Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Matrix Biol ; 67: 1-14, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29519758

RESUMO

Microfibril-associated glycoprotein-1 (MAGP1) is an extracellular matrix protein that interacts with fibrillin and is involved in regulating the bioavailability of signaling molecules such as TGFß. Mice with germline MAGP1 deficiency (Mfap2-/-) develop increased adiposity, hyperglycemia, insulin resistance, bone marrow adipose tissue expansion, reduced cancellous bone mass, cortical bone thinning and bone fragility. The goal of this study was to assess whether the Mfap2-/- bone phenotypes were due to loss of MAGP1 locally or secondary to a change in whole body physiology (metabolic dysfunction). To do this, mice with conditional deletion of MAGP1 in the limb skeleton were generated by crossing MAGP1-flox mice (Mfap2lox/lox) with Prx1-Cre mice. Mfap2Prx-/- mice did not show any changes in peripheral adiposity, hyperglycemia or insulin sensitivity, but did have increased bone length and cancellous bone loss that was comparable to the germline Mfap2-/- knockout. Unlike the germline knockout, marrow adiposity, cortical bone thickness and bone strength in Mfap2Prx-/- mice were normal. These findings implicate systemic metabolic dysfunction in the development of bone fragility in germline Mfap2-/- mice. An unexpected finding of this study was the detection of MAGP1 protein in the Mfap2Prx-/- hematopoietic bone marrow, despite the absence of MAGP1 protein in osseous bone matrix and absent Mfap2 transcript expression at both sites. This suggests MAGP1 from a secondary site may accumulate in the bone marrow, but not be incorporated into the bone matrix, during times of regional MAGP1 depletion.


Assuntos
Osso e Ossos/patologia , Proteínas Contráteis/deficiência , Proteínas da Matriz Extracelular/deficiência , Proteínas de Homeodomínio/metabolismo , Doenças Metabólicas/genética , Adipócitos/metabolismo , Animais , Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Modelos Animais de Doenças , Mutação em Linhagem Germinativa , Proteínas de Homeodomínio/genética , Doenças Metabólicas/metabolismo , Camundongos , Fatores de Processamento de RNA , Transdução de Sinais
2.
Diabetes ; 63(6): 1920-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24458361

RESUMO

Microfibril-associated glycoprotein 1 (MAGP1) is a component of extracellular matrix microfibrils. Here we show that MAGP1 expression is significantly altered in obese humans, and inactivation of the MAGP1 gene (Mfap2(-/-)) in mice results in adipocyte hypertrophy and predisposition to metabolic dysfunction. Impaired thermoregulation was evident in Mfap2(-/-) mice prior to changes in adiposity, suggesting a causative role for MAGP1 in the increased adiposity and predisposition to diabetes. By 5 weeks of age, Mfap2(-/-) mice were maladaptive to cold challenge, uncoupling protein-1 expression was attenuated in the brown adipose tissue, and there was reduced browning of the subcutaneous white adipose tissue. Levels of transforming growth factor-ß (TGF-ß) activity were elevated in Mfap2(-/-) adipose tissue, and the treatment of Mfap2(-/-) mice with a TGF-ß-neutralizing antibody improved their body temperature and prevented the increased adiposity phenotype. Together, these findings indicate that the regulation of TGF-ß by MAGP1 is protective against the effects of metabolic stress, and its absence predisposes individuals to metabolic dysfunction.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Proteínas Contráteis/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Obesidade/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteínas Contráteis/deficiência , Diabetes Mellitus Tipo 2/genética , Proteínas da Matriz Extracelular/deficiência , Pleiotropia Genética , Predisposição Genética para Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Fenótipo , Fatores de Processamento de RNA , Transdução de Sinais , Termogênese/genética
3.
Invest Ophthalmol Vis Sci ; 54(12): 7163-73, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24130178

RESUMO

PURPOSE: Fibrillin-2 (Fbn2) is the dominant fibrillin isoform expressed during development of the mouse eye. To test its role in morphogenesis, we examined the ocular phenotype of Fbn2(-/-) mice. METHODS: Ocular morphology was assessed by confocal microscopy using antibodies against microfibril components. RESULTS: Fbn2(-/-) mice had a high incidence of anterior segment dysgenesis. The iris was the most commonly affected tissue. Complete iridal coloboma was present in 37% of eyes. Dyscoria, corectopia and pseudopolycoria were also common (43% combined incidence). In wild-type (WT) mice, fibrillin-2-rich microfibrils are prominent in the pupillary membrane (PM) during development. In Fbn2-null mice, the absence of Fbn2 was partially compensated for by increased expression of fibrillin-1, although the resulting PM microfibrils were disorganized, compared with WTs. In colobomatous adult Fbn2(-/-) eyes, the PM failed to regress normally, especially beneath the notched region of the iris. Segments of the ciliary body were hypoplastic, and zonular fibers, although relatively plentiful, were unevenly distributed around the lens equator. In regions where the zonular fibers were particularly disturbed, the synchronous differentiation of the underlying lens fiber cells was affected. CONCLUSIONS: Fbn2 has an indispensable role in ocular morphogenesis in mice. The high incidence of iris coloboma in Fbn2-null animals implies a previously unsuspected role in optic fissure closure. The observation that fiber cell differentiation was disturbed in Fbn2(-/-) mice raises the possibility that the attachment of zonular fibers to the lens surface may help specify the equatorial margin of the lens epithelium.


Assuntos
Coloboma/patologia , Iris/anormalidades , Proteínas dos Microfilamentos/deficiência , Animais , Proteínas Contráteis/deficiência , Proteínas Contráteis/fisiologia , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Camundongos , Camundongos Endogâmicos C57BL , Microfibrilas/patologia , Proteínas dos Microfilamentos/fisiologia , Fenótipo , Distúrbios Pupilares/etiologia
4.
J Biol Chem ; 288(40): 28869-80, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23963447

RESUMO

Microfibril-associated glycoprotein (MAGP) 1 and 2 are evolutionarily related but structurally divergent proteins that are components of microfibrils of the extracellular matrix. Using mice with a targeted inactivation of Mfap5, the gene for MAGP2 protein, we demonstrate that MAGPs have shared as well as unique functions in vivo. Mfap5(-/-) mice appear grossly normal, are fertile, and have no reduction in life span. Cardiopulmonary development is typical. The animals are normotensive and have vascular compliance comparable with age-matched wild-type mice, which is indicative of normal, functional elastic fibers. Loss of MAGP2 alone does not significantly alter bone mass or architecture, and loss of MAGP2 in tandem with loss of MAGP1 does not exacerbate MAGP1-dependent osteopenia. MAGP2-deficient mice are neutropenic, which contrasts with monocytopenia described in MAGP1-deficient animals. This suggests that MAGP1 and MAGP2 have discrete functions in hematopoiesis. In the cardiovascular system, MAGP1;MAGP2 double knockout mice (Mfap2(-/-);Mfap5(-/-)) show age-dependent aortic dilation. These findings indicate that MAGPs have shared primary functions in maintaining large vessel integrity. In solid phase binding assays, MAGP2 binds active TGFß1, TGFß2, and BMP2. Together, these data demonstrate that loss of MAGP2 expression in vivo has pleiotropic effects potentially related to the ability of MAGP2 to regulate growth factors or participate in cell signaling.


Assuntos
Proteínas Contráteis/deficiência , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/metabolismo , Pleiotropia Genética , Alelos , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Densidade Óssea , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Movimento Celular , Proteínas Contráteis/química , Éxons/genética , Proteínas da Matriz Extracelular/química , Marcação de Genes , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neutropenia/metabolismo , Neutropenia/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Tamanho do Órgão , Ligação Proteica , Fatores de Processamento de RNA , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
5.
Arch Dis Child ; 98(2): 136-40, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23220206

RESUMO

Pulmonary air leaks in children are most commonly due to infection or barotrauma. While cases of severe barotrauma are falling because of advances in neonatal care, the incidence of necrotising pneumonia is rising. The majority of air leaks can be managed conservatively, but more severe cases pose a significant challenge to the clinician. The use of occlusive endobronchial balloons is an established anaesthetic technique for a number of indications, but is not widely used in children. We conducted a review over a 12-year period, and report six cases of complex air leaks in which balloon occlusion was used. Balloon occlusion was successful in both cases of bronchopleural fistulae (secondary to severe necrotising pneumonia) and half of the cases of intrapulmonary air leak (due to barotrauma). In the other two cases (due to barotrauma and filamin A deficiency), it was transiently effective. No serious adverse effects or complications were encountered. In selected cases, endobronchial balloons are a useful adjunct in the management of life-threatening bronchopleural fistulae and cystic lung disease. The procedure is non-operative, minimally invasive and reversible. With the increasing incidence of bronchopleural fistulae, this may become an increasingly important therapy.


Assuntos
Oclusão com Balão/métodos , Barotrauma/terapia , Fístula Brônquica/terapia , Proteínas Contráteis/deficiência , Proteínas dos Microfilamentos/deficiência , Doenças Pleurais/terapia , Doenças Respiratórias/terapia , Colúmbia Britânica , Criança , Pré-Escolar , Feminino , Filaminas , Humanos , Lactente , Masculino , Pneumotórax/terapia , Doenças da Traqueia/terapia
6.
J Neurosci ; 32(22): 7672-84, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649246

RESUMO

Cytoskeleton-associated proteins play key roles not only in regulating cell morphology and migration but also in proliferation. Mutations in the cytoskeleton-associated gene filamin A (FlnA) cause the human disorder periventricular heterotopia (PH). PH is a disorder of neural stem cell development that is characterized by disruption of progenitors along the ventricular epithelium and subsequent formation of ectopic neuronal nodules. FlnA-dependent regulation of cytoskeletal dynamics is thought to direct neural progenitor migration and proliferation. Here we show that embryonic FlnA-null mice exhibited a reduction in brain size and decline in neural progenitor numbers over time. The drop in the progenitor population was not attributable to cell death or changes in premature differentiation, but to prolonged cell cycle duration. Suppression of FlnA led to prolongation of the entire cell cycle length, principally in M phase. FlnA loss impaired degradation of cyclin B1-related proteins, thereby delaying the onset and progression through mitosis. We found that the cdk1 kinase Wee1 bound FlnA, demonstrated increased expression levels after loss of FlnA function, and was associated with increased phosphorylation of cdk1. Phosphorylation of cdk1 inhibited activation of the anaphase promoting complex degradation system, which was responsible for cyclin B1 degradation and progression through mitosis. Collectively, our results demonstrate a molecular mechanism whereby FlnA loss impaired G2 to M phase entry, leading to cell cycle prolongation, compromised neural progenitor proliferation, and reduced brain size.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Córtex Cerebral/fisiologia , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Proteína Quinase CDC2/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Proteínas Contráteis/deficiência , Ciclina B1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Filaminas , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica/genética , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67 , Camundongos , Camundongos Transgênicos , Microcefalia/genética , Proteínas dos Microfilamentos/deficiência , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/patologia , Fosforilação/genética , Proteínas com Domínio T/metabolismo , Tirosina/metabolismo
7.
J Cell Sci ; 125(Pt 16): 3858-69, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595522

RESUMO

Filamins are an important family of actin-binding proteins that, in addition to bundling actin filaments, link cell surface adhesion proteins, signaling receptors and channels to the actin cytoskeleton, and serve as scaffolds for an array of intracellular signaling proteins. Filamins are known to regulate the actin cytoskeleton, act as mechanosensors that modulate tissue responses to matrix density, control cell motility and inhibit activation of integrin adhesion receptors. In this study, we extend the repertoire of filamin activities to include control of extracellular matrix (ECM) degradation. We show that knockdown of filamin increases matrix metalloproteinase (MMP) activity and induces MMP2 activation, enhancing the ability of cells to remodel the ECM and increasing their invasive potential, without significantly altering two-dimensional random cell migration. We further show that within filamin A, the actin-binding domain is necessary, but not sufficient, to suppress the ECM degradation seen in filamin-A-knockdown cells and that dimerization and integrin binding are not required. Filamin mutations are associated with neuronal migration disorders and a range of congenital malformations characterized by skeletal dysplasia and various combinations of cardiac, craniofacial and intestinal anomalies. Furthermore, in breast cancers loss of filamin A has been correlated with increased metastatic potential. Our data suggest that effects on ECM remodeling and cell invasion should be considered when attempting to provide cellular explanations for the physiological and pathological effects of altered filamin expression or filamin mutations.


Assuntos
Proteínas Contráteis/metabolismo , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Metaloproteinase 2 da Matriz/metabolismo , Proteínas dos Microfilamentos/metabolismo , Actinas/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas Contráteis/deficiência , Proteínas Contráteis/genética , Ativação Enzimática , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrossarcoma/enzimologia , Fibrossarcoma/genética , Filaminas , Técnicas de Silenciamento de Genes , Humanos , Integrinas/metabolismo , Metaloproteinase 14 da Matriz , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Invasividade Neoplásica , Fenótipo , Estrutura Terciária de Proteína
8.
J Cell Biochem ; 113(1): 93-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898536

RESUMO

Microfibril-associated glycoprotein-1 (MAGP1), together with the fibrillins, are constitutive components of vertebrate microfibrils. Mice deficient in MAGP1 (murine MAGP1 knockout animals (Mfap2(-/-)); MAGP1Δ) is appropriate develop progressive osteopenia and reduced whole bone strength, and have elevated numbers of osteoclasts lining the bone surface. Our previous studies suggested that the increased osteoclast population was associated with elevated levels of receptor activator of NF-κB ligand (RANKL), a positive regulator of osteoclast differentiation. To explore the relationship between RANKL expression and osteoclast differentiation in MAGP1 deficiency, oophorectomy (OVX) was used to stimulate RANKL expression in both WT and MAGP1Δ animals. Bone loss following OVX was monitored using whole body DEXA and in vivo µCT. While WT mice exhibited significant bone loss following OVX, percent bone loss was reduced in MAGP1Δ mice. Further, serum RANKL levels rose significantly in OVX WT mice, whereas, there was only a modest increase in RANKL following OVX in the mutant mice due to already high baseline levels. Elevated RANKL expression was normalized when cultured MAGP1Δ osteoblasts were treated with a neutralizing antibody targeting free TGFß. These studies provide support for increased RANKL expression associated with MAGP1 deficiency and provide a link to altered TGF-ß signaling as a possible causative signaling pathway regulating RANKL expression in MAGP1Δ osteoblasts.


Assuntos
Reabsorção Óssea/metabolismo , Proteínas Contráteis/deficiência , Proteínas da Matriz Extracelular/deficiência , Ligante RANK/metabolismo , Animais , Doenças Ósseas Metabólicas/metabolismo , Reabsorção Óssea/genética , Diferenciação Celular , Células Cultivadas , Proteínas Contráteis/genética , Proteínas da Matriz Extracelular/genética , Feminino , Camundongos , Camundongos Knockout , Microfibrilas/fisiologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Ovariectomia , Fatores de Processamento de RNA , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
9.
DNA Repair (Amst) ; 11(2): 192-200, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22051193

RESUMO

Filamin-A, also called actin binding protein 280 (ABP-280), cross-links the actin filaments into dynamic orthogonal network to serve as scaffolds in multiple signaling pathways. It has been reported that filamin-A interacts with DNA damage response proteins BRCA1 and BRCA2. Defects of filamin-A impair the repair of DNA double strand breaks (DSBs), resulting in sensitization of cells to ionizing radiation. In this study, we sought to test the hypothesis that filamin-A can be used as a target for cancer chemotherapy and as a biomarker to predict cancer response to therapeutic DNA damage. We found that reduction of filamin-A sensitizes cancer cells to chemotherapy reagents bleomycin and cisplatin, delays the repair of not only DSBs but also single strand breaks (SSBs) and interstrand crosslinks (ICLs), and increases chromosome breaks after the drug treatment. By treating a panel of human melanoma cell lines with variable filamin-A expression, we observed a correlation between expression level of filamin-A protein and drug IC(50). We further inhibited the expression of filamin-A in melanoma cells, and found that this confers an increased sensitivity to bleomycin and cisplatin treatment in a mouse xenograft tumor model. These results suggest that filamin-A plays a role in repair of a variety of DNA damage, that lack of filamin-A is a prognostic marker for a better outcome after DNA damage based treatment, and filamin-A can be inhibited to sensitize filamin-A positive cancer cells to therapeutic DNA damage. Thus filamin-A can be used as a biomarker and a target for DNA damage based cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Proteínas Contráteis/metabolismo , Dano ao DNA , Melanoma/patologia , Proteínas dos Microfilamentos/metabolismo , Terapia de Alvo Molecular/métodos , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/deficiência , Bleomicina/farmacologia , Bleomicina/uso terapêutico , Linhagem Celular Tumoral , Instabilidade Cromossômica/efeitos dos fármacos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas Contráteis/deficiência , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Filaminas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Camundongos , Proteínas dos Microfilamentos/deficiência , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Bone Miner Res ; 25(5): 1077-91, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19929439

RESUMO

Osteoclastogenesis (OCG) results from the fusion of monocytes after stimulation with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-kappaB ligand (RANKL). Migration of monocytes into close proximity precedes critical fusion events that are required for osteoclast formation. Cellular migration requires leading-edge actin cytoskeleton assembly that drives cellular locomotion. Filamin A (FLNa) cross-links F-actin filaments in the leading edge of migrating cells and also has been shown to regulate signal transduction during cell migration. However, little is known about the possible role of FLNa in osteoclastogenesis. Our objective in this study was to investigate the role of FLNa in osteoclastogenesis. Bone marrow monocytes isolated from the tibiae and femora of wild type (WT) and Flna-null mice were cultured for 6 days with M-CSF and RANKL, and osteoclasts were identified by tartrate-resistant acid phosphatase (TRACP) staining. The Flna-null mouse skeletal phenotype was characterized using dual-energy X-ray absorptiometry (DXA) to analyze the skeleton, as well as tests on blood chemistry. Osteoclast levels in vivo were quantified by counting of TRACP-stained histologic sections of distal femora. To elucidate the mechanisms by which Flna regulates osteoclastogenesis, migration, actin polymerization, and activation of Rho GTPases, Rac1, Cdc42, and RhoA were assessed in monocytes during in vitro OCG. Deficiencies in migration were rescued using constitutively active Rac1 and Cdc42 TAT fusion proteins. The RANKL signaling pathway was evaluated for activation by monitoring nuclear translocation of NF kappaB and c-jun and expression of key osteoclast genes using quantitative real-time polymerase chain reaction (qRT-PCR). Our results show that Flna-null monocytes formed fewer osteoclasts in vitro, and those that were formed were smaller with fewer nuclei. Decreased OCG was reflected in vivo in TRACP-stained histologic bone sections. Flna-null monocytes experienced impaired migratory ability. When OCG was performed at increasing starting cellular plating densities in order to decrease intercellular distances, there was progressive rescue of Flna-null osteoclast formation comparable with WT levels, confirming that Flna regulates monocyte migration prefusion. Activation of the actin cytoskeleton regulators Rac1, Cdc42, and RhoA and actin free-barbed end generation were partially or completely abrogated in Flna-null monocytes; however, monocyte migration was restored on rescuing with constitutively active Rac1 and Cdc42 TAT fusion proteins. We conclude that filamin A is required for osteoclastogenesis by regulating actin dynamics via Rho GTPases that control monocyte migration.


Assuntos
Actinas/metabolismo , Movimento Celular/efeitos dos fármacos , Proteínas Contráteis/fisiologia , Proteínas dos Microfilamentos/fisiologia , Monócitos/citologia , Osteoclastos/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Animais , Movimento Celular/fisiologia , Proteínas Contráteis/deficiência , Filaminas , Camundongos , Proteínas dos Microfilamentos/deficiência , Osteoclastos/citologia , Ligante RANK/fisiologia , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
11.
Cell Death Differ ; 16(6): 921-32, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19300455

RESUMO

Ubiquitin-mediated protein degradation is the main mechanism for controlled proteolysis, which is crucial for muscle development and maintenance. The ankyrin repeat-containing protein with a suppressor of cytokine signaling box 2 gene (ASB2) encodes the specificity subunit of an E3 ubiquitin ligase complex involved in differentiation of hematopoietic cells. Here, we provide the first evidence that a novel ASB2 isoform, ASB2beta, is important for muscle differentiation. ASB2beta is expressed in muscle cells during embryogenesis and in adult tissues. ASB2beta is part of an active E3 ubiquitin ligase complex and targets the actin-binding protein filamin B (FLNb) for proteasomal degradation. Thus, ASB2beta regulates FLNb functions by controlling its degradation. Knockdown of endogenous ASB2beta by shRNAs during induced differentiation of C2C12 cells delayed FLNb degradation as well as myoblast fusion and expression of muscle contractile proteins. Finally, knockdown of FLNb in ASB2beta knockdown cells restores myogenic differentiation. Altogether, our results suggest that ASB2beta is involved in muscle differentiation through the targeting of FLNb to destruction by the proteasome.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular , Galinhas , Proteínas Contráteis/deficiência , Filaminas , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Proteínas dos Microfilamentos/deficiência , Interferência de RNA , RNA Mensageiro/metabolismo , Proteínas Supressoras da Sinalização de Citocina/deficiência , Proteínas Supressoras da Sinalização de Citocina/genética
12.
J Immunol ; 180(6): 3938-45, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322202

RESUMO

Filamin A, or actin-binding protein 280, is a ubiquitously expressed cytosolic protein that interacts with intracellular domains of multiple receptors to control their subcellular distribution, and signaling capacity. In this study, we document interaction between FcgammaRI, a high-affinity IgG receptor, and filamin A by yeast two-hybrid techniques and coimmunoprecipitation. Both proteins colocalized at the plasma membrane in monocytes, but dissociated upon FcgammaRI triggering. The filamin-deficient cell line M2 and a filamin-reconstituted M2 subclone (A7), were used to further study FcgammaRI-filamin interactions. FcgammaRI transfection in A7 cells with filamin resulted in high plasma membrane expression levels. In filamin-deficient M2 cells and in filamin RNA-interference studies, FcgammaRI surface expression was consistently reduced. FcgammaRI localized to LAMP-1-positive vesicles in the absence of filamin as shown by confocal microscopy indicative for lysosomal localization. Mouse IgG2a capture experiments suggested a transient membrane expression of FcgammaRI before being transported to the lysosomes. These data support a pivotal role for filamin in FcgammaRI surface expression via retention of FcgammaRI from a default lysosomal pathway.


Assuntos
Proteínas Contráteis/fisiologia , Lisossomos/metabolismo , Proteínas dos Microfilamentos/fisiologia , Receptores de IgG/biossíntese , Receptores de IgG/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Células Clonais , Proteínas Contráteis/deficiência , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Filaminas , Humanos , Lisossomos/genética , Lisossomos/imunologia , Camundongos , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Transporte Proteico/genética , Transporte Proteico/imunologia , Receptores de IgG/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Frações Subcelulares/imunologia , Frações Subcelulares/metabolismo , Células U937
13.
Blood ; 111(8): 4137-44, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281502

RESUMO

Mice lacking the extracellular matrix protein microfibril-associated glycoprotein-1 (MAGP1) display delayed thrombotic occlusion of the carotid artery following injury as well as prolonged bleeding from a tail vein incision. Normal occlusion times were restored when recombinant MAGP1 was infused into deficient animals prior to vessel wounding. Blood coagulation was normal in these animals as assessed by activated partial thromboplastin time and prothrombin time. Platelet number was lower in MAGP1-deficient mice, but the platelets showed normal aggregation properties in response to various agonists. MAGP1 was not found in normal platelets or in the plasma of wild-type mice. In ligand blot assays, MAGP1 bound to fibronectin, fibrinogen, and von Willebrand factor, but von Willebrand factor was the only protein of the 3 that bound to MAGP1 in surface plasmon resonance studies. These findings show that MAGP1, a component of microfibrils and vascular elastic fibers, plays a role in hemostasis and thrombosis.


Assuntos
Artérias Carótidas/patologia , Proteínas Contráteis/deficiência , Proteínas da Matriz Extracelular/deficiência , Trombose/patologia , Animais , Tempo de Sangramento , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/fisiopatologia , Bovinos , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrinogênio/metabolismo , Fibronectinas/metabolismo , Humanos , Imuno-Histoquímica , Injeções , Camundongos , Camundongos Endogâmicos C57BL , Testes de Função Plaquetária , Ligação Proteica/efeitos dos fármacos , Fatores de Processamento de RNA , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Ressonância de Plasmônio de Superfície , Fator de von Willebrand/metabolismo
14.
Hum Mol Genet ; 17(5): 631-41, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17635842

RESUMO

Spondylocarpotarsal synostosis syndrome (SCT) is an autosomal recessive disease that is characterized by short stature, and fusions of the vertebrae and carpal and tarsal bones. SCT results from homozygosity or compound heterozygosity for nonsense mutations in FLNB. FLNB encodes filamin B, a multifunctional cytoplasmic protein that plays a critical role in skeletal development. Protein extracts derived from cells of SCT patients with nonsense mutations in FLNB did not contain filamin B, demonstrating that SCT results from absence of filamin B. To understand the role of filamin B in skeletal development, an Flnb-/- mouse model was generated. The Flnb-/- mice were phenotypically similar to individuals with SCT as they exhibited short stature and similar skeletal abnormalities. Newborn Flnb-/- mice had fusions between the neural arches of the vertebrae in the cervical and thoracic spine. At postnatal day 60, the vertebral fusions were more widespread and involved the vertebral bodies as well as the neural arches. In addition, fusions were seen in sternum and carpal bones. Analysis of the Flnb-/- mice phenotype showed that an absence of filamin B causes progressive vertebral fusions, which is contrary to the previous hypothesis that SCT results from failure of normal spinal segmentation. These findings suggest that spinal segmentation can occur normally in the absence of filamin B, but the protein is required for maintenance of intervertebral, carpal and sternal joints, and the joint fusion process commences antenatally.


Assuntos
Anormalidades Múltiplas/genética , Proteínas Contráteis/genética , Proteínas dos Microfilamentos/genética , Mutação , Osteocondrodisplasias/genética , Sinostose/genética , Animais , Animais Recém-Nascidos , Tornozelo/anormalidades , Códon sem Sentido , Proteínas Contráteis/química , Proteínas Contráteis/deficiência , Cruzamentos Genéticos , Dimerização , Modelos Animais de Doenças , Embrião de Mamíferos , Filaminas , Regulação da Expressão Gênica no Desenvolvimento , Genes Recessivos , Heterozigoto , Homozigoto , Humanos , Metacarpo/anormalidades , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/deficiência , Modelos Biológicos , Modelos Genéticos , Peso Molecular , Fenótipo , Estrutura Terciária de Proteína , Coluna Vertebral/anormalidades , Síndrome
15.
Brain Res ; 1177: 9-18, 2007 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17897634

RESUMO

We investigated the effects of morphine and other agonists on the human mu opioid receptor (MOP) expressed in M2 melanoma cells, lacking the actin cytoskeleton protein filamin A and in A7, a subclone of the M2 melanoma cells, stably transfected with filamin A cDNA. The results of binding experiments showed that after chronic morphine treatment (24 h) of A7 cells, MOP-binding sites were down-regulated to 63% of control, whereas, unexpectedly, in M2 cells, MOP binding was up-regulated to 188% of control naive cells. Similar up-regulation was observed with the agonists methadone and levorphanol. The presence of antagonists (naloxone or CTAP) during chronic morphine treatment inhibited MOP down-regulation in A7 cells. In contrast, morphine-induced up-regulation of MOP in M2 cells was further increased by these antagonists. Chronic morphine desensitized MOP in A7 cells, i.e., it decreased DAMGO-induced stimulation of GTPgammaS binding. In M2 cells DAMGO stimulation of GTPgammaS binding was significantly greater than in A7 cells and was not desensitized by chronic morphine. Pertussis toxin treatment abolished morphine-induced receptor up-regulation in M2 cells, whereas it had no effect on morphine-induced down-regulation in A7 cells. These results indicate that, in the absence of filamin A, chronic treatment with morphine, methadone or levorphanol leads to up-regulation of MOP, to our knowledge, the first instance of opioid receptor up-regulation by agonists in cell culture.


Assuntos
Proteínas Contráteis/fisiologia , Proteínas dos Microfilamentos/fisiologia , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Contráteis/deficiência , Interpretação Estatística de Dados , Diprenorfina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Filaminas , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Ligantes , Melanoma/genética , Melanoma/patologia , Proteínas dos Microfilamentos/deficiência , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Toxina Pertussis/farmacologia , Ensaio Radioligante , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Tubulina (Proteína)/farmacologia , Regulação para Cima/efeitos dos fármacos
16.
J Cell Biol ; 178(1): 121-8, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17606870

RESUMO

FILAMIN B, which encodes a cytoplasmic actin binding protein, is mutated in several skeletal dysplasias. To further investigate how an actin binding protein influences skeletogenesis, we generated mice lacking intact Filamin B. As observed in spondylocarpotarsal synostosis syndrome patients, Filamin B mutant mice display ectopic mineralization in many cartilaginous elements. This aberrant mineralization is due to ectopic chondrocyte hypertrophy similar to that seen in mice expressing Runx2 in chondrocytes. Accordingly, removing one copy of Runx2 rescues the Filamin B mutant phenotype, indicating that Filamin B is a regulator of Runx2 function during chondrocyte differentiation. Filamin B binds Smad3, which is known to interact with Runx2. Smad3 phosphorylation is increased in the mutant mice. Thus, Filamin B inhibits Runx2 activity, at least in part, through the Smad3 pathway. Our results uncover the involvement of actin binding proteins during chondrogenesis and provide a molecular basis to a human genetic disease.


Assuntos
Condrócitos/fisiologia , Proteínas Contráteis/deficiência , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas dos Microfilamentos/deficiência , Proteína Smad3/metabolismo , Animais , Condrócitos/citologia , Proteínas Contráteis/genética , Filaminas , Glutationa Transferase/metabolismo , Hipertrofia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , Modelos Biológicos , Mutação , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
17.
Proc Natl Acad Sci U S A ; 104(10): 3919-24, 2007 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17360453

RESUMO

Mutations in filamin B (FLNB), a gene encoding a cytoplasmic actin-binding protein, have been found in human skeletal disorders, including boomerang dysplasia, spondylocarpotarsal syndrome, Larsen syndrome, and atelosteogenesis phenotypes I and III. To examine the role of FLNB in vivo, we generated mice with a targeted disruption of Flnb. Fewer than 3% of homozygous embryos reached term, indicating that Flnb is important in embryonic development. Heterozygous mutant mice were indistinguishable from their wild-type siblings. Flnb was ubiquitously expressed; strong expression was found in endothelial cells and chondrocytes. Flnb-deficient fibroblasts exhibited more disorganized formation of actin filaments and reduced ability to migrate compared with wild-type controls. Flnb-deficient embryos exhibited impaired development of the microvasculature and skeletal system. The few Flnb-deficient mice that were born were very small and had severe skeletal malformations, including scoliotic and kyphotic spines, lack of intervertebral discs, fusion of vertebral bodies, and reduced hyaline matrix in extremities, thorax, and vertebrae. These mice died or had to be euthanized before 4 weeks of age. Thus, the phenotypes of Flnb-deficient mice closely resemble those of human skeletal disorders with mutations in FLNB.


Assuntos
Osso e Ossos/fisiologia , Proteínas Contráteis/genética , Proteínas Contráteis/fisiologia , Microcirculação , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Actinas/metabolismo , Animais , Movimento Celular , Condrócitos/metabolismo , Proteínas Contráteis/deficiência , Citoplasma/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Filaminas , Humanos , Cifose/genética , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Escoliose/genética
18.
Proc Natl Acad Sci U S A ; 103(52): 19836-41, 2006 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-17172441

RESUMO

Mutations in the human Filamin A (FLNA) gene disrupt neuronal migration to the cerebral cortex and cause cardiovascular defects. Complete loss of Flna in mice results in embryonic lethality with severe cardiac structural defects involving ventricles, atria, and outflow tracts, as well as widespread aberrant vascular patterning. Despite these widespread developmental defects, migration and motility of many cell types does not appear to be affected. Instead, Flna-null embryos display abnormal epithelial and endothelial organization and aberrant adherens junctions in developing blood vessels, heart, brain, and other tissues. Essential roles for FLNA in intercellular junctions provide a mechanism for the diverse developmental defects seen in patients with FLNA mutations.


Assuntos
Comunicação Celular , Proteínas Contráteis/metabolismo , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/metabolismo , Coração/embriologia , Proteínas dos Microfilamentos/metabolismo , Miocárdio/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Movimento Celular , Células Cultivadas , Proteínas Contráteis/deficiência , Proteínas Contráteis/genética , Embrião de Mamíferos/embriologia , Células Endoteliais/metabolismo , Filaminas , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Mutação/genética , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo
19.
Mol Cell Biol ; 26(17): 6522-34, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16914736

RESUMO

FilaminC (FLNc) is the muscle-specific member of a family of actin binding proteins. Although it interacts with many proteins involved in muscular dystrophies, its unique role in muscle is poorly understood. To address this, two models were developed. First, FLNc expression was stably reduced in C2C12 myoblasts by RNA interference. While these cells start differentiation normally, they display defects in differentiation and fusion ability and ultimately form multinucleated "myoballs" rather than maintain elongated morphology. Second, a mouse model carrying a deletion of last 8 exons of Flnc was developed. FLNc-deficient mice die shortly after birth, due to respiratory failure, and have severely reduced birth weights, with fewer muscle fibers and primary myotubes, indicating defects in primary myogenesis. They exhibit variation in fiber size, fibers with centrally located nuclei, and some rounded fibers resembling the in vitro phenotype. The similarity of the phenotype of FLNc-deficient mice to the filamin-interacting TRIO null mice was further confirmed by comparing FLNc-deficient C2C12 cells to TRIO-deficient cells. These data provide the first evidence that FLNc has a crucial role in muscle development and maintenance of muscle structural integrity and suggest the presence of a TRIO-FLNc-dependent pathway in maintaining proper myotube structure.


Assuntos
Proteínas Contráteis/deficiência , Proteínas dos Microfilamentos/deficiência , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Fusão Celular , Proteínas Contráteis/genética , Cruzamentos Genéticos , Feminino , Filaminas , Regulação da Expressão Gênica , Marcação de Genes , Genótipo , Fatores de Troca do Nucleotídeo Guanina/deficiência , Humanos , Masculino , Camundongos , Proteínas dos Microfilamentos/genética , Músculo Esquelético/anormalidades , Músculo Esquelético/ultraestrutura , Mioblastos/citologia , Tamanho do Órgão , Fenótipo , Fosfoproteínas/deficiência , Proteínas Serina-Treonina Quinases/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
20.
J Immunol ; 177(3): 1721-8, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16849481

RESUMO

Induction of T cell responses following engagement of the Ag-specific TCR depends on TCR-initiated rearrangements of the cellular actin cytoskeleton and highly coordinated and tightly regulated interactions and of diverse intracellular signaling proteins. In this study, we show that filamin A (FLNa), an actin-binding and signal mediator scaffolding protein, is required for T cell activation. Following Ag stimulation, FLNa was recruited to the T cell-APC contact area, where it colocalized with protein kinase C-theta (PKCtheta). Depletion of FLNa by RNA interference did not affect TCR-induced early tyrosine phosphorylation or actin polymerization but, nevertheless, resulted in impaired IL-2 expression by human primary T cells and reduced activation of NF-kappaB, AP-1, and NFAT reporter genes in transfected T cells. TCR stimulation induced stable physical association of FLNa with PKCtheta. Furthermore, the TCR/CD28-induced membrane translocation of PKCtheta was inhibited in FLNa-depleted T cells. These results reveal novel role for FLNa in the TCR/CD28 signaling pathway leading to transcription factor activation and IL-2 production, and suggest that this role is mediated, in part, through the inducible interaction of FLNa with PKCtheta.


Assuntos
Proteínas Contráteis/fisiologia , Isoenzimas/fisiologia , Ativação Linfocitária , Proteínas dos Microfilamentos/fisiologia , Proteína Quinase C/fisiologia , Linfócitos T/enzimologia , Linfócitos T/imunologia , Células Apresentadoras de Antígenos/enzimologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD28/fisiologia , Comunicação Celular/imunologia , Membrana Celular/enzimologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Proteínas Contráteis/deficiência , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Citosol/enzimologia , Citosol/imunologia , Citosol/metabolismo , Filaminas , Humanos , Interleucina-2/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Células Jurkat , Ativação Linfocitária/imunologia , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C-theta , Transporte Proteico , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...