Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Clin Transl Oncol ; 22(4): 522-531, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31190277

RESUMO

BACKGROUND: The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS: Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS: Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION: CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/fisiologia , Proteínas Contráteis/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Receptor Notch1/fisiologia , Fatores de Transcrição da Família Snail/fisiologia , Movimento Celular , Transição Epitelial-Mesenquimal , Feminino , Humanos , Células MCF-7 , Invasividade Neoplásica , Transdução de Sinais/fisiologia , Microambiente Tumoral
2.
PLoS Comput Biol ; 15(10): e1007431, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31652274

RESUMO

Non-muscle myosin II (NMII)-induced multicellular contractility is essential for development, maintenance and remodeling of tissue morphologies. Dysregulation of the cytoskeleton can lead to birth defects or enable cancer progression. We demonstrate that the Matrigel patterning assay, widely used to characterize endothelial cells, is a highly sensitive tool to evaluate cell contractility within a soft extracellular matrix (ECM) environment. We propose a computational model to explore how cell-exerted contractile forces can tear up the cell-Matrigel composite material and gradually remodel it into a network structure. We identify measures that are characteristic for cellular contractility and can be obtained from image analysis of the recorded patterning process. The assay was calibrated by inhibition of NMII activity in A431 epithelial carcinoma cells either directly with blebbistatin or indirectly with Y27632 Rho kinase inhibitor. Using Matrigel patterning as a bioassay, we provide the first functional demonstration that overexpression of S100A4, a calcium-binding protein that is frequently overexpressed in metastatic tumors and inhibits NMIIA activity by inducing filament disassembly, effectively reduces cell contractility.


Assuntos
Bioensaio/métodos , Colágeno/fisiologia , Proteínas Contráteis/fisiologia , Laminina/fisiologia , Proteoglicanas/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Simulação por Computador , Citoesqueleto/metabolismo , Combinação de Medicamentos , Células Epiteliais/fisiologia , Humanos , Camundongos , Microtúbulos/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo
3.
Mol Microbiol ; 112(6): 1718-1730, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515877

RESUMO

The flagellated eukaryote Trypanosoma brucei alternates between the insect vector and the mammalian host and proliferates through an unusual mode of cell division. Cell division requires flagellum motility-generated forces, but flagellum motility exerts distinct effects between different life cycle forms. Motility is required for the final cell abscission of the procyclic form in the insect vector, but is necessary for the initiation of cell division of the bloodstream form in the mammalian host. The underlying mechanisms remain elusive. Here we carried out functional analyses of a flagellar axonemal inner-arm dynein complex in the bloodstream form and investigated its mechanistic role in cytokinesis initiation. We showed that the axonemal inner-arm dynein heavy chain TbIAD5-1 and TbCentrin3 form a complex, localize to the flagellum, and are required for viability in the bloodstream form. We further demonstrated the interdependence between TbIAD5-1 and TbCentrin3 for maintenance of protein stability. Finally, we showed that depletion of TbIAD5-1 and TbCentrin3 arrested cytokinesis initiation and disrupted the localization of multiple cytokinesis initiation regulators. These findings identified the essential role of an axonemal inner-arm dynein complex in cell division, and provided molecular insights into the flagellum motility-mediated cytokinesis initiation in the bloodstream form of T. brucei.


Assuntos
Dineínas do Axonema/metabolismo , Proteínas Contráteis/metabolismo , Citocinese/fisiologia , Proteínas de Protozoários/metabolismo , Dineínas do Axonema/fisiologia , Axonema/metabolismo , Divisão Celular/genética , Divisão Celular/fisiologia , Linhagem Celular , Movimento Celular , Proteínas Contráteis/genética , Proteínas Contráteis/fisiologia , Dineínas/metabolismo , Dineínas/fisiologia , Flagelos/metabolismo , Flagelos/fisiologia , Estágios do Ciclo de Vida , Proteínas de Protozoários/genética , Proteínas de Protozoários/fisiologia , Interferência de RNA , Trypanosoma brucei brucei/metabolismo
4.
J Proteome Res ; 17(6): 2045-2059, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29681158

RESUMO

Bidirectional communication between cells and their microenvironment is crucial for both normal tissue homeostasis and tumor growth. During the development of oral tongue squamous cell carcinoma (OTSCC), cancer-associated fibroblasts (CAFs) create a supporting niche by maintaining a bidirectional crosstalk with cancer cells, mediated by classically secreted factors and various nanometer-sized vesicles, termed as extracellular vesicles (EVs). To better understand the role of CAFs within the tumor stroma and elucidate the mechanism by which secreted proteins contribute to OTSCC progression, we isolated and characterized patient-derived CAFs from resected tumors with matched adjacent tissue fibroblasts (AFs). Our strategy employed shotgun proteomics to comprehensively characterize the proteomes of these matched fibroblast populations. Our goals were to identify CAF-secreted factors (EVs and soluble) that can functionally modulate OTSCC cells in vitro and to identify novel CAF-associated biomarkers. Comprehensive proteomic analysis identified 4247 proteins, the most detailed description of a pro-tumorigenic stroma to date. We demonstrated functional effects of CAF secretomes (EVs and conditioned media) on OTSCC cell growth and migration. Comparative proteomics identified novel proteins associated with a CAF-like state. Specifically, MFAP5, a protein component of extracellular microfibrils, was enriched in CAF secretomes. Using in vitro assays, we demonstrated that MFAP5 activated OTSCC cell growth and migration via activation of MAPK and AKT pathways. Using a tissue microarray of richly annotated primary human OTSCCs, we demonstrated an association of MFAP5 expression with patient survival. In summary, our proteomics data of patient-derived stromal fibroblasts provide a useful resource for future mechanistic and biomarker studies.


Assuntos
Fibroblastos Associados a Câncer/química , Proteínas Contráteis/fisiologia , Glicoproteínas/fisiologia , Neoplasias de Cabeça e Pescoço/patologia , Comunicação Parácrina , Proteômica , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Biomarcadores , Fibroblastos Associados a Câncer/metabolismo , Movimento Celular , Proliferação de Células , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/mortalidade , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Análise de Sobrevida , Neoplasias da Língua
5.
Matrix Biol ; 47: 13-33, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25963142

RESUMO

The microfibril-associated glycoproteins MAGP-1 and MAGP-2 are extracellular matrix proteins that interact with fibrillin to influence microfibril function. The two proteins are related through a 60 amino acid matrix-binding domain but their sequences differ outside of this region. A distinguishing feature of both proteins is their ability to interact with TGFß family growth factors, Notch and Notch ligands, and multiple elastic fiber proteins. MAGP-2 can also interact with αvß3 integrins via a RGD sequence that is not found in MAGP-1. Morpholino knockdown of MAGP-1 expression in zebrafish resulted in abnormal vessel wall architecture and altered vascular network formation. In the mouse, MAGP-1 deficiency had little effect on elastic fibers in blood vessels and lung but resulted in numerous unexpected phenotypes including bone abnormalities, hematopoietic changes, increased fat deposition, diabetes, impaired wound repair, and a bleeding diathesis. Inactivation of the gene for MAGP-2 in mice produced a neutropenia yet had minimal effects on bone or adipose homeostasis. Double knockouts had phenotypes characteristic of each individual knockout as well as several additional traits only seen when both genes are inactivated. A common mechanism underlying all of the traits associated with the knockout phenotypes is altered TGFß signaling. This review summarizes our current understanding of the function of the MAGPs and discusses ideas related to their role in growth factor regulation.


Assuntos
Proteínas Contráteis/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Microfibrilas/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Desenvolvimento Ósseo , Fibrilinas , Expressão Gênica , Glicoproteínas/fisiologia , Hematopoese , Humanos , Proteínas dos Microfilamentos/fisiologia , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Fatores de Processamento de RNA , Cicatrização
6.
Ann Thorac Surg ; 95(5): 1619-25, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523189

RESUMO

BACKGROUND: Contractility in the borderzone (BZ) after anteroapical myocardial infarction (MI) is depressed. We tested the hypothesis that BZ contractility is also decreased after posterolateral MI. METHODS: Five sheep underwent posterolateral MI. Magnetic resonance imaging (MRI) was performed 2 weeks before and 16 weeks after MI, and left ventricular (LV) volume and regional strain were measured. Finite element (FE) models were constructed, and the systolic material parameter, Tmax, was calculated in the BZ and remote myocardium by minimizing the difference between experimentally measured and calculated LV strain and volume. Sheep were sacrificed 17 weeks after MI, and myocardial muscle fibers were taken from the BZ and remote myocardium. Fibers were chemically demembranated, and isometric developed force, Fmax, was measured at supramaximal [Ca(2+)]. Routine light microscopy was also performed. RESULTS: There was no difference in Tmax in the remote myocardium before and 16 weeks after MI. However, there was a large decrease (63.3%, p = 0.005) in Tmax in the BZ when compared with the remote myocardium 16 weeks after MI. In addition, there was a significant reduction of BZ Fmax for all samples (18.9%, p = 0.0067). Myocyte cross-sectional area increased by 61% (p = 0.021) in the BZ, but there was no increase in fibrosis. CONCLUSIONS: Contractility in the BZ is significantly depressed relative to the remote myocardium after posterolateral MI. The reduction in contractility is due at least in part to a decrease in contractile protein function.


Assuntos
Contração Miocárdica/fisiologia , Infarto do Miocárdio/fisiopatologia , Função Ventricular Esquerda/fisiologia , Animais , Proteínas Contráteis/fisiologia , Análise de Elementos Finitos , Ovinos
7.
Reprod Sci ; 20(4): 463-75, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23012315

RESUMO

Infection during pregnancy triggers inflammation, which can increase myometrial contractions and the risk of premature labor and delivery. In this study, we assessed the effects of vitamin D, an anti-inflammatory ligand on cytokines, chemokines, toll-like receptors, and contractile-associated proteins on immortalized human myometrial smooth muscle (UtSM) cells stimulated with lipopolysaccharide (LPS), a bacterial endotoxin, or interleukin (IL)-1ß and measured Toll-like receptor (TLR)-10 expression in pregnant myometrial tissues. A superarray analysis revealed downregulation of the chemokines monocyte chemoattractant protein (MCP)-1, Chemokine (C-X-C motif) ligand (CXCL)-10, CXCL-11, and chemokine (C-X3-C motif) ligand (CX3CL)-1; the proinflammatory cytokines IL-13 and tumor necrosis factor (TNF)-α; the TLR-4 and -5 and triggering receptor expressed on myeloid cells (TREM)-2 and upregulation of the anti-inflammatory cytokine IL-10, as well as Toll interacting protein (TOLLIP) and TREM-1 in vitamin D-treated UtSM cells. In the presence of LPS, vitamin D caused dose-dependent decreases in the messenger RNA expression of MCP-1, IL-1ß, IL-13, TNF-α, TLR-4, and TLR-5, the contractile-associated proteins connexin 43, the oxytocin receptor, and the prostaglandin receptor but caused increases in IL-10 and TLR-10 in UtSM cells. The TLR-10 expression was higher in human myometrial tissue obtained from women at term not in labor compared to labor. Vitamin D also attenuated IL-1ß-induced MCP-1, IL-6, connexin 43, cyclooxygenase (COX)-2, and prostaglandin receptor expression. Western analysis showed that vitamin D decreased MCP-1, TLR-4, and connexin 43 in the presence of LPS and decreased connexin 43 in the presence of IL-1ß. Our results suggest that vitamin D can potentially decrease infection-induced increases in cytokines and contractile-associated proteins in the myometrium.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Proteínas Contráteis/antagonistas & inibidores , Proteínas Contráteis/fisiologia , Miométrio/fisiologia , Receptores Toll-Like/metabolismo , Vitamina D/farmacologia , Linhagem Celular Transformada , Células Cultivadas , Feminino , Humanos , Miométrio/citologia , Miométrio/efeitos dos fármacos , Gravidez , Receptores Toll-Like/biossíntese , Receptores Toll-Like/fisiologia
8.
Dev Cell ; 23(1): 137-52, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22771033

RESUMO

Disruption of the BRCA2 tumor suppressor is associated with structural and numerical chromosomal defects. The numerical abnormalities in BRCA2-deficient cells may partly result from aberrations in cell division caused by disruption of BRCA2 during cytokinesis. Here we show that BRCA2 is a component of the midbody that is recruited through an interaction with Filamin A actin-binding protein. At the midbody, BRCA2 influences the recruitment of endosomal sorting complex required for transport (ESCRT)-associated proteins, Alix and Tsg101, and formation of CEP55-Alix and CEP55-Tsg101 complexes during abscission. Disruption of these BRCA2 interactions by cancer-associated mutations results in increased cytokinetic defects but has no effect on BRCA2-dependent homologous recombination repair of DNA damage. These findings identify a specific role for BRCA2 in the regulation of midbody structure and function, separate from DNA damage repair, that may explain in part the whole-chromosomal instability in BRCA2-deficient tumors.


Assuntos
Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/fisiologia , Proteínas Contráteis/fisiologia , Citocinese/fisiologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Proteínas dos Microfilamentos/fisiologia , Proteínas Nucleares/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteína BRCA2/química , Proteína BRCA2/genética , Proteína BRCA2/fisiologia , Proteínas de Ligação a DNA , Complexos Endossomais de Distribuição Requeridos para Transporte , Feminino , Filaminas , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Mutantes , Transporte Proteico/genética , Fatores de Transcrição
9.
PLoS One ; 7(7): e40448, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22802962

RESUMO

Filamins are important actin cross-linking proteins implicated in scaffolding, membrane stabilization and signal transduction, through interaction with ion channels, receptors and signaling proteins. Here we report the physical and functional interaction between filamins and polycystin-2, a TRP-type cation channel mutated in 10-15% patients with autosomal dominant polycystic kidney disease. Yeast two-hybrid and GST pull-down experiments demonstrated that the C-termini of filamin isoforms A, B and C directly bind to both the intracellular N- and C-termini of polycystin-2. Reciprocal co-immunoprecipitation experiments showed that endogenous polycystin-2 and filamins are in the same complexes in renal epithelial cells and human melanoma A7 cells. We then examined the effect of filamin on polycystin-2 channel function by electrophysiology studies with a lipid bilayer reconstitution system and found that filamin-A substantially inhibits polycystin-2 channel activity. Our study indicates that filamins are important regulators of polycystin-2 channel function, and further links actin cytoskeletal dynamics to the regulation of this channel protein.


Assuntos
Proteínas Contráteis/química , Proteínas dos Microfilamentos/química , Canais de Cátion TRPP/fisiologia , Animais , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Contráteis/fisiologia , Filaminas , Humanos , Imunoprecipitação , Bicamadas Lipídicas , Proteínas dos Microfilamentos/fisiologia , Isoformas de Proteínas/metabolismo , Técnicas do Sistema de Duplo-Híbrido
10.
Adv Exp Med Biol ; 740: 103-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453940

RESUMO

The extracellular calcium ([Formula: see text])-sensing receptor (CaSR) was the first GPCR identified whose principal physiological ligand is an ion, namely extracellular Ca(2+). It maintains the near constancy of [Formula: see text] that complex organisms require to ensure normal cellular function. A wealth of information has accumulated over the past two decades about the CaSR's structure and function, its role in diseases and CaSR-based therapeutics. This review briefly describes the CaSR and key features of its structure and function, then discusses the extracellular signals modulating its activity, provides an overview of the intracellular signaling pathways that it controls, and, finally, briefly describes CaSR signaling both in tissues participating in [Formula: see text] homeostasis as well as those that do not. Factors controlling CaSR signaling include various factors affecting the expression of the CaSR gene as well as modulation of its trafficking to and from the cell surface. The dimeric cell surface CaSR, in turn, links to various heterotrimeric and small molecular weight G proteins to regulate intracellular second messengers, lipid kinases, various protein kinases, and transcription factors that are part of the machinery enabling the receptor to modulate the functions of the wide variety of cells in which it is expressed. CaSR signaling is impacted by its interactions with several binding partners in addition to signaling elements per se (i.e., G proteins), including filamin-A and caveolin-1. These latter two proteins act as scaffolds that bind signaling components and other key cellular elements (e.g., the cytoskeleton). Thus CaSR signaling likely does not take place randomly throughout the cell, but is compartmentalized and organized so as to facilitate the interaction of the receptor with its various signaling pathways.


Assuntos
Sinalização do Cálcio/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Animais , Cálcio/metabolismo , Caveolina 1/fisiologia , Proteínas Contráteis/fisiologia , Filaminas , Homeostase , Humanos , Rim/metabolismo , Proteínas dos Microfilamentos/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neoplasias/metabolismo , Receptores de Detecção de Cálcio/química
11.
Hepatogastroenterology ; 58(109): 1182-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21937374

RESUMO

BACKGROUND/AIMS: Liver metastasis is a major factor associated with poor prognosis in patients with colorectal cancer. The objective of this study is to determine the possible indicators in identifying the risk of liver metastasis. METHODOLOGY: We randomly selected 114 colorectal cancer patients with liver metastases and 114 patients without liver metastasis. Several clinicopathological factors were analyzed for the correlation with liver metastasis. PTEN, CapG, MFAP3L, EGFR and HSP-70, chosen from PI3K/ AKT pathway, were evaluated by immunohistochemistry staining. The predictive value of those 5 markers for colorectal liver metastasis was evaluated. RESULTS: Univariate analysis showed that histological type, invasion depth, microscopic tumor embolus and lymph node metastasis were associated with liver metastasis. In multivariate analysis, only microscopic tumor embolus and lymph node metastasis were associated with liver metastasis. PTEN, EGFR, MFAP3L, CapG and HSP-70 were significantly correlated with colorectal liver metastasis. Grouped as a marker set, EGFR, MFAP3L and CapG make an effective predictor for colorectal liver metastasis, wherein the sensitivity and specificity are 92.38% and 94.39%, respectively. CONCLUSIONS: Microscopic tumor embolus and lymph node metastasis are risk factors for colorectal liver metastasis. As a molecular marker set, EGFR, MFAP3L and CapG exhibited both high sensitivity and high specificity in predicting colorectal liver metastasis.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Hepáticas/secundário , Adulto , Idoso , Proteínas Contráteis/fisiologia , Receptores ErbB/fisiologia , Feminino , Proteínas de Choque Térmico HSP70/fisiologia , Humanos , Masculino , Proteínas dos Microfilamentos/fisiologia , Pessoa de Meia-Idade , Proteínas Nucleares/fisiologia , PTEN Fosfo-Hidrolase/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais
12.
PLoS One ; 6(2): e17218, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21359179

RESUMO

BACKGROUND: Androgen receptor (AR) controls male morphogenesis, gametogenesis and prostate growth as well as development of prostate cancer. These findings support a role for AR in cell migration and invasiveness. However, the molecular mechanism involved in AR-mediated cell migration still remains elusive. METHODOLOGY/PRINCIPAL FINDINGS: Mouse embryo NIH3T3 fibroblasts and highly metastatic human fibrosarcoma HT1080 cells harbor low levels of transcriptionally incompetent AR. We now report that, through extra nuclear action, AR triggers migration of both cell types upon stimulation with physiological concentrations of the androgen R1881. We analyzed the initial events leading to androgen-induced cell migration and observed that challenging NIH3T3 cells with 10 nM R1881 rapidly induces interaction of AR with filamin A (FlnA) at cytoskeleton. AR/FlnA complex recruits integrin beta 1, thus activating its dependent cascade. Silencing of AR, FlnA and integrin beta 1 shows that this ternary complex controls focal adhesion kinase (FAK), paxillin and Rac, thereby driving cell migration. FAK-null fibroblasts migrate poorly and Rac inhibition by EHT impairs motility of androgen-treated NIH3T3 cells. Interestingly, FAK and Rac activation by androgens are independent of each other. Findings in human fibrosarcoma HT1080 cells strengthen the role of Rac in androgen signaling. The Rac inhibitor significantly impairs androgen-induced migration in these cells. A mutant AR, deleted of the sequence interacting with FlnA, fails to mediate FAK activation and paxillin tyrosine phosphorylation in androgen-stimulated cells, further reinforcing the role of AR/FlnA interaction in androgen-mediated motility. CONCLUSIONS/SIGNIFICANCE: The present report, for the first time, indicates that the extra nuclear AR/FlnA/integrin beta 1 complex is the key by which androgen activates signaling leading to cell migration. Assembly of this ternary complex may control organ development and prostate cancer metastasis.


Assuntos
Androgênios/farmacologia , Movimento Celular/efeitos dos fármacos , Proteínas Contráteis/metabolismo , Proteínas Contráteis/fisiologia , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/fisiologia , Receptores Androgênicos/metabolismo , Receptores Androgênicos/fisiologia , Células 3T3-L1 , Animais , Células COS , Carcinoma/metabolismo , Carcinoma/patologia , Células Cultivadas , Chlorocebus aethiops , Filaminas , Humanos , Integrina beta1/metabolismo , Integrina beta1/fisiologia , Masculino , Metribolona/farmacologia , Camundongos , Células NIH 3T3 , Metástase Neoplásica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
13.
J Invest Dermatol ; 131(5): 1119-28, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21209619

RESUMO

Extracellular Ca(2+) (Ca(2+)(o)) functioning through the calcium-sensing receptor (CaR) induces E-cadherin-mediated cell-cell adhesion and cellular signals mediating cell differentiation in epidermal keratinocytes. Previous studies indicate that CaR regulates cell-cell adhesion through Fyn/Src tyrosine kinases. In this study, we investigate whether Rho GTPase is a part of the CaR-mediated signaling cascade regulating cell adhesion and differentiation. Suppressing endogenous Rho A expression by small interfering RNA (siRNA)-mediated gene silencing blocked the Ca(2+)(o)-induced association of Fyn with E-cadherin and suppressed the Ca(2+)(o)-induced tyrosine phosphorylation of ß-, γ-, and p120-catenin and formation of intercellular adherens junctions. Rho A silencing also decreased the Ca(2+)(o)-stimulated expression of terminal differentiation markers. Elevating the Ca(2+)(o) level induced interactions among CaR, Rho A, E-cadherin, and the scaffolding protein filamin A at the cell membrane. Inactivation of CaR expression by adenoviral expression of a CaR antisense complementary DNA inhibited Ca(2+)(o)-induced activation of endogenous Rho. Ca(2+)(o) activation of Rho required a direct interaction between CaR and filamin A. Interference of CaR-filamin interaction inhibited Ca(2+)(o)-induced Rho activation and the formation of cell-cell junctions. These results indicate that Rho is a downstream mediator of CaR in the regulation of Ca(2+)(o)-induced E-cadherin-mediated cell-cell adhesion and keratinocyte differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Contráteis/fisiologia , Queratinócitos/fisiologia , Proteínas dos Microfilamentos/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Junções Aderentes/metabolismo , Caderinas/metabolismo , Cateninas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Contráteis/metabolismo , Epiderme/efeitos dos fármacos , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Filaminas , Inativação Gênica/efeitos dos fármacos , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-fyn/metabolismo , RNA Interferente Pequeno/farmacologia , Quinases da Família src/metabolismo
14.
Int J Cancer ; 128(4): 839-46, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473907

RESUMO

Deregulated hepatocyte growth factor (HGF)/c-MET axis has been correlated with poor clinical outcome and drug resistance in many human cancers. Identification of novel regulatory mechanisms influencing HGF/c-MET signaling may therefore be necessary to develop more effective cancer therapies. In our study, we show that multiple human cancer tissues and cells express filamin A (FLNA), a large cytoskeletal actin-binding protein, and expression of c-MET is significantly reduced in human tumor cells deficient for FLNA. The FLNA-deficient tumor cells exhibited poor migrative and invasive ability in response to HGF. On the other hand, the anchorage-dependent and independent tumor cell proliferation was not altered by HGF. The FLNA-deficiency specifically attenuated the activation of the c-MET downstream signaling molecule AKT in response to HGF stimulation. Furthermore, FLNA enhanced c-MET promoter activity by its binding to SMAD2. The impact of FLNA deficiency on c-MET expression and HGF-mediated cell migration in human tumor cells was confirmed in primary mouse embryonic fibroblasts deficient for Flna. These data suggest that FLNA is one of the important regulators of c-MET signaling and HGF-induced tumor cell migration.


Assuntos
Movimento Celular , Proteínas Contráteis/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Proteínas dos Microfilamentos/fisiologia , Neoplasias/patologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Filaminas , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Luciferases/metabolismo , Camundongos , Neoplasias/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Análise Serial de Tecidos , Células Tumorais Cultivadas
15.
J Appl Physiol (1985) ; 110(3): 705-16, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21030671

RESUMO

Oxidative modification of contractile proteins is thought to be a key factor in muscle weakness observed in many pathophysiological conditions. In particular, peroxynitrite (ONOO(-)), a potent short-lived oxidant, is a likely candidate responsible for this contractile dysfunction. In this study ONOO(-) or 3-morpholinosydnonimine (Sin-1, a ONOO(-) donor) was applied to rat skinned muscle fibers to characterize the effects on contractile properties. Both ONOO(-) and Sin-1 exposure markedly reduced maximum force in slow-twitch fibers but had much less effect in fast-twitch fibers. The rate of isometric force development was also reduced without change in the number of active cross bridges. Sin-1 exposure caused a disproportionately large decrease in Ca(2+) sensitivity, evidently due to coproduction of superoxide, as it was prevented by Tempol, a superoxide dismutase mimetic. The decline in maximum force with Sin-1 and ONOO(-) treatments could be partially reversed by DTT, provided it was applied before the fiber was activated. Reversal by DTT indicates that the decrease in maximum force was due at least in part to oxidation of cysteine residues. Ascorbate caused similar reversal, further suggesting that the cysteine residues had undergone S-nitrosylation. The reduction in Ca(2+) sensitivity, however, was not reversed by either DTT or ascorbate. Western blot analysis showed cross-linking of myosin heavy chain (MHC) I, appearing as larger protein complexes after ONOO(-) exposure. The findings suggest that ONOO(-) initially decreases maximum force primarily by oxidation of cysteine residues on the myosin heads, and that the accompanying decrease in Ca(2+) sensitivity is likely due to other, less reversible actions of hydroxyl or related radicals.


Assuntos
Proteínas Contráteis/fisiologia , Contração Muscular/fisiologia , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Força Muscular/fisiologia , Doadores de Óxido Nítrico/administração & dosagem , Ácido Peroxinitroso/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Masculino , Contração Muscular/efeitos dos fármacos , Fibras Musculares de Contração Rápida/efeitos dos fármacos , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Ratos , Ratos Long-Evans
16.
Adv Exp Med Biol ; 682: 77-103, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20824521

RESUMO

Although it has been well established that muscle contraction results from cyclic attachment-detachment between the cross-bridges extending from the thick filaments and the sites on the thin filaments, the movement of the cross-bridges coupled with ATP hydrolysis still remains to be a matter for debate and speculation. The most straightforward way to elucidate this mystery is to record individual cross-bridge movement in response to ATP. Using a gas environmental chamber (EC, or hydration chamber), with which biological specimens retaining their physiological function can be observed under an electron microscope, my coworkers and I succeeded in recording the ATP-induced individual cross-bridge movement in two different kinds of synthetic thick filaments in 1997 and 2008. In the synthetic bipolar filaments consisting of rabbit skeletal muscle myosin, the amplitude of cross-bridge movement exhibits a peak at 5-7.5 nm, and the direction of cross-bridge movement is away from, but not towards, the filament bare region in the absence of thin filaments. After exhaustion of ATP, the cross-bridges return towards their initial position, indicating that the initial cross-bridge state may be analogous to that after completion of power stroke. These results constitute the first visualization of the cross-bridge recovery stroke, indicating that the EC is a powerful tool to open new horizons in the research fields of life sciences.


Assuntos
Citoesqueleto de Actina/fisiologia , Trifosfato de Adenosina/metabolismo , Músculo Esquelético/fisiologia , Citoesqueleto de Actina/ultraestrutura , Difosfato de Adenosina/metabolismo , Animais , Astacoidea/fisiologia , Proteínas Contráteis/fisiologia , Hidrólise , Microscopia Eletrônica/métodos , Modelos Teóricos , Contração Muscular/fisiologia , Músculo Esquelético/ultraestrutura , Miofibrilas/fisiologia , Miofibrilas/ultraestrutura , Miosinas/fisiologia , Mytilus edulis , Coelhos , Tropomiosina/fisiologia
17.
Carcinogenesis ; 31(10): 1710-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20656791

RESUMO

Resistance to transforming growth factor (TGF) ß-mediated tumor suppression in melanoma appears to be a crucial step in tumor aggressiveness since it is usually coupled with the ability of TGFß to drive the oncogenic process via autocrine and paracrine effects. In this review, we will focus mainly on the mechanisms of escape from TGFß-induced cell cycle arrest because the mechanisms of resistance to TGFß-mediated apoptosis are still essentially speculative. As expected, some of these mechanisms can directly affect the function of the main downstream effectors of TGFß, Smad2 and Smad3, resulting in compromised Smad-mediated antiproliferative activity. Other mechanisms can counteract or overcome TGFß-mediated cell cycle arrest independently of the Smads. In melanoma, some models of resistance to TGFß have been suggested and will be described. In addition, we propose additional models of resistance taking into consideration the information available on the dysregulation of fundamental cellular effectors and signaling pathways in melanoma.


Assuntos
Melanoma/patologia , Fator de Crescimento Transformador beta/fisiologia , Apoptose , Ciclo Celular , Proteínas Contráteis/fisiologia , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/análise , Progressão da Doença , Filaminas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/fisiologia , Genes myc , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Melanoma/prevenção & controle , Proteínas dos Microfilamentos/fisiologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia
18.
PLoS One ; 5(6): e11269, 2010 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-20585650

RESUMO

BACKGROUND: Changes in cell adhesion and migration in the tumor microenvironment are key in the initiation and progression of metastasis. R-Ras is one of several small GTPases that regulate cell adhesion and migration on the extracellular matrix, however the mechanism has not been completely elucidated. Using a yeast two-hybrid approach we sought to identify novel R-Ras binding proteins that might mediate its effects on integrins. METHODS AND FINDINGS: We identified Filamin A (FLNa) as a candidate interacting protein. FLNa is an actin-binding scaffold protein that also binds to integrin beta1, beta2 and beta7 tails and is associated with diverse cell processes including cell migration. Indeed, M2 melanoma cells require FLNa for motility. We further show that R-Ras and FLNa interact in co-immunoprecipitations and pull-down assays. Deletion of FLNa repeat 3 (FLNaDelta3) abrogated this interaction. In M2 melanoma cells active R-Ras co-localized with FLNa but did not co-localize with FLNa lacking repeat 3. Thus, activated R-Ras binds repeat 3 of FLNa. The functional consequence of this interaction was that active R-Ras and FLNa coordinately increased cell migration. In contrast, co-expression of R-Ras and FLNaDelta3 had a significantly reduced effect on migration. While there was enhancement of integrin activation and fibronectin matrix assembly, cell adhesion was not altered. Finally, siRNA knockdown of endogenous R-Ras impaired FLNa-dependent fibronectin matrix assembly. CONCLUSIONS: These data support a model in which R-Ras functionally associates with FLNa and thereby regulates integrin-dependent migration. Thus in melanoma cells R-Ras and FLNa may cooperatively promote metastasis by enhancing cell migration.


Assuntos
Movimento Celular/fisiologia , Proteínas Contráteis/fisiologia , Melanoma/patologia , Proteínas dos Microfilamentos/fisiologia , Proteínas ras/fisiologia , Sequência de Bases , Western Blotting , Primers do DNA , Filaminas , Humanos , Microscopia de Fluorescência
19.
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
20.
PLoS One ; 4(11): e7830, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19915675

RESUMO

Mammalian filamins (FLNs) are a family of three large actin-binding proteins. FLNa, the founding member of the family, was implicated in migration by cell biological analyses and the identification of FLNA mutations in the neuronal migration disorder periventricular heterotopia. However, recent knockout studies have questioned the relevance of FLNa to cell migration. Here we have used shRNA-mediated knockdown of FLNa, FLNb or FLNa and FLNb, or, alternatively, acute proteasomal degradation of all three FLNs, to generate FLN-deficient cells and assess their ability to migrate. We report that loss of FLNa or FLNb has little effect on migration but that knockdown of FLNa and FLNb, or proteolysis of all three FLNs, impairs migration. The observed defect is primarily a deficiency in initiation of motility rather than a problem with maintenance of locomotion speed. FLN-deficient cells are also impaired in spreading. Re-expression of full length FLNa, but not re-expression of a mutated FLNa lacking immunoglobulin domains 19 to 21, reverts both the spreading and the inhibition of initiation of migration.Our results establish a role for FLNs in cell migration and spreading and suggest that compensation by other FLNs may mask phenotypes in single knockout or knockdown cells. We propose that interactions between FLNs and transmembrane or signalling proteins, mediated at least in part by immunoglobulin domains 19 to 21 are important for both cell spreading and initiation of migration.


Assuntos
Proteínas Contráteis/fisiologia , Proteínas dos Microfilamentos/fisiologia , Actinas/química , Animais , Linhagem Celular Tumoral , Movimento Celular , Proteínas Contráteis/metabolismo , Filaminas , Humanos , Imunoglobulinas/química , Células Jurkat , Proteínas dos Microfilamentos/metabolismo , Modelos Biológicos , Mutação , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA