-cadherin activation could be a potential strategy for limiting inflammation in UC.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Animais , Caderinas/metabolismo , Colite/metabolismo , Colite/patologia , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Camundongos
5.
Am J Physiol Heart Circ Physiol ; 320(4): H1403-H1410, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33577432

RESUMO

Excessive vascular permeability occurs in inflammatory disease processes. Vascular endothelial cadherin (VE-cadherin) is an adhesion protein that controls vascular permeability. We identified monoclonal antibodies (mAbs) to human VE-cadherin that activate cell adhesion and inhibit the increased permeability of endothelial cell monolayers induced by thrombin receptor activator peptide-6 (TRAP-6). Two mAbs, 8A12c and 3A5a, reduce permeability, whereas an inhibitory mAb, 2E11d, enhances permeability. Activating mAbs also reduce permeability induced by tumor necrosis factor-α (TNF-α) and vascular endothelial cell growth factor (VEGF). The activating mAbs also stabilize the organization of the adherens junctions that are disrupted by TRAP-6, VEGF, or TNF-α. The activating mAbs act directly on the adhesive function of VE-cadherin because they did not block the accumulation of actin filaments stimulated by TRAP-6 and enhance physical cell-cell adhesion of VE-cadherin-expressing tissue culture cells. Therefore, VE-cadherin function can be regulated at the cell surface to control endothelial permeability.NEW & NOTEWORTHY Excessive vascular permeability is a serious complication of many inflammatory disease conditions. We have developed monoclonal antibodies that inhibit increases in endothelial monolayer permeability induced by several signaling factors by activating VE-cadherin mediated adhesion and stabilizing cell junctions. These antibodies and/or the mechanisms they reveal may lead to important therapeutics to treat vascular leakiness and inflammation.


Assuntos
Junções Aderentes/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Caderinas/agonistas , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Junções Aderentes/metabolismo , Antígenos CD/metabolismo , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Nocodazol/farmacologia , Oligopeptídeos/farmacologia , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transdução de Sinais , Moduladores de Tubulina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Mol Vis ; 26: 576-587, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32863706

RESUMO

Purpose: The cornea is highly enriched in sensory neurons expressing the thermal TRP channels TRPV1, TRPA1, and TRPM8, and is an accessible tissue for study and experimental manipulation. The aim of this work was to provide a concise characterization of the expression patterns of various TRP channels and vesicular proteins in the mammalian cornea. Methods: Immunohistochemistry (IHC) was performed using wholemount and cryostat tissue preparations of mouse and monkey corneas. The expression patterns of TRPV1 and TRPA1 were determined using specific antisera, and further colocalization was performed with antibodies directed against calcitonin-related gene protein (CGRP), neurofilament protein NF200, and the secretogranins ScgII and SCG3. The expression of TRPM8 was determined using corneas from mice expressing EGFP under the direction of a TRPM8 promoter (TRPM8EGFP mice). Laser scanning confocal microscopy and image analysis were performed. Results: In the mouse cornea, TRPV1 and TRPM8 were expressed in distinct populations of small diameter C fibers extending to the corneal surface and ending either as simple or ramifying terminals, or in the case of TRPM8, as complex terminals. TRPA1 was expressed in large-diameter NF200-positive Aδ axons. TRPV1 and TRPA1 appeared to localize to separate intracellular vesicular structures and were primarily found in axons containing components of large dense vesicles with TRPV1 colocalizing with CGRP and ScgII, and TRPA1 colocalizing with SCG3. Monkey corneas showed similar colocalization of CGRP and TRPV1 on small-diameter axons extending to the epithelial surface. Conclusions: The mouse cornea is abundant in sensory neurons expressing TRPV1, TRPM8, and TRPA1, and provides an accessible tissue source for implementing a live tissue preparation useful for further exploration of the molecular mechanisms of hyperalgesia. This study showed that surprisingly, these TRP channels localize to separate neurons in the mouse cornea and likely have unique physiological functions. The similar TRPV1 expression pattern we observed in the mouse and monkey corneas suggests that mice provide a reasonable initial model for understanding the role of these ion channels in higher mammalian corneal physiology.


Assuntos
Axônios/metabolismo , Córnea/metabolismo , Células Receptoras Sensoriais/metabolismo , Canal de Cátion TRPA1/genética , Canais de Cátion TRPM/genética , Canais de Cátion TRPV/genética , Animais , Axônios/ultraestrutura , Cromograninas/genética , Cromograninas/metabolismo , Sequência Conservada , Córnea/anatomia & histologia , Córnea/ultraestrutura , Expressão Gênica , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Imuno-Histoquímica , Macaca nemestrina , Camundongos , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/genética , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo , Secretogranina II/genética , Secretogranina II/metabolismo , Células Receptoras Sensoriais/ultraestrutura , Transmissão Sináptica/genética , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(11): 5931-5937, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32127478

RESUMO

E-cadherin is a tumor suppressor protein, and the loss of its expression in association with the epithelial mesenchymal transition (EMT) occurs frequently during tumor metastasis. However, many metastases continue to express E-cadherin, and a full EMT is not always necessary for metastasis; also, positive roles for E-cadherin expression in metastasis have been reported. We hypothesize instead that changes in the functional activity of E-cadherin expressed on tumor cells in response to environmental factors is an important determinant of the ability of the tumor cells to metastasize. We find that E-cadherin expression persists in metastatic lung nodules and circulating tumor cells (CTCs) in two mouse models of mammary cancer: genetically modified MMTV-PyMT mice and orthotopically grafted 4T1 tumor cells. Importantly, monoclonal antibodies that bind to and activate E-cadherin at the cell surface reduce lung metastasis from endogenous genetically driven tumors and from tumor cell grafts. E-cadherin activation inhibits metastasis at multiple stages, including the accumulation of CTCs from the primary tumor and the extravasation of tumor cells from the vasculature. These activating mAbs increase cell adhesion and reduce cell invasion and migration in both cell culture and three-dimensional spheroids grown from primary tumors. Moreover, activating mAbs increased the frequency of apoptotic cells without affecting proliferation. Although the growth of the primary tumors was unaffected by activating mAbs, CTCs and tumor cells in metastatic nodules exhibited increased apoptosis. Thus, the functional state of E-cadherin is an important determinant of metastatic potential beyond whether the gene is expressed.


Assuntos
Neoplasias da Mama/metabolismo , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Neoplasias Pulmonares/metabolismo , Metástase Neoplásica , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caderinas/genética , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica/tratamento farmacológico , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Processos Neoplásicos
8.
Mol Vis ; 26: 392-404, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-38860239

RESUMO

Purpose: The cornea is highly enriched in sensory neurons expressing the thermal TRP channels TRPV1, TRPA1, and TRPM8, and is an accessible tissue for study and experimental manipulation. The aim of this work was to provide a concise characterization of the expression patterns of various TRP channels and vesicular proteins in the mammalian cornea. Methods: Immunohistochemistry (IHC) was performed using wholemount and cryostat tissue preparations of mouse and monkey corneas. The expression patterns of TRPV1 and TRPA1 were determined using specific antisera, and further colocalization was performed with antibodies directed against calcitonin-related gene protein (CGRP), neurofilament protein NF200, and the secretogranins ScgII and SCG3. The expression of TRPM8 was determined using corneas from mice expressing EGFP under the direction of a TRPM8 promoter (TRPM8EGFP mice). Laser scanning confocal microscopy and image analysis were performed. Results: In the mouse cornea, TRPV1 and TRPM8 were expressed in distinct populations of small diameter C fibers extending to the corneal surface and ending either as simple or ramifying terminals, or in the case of TRPM8, as complex terminals. TRPA1 was expressed in large-diameter NF200-positive Aδ axons. TRPV1 and TRPA1 appeared to localize to separate intracellular vesicular structures and were primarily found in axons containing components of large dense vesicles with TRPV1 colocalizing with CGRP and ScgII, and TRPA1 colocalizing with SCG3. Monkey corneas showed similar colocalization of CGRP and TRPV1 on small-diameter axons extending to the epithelial surface. Conclusions: The mouse cornea is abundant in sensory neurons expressing TRPV1, TRPM8, and TRPA1, and provides an accessible tissue source for implementing a live tissue preparation useful for further exploration of the molecular mechanisms of hyperalgesia. This study showed that surprisingly, these TRP channels localize to separate neurons in the mouse cornea and likely have unique physiological functions. The similar TRPV1 expression pattern we observed in the mouse and monkey corneas suggests that mice provide a reasonable initial model for understanding the role of these ion channels in higher mammalian corneal physiology.

9.
J Neurosci ; 38(29): 6445-6460, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29950504

RESUMO

A global loss of the fragile X mental retardation protein (FMRP; encoded by the Fmr1 gene) leads to sensory dysfunction and intellectual disabilities. One underlying mechanism of these phenotypes is structural and functional deficits in synapses. Here, we determined the autonomous function of postsynaptic FMRP in circuit formation, synaptogenesis, and synaptic maturation. In normal cochlea nucleus, presynaptic auditory axons form large axosomatic endbulb synapses on cell bodies of postsynaptic bushy neurons. In ovo electroporation of drug-inducible Fmr1-shRNA constructs produced a mosaicism of FMRP expression in chicken (either sex) bushy neurons, leading to reduced FMRP levels in transfected, but not neighboring nontransfected, neurons. Structural analyses revealed that postsynaptic FMRP reduction led to smaller size and abnormal morphology of individual presynaptic endbulbs at both early and later developmental stages. We further examined whether FMRP reduction affects dendritic development, as a potential mechanism underlying defective endbulb formation. Normally, chicken bushy neurons grow extensive dendrites at early stages and retract these dendrites when endbulbs begin to form. Neurons transfected with Fmr1 shRNA exhibited a remarkable delay in branch retraction, failing to provide necessary somatic surface for timely formation and growth of large endbulbs. Patch-clamp recording verified functional consequences of dendritic and synaptic deficits on neurotransmission, showing smaller amplitudes and slower kinetics of spontaneous and evoked EPSCs. Together, these data demonstrate that proper levels of postsynaptic FMRP are required for timely maturation of somatodendritic morphology, a delay of which may affect synaptogenesis and thus contribute to long-lasting deficits of excitatory synapses.SIGNIFICANCE STATEMENT Fragile X mental retardation protein (FMRP) regulates a large variety of neuronal activities. A global loss of FMRP affects neural circuit development and synaptic function, leading to fragile X syndrome (FXS). Using temporally and spatially controlled genetic manipulations, this study provides the first in vivo report that autonomous FMRP regulates multiple stages of dendritic development, and that selective reduction of postsynaptic FMRP leads to abnormal development of excitatory presynaptic terminals and compromised neurotransmission. These observations demonstrate secondary influence of developmentally transient deficits in neuronal morphology and connectivity to the development of long-lasting synaptic pathology in FXS.


Assuntos
Núcleo Coclear/embriologia , Núcleo Coclear/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurogênese/fisiologia , Sinapses/fisiologia , Animais , Embrião de Galinha , Feminino , Masculino , Neurônios/fisiologia , Transmissão Sináptica/fisiologia
10.
Mol Biol Cell ; 27(21): 3233-3244, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582386

RESUMO

The loss of E-cadherin expression in association with the epithelial-mesenchymal transition (EMT) occurs frequently during tumor metastasis. However, metastases often retain E-cadherin expression, an EMT is not required for metastasis, and metastases can arise from clusters of tumor cells. We demonstrate that the regulation of the adhesive activity of E-cadherin present at the cell surface by an inside-out signaling mechanism is important in cancer. First, we find that the metastasis of an E-cadherin-expressing mammary cell line from the mammary gland to the lung depends on reduced E-cadherin adhesive function. An activating monoclonal antibody to E-cadherin that induces a high adhesive state significantly reduced the number of cells metastasized to the lung without affecting the growth in size of the primary tumor in the mammary gland. Second, we find that many cancer-associated germline missense mutations in the E-cadherin gene in patients with hereditary diffuse gastric cancer selectively affect the mechanism of inside-out cell surface regulation without inhibiting basic E-cadherin adhesion function. This suggests that genetic deficits in E-cadherin cell surface regulation contribute to cancer progression. Analysis of these mutations also provides insights into the molecular mechanisms underlying cadherin regulation at the cell surface.


Assuntos
Caderinas/genética , Caderinas/metabolismo , Animais , Caderinas/fisiologia , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Receptores de Superfície Celular , Transdução de Sinais , Fatores de Transcrição/metabolismo
11.
J Comp Neurol ; 521(1): 5-23, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22806400

RESUMO

Research performed on transgenic animals has led to numerous advances in biological research. However, using traditional retroviral methods to generate transgenic avian research models has proved problematic. As a result, experiments aimed at genetic manipulations on birds have remained difficult for this popular research tool. Recently, lentiviral methods have allowed the production of transgenic birds, including a transgenic Japanese quail (Coturnix coturnix japonica) line showing neuronal specificity and stable expression of enhanced green fluorescent protein (eGFP) across generations (termed here GFP quail). To test whether the GFP quail may serve as a viable alternative to the popular chicken model system, with the additional benefit of genetic manipulation, we compared the development, organization, structure, and function of a specific neuronal circuit in chicken (Gallus gallus domesticus) with that of the GFP quail. This study focuses on a well-defined avian brain region, the principal nuclei of the sound localization circuit in the auditory brainstem, nucleus magnocellularis (NM), and nucleus laminaris (NL). Our results demonstrate that structural and functional properties of NM and NL neurons in the GFP quail, as well as their dynamic properties in response to changes in the environment, are nearly identical to those in chickens. These similarities demonstrate that the GFP quail, as well as other transgenic quail lines, can serve as an attractive avian model system, with the advantage of being able to build on the wealth of information already available from the chicken.


Assuntos
Tronco Encefálico , Regulação da Expressão Gênica no Desenvolvimento/genética , Modelos Animais , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Tronco Encefálico/citologia , Tronco Encefálico/embriologia , Tronco Encefálico/crescimento & desenvolvimento , Embrião de Galinha , Cóclea/metabolismo , Cóclea/cirurgia , Coturnix , Estimulação Elétrica , Embrião não Mamífero , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Fluoxetina/farmacologia , Lateralidade Funcional , Antagonistas GABAérgicos/farmacologia , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Técnicas In Vitro , Canal de Potássio Kv1.3/metabolismo , Lentivirus/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Vias Neurais/fisiologia , Ácido Okadáico/análogos & derivados , Técnicas de Patch-Clamp , Picrotoxina/farmacologia , Piranos/farmacocinética , Quinoxalinas/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sinapsinas/genética , Sinapsinas/metabolismo , Transgenes , Valina/análogos & derivados , Valina/farmacologia
12.
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
13.
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
14.
PLoS One ; 5(2): e9135, 2010 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-20161760

RESUMO

BACKGROUND: The familial and sporadic forms of Alzheimer's disease (AD) have an identical pathology with a severe disparity in the time of onset [1]. The pathological similarity suggests that epigenetic processes may phenocopy the Familial Alzheimer's disease (FAD) mutations within sporadic AD. Numerous groups have demonstrated that FAD mutations in presenilin result in 'loss of function' of gamma-secretase mediated APP cleavage [2], [3], [4], [5]. Accordingly, ER stress is prominent within the pathologically impacted brain regions in AD patients [6] and is reported to inhibit APP trafficking through the secretory pathway [7], [8]. As the maturation of APP and the cleaving secretases requires trafficking through the secretory pathway [9], [10], [11], we hypothesized that ER stress may block trafficking requisite for normal levels of APP cleavage and that the small molecular chaperone 4-phenylbutyrate (PBA) may rescue the proteolytic deficit. METHODOLOGY/PRINCIPAL FINDINGS: The APP-Gal4VP16/Gal4-reporter screen was stably incorporated into neuroblastoma cells in order to assay gamma-secretase mediated APP proteolysis under normal and pharmacologically induced ER stress conditions. Three unrelated pharmacological agents (tunicamycin, thapsigargin and brefeldin A) all repressed APP proteolysis in parallel with activation of unfolded protein response (UPR) signaling-a biochemical marker of ER stress. Co-treatment of the gamma-secretase reporter cells with PBA blocked the repressive effects of tunicamycin and thapsigargin upon APP proteolysis, UPR activation, and apoptosis. In unstressed cells, PBA stimulated gamma-secretase mediated cleavage of APP by 8-10 fold, in the absence of any significant effects upon amyloid production, by promoting APP trafficking through the secretory pathway and the stimulation of the non-pathogenic alpha/gamma-cleavage. CONCLUSIONS/SIGNIFICANCE: ER stress represses gamma-secretase mediated APP proteolysis, which replicates some of the proteolytic deficits associated with the FAD mutations. The small molecular chaperone PBA can reverse ER stress induced effects upon APP proteolysis, trafficking and cellular viability. Pharmaceutical agents, such as PBA, that stimulate alpha/gamma-cleavage of APP by modifying intracellular trafficking should be explored as AD therapeutics.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apoptose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Fenilbutiratos/farmacologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Western Blotting , Brefeldina A/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Tapsigargina/farmacologia , Transfecção , Tunicamicina/farmacologia
15.
Dev Neurobiol ; 70(5): 332-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20186712

RESUMO

Neurotrophins are important regulators of embryonic development and adult function of most populations of neurons in vertebrate nervous systems. This signaling system regulates many diverse activities, including survival, axon outgrowth, and synaptic plasticity. In mammals, neurotrophin action is mediated by four receptors, p75(NTR), TrkA, TrkB, and TrkC. Although early studies viewed these receptors as solitary agents in the cells outer membrane, recent discoveries reveal that the cell outer membrane is a crowded and highly interactive neighborhood. Neurotrophin receptors partner with a diverse array of membrane proteins, dramatically expanding their functional repertoire. This review will focus on some of the most recent discoveries concerning the promiscuous partnering of neurotrophin receptors.


Assuntos
Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Humanos , Neurônios/fisiologia
16.
Sci Signal ; 1(47): pe50, 2008 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19036712

RESUMO

The functions of the 75-kilodalton neurotrophin receptor p75(NTR) have remained enigmatic despite nearly three decades of study. Recent studies reveal that p75(NTR) is a versatile co-receptor that controls signaling by receptors for multiple ligands that provide repellant guidance cues to developing axons.


Assuntos
Axônios/ultraestrutura , Receptor de Fator de Crescimento Neural/fisiologia , Animais , Humanos , Transdução de Sinais
17.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...