Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 15(1): 242-255, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32619491

RESUMO

Individuals with the genetic disorder alpha-1 antitrypsin deficiency (AATD) are at risk of developing lung and liver disease. Patient induced pluripotent stem cells (iPSCs) have been found to model features of AATD pathogenesis but only a handful of AATD patient iPSC lines have been published. To capture the significant phenotypic diversity of the patient population, we describe here the establishment and characterization of a curated repository of AATD iPSCs with associated disease-relevant clinical data. To highlight the utility of the repository, we selected a subset of iPSC lines for functional characterization. Selected lines were differentiated to generate both hepatic and lung cell lineages and analyzed by RNA sequencing. In addition, two iPSC lines were targeted using CRISPR/Cas9 editing to accomplish scarless repair. Repository iPSCs are available to investigators for studies of disease pathogenesis and therapeutic discovery.


Assuntos
Acesso à Informação , Bases de Dados como Assunto , Células-Tronco Pluripotentes Induzidas/patologia , Deficiência de alfa 1-Antitripsina/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sistemas CRISPR-Cas/genética , Diferenciação Celular , Linhagem da Célula , Endoderma/patologia , Feminino , Edição de Genes , Loci Gênicos , Genótipo , Hepatócitos/patologia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Mutação/genética , Fenótipo , Transcriptoma/genética , alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/diagnóstico por imagem , Deficiência de alfa 1-Antitripsina/genética
2.
Amyloid ; 25(3): 148-155, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30032658

RESUMO

Hereditary transthyretin amyloidosis (ATTR amyloidosis) is an autosomal dominant protein-folding disorder caused by over 100 distinct mutations in the transthyretin (TTR) gene. In ATTR amyloidosis, protein secreted from the liver aggregates and forms amyloid fibrils in downstream target organs, chiefly the heart and peripheral nervous system. Few animal models of ATTR amyloidosis exist and none recapitulate the multisystem complexity and clinical variability associated with disease pathogenesis in patients. Induced pluripotent stem cells (iPSCs) stand to revolutionize the way we study human development, model disease, and perhaps treat patients afflicted with highly variable multisystem diseases such as ATTR amyloidosis. Here, we fully characterize six representative iPSC lines from a library of previously reprogrammed iPSC lines and reprogrammable blood samples derived from ATTR amyloidosis patients. This unique resource, described herein, can be harnessed to study diverse disorder.


Assuntos
Neuropatias Amiloides Familiares/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Citometria de Fluxo , Humanos , Mutação/genética
3.
Neurology ; 88(20): 1968-1975, 2017 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-28507260

RESUMO

Gulf War illness (GWI), which afflicts at least 25% of veterans who served in the 1990-1991 war in the Persian Gulf, is thought to be caused by deployment exposures to various neurotoxicants, including pesticides, anti-nerve gas pills, and low-level nerve agents including sarin/cyclosarin. GWI is a multisymptom disorder characterized by fatigue, joint pain, cognitive problems, and gastrointestinal complaints. The most prominent symptoms of GWI (memory problems, poor attention/concentration, chronic headaches, mood alterations, and impaired sleep) suggest that the disease primarily affects the CNS. Development of urgently needed treatments depends on experimental models appropriate for testing mechanistic hypotheses and for screening therapeutic compounds. Rodent models have been useful thus far, but are limited by their inability to assess the contribution of genetic or epigenetic background to the disease, and because disease-vulnerable proteins and pathways may be different in humans relative to rodents. As of yet, no postmortem tissue from the veterans has become available for research. We are moving forward with a paradigm shift in the study of GWI, which utilizes contemporary stem cell technology to convert somatic cells from Gulf War veterans into pluripotent cell lines that can be differentiated into various cell types, including neurons, glia, muscle, or other relevant cell types. Such cell lines are immortal and will be a resource for GWI researchers to pursue mechanistic hypotheses and therapeutics.


Assuntos
Reprogramação Celular , Guerra do Golfo , Neurônios , Síndrome do Golfo Pérsico/patologia , Síndrome do Golfo Pérsico/fisiopatologia , Veteranos , Animais , Córtex Cerebral , Humanos , Células-Tronco Pluripotentes Induzidas , Camundongos , Neurilema , Projetos de Pesquisa
4.
Neuro Oncol ; 18(9): 1278-87, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26951380

RESUMO

BACKGROUND: Higher-grade meningiomas (HGMs; World Health Organization grades II and III) pose a clinical problem due to high recurrence rates and the absence of effective therapy. Preclinical development of novel therapeutics requires a disease model that recapitulates the genotype and phenotype of patient HGM. Oncolytic herpes simplex virus (oHSV) has shown efficacy and safety in cancers in preclinical and clinical studies, but its utility for HGM has not been well characterized. METHODS: Tumorsphere cultures and serial orthotopic xenografting in immunodeficient mice were used to establish a patient-derived HGM model. The model was pathologically and molecularly characterized by immunohistochemistry, western blot, and genomic DNA sequencing and compared with the patient tumor. Anti-HGM effects of oHSV G47Δ were assessed using cell viability and virus replication assays in vitro and animal survival analysis following intralesional injections of G47Δ. RESULTS: We established a serially transplantable orthotopic malignant meningioma model, MN3, which was lethal within 3 months after tumorsphere implantation. MN3 xenografts exhibited the pathological hallmarks of malignant meningioma such as high Ki67 and vimentin expression. Both the patient tumor and xenografts were negative for neurofibromin 2 (merlin) and had the identical NF2 mutation. Oncolytic HSV G47Δ efficiently spread and killed MN3 cells, as well as other patient-derived HGM lines in vitro. Treatment with G47Δ significantly extended the survival of mice bearing subdural MN3 tumors. CONCLUSIONS: We established a new patient-derived meningioma model that will enable the study of targeted therapeutic approaches for HGM. Based on these studies, it is reasonable to consider a clinical trial of G47Δ for HGM.


Assuntos
Vetores Genéticos/administração & dosagem , Neoplasias Meníngeas/terapia , Meningioma/terapia , Terapia Viral Oncolítica , Simplexvirus/genética , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patologia , Meningioma/genética , Meningioma/patologia , Camundongos , Camundongos SCID , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
5.
PLoS One ; 10(10): e0140192, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26445504

RESUMO

The multi-subunit mammalian Mediator complex acts as an integrator of transcriptional regulation by RNA Polymerase II, and has emerged as a master coordinator of development and cell fate determination. We previously identified the Mediator subunit, MED28, as a cytosolic binding partner of merlin, the Neurofibromatosis 2 (NF2) tumor suppressor, and thus MED28 is distinct in having a cytosolic role as an NF2 interacting protein as well as a nuclear role as a Mediator complex subunit. Although limited in vitro studies have been performed on MED28, its in vivo function remains unknown. Employing a knockout mouse model, we describe for the first time the requirement for Med28 in the developing mouse embryo. Med28-deficiency causes peri-implantation lethality resulting from the loss of pluripotency of the inner cell mass accompanied by reduced expression of key pluripotency transcription factors Oct4 and Nanog. Further, overexpression of Med28 in mouse embryonic fibroblasts enhances the efficiency of their reprogramming to pluripotency. Cre-mediated inactivation of Med28 in induced pluripotent stem cells shows that Med28 is required for their survival. Intriguingly, heterozygous loss of Med28 results in differentiation of induced pluripotent stem cells into extraembryonic trophectoderm and primitive endoderm lineages. Our findings document the essential role of Med28 in the developing embryo as well as in acquisition and maintenance of pluripotency during reprogramming.


Assuntos
Implantação do Embrião , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes Induzidas/metabolismo , Complexo Mediador/metabolismo , Animais , Diferenciação Celular , Reprogramação Celular , Perda do Embrião/genética , Perda do Embrião/metabolismo , Deleção de Genes , Células-Tronco Pluripotentes Induzidas/citologia , Complexo Mediador/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Oncotarget ; 6(19): 16981-97, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26219339

RESUMO

Meningiomas are the most common primary intracranial adult tumor. All Neurofibromatosis 2 (NF2)-associated meningiomas and ~60% of sporadic meningiomas show loss of NF2 tumor suppressor protein. There are no effective medical therapies for progressive and recurrent meningiomas. Our previous work demonstrated aberrant activation of mTORC1 signaling that led to ongoing clinical trials with rapamycin analogs for NF2 and sporadic meningioma patients. Here we performed a high-throughput kinome screen to identify kinases responsible for mTORC1 pathway activation in NF2-deficient meningioma cells. Among the emerging top candidates were the mTORC2-specific target serum/glucocorticoid-regulated kinase 1 (SGK1) and p21-activated kinase 1 (PAK1). In NF2-deficient meningioma cells, inhibition of SGK1 rescues mTORC1 activation, and SGK1 activation is sensitive to dual mTORC1/2 inhibitor AZD2014, but not to rapamycin. PAK1 inhibition also leads to attenuated mTORC1 but not mTORC2 signaling, suggesting that mTORC2/SGK1 and Rac1/PAK1 pathways are independently responsible for mTORC1 activation in NF2-deficient meningiomas. Using CRISPR-Cas9 genome editing, we generated isogenic human arachnoidal cell lines (ACs), the origin cell type for meningiomas, expressing or lacking NF2. NF2-null CRISPR ACs recapitulate the signaling of NF2-deficient meningioma cells. Interestingly, we observe increased SGK1 transcription and protein expression in NF2-CRISPR ACs and in primary NF2-negative meningioma lines. Moreover, we demonstrate that the dual mTORC1/mTORC2 inhibitor, AZD2014 is superior to rapamycin and PAK inhibitor FRAX597 in blocking proliferation of meningioma cells. Importantly, AZD2014 is currently in use in several clinical trials of cancer. Therefore, we believe that AZD2014 may provide therapeutic advantage over rapalogs for recurrent and progressive meningiomas.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Neoplasias Meníngeas/enzimologia , Meningioma/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Immunoblotting , Neoplasias Meníngeas/genética , Meningioma/genética , Morfolinas/farmacologia , Neurofibromatose 2/genética , Reação em Cadeia da Polimerase , Pirimidinas , Transdução de Sinais/efeitos dos fármacos
7.
Mol Cancer Res ; 10(5): 649-59, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22426462

RESUMO

Inactivating mutations in the neurofibromatosis 2 (NF2) tumor suppressor gene results in the development of schwannomas and meningiomas. Using NF2-deficient meningioma cells and tumors, together with the normal cellular counterparts that meningiomas derive, arachnoid cells, we identified merlin as a novel negative regulator of mTOR complex 1 (mTORC1). We now show that merlin positively regulates the kinase activity of mTORC2, a second functionally distinct mTOR complex, and that downstream phosphorylation of mTORC2 substrates, including Akt, is reduced upon acute merlin deficiency in cells. In response to general growth factor stimulation, Akt signaling is attenuated in merlin RNA interference-suppressed human arachnoid and Schwann cells by mechanisms mediated by hyperactive mTORC1 and impaired mTORC2. Moreover, Akt signaling is impaired differentially in a cell type-dependent manner in response to distinct growth factor stimuli. However, contrary to activation of mTORC1, the attenuated mTORC2 signaling profiles exhibited by normal arachnoid and Schwann cells in response to acute merlin loss were not consistently reflected in NF2-deficient meningiomas and schwannomas, suggesting additional genetic events may have been acquired in tumors after initial merlin loss. This finding contrasts with another benign tumor disorder, tuberous sclerosis complex, which exhibits attenuated mTORC2 signaling profiles in both cells and tumors. Finally, we examined rapamycin, as well as the mTOR kinase inhibitor, Torin1, targeting both mTOR complexes to identify the most efficacious class of compounds for blocking mTOR-mediated signaling and proliferation in merlin-deficient meningioma cells. These studies may ultimately aid in the development of suitable therapeutics for NF2-associated tumors.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neurofibromina 2/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/metabolismo , Meningioma/genética , Meningioma/metabolismo , Complexos Multiproteicos , Naftiridinas/farmacologia , Neurilemoma/genética , Neurilemoma/metabolismo , Neurofibromina 2/deficiência , Neurofibromina 2/genética , Proteína Oncogênica v-akt/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Interferência de RNA , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Esclerose Tuberosa/genética , Esclerose Tuberosa/metabolismo
8.
Mol Cell Biol ; 29(21): 5923-40, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19703993

RESUMO

Meningiomas, one of the most common human brain tumors, are derived from arachnoidal cells associated with brain meninges, are usually benign, and are frequently associated with neurofibromatosis type 2. Here, we define a typical human meningioma microRNA (miRNA) profile and characterize the effects of one downregulated miRNA, miR-200a, on tumor growth. Elevated levels of miR-200a inhibited meningioma cell growth in culture and in a tumor model in vivo. Upregulation of miR-200a decreased the expression of transcription factors ZEB1 and SIP1, with consequent increased expression of E-cadherin, an adhesion protein associated with cell differentiation. Downregulation of miR-200a in meningiomas and arachnoidal cells resulted in increased expression of beta-catenin and cyclin D1 involved in cell proliferation. miR-200a was found to directly target beta-catenin mRNA, thereby inhibiting its translation and blocking Wnt/beta-catenin signaling, which is frequently involved in cancer. A direct correlation was found between the downregulation of miR-200a and the upregulation of beta-catenin in human meningioma samples. Thus, miR-200a appears to act as a multifunctional tumor suppressor miRNA in meningiomas through effects on the E-cadherin and Wnt/beta-catenin signaling pathways. This reveals a previously unrecognized signaling cascade involved in meningioma tumor development and highlights a novel molecular interaction between miR-200a and Wnt signaling, thereby providing insights into novel therapies for meningiomas.


Assuntos
Caderinas/metabolismo , Regulação para Baixo/genética , Neoplasias Meníngeas/genética , Meningioma/genética , MicroRNAs/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Apoptose , Sequência de Bases , Proliferação de Células , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Hibridização Genômica Comparativa , Ciclina D1/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Meníngeas/patologia , Meningioma/patologia , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas , Regulação para Cima/genética , beta Catenina/genética
9.
Mol Cell Biol ; 29(15): 4250-61, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19451225

RESUMO

Inactivating mutations of the neurofibromatosis 2 (NF2) gene, NF2, result predominantly in benign neurological tumors, schwannomas and meningiomas, in humans; however, mutations in murine Nf2 lead to a broad spectrum of cancerous tumors. The tumor-suppressive function of the NF2 protein, merlin, a membrane-cytoskeleton linker, remains unclear. Here, we identify the mammalian target of rapamycin complex 1 (mTORC1) as a novel mediator of merlin's tumor suppressor activity. Merlin-deficient human meningioma cells and merlin knockdown arachnoidal cells, the nonneoplastic cell counterparts of meningiomas, exhibit rapamycin-sensitive constitutive mTORC1 activation and increased growth. NF2 patient tumors and Nf2-deficient mouse embryonic fibroblasts demonstrate elevated mTORC1 signaling. Conversely, the exogenous expression of wild-type merlin isoforms, but not a patient-derived L64P mutant, suppresses mTORC1 signaling. Merlin does not regulate mTORC1 via the established mechanism of phosphoinositide 3-kinase-Akt or mitogen-activated protein kinase/extracellular signal-regulated kinase-mediated TSC2 inactivation and may instead regulate TSC/mTOR signaling in a novel fashion. In conclusion, the deregulation of mTORC1 activation underlies the aberrant growth and proliferation of NF2-associated tumors and may restrain the growth of these lesions through negative feedback mechanisms, suggesting that rapamycin in combination with phosphoinositide 3-kinase inhibitors may be therapeutic for NF2.


Assuntos
Proliferação de Células , Neurofibromina 2/metabolismo , Fatores de Transcrição/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Aracnoide-Máter/citologia , Aracnoide-Máter/efeitos dos fármacos , Aracnoide-Máter/metabolismo , Ciclo Celular , Linhagem Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Alvo Mecanístico do Complexo 1 de Rapamicina , Meningioma/genética , Meningioma/metabolismo , Meningioma/patologia , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Neurilemoma/genética , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurofibromina 2/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas
10.
Neurobiol Dis ; 29(2): 278-92, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17962031

RESUMO

Meningiomas, common tumors arising from arachnoidal cells of the meninges, may occur sporadically, or in association with the inherited disorder, neurofibromatosis 2 (NF2). Most sporadic meningiomas result from NF2 inactivation, resulting in loss of tumor suppressor merlin, implicated in regulating membrane-cytoskeletal organization. To investigate merlin function in an authentic target cell type for NF2 tumor formation, we established primary cultures from genetically-matched meningioma and normal arachnoidal tissues. Our studies revealed novel and distinct cell biological and biochemical properties unique to merlin-deficient meningioma cells compared to merlin-expressing arachnoidal and meningioma cells, and other NF2-deficient cell types. Merlin-deficient meningioma cells displayed cytoskeletal and cell contact defects, altered cell morphology and growth properties, most notably cell senescence, implicating the activation of senescence pathways in limiting benign meningioma growth. Merlin suppression by RNAi in arachnoidal cells replicated merlin-deficient meningioma features, thus establishing these cell systems as disease-relevant models for studying NF2 tumorigenesis.


Assuntos
Aracnoide-Máter/citologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes da Neurofibromatose 2/fisiologia , Neoplasias Meníngeas/patologia , Meningioma/patologia , RNA Interferente Pequeno/farmacologia , Actinas/metabolismo , Aracnoide-Máter/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Cateninas/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Membrana/metabolismo , Mutação , Proteínas de Neoplasias/metabolismo , Neurofibromina 2/deficiência
11.
Proc Natl Acad Sci U S A ; 102(24): 8573-8, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15937108

RESUMO

Loss-of-function mutations in the NF1 tumor suppressor gene underlie the familial cancer syndrome neurofibromatosis type I (NF1). The NF1-encoded protein, neurofibromin, functions as a Ras-GTPase activating protein (RasGAP). Accordingly, deregulation of Ras is thought to contribute to NF1 development. However, the critical effector pathways involved in disease pathogenesis are still unknown. We show here that the mTOR pathway is tightly regulated by neurofibromin. mTOR is constitutively activated in both NF1-deficient primary cells and human tumors in the absence of growth factors. This aberrant activation depends on Ras and PI3 kinase, and is mediated by the phosphorylation and inactivation of the TSC2-encoded protein tuberin by AKT. Importantly, tumor cell lines derived from NF1 patients, and a genetically engineered cell system that requires Nf1-deficiency for transformation, are highly sensitive to the mTOR inhibitor rapamycin. Furthermore, while we show that the activation of endogenous Ras leads to constitutive mTOR signaling in this disease state, we also demonstrate that in normal cells Ras is differentially required for mTOR signaling in response to various growth factors. Thus, these findings identify the NF1 tumor suppressor as an indispensable regulator of TSC2 and mTOR. Furthermore, our results also demonstrate that Ras plays a critical role in the activation of mTOR in both normal and tumorigenic settings. Finally, these data suggest that rapamycin, or its derivatives, may represent a viable therapy for NF1.


Assuntos
Neurofibromatose 1/metabolismo , Neurofibromina 1/metabolismo , Proteínas Quinases/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Vetores Genéticos , Imunoprecipitação , Camundongos , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR , Transfecção , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas ras/metabolismo
12.
J Biol Chem ; 280(13): 12517-22, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15699051

RESUMO

The function of the NF2 tumor suppressor merlin has remained elusive despite increasing evidence for its role in actin cytoskeleton reorganization. The closely related ERM proteins (ezrin, radixin, and moesin) act as linkers between the cell membrane and cytoskeleton, and have also been implicated as active actin reorganizers. We report here that merlin and the ERMs can interact with and regulate N-WASP, a critical regulator of actin dynamics. Merlin and moesin were found to inhibit N-WASP-mediated actin assembly in vitro, a function that appears independent of their ability to bind actin. Furthermore, exogenous expression of a constitutively active ERM inhibits N-WASP-dependent Shigella tail formation, suggesting that the ERMs may function as inhibitors of N-WASP function in vivo. This novel function of merlin and the ERMs illustrates a mechanism by which these proteins directly exert their effects on actin reorganization and also provides new insight into N-WASP regulation.


Assuntos
Actinas/química , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/química , Neurofibromina 2/química , Actinas/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Proteínas Sanguíneas/metabolismo , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Camundongos , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Neurofibromina 2/metabolismo , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Shigella/metabolismo , Transdução de Sinais , Fatores de Tempo , Proteína Neuronal da Síndrome de Wiskott-Aldrich
13.
J Cell Sci ; 117(Pt 14): 2951-61, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15161943

RESUMO

The Na(+)/H(+) exchanger regulatory factor, NHERF, is a multifunctional adapter protein involved in a wide range of physiological activities. NHERF associates with merlin and the ezrin/radixin/moesin (MERM) family of membrane-actin cytoskeletal linker proteins through its C-terminus and is capable of interacting via its PDZ1 domain to the betaPDGF receptor (betaPDGFR). Thus, NHERF, potentially links the betaPDGFR to the actin cytoskeleton through its interaction with MERM proteins. In the present study, we have examined whether abolishing the interaction of betaPDGFR with NHERF results in actin cytoskeletal rearrangements. We have stably expressed a wild-type betaPDGFR, a mutant betaPDGFR (L1106A) that is incapable of interacting with NHERF, as well as a kinase defective mutant receptor (K634R), in PDGFR-deficient mouse embryonic fibroblasts. Our observations indicate that cells expressing betaPDGFR (L1106A) were impaired in their ability to spread and migrate on fibronectin compared with wild-type and K634R cells. L1106A mutant cells also revealed an increased number of focal adhesions, a condensed F-actin ring at the cell periphery and a decrease in total focal adhesion kinase (FAK) tyrosine phosphorylation. Further, we show that NHERF and MERM proteins could act as intermediary bridging proteins between betaPDGFR and FAK. Thus, the interaction of betaPDGFR with NHERF may provide an essential link between the cell membrane and the cortical actin cytoskeleton independent of receptor activity.


Assuntos
Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Actinas/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Forma Celular/fisiologia , Adesões Focais/metabolismo , Humanos , Camundongos , Mutação , Neurofibromina 2/metabolismo , Fosforilação , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Trocadores de Sódio-Hidrogênio , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...