Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Intervalo de año de publicación
1.
Neuroendocrinology ; 110(7-8): 642-652, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31574507

RESUMEN

The high expression of somatostatin receptor 2 (SST2) in growth hormone (GH)-secreting tumors represents the rationale for the clinical use of somatostatin analogs (SSAs) in acromegaly. Recently, the cytoskeletal protein Filamin A (FLNA) has emerged as key modulator of the responsiveness of GH-secreting pituitary tumors to SSAs by regulating SST2 signaling and expression. The aim of this study was to explore FLNA involvement in SST2 intracellular trafficking in tumor somatotroph cells. By biotinylation assay, we found that FLNA silencing abolished octreotide-mediated SST2 internalization in rat GH3 cell line (28.0 ± 2.7 vs. 4 ± 4.3% SST2 internalization, control versus FLNA small interfering RNAs (siRNA) cells, respectively, p < 0.001) and human GH-secreting primary cultured cells (70.3 ± 21.1 vs. 24 ± 19.2% SST2 internalization, control versus FLNA siRNA cells, respectively, p < 0.05). In addition, confocal imaging revealed impaired SST2 recycling to the plasma membrane in FLNA silenced GH3 cells. Coimmunoprecipitation and immunofluorescence experiments showed that FLNA, as well as ß-arrestin2, is timely dependent recruited to octreotide-stimulated SST2 receptors both in rat and human tumor somatotroph cells. Although FLNA expression knock down did not prevent the formation of ß-arrestin2-SST2 complex in GH3 cells, it significantly impaired efficient SST2 loading into cytosolic vesicles positive for the early endocytic and recycling markers Rab5 and 4, respectively (33.7 ± 8.9% down to 25.9 ± 6.9%, p < 0.05, and 28.4 ± 7.4% down to 17.6 ± 5.7%, p < 0.01, for SST2-Rab5 and SST2-Rab4 colocalization, respectively, in control versus FLNA siRNA cells). Altogether these data support an important role for FLNA in the mediation of octreotide-induced SST2 trafficking in GH-secreting pituitary tumor cells through Rab5 and 4 sorting endosomes.


Asunto(s)
Adenoma/metabolismo , Endosomas/fisiología , Filaminas/fisiología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Receptores de Somatostatina/metabolismo , Adenoma/patología , Animales , Células Cultivadas , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Humanos , Octreótido/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo , Somatotrofos/patología , Proteínas de Unión al GTP rab4/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
2.
Int. braz. j. urol ; 45(5): 916-924, Sept.-Dec. 2019. graf
Artículo en Inglés | LILACS | ID: biblio-1040072

RESUMEN

ABSTRACT Objective This study aims to investigate the association of filamin A with the function and morphology of prostate cancer (PCa) cells, and explore the role of filamin A in the development of PCa, in order to analyze its significance in the evolvement of PCa. Materials and Methods A stably transfected cell line, in which filamin A expression was suppressed by RNA interference, was first established. Then, the effects of the suppression of filamin A gene expression on the biological characteristics of human PCa LNCaP cells were observed through cell morphology, in vitro cell growth curve, soft agar cloning assay, and scratch test. Results A cell line model with a low expression of filamin A was successfully constructed on the basis of LNCaP cells. The morphology of cells transfected with plasmid pSilencer-filamin A was the following: Cells were loosely arranged, had less connection with each other, had fewer tentacles, and presented a fibrous look. The growth rate of LNCap cells was faster than cells transfected with plasmid pSilencer-filamin A (P <0.05). The clones of LNCap cells in the soft agar cloning assay was significantly fewer than that of cells stably transfected with plasmid pSilencer-filamin A (P <0.05). Cells stably transfected with plasmid pSilencer-filamin A presented with a stronger healing and migration ability compared to LNCap cells (healing rate was 32.2% and 12.1%, respectively; P <0.05). Conclusion The expression of the filamin A gene inhibited the malignant development of LNCap cells. Therefore, the filamin A gene may be a tumor suppressor gene.


Asunto(s)
Humanos , Masculino , Neoplasias de la Próstata/patología , Filaminas/análisis , Filaminas/fisiología , Plásmidos , Neoplasias de la Próstata/genética , Sales de Tetrazolio , Factores de Tiempo , Cicatrización de Heridas/fisiología , Transfección/métodos , Células Cultivadas , Western Blotting , Colorimetría/métodos , Línea Celular Tumoral , Proliferación Celular , Filaminas/genética , Formazáns
3.
Int Braz J Urol ; 45(5): 916-924, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31268639

RESUMEN

OBJECTIVE: This study aims to investigate the association of filamin A with the function and morphology of prostate cancer (PCa) cells, and explore the role of filamin A in the development of PCa, in order to analyze its significance in the evolvement of PCa. MATERIALS AND METHODS: A stably transfected cell line, in which filamin A expression was suppressed by RNA interference, was first established. Then, the effects of the suppression of filamin A gene expression on the biological characteristics of human PCa LNCaP cells were observed through cell morphology, in vitro cell growth curve, soft agar cloning assay, and scratch test. RESULTS: A cell line model with a low expression of filamin A was successfully constructed on the basis of LNCaP cells. The morphology of cells transfected with plasmid pSilencer-filamin A was the following: Cells were loosely arranged, had less connection with each other, had fewer tentacles, and presented a fibrous look. The growth rate of LNCap cells was faster than cells transfected with plasmid pSilencer-filamin A (P<0.05). The clones of LNCap cells in the soft agar cloning assay was significantly fewer than that of cells stably transfected with plasmid pSilencer-filamin A (P<0.05). Cells stably transfected with plasmid pSilencer-filamin A presented with a stronger healing and migration ability compared to LNCap cells (healing rate was 32.2% and 12.1%, respectively; P<0.05). CONCLUSION: The expression of the filamin A gene inhibited the malignant development of LNCap cells. Therefore, the filamin A gene may be a tumor suppressor gene.


Asunto(s)
Filaminas/análisis , Filaminas/fisiología , Neoplasias de la Próstata/patología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Colorimetría/métodos , Filaminas/genética , Formazáns , Humanos , Masculino , Plásmidos , Neoplasias de la Próstata/genética , Sales de Tetrazolio , Factores de Tiempo , Transfección/métodos , Cicatrización de Heridas/fisiología
4.
Sci Adv ; 5(5): eaav8421, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31131323

RESUMEN

Mechanical force-induced conformational changes in proteins underpin a variety of physiological functions, typified in muscle contractile machinery. Mutations in the actin-binding protein filamin C (FLNC) are linked to musculoskeletal pathologies characterized by altered biomechanical properties and sometimes aggregates. HspB1, an abundant molecular chaperone, is prevalent in striated muscle where it is phosphorylated in response to cues including mechanical stress. We report the interaction and up-regulation of both proteins in three mouse models of biomechanical stress, with HspB1 being phosphorylated and FLNC being localized to load-bearing sites. We show how phosphorylation leads to increased exposure of the residues surrounding the HspB1 phosphosite, facilitating their binding to a compact multidomain region of FLNC proposed to have mechanosensing functions. Steered unfolding of FLNC reveals that its extension trajectory is modulated by the phosphorylated region of HspB1. This may represent a posttranslationally regulated chaperone-client protection mechanism targeting over-extension during mechanical stress.


Asunto(s)
Filaminas/fisiología , Proteínas de Choque Térmico/fisiología , Chaperonas Moleculares/fisiología , Animales , Sitios de Unión , Filaminas/genética , Corazón/fisiología , Proteínas de Choque Térmico/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Mutación , Miocardio/metabolismo , Fosforilación , Desnaturalización Proteica , Dominios Proteicos , Pliegue de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes , Estrés Mecánico
5.
Cereb Cortex ; 29(3): 1280-1290, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29462287

RESUMEN

Neural progenitor proliferation and cell fate decision from self-renewal to differentiation are crucial factors in determining brain size and morphology. The cytoskeletal dependent regulation of these processes is not entirely known. The actin-binding filamin A (FlnA) was shown to regulate proliferation of progenitors by directing changes in cell cycles proteins such as Cdk1 during G2/M phase. Here we report that functional loss of FlnA not only affects the rate of proliferation by altering cell cycle length but also causes a defect in early differentiation through changes in cell fate specification. FlnA interacts with Rho GTPase RhoA, and FlnA loss impairs RhoA activation. Disruption of either of these cytoskeletal associated proteins delays neurogenesis and promotes neural progenitors to remain in proliferative states. Aurora kinase B (Aurkb) has been implicated in cytokinesis, and peaks in expression during the G2/M phase. Inhibition of FlnA or RhoA impairs Aurkb degradation and alters its localization during mitosis. Overexpression of Aurkb replicates the same delay in neurogenesis seen with loss of FlnA or RhoA. Our findings suggest that shared cytoskeletal processes can direct neural progenitor proliferation by regulating the expression and localization of proteins that are implicated in the cell cycle progression and cell fate specification.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Citoesqueleto/fisiología , Filaminas/fisiología , Mitosis/fisiología , Células-Madre Neurales/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Aurora Quinasa B/fisiología , Diferenciación Celular , Proliferación Celular , Ratones , Neurogénesis
6.
Anat Rec (Hoboken) ; 302(1): 117-124, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30288957

RESUMEN

Mitral valve prolapse (MVP) affects 2.4% of the population and has poorly understood etiology. Recent genetic studies have begun to unravel the complexities of MVP and through these efforts, mutations in the FLNA (Filamin-A) gene were identified as disease causing. Our in vivo and in vitro studies have validated these genetic findings and have revealed FLNA as a central regulator of valve morphogenesis. The mechanisms by which FLNA mutations result in myxomatous mitral valve disease are currently unknown, but may involve proteins previously associated with mutated regions of the FLNA protein, such as the small GTPase signaling protein, R-Ras. Herein, we report that Filamin-A is required for R-Ras expression and activation of the Ras-Mek-Erk pathway. Loss of the Ras/Erk pathway correlated with hyperactivation of pSmad2/3, increased extracellular matrix (ECM) production and enlarged mitral valves. Analyses of integrin receptors in the mitral valve revealed that Filamin-A was required for ß1-integrin expression and provided a potential mechanism for impaired ECM compaction and valve enlargement. Our data support Filamin-A as a protein that regulates the balance between Erk and Smad activation and an inability of Filamin-A deficient valve interstitial cells to effectively remodel the increased ECM production through a ß1-integrin mechanism. As a consequence, loss of Filamin-A function results in increased ECM production and generation of a myxomatous phenotype characterized by improperly compacted mitral valve tissue. Anat Rec, 302:117-124, 2019. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Matriz Extracelular/metabolismo , Filaminas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Válvula Mitral/metabolismo , Organogénesis , Proteína smad3/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Válvula Mitral/citología , Fenotipo
8.
PLoS One ; 12(12): e0189285, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29240780

RESUMEN

The effects of actin dependent molecular mechanisms in coordinating cellular proliferation, migration and differentiation during embryogenesis are not well-understood. We have previously shown that actin-binding Filamin A (FlnA) and actin-nucleating Formin 2 (Fmn2) influence the development of the brain causing microcephaly in mice. In this study, we broaden this phenotype to explore the effects of these two proteins in the development of extra-CNS organ systems, including the gut, muscle, and skeleton. We observed defects in rib and sternum midline closure leading to thoracoabdominal schisis in FlnA+Fmn2 knockout mice, reminiscent of the pentalogy of Cantrell syndrome. These mice exhibit shortened guts, as well as thinned thoracic muscle mass. Immunostaining showed these changes are partially caused by a decrease in the number of presumptive mesenchymal proliferating cells with loss of either FlnA or FlnA+Fmn2. This proliferation defect appears to be in part due to delayed differentiation in these regions. While both FlnA and FlnA+Fmn2 mice show reduced cell death relative to WT control, increased caspase staining was seen in the double null relative to FlnA null suggesting that this could also contribute to the FlnA+Fmn2 phenotype. Therefore FlnA and Fmn2 are likely essential to cell proliferation, differentiation and cell death in a variety of tissues and organs, further reiterating the importance of vesicle trafficking in regulation of development.


Asunto(s)
Huesos/citología , Proliferación Celular/fisiología , Filaminas/fisiología , Intestinos/citología , Células Madre Mesenquimatosas/citología , Proteínas de Microfilamentos/fisiología , Músculo Esquelético/citología , Proteínas Nucleares/fisiología , Animales , Forminas , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso
9.
Mol Biol Cell ; 28(22): 3013-3028, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28855378

RESUMEN

Changes in cell morphology and the physical forces that occur during migration are generated by a dynamic filamentous actin cytoskeleton. The ADP-ribosylation factor-like 4C (Arl4C) small GTPase acts as a molecular switch to regulate morphological changes and cell migration, although the mechanism by which this occurs remains unclear. Here we report that Arl4C functions with the actin regulator filamin-A (FLNa) to modulate filopodium formation and cell migration. We found that Arl4C interacted with FLNa in a GTP-dependent manner and that FLNa IgG repeat 22 is both required and sufficient for this interaction. We also show that interaction between FLNa and Arl4C is essential for Arl4C-induced filopodium formation and increases the association of FLNa with Cdc42-GEF FGD6, promoting cell division cycle 42 (Cdc42) GTPase activation. Thus our study revealed a novel mechanism, whereby filopodium formation and cell migration are regulated through the Arl4C-FLNa-mediated activation of Cdc42.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Filaminas/metabolismo , Seudópodos/metabolismo , ADP-Ribosilación , Factores de Ribosilacion-ADP/fisiología , Citoesqueleto de Actina/metabolismo , Actinas , Animales , Movimiento Celular/fisiología , Filaminas/fisiología , Células HeLa , Humanos , Proteínas de Microfilamentos/metabolismo , Metástasis de la Neoplasia/fisiopatología , Neoplasias/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
10.
Proc Biol Sci ; 284(1854)2017 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-28469022

RESUMEN

The indirect flight muscles (IFMs) of Drosophila and other insects with asynchronous flight muscles are characterized by a crystalline myofilament lattice structure. The high-order lattice regularity is considered an adaptation for enhanced power output, but supporting evidence for this claim is lacking. We show that IFMs from transgenic flies expressing flightin with a deletion of its poorly conserved N-terminal domain (flnΔN62 ) have reduced inter-thick filament spacing and a less regular lattice. This resulted in a decrease in flight ability by 33% and in skinned fibre oscillatory power output by 57%, but had no effect on wingbeat frequency or frequency of maximum power output, suggesting that the underlying actomyosin kinetics is not affected and that the flight impairment arises from deficits in force transmission. Moreover, we show that flnΔN62 males produced an abnormal courtship song characterized by a higher sine song frequency and a pulse song with longer pulses and longer inter-pulse intervals (IPIs), the latter implicated in male reproductive success. When presented with a choice, wild-type females chose control males over mutant males in 92% of the competition events. These results demonstrate that flightin N-terminal domain is required for optimal myofilament lattice regularity and IFM activity, enabling powered flight and courtship song production. As the courtship song is subject to female choice, we propose that the low amino acid sequence conservation of the N-terminal domain reflects its role in fine-tuning species-specific courtship songs.


Asunto(s)
Cortejo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Filaminas/fisiología , Vuelo Animal , Proteínas Musculares/fisiología , Miofibrillas/fisiología , Animales , Femenino , Masculino
11.
J Physiol ; 595(7): 2271-2284, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27779751

RESUMEN

KEY POINTS: Ion channels are transmembrane proteins that are synthesized within the cells but need to be trafficked to the cell membrane for the channels to function. Small-conductance, Ca2+ -activated K+ channels (SK, KCa 2) are unique subclasses of K+ channels that are regulated by Ca2+ inside the cells; they are expressed in human atrial myocytes and responsible for shaping atrial action potentials. We have previously shown that interacting proteins of SK2 channels are important for channel trafficking to the membrane. Using total internal reflection fluorescence (TIRF) and confocal microscopy, we studied the mechanisms by which the surface membrane localization of SK2 (KCa 2.2) channels is regulated by their interacting proteins. Understanding the mechanisms of SK channel trafficking may provide new insights into the regulation controlling the repolarization of atrial myocytes. ABSTRACT: The normal function of ion channels depends critically on the precise subcellular localization and the number of channel proteins on the cell surface membrane. Small-conductance, Ca2+ -activated K+ channels (SK, KCa 2) are expressed in human atrial myocytes and are responsible for shaping atrial action potentials. Understanding the mechanisms of SK channel trafficking may provide new insights into the regulation controlling the repolarization of atrial myocytes. We have previously demonstrated that the C- and N-termini of SK2 channels interact with the actin-binding proteins α-actinin2 and filamin A, respectively. However, the roles of the interacting proteins on SK2 channel trafficking remain incompletely understood. Using total internal reflection fluorescence (TIRF) microscopy, we studied the mechanisms of surface membrane localization of SK2 (KCa 2.2) channels. When SK2 channels were co-expressed with filamin A or α-actinin2, the membrane fluorescence intensity of SK2 channels increased significantly. We next tested the effects of primaquine and dynasore on SK2 channels expression. Treatment with primaquine significantly reduced the membrane expression of SK2 channels. In contrast, treatment with dynasore failed to alter the surface membrane expression of SK2 channels. Further investigations using constitutively active or dominant-negative forms of Rab GTPases provided additional insights into the distinct roles of the two cytoskeletal proteins on the recycling processes of SK2 channels from endosomes. α-Actinin2 facilitated recycling of SK2 channels from both early and recycling endosomes while filamin A probably aids the recycling of SK2 channels from recycling endosomes.


Asunto(s)
Actinina/fisiología , Filaminas/fisiología , Miocitos Cardíacos/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Endosomas/metabolismo , Células HEK293 , Atrios Cardíacos/citología , Humanos , Hidrazonas/farmacología , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Primaquina/farmacología
12.
Biochim Biophys Acta ; 1863(8): 2072-83, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27188791

RESUMEN

Filamin A (FLNA) is known to act as platform for the signaling and intracellular trafficking of various GPCRs including dopamine D2 and D3 receptors (D2R, D3R). To understand molecular mechanisms involved in the FLNA-mediated regulation of D2R and D3R, comparative studies were conducted on the signaling and intracellular trafficking of the D2R and D3R in FLNA-knockdown cells, with a specific focus on the roles of the proteins that interact with FLNA and the D2R and D3R. Lowering the level of cellular FLNA caused an elevation in RalA activity and resulted in selective interference with the normal intracellular trafficking and signaling of the D2R and D3R, through GRK2 and ß-arrestins, respectively. Knockdown of FLNA or coexpression of active RalA interfered with the recycling of the internalized D2R and resulted in the development of receptor tolerance. Active RalA was found to interact with GRK2 to sequester it from D2R. Knockdown of FLNA or coexpression of active RalA prevented D3R from coupling with G protein. The selective involvement of GRK2- and ß-arrestins in the RalA-mediated cellular processes of the D2R and D3R was achieved via their different modes of interactions with the receptor and their distinct functional roles in receptor regulation. Our results show that FLNA is a multi-functional protein that acts as a platform on which D2R and D3R can interact with various proteins, through which selective regulation of these receptors occurs in combination with GRK2 and ß-arrestins.


Asunto(s)
Filaminas/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , beta-Arrestina 1/fisiología , Arrestina beta 2/fisiología , Proteínas de Unión al GTP ral/fisiología , Adenosina Trifosfato/metabolismo , Membrana Celular/metabolismo , AMP Cíclico/biosíntesis , Agonistas de Dopamina/farmacología , Genes Reporteros , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Transporte de Proteínas/fisiología , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Dopamina D3/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Proteínas de Unión al GTP ral/antagonistas & inhibidores
13.
Hum Mol Genet ; 25(13): 2776-2788, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206985

RESUMEN

Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.


Asunto(s)
Filaminas/genética , Filaminas/metabolismo , Miofibrillas/patología , Actinas/metabolismo , Animales , Técnicas de Cultivo de Célula , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Filaminas/fisiología , Humanos , Ratones , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Mutación , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Unión Proteica
14.
Cell Rep ; 14(9): 2050-2058, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923587

RESUMEN

Mutations in the filamin A (FlnA) gene are frequently associated with severe arterial abnormalities, although the physiological role for this cytoskeletal element remains poorly understood in vascular cells. We used a conditional mouse model to selectively delete FlnA in smooth muscle (sm) cells at the adult stage, thus avoiding the developmental effects of the knockout. Basal blood pressure was significantly reduced in conscious smFlnA knockout mice. Remarkably, pressure-dependent tone of the resistance caudal artery was lost, whereas reactivity to vasoconstrictors was preserved. Impairment of the myogenic behavior was correlated with a lack of calcium influx in arterial myocytes upon an increase in intraluminal pressure. Notably, the stretch activation of CaV1.2 was blunted in the absence of smFlnA. In conclusion, FlnA is a critical upstream element of the signaling cascade underlying the myogenic tone. These findings allow a better understanding of the molecular basis of arterial autoregulation and associated disease states.


Asunto(s)
Arterias/fisiología , Filaminas/fisiología , Animales , Presión Sanguínea , Señalización del Calcio , Células Cultivadas , Femenino , Riñón/irrigación sanguínea , Masculino , Mecanotransducción Celular , Ratones Noqueados , Desarrollo de Músculos , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología
15.
Biochem Biophys Res Commun ; 467(4): 730-5, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26482849

RESUMEN

Dictyostelium discoideum cells resemble in many aspects human leukocytes and serve as a model to study actin cytoskeleton dynamics and cell migration of highly motile cells. Dictyostelium cells deficient in the actin-binding protein filamin (ddFLN) showed a surprisingly subtle change in phenotype with no or only minor effects in single cell motility. These findings were in contrast to the strong actin-crosslinking activities measured for filamin in vitro. In the present study, we set out to revisit the role of ddFLN in cell migration. For this purpose, we examined migration of wild-type, ddFLN-null and ddFLN-overexpressing cells under different conditions. In addition to cyclic-AMP chemotaxis assays using micropipettes, we explored cell migration under more confined conditions: an under-agarose 2D assay and a 3D assay employing a collagen matrix that was adapted from assays for leukocytes. Using 3D migration conditions, cells deficient in ddFLN displayed only a minor impairment of motility, similar to the results obtained for migration in 2D. However, cells overexpressing ddFLN showed a remarkable decrease in the speed of migration in particular in 3D environments. We suggest that these results are in line with an increased stiffening of the cortex due to the crosslinking activity of overexpressed ddFLN. Our conclusion is that the absolute level of ddFLN is critical for efficient migration. Furthermore, our results show that under conditions of increased mechanical stress, Dictyostelium cells, like leukocytes, switch to a bleb-based mode of movement.


Asunto(s)
Quimiotaxis , Dictyostelium/fisiología , Dictyostelium/citología , Filaminas/fisiología
16.
Soft Matter ; 11(27): 5435-46, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26059185

RESUMEN

The actin cytoskeleton plays a key role in the deformability of the cell and in mechanosensing. Here we analyze the contributions of three major actin cross-linking proteins, myosin II, α-actinin and filamin, to cell deformability, by using micropipette aspiration of Dictyostelium cells. We examine the applicability of three simple mechanical models: for small deformation, linear viscoelasticity and drop of liquid with a tense cortex; and for large deformation, a Newtonian viscous fluid. For these models, we have derived linearized equations and we provide a novel, straightforward methodology to analyze the experiments. This methodology allowed us to differentiate the effects of the cross-linking proteins in the different regimes of deformation. Our results confirm some previous observations and suggest important relations between the molecular characteristics of the actin-binding proteins and the cell behavior: the effect of myosin is explained in terms of the relation between the lifetime of the bond to actin and the resistive force; the presence of α-actinin obstructs the deformation of the cytoskeleton, presumably mainly due to the higher molecular stiffness and to the lower dissociation rate constants; and filamin contributes critically to the global connectivity of the network, possibly by rapidly turning over cross-links during the remodeling of the cytoskeletal network, thanks to the higher rate constants, flexibility and larger size. The results suggest a sophisticated relationship between the expression levels of actin-binding proteins, deformability and mechanosensing.


Asunto(s)
Actinina/fisiología , Dictyostelium/citología , Filaminas/fisiología , Miosina Tipo II/fisiología , Proteínas Protozoarias/fisiología , Algoritmos , Dictyostelium/fisiología , Modelos Lineales , Mecanotransducción Celular , Modelos Biológicos , Viscosidad
17.
Blood ; 126(1): 80-8, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25838348

RESUMEN

Bin-Amphiphysin-Rvs (BAR) and Fes-CIP4 homology BAR (F-BAR) proteins generate tubular membrane invaginations reminiscent of the megakaryocyte (MK) demarcation membrane system (DMS), which provides membranes necessary for future platelets. The F-BAR protein PACSIN2 is one of the most abundant BAR/F-BAR proteins in platelets and the only one reported to interact with the cytoskeletal and scaffold protein filamin A (FlnA), an essential regulator of platelet formation and function. The FlnA-PACSIN2 interaction was therefore investigated in MKs and platelets. PACSIN2 associated with FlnA in human platelets. The interaction required FlnA immunoglobulin-like repeat 20 and the tip of PACSIN2 F-BAR domain and enhanced PACSIN2 F-BAR domain membrane tubulation in vitro. Most human and wild-type mouse platelets had 1 to 2 distinct PACSIN2 foci associated with cell membrane GPIbα, whereas Flna-null platelets had 0 to 4 or more foci. Endogenous PACSIN2 and transfected enhanced green fluorescent protein-PACSIN2 were concentrated in midstage wild-type mouse MKs in a well-defined invagination of the plasma membrane reminiscent of the initiating DMS and dispersed in the absence of FlnA binding. The DMS appeared less well defined, and platelet territories were not readily visualized in Flna-null MKs. We conclude that the FlnA-PACSIN2 interaction regulates membrane tubulation in MKs and platelets and likely contributes to DMS formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Plaquetas , Membrana Celular/ultraestructura , Filaminas/metabolismo , Megacariocitos , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Plaquetas/metabolismo , Plaquetas/ultraestructura , Membrana Celular/metabolismo , Células Cultivadas , Filaminas/fisiología , Células HEK293 , Humanos , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Seudópodos/metabolismo
18.
Sci Rep ; 5: 8622, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25722249

RESUMEN

Mechanotransduction has been divided into mechanotransmission, mechanosensing, and mechanoresponse, although how a cell performs all three functions using the same set of structural components is still highly debated. Here, we bridge the gap between emerging molecular and systems-level understandings of mechanotransduction through a multiscale model linking these three phases. Our model incorporates a discrete network of actin filaments and associated proteins that responds to stretching through geometric relaxation. We assess three potential activating mechanisms at mechanosensitive crosslinks as inputs to a mixture model of molecular release and benchmark each using experimental data of mechanically-induced Rho GTPase FilGAP release from actin-filamin crosslinks. Our results suggest that filamin-FilGAP mechanotransduction response is best explained by a bandpass mechanism favoring release when crosslinking angles fall outside of a specific range. Our model further investigates the difference between ordered versus disordered networks and finds that a more disordered actin network may allow a cell to more finely tune control of molecular release enabling a more robust response.


Asunto(s)
Mecanotransducción Celular , Citoesqueleto de Actina/fisiología , Simulación por Computador , Proteínas del Citoesqueleto/fisiología , Filaminas/fisiología , Proteínas Activadoras de GTPasa/fisiología , Modelos Biológicos
19.
Nat Commun ; 5: 4656, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25120197

RESUMEN

Endogenously and externally generated mechanical forces influence diverse cellular activities, a phenomenon defined as mechanotransduction. Deformation of protein domains by application of stress, previously documented to alter macromolecular interactions in vitro, could mediate these effects. We engineered a photon-emitting system responsive to unfolding of two repeat domains of the actin filament (F-actin) crosslinker protein filamin A (FLNA) that binds multiple partners involved in cell signalling reactions and validated the system using F-actin networks subjected to myosin-based contraction. Expressed in cultured cells, the sensor-containing FLNA construct reproducibly reported FLNA domain unfolding strikingly localized to dynamic, actively protruding, leading cell edges. The unfolding signal depends upon coherence of F-actin-FLNA networks and is enhanced by stimulating cell contractility. The results establish protein domain distortion as a bona fide mechanism for mechanotransduction in vivo.


Asunto(s)
Movimiento Celular/fisiología , Filaminas/química , Filaminas/fisiología , Riñón/fisiología , Mecanotransducción Celular/fisiología , Actinas/fisiología , Animales , Fenómenos Biomecánicos/fisiología , Células COS , Células Cultivadas , Chlorocebus aethiops , Transferencia Resonante de Energía de Fluorescencia/métodos , Técnicas In Vitro , Riñón/citología , Óptica y Fotónica/métodos
20.
Endocrinology ; 155(8): 2932-41, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24828612

RESUMEN

Somatostatin receptor type 2 (SST2) is the main pharmacological target of medical therapy for GH-secreting pituitary tumors, but molecular mechanisms regulating its expression and signaling are largely unknown. The aim of this study was to investigate the role of cytoskeleton protein filamin A (FLNA) in SST2 expression and signaling in somatotroph tumor cells. We found a highly variable expression of FLNA in human GH-secreting tumors, without a correlation with SST2 levels. FLNA silencing in human tumoral cells did not affect SST2 expression and localization but abolished the SST2-induced reduction of cyclin D1 (-37% ± 15% in control cells, P < .05 vs basal) and caspase-3/7 activation (+63% ± 31% in control cells, P < .05 vs basal). Overexpression of a FLNA dominant-negative mutant that specifically prevents SST2-FLNA binding reduced SST2 expression after prolonged agonist exposure (-55% ± 5%, P < .01 vs untreated cells) in GH3 cells. Moreover, SST2-induced apoptotic effect (77% ± 54% increase of caspase activity, P < .05 vs basal) and SST2-mediated ERK1/2 inhibition (48% ± 17% reduction of ERK1/2 phosphorylation, P < .01 vs basal) were abrogated in cells overexpressing another FLNA mutant that prevents FLNA interaction with partner proteins but not with SST2, suggesting a scaffold function of FLNA in somatotrophs. In conclusion, these data demonstrate that FLNA is involved in SST2 stabilization and signaling in tumoral somatotrophs, playing both a structural and functional role.


Asunto(s)
Filaminas/fisiología , Receptores de Somatostatina/fisiología , Transducción de Señal/fisiología , Somatotrofos/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Estabilidad Proteica , Ratas , Receptores de Somatostatina/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA