Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Exp Cell Res ; 403(2): 112613, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33901448

RESUMO

The Hippo signaling pathway is a tumor suppressor pathway that plays an important role in tissue homeostasis and organ size control. KIBRA is one of the many upstream regulators of the Hippo pathway. It functions as a tumor suppressor by positively regulating the core Hippo kinase cascade. However, there are accumulating shreds of evidence showing that KIBRA has an oncogenic function, which we speculate may arise from its functions away from the Hippo pathway. In this review, we have attempted to provide an overview of the Hippo signaling with a special emphasis on evidence showing the paradoxical role of KIBRA in cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Junções Aderentes/metabolismo , Junções Aderentes/ultraestrutura , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Via de Sinalização Hippo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Serina-Treonina Quinase 3 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
2.
Structure ; 29(5): 488-498.e4, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33476550

RESUMO

The actin cytoskeleton plays a fundamental role in numerous cellular processes, such as cell motility, cytokinesis, and adhesion to the extracellular matrix. Revealing the polarity of individual actin filaments in intact cells would foster an unprecedented understanding of cytoskeletal processes and their associated mechanical forces. Cryo-electron tomography provides the means for high-resolution structural imaging of cells. However, the low signal-to-noise ratio of cryo-tomograms obscures the high frequencies, and therefore the polarity of actin filaments cannot be directly measured. Here, we developed a method that enables us to determine the polarity of actin filaments in cellular cryo-tomograms. We applied it to reveal the actin polarity distribution in focal adhesions, and show a linear relation between actin polarity and distance from the apical boundary of the adhesion site.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Adesões Focais/ultraestrutura , Actinas/química , Animais , Linhagem Celular , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Camundongos
3.
Methods Mol Biol ; 2217: 27-37, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33215374

RESUMO

Focal adhesions in planar substrates constitute an excellent cellular resource to evaluate different parameters related to cell morphology, cytoskeletal organization, and adhesive strength. However, their intrinsic heterogeneity in terms of size, molecular composition, orientation, and so on complicates their analysis. Here, we describe a simple and straightforward ImageJ/Fiji-based method to quantify several parameters that describe the morphology and relative composition of focal adhesions. This type of analysis can be implemented in various ways and become useful for drug and shRNA screenings.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Matriz Extracelular/ultraestrutura , Adesões Focais/ultraestrutura , Processamento de Imagem Assistida por Computador/estatística & dados numéricos , Imagem Molecular/métodos , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Animais , Células CHO , Adesão Celular , Linhagem Celular Tumoral , Cricetulus , Matriz Extracelular/metabolismo , Fibronectinas/química , Fibronectinas/metabolismo , Adesões Focais/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Faloidina/química
4.
Methods Mol Biol ; 2217: 71-81, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33215378

RESUMO

The in situ proximity ligation assay (PLA) is capable of detecting single protein events such as protein protein-interactions and posttranslational modifications (e.g., protein phosphorylation) in tissue and cell samples prepared for analysis by immunofluorescent or immunohistochemical microscopy. The targets are detected using two primary antibodies which must be from different host species. A pair of secondary antibodies (PLA probes) conjugated to complementary oligonucleotides is applied to the sample, and a signal is generated only when the two PLA probes are in close proximity by their binding to the two primary antibodies that have bound to their targets in close proximity. The signal from each pair of PLA probes is visualized as an individual fluorescent spot. These PLA signals can be quantified (counted) using image analysis software (ImageJ), and also assigned to a specific subcellular location based on microscopy image overlays. In principle, in situ PLA offers a relatively simple and sensitive technique to analyze interactions among any proteins for which suitable antibodies are available. Integrin-mediated focal adhesions (FAs) are large multiprotein complexes consisting of more than 150 proteins, also known as the integrin adhesome, which link the extracellular matrix (ECM) to the actin cytoskeleton and regulate the functioning of mechanosignaling pathways. The in situ PLA approach is well suited for examining the spatiotemporal aspects of protein posttranslational modifications and protein interactions occurring in dynamic multiprotein complexes such as integrin mediated focal adhesions.


Assuntos
Adesões Focais/metabolismo , Imuno-Histoquímica/métodos , Cadeias alfa de Integrinas/metabolismo , Integrina beta1/metabolismo , Complexos Multiproteicos/metabolismo , Oligonucleotídeos/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Anticorpos/química , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Adesões Focais/ultraestrutura , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestrutura , Humanos , Processamento de Imagem Assistida por Computador , Cadeias alfa de Integrinas/química , Integrina beta1/química , Microscopia de Fluorescência , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Complexos Multiproteicos/química , Músculo Liso/metabolismo , Músculo Liso/ultraestrutura , Oligonucleotídeos/síntese química , Ligação Proteica
5.
Methods Mol Biol ; 2217: 183-195, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33215382

RESUMO

Surface nanopatterning allows for the creation of spatially controlled binding sites for extracellular matrix ligands and the modulation of receptor binding sites. Here we describe the preparation of gold nanopatterned substrates using diblock micellar nanolithography to immobilize integrin ligands at defined spacing and combined with molecular tension sensors to measure molecular forces as function of integrin lateral clustering.


Assuntos
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Adesões Focais/metabolismo , Integrinas/química , Oligodesoxirribonucleotídeos/química , Estereolitografia , Animais , Sítios de Ligação , Adesão Celular , Movimento Celular , Cloretos/química , Matriz Extracelular/ultraestrutura , Fibroblastos/ultraestrutura , Fibronectinas/química , Fibronectinas/metabolismo , Adesões Focais/ultraestrutura , Compostos de Ouro/química , Integrinas/metabolismo , Ligantes , Camundongos , Micelas , Microscopia de Fluorescência/métodos , Nanoestruturas/química , Polietilenoglicóis/química , Polimerização , Ligação Proteica , Piridinas/química , Propriedades de Superfície
6.
FEBS Lett ; 595(4): 532-547, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33314143

RESUMO

Integrin-mediated adhesion regulates cellular responses to changes in the mechanical and biochemical properties of the extracellular matrix. Cell-matrix adhesion regulates caveolar endocytosis, dependent on caveolin 1 (Cav1) Tyr14 phosphorylation (pY14Cav1), to control anchorage-dependent signaling. We find that cell-matrix adhesion regulates pY14Cav1 levels in mouse fibroblasts. Biochemical fractionation reveals endogenous pY14Cav1 to be present in caveolae and focal adhesions (FA). Adhesion does not affect caveolar pY14Cav1, supporting its regulation at FA, in which PF-228-mediated inhibition of focal adhesion kinase (FAK) disrupts. Cell adhesion on 2D polyacrylamide matrices of increasing stiffness stimulates Cav1 phosphorylation, which is comparable to the phosphorylation of FAK. Inhibition of FAK across varying stiffnesses shows it regulates pY14Cav1 more prominently at higher stiffness. Taken together, these studies reveal the presence of FAK-pY14Cav1 crosstalk at FA, which is regulated by cell-matrix adhesion.


Assuntos
Caveolina 1/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Quinase 1 de Adesão Focal/genética , Processamento de Proteína Pós-Traducional , Tirosina/metabolismo , Animais , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Cavéolas/ultraestrutura , Caveolina 1/deficiência , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Embrião de Mamíferos , Endocitose/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/ultraestrutura , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Mecanotransdução Celular , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Sulfonas/farmacologia
7.
Biomolecules ; 10(12)2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33321985

RESUMO

Barrier function is a vital homeostatic mechanism employed by epithelial and endothelial tissue. Diseases across a wide range of tissue types involve dynamic changes in transcellular junctional complexes and the actin cytoskeleton in the regulation of substance exchange across tissue compartments. In this review, we focus on the contribution of the gap junction protein, Cx43, to the biophysical and biochemical regulation of barrier function. First, we introduce the structure and canonical channel-dependent functions of Cx43. Second, we define barrier function and examine the key molecular structures fundamental to its regulation. Third, we survey the literature on the channel-dependent roles of connexins in barrier function, with an emphasis on the role of Cx43 and the actin cytoskeleton. Lastly, we discuss findings on the channel-independent roles of Cx43 in its associations with the actin cytoskeleton and focal adhesion structures highlighted by PI3K signaling, in the potential modulation of cellular barriers. Mounting evidence of crosstalk between connexins, the cytoskeleton, focal adhesion complexes, and junctional structures has led to a growing appreciation of how barrier-modulating mechanisms may work together to effect solute and cellular flux across tissue boundaries. This new understanding could translate into improved therapeutic outcomes in the treatment of barrier-associated diseases.


Assuntos
Citoesqueleto de Actina/metabolismo , Conexina 43/metabolismo , Doença da Artéria Coronariana/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Esclerose Múltipla/metabolismo , Síndrome do Desconforto Respiratório/metabolismo , Acidente Vascular Cerebral/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Transporte Biológico , Conexina 43/genética , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Regulação da Expressão Gênica , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/patologia , Transdução de Sinais , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
8.
Cells ; 9(8)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32751580

RESUMO

Endothelial cellular stiffening has been observed not only in inflamed cultured endothelial cells but also in the endothelium of atherosclerotic regions, which is an underlying cause of monocyte adhesion and accumulation. Although recombinant soluble thrombomodulin (rsTM) has been reported to suppress the inflammatory response of endothelial cells, its role in regulating endothelial cellular stiffness remains unclear. The purpose of this study was to investigate the impact of anticoagulant rsTM on lipopolysaccharide (LPS)-induced endothelial cellular stiffening. We show that LPS increases endothelial cellular stiffness by using atomic force microscopy and that rsTM reduces LPS-induced cellular stiffening not only through the attenuation of actin fiber and focal adhesion formation but also via the improvement of gap junction functionality. Moreover, post-administration of rsTM, after LPS stimulation, attenuated LPS-induced cellular stiffening. We also found that endothelial cells regulate leukocyte adhesion in a substrate- and cellular stiffness-dependent manner. Our result show that LPS-induced cellular stiffening enhances monocytic THP-1 cell line adhesion, whereas rsTM suppresses THP-1 cell adhesion to inflamed endothelial cells by reducing cellular stiffness. Endothelial cells increase cellular stiffness in reaction to inflammation, thereby promoting monocyte adhesion. Treatment of rsTM reduced LPS-induced cellular stiffening and suppressed monocyte adhesion in a cellular stiffness-dependent manner.


Assuntos
Actinas/ultraestrutura , Adesão Celular/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/metabolismo , Trombomodulina/administração & dosagem , Trombomodulina/química , Anticoagulantes/administração & dosagem , Anticoagulantes/química , Aterosclerose/metabolismo , Adesões Focais/ultraestrutura , Junções Comunicantes/ultraestrutura , Humanos , Inflamação/tratamento farmacológico , Microscopia de Força Atômica , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Células THP-1
9.
J Cell Biol ; 219(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32525992

RESUMO

The members of the Rab family of small GTPases are molecular switches that regulate distinct steps in different membrane traffic pathways. In addition to this canonical function, Rabs can play a role in other processes, such as cell adhesion and motility. Here, we reveal the role of the small GTPase Rab18 as a positive regulator of directional migration in chemotaxis, and the underlying mechanism. We show that knockdown of Rab18 reduces the size of focal adhesions (FAs) and influences their dynamics. Furthermore, we found that Rab18, by directly interacting with the endoplasmic reticulum (ER)-resident protein kinectin-1, controls the anterograde kinesin-1-dependent transport of the ER required for the maturation of nascent FAs and protrusion orientation toward a chemoattractant. Altogether, our data support a model in which Rab18 regulates kinectin-1 transport toward the cell surface to form ER-FA contacts, thus promoting FA growth and cell migration during chemotaxis.


Assuntos
Membrana Celular/metabolismo , Quimiotaxia/genética , Retículo Endoplasmático/metabolismo , Adesões Focais/metabolismo , Proteínas de Membrana/genética , Proteínas rab de Ligação ao GTP/genética , Transporte Biológico , Adesão Celular , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/ultraestrutura , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Fosforilação , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Proteínas rab de Ligação ao GTP/antagonistas & inibidores , Proteínas rab de Ligação ao GTP/metabolismo
10.
PLoS One ; 15(6): e0234430, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32511274

RESUMO

Excess presence of the human epidermal growth factor receptor 2 (HER2) as well as of the focal adhesion protein complexes are associated with increased proliferation, migratory, and invasive behavior of cancer cells. A cross-regulation between HER2 and integrin signaling pathways has been found, but the exact mechanism remains elusive. Here, we investigated whether HER2 colocalizes with focal adhesion complexes on breast cancer cells overexpressing HER2. For this purpose, vinculin or talin green fluorescent protein (GFP) fusion proteins, both key constituents of focal adhesions, were expressed in breast cancer cells. HER2 was either extracellularly or intracellularly labeled with fluorescent quantum dots nanoparticles (QDs). The cell-substrate interface was analyzed at the location of the focal adhesions by means of total internal reflection fluorescent microscopy or correlative fluorescence- and scanning transmission electron microscopy. Expression of HER2 at the cell-substrate interface was only observed upon intracellular labeling, and was heterogeneous with both HER2-enriched and -low regions. In contrast to an expected enrichment of HER2 at focal adhesions, an anti-correlated expression pattern was observed for talin and HER2. Our findings suggest a spatial anti-correlation between HER2 and focal adhesion complexes for adherent cells.


Assuntos
Membrana Celular/metabolismo , Adesões Focais/metabolismo , Receptor ErbB-2/metabolismo , Análise Espacial , Adesão Celular , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Adesões Focais/ultraestrutura , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia Eletrônica de Transmissão e Varredura , Microscopia de Fluorescência , Receptor ErbB-2/análise , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Talina/análise , Talina/genética , Talina/metabolismo , Vinculina/análise , Vinculina/genética , Vinculina/metabolismo
11.
Adv Biosyst ; 4(8): e2000092, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32500640

RESUMO

Nanotopography mimicking extracellular environments reportedly impact cell morphological changes; however, elucidating this relationship has been challenging. To control cellular responses using nanostructures, in this study, the quantitative relationship between nanotopography and cell spreading mediated by focal adhesions (FAs) is demonstrated using adipose-derived stem cells (ASCs). The spreading of ASCs and area of FAs are analyzed for the distribution of filamentous actin and vinculin, respectively, using fluorescent images. FAs require a specific area for adhesion (herein defined as effective contact area [ECA]) to maintain cell attachment on nanopillar arrays. An ECA is the area of FAs supported by nanopillars, multiplying the area fraction (AF) of their top surface. Regarding the spreading of cells, the mean area of ASCs linearly decreases as the mean area of FAs increases. Because the area of FAs is inversely correlated to the AF of the nanopillar arrays, the spreading of cells can be quantitatively correlated with nanotopography. The results provide a conceptual framework for controlling cell behaviors to design artificial substrates for tissue-engineering applications.


Assuntos
Adipócitos/citologia , Fluorocarbonos/farmacologia , Adesões Focais/efeitos dos fármacos , Silanos/farmacocinética , Células-Tronco/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/fisiologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Ácidos Graxos , Adesões Focais/ultraestrutura , Humanos , Nanoestruturas/química , Nanoestruturas/ultraestrutura , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Engenharia Tecidual/métodos
12.
Cell Signal ; 72: 109646, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32311505

RESUMO

In autosomal dominant polycystic kidney disease (ADPKD), the inexorable growth of numerous fluid-filled cysts leads to massively enlarged kidneys, renal interstitial damage, inflammation, and fibrosis, and progressive decline in kidney function. It has long been recognized that interstitial fibrosis is the most important manifestation associated with end-stage renal disease; however, the role of abnormal extracellular matrix (ECM) production on ADPKD pathogenesis is not fully understood. Early evidence showed that cysts in end-stage human ADPKD kidneys had thickened and extensively laminated cellular basement membranes, and abnormal regulation of gene expression of several basement membrane components, including collagens, laminins, and proteoglycans by cyst epithelial cells. These basement membrane changes were also observed in dilated tubules and small cysts of early ADPKD kidneys, indicating that ECM alterations were early features of cyst development. Renal cystic cells were also found to overexpress several integrins and their ligands, including ECM structural components and soluble matricellular proteins. ECM ligands binding to integrins stimulate focal adhesion formation and can promote cell attachment and migration. Abnormal expression of laminin-332 (laminin-5) and its receptor α6ß4 stimulated cyst epithelial cell proliferation; and mice that lacked laminin α5, a component of laminin-511 normally expressed by renal tubules, had an overexpression of laminin-332 that was associated with renal cyst formation. Periostin, a matricellular protein that binds αVß3- and αVß5-integrins, was found to be highly overexpressed in the kidneys of ADPKD and autosomal recessive PKD patients, and several rodent models of PKD. αVß3-integrin is also overexpressed by cystic epithelial cells, and the binding of periostin to αVß3-integrin activates the integrin-linked kinase and downstream signal transduction pathways involved in tissue repair promoting cyst growth, ECM synthesis, and tissue fibrosis. This chapter reviews the roles of the ECM, integrins, and focal adhesion signaling in cyst growth and fibrosis in PKD.


Assuntos
Matriz Extracelular/metabolismo , Adesões Focais/metabolismo , Integrinas/metabolismo , Doenças Renais Policísticas/metabolismo , Transdução de Sinais , Animais , Matriz Extracelular/ultraestrutura , Adesões Focais/ultraestrutura , Humanos , Modelos Biológicos
13.
Am J Physiol Endocrinol Metab ; 318(6): E930-E942, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32343611

RESUMO

Preterm birth is one of the most common complications during human pregnancy and is associated with a dramatic switch within the uterus from quiescence to contractility. However, the mechanisms underlying uterine remodeling are largely unknown. Protein kinases and phosphatases play critical roles in regulating the phosphorylation of proteins involved in the smooth muscle cell functions. In the present study, we found that Src-homology phosphatase type-1 (SHP-1, PTPN6) was significantly decreased in human myometrium in labor compared with that not in labor. Timed-pregnant mice injected intraperitoneally with the specific SHP-1 inhibitor protein tyrosine phosphatase inhibitor I (PTPI-1) manifested significantly preterm labor, with enriched plasmalemmal dense plaques between myometrial cells and increased phosphorylation at Tyr397 and Tyr576/577 sites of focal adhesion kinase (FAK) in myometrial cells, which remained to the time of labor, whereas the phosphorylation levels of ERK1/2 and phosphatidylinositol 3 kinase (PI3K) showed a rapid increase upon PTPI-1 injection but fell back to normal at the time of labor. The Tyr576/577 in FAK played an important role in the interaction between FAK and SHP-1. Knockdown of SHP-1 dramatically increased the spontaneous contraction of human uterine smooth muscle cells (HUSMCs), which was reversed by coinfection of a FAK-knockdown lentivirus. PGF2α downregulated SHP-1 via PLCß-PKC-NF-κB or PI3K-NF-κB pathways, suggesting the regenerative downregulation of SHP-1 enhances the uterine remodeling and plasticity by activating FAK and subsequent focal adhesion pathway, which eventually facilitates myometrium contraction and leads to labor. The study sheds new light on understanding of mechanisms that underlie the initiation of labor, and interventions for modulation of SHP-1 may provide a potential strategy for preventing preterm birth.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Trabalho de Parto/metabolismo , Miócitos de Músculo Liso/metabolismo , Miométrio/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Adulto , Animais , Dinoprosta/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Adesões Focais/ultraestrutura , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/ultraestrutura , Miométrio/citologia , Miométrio/efeitos dos fármacos , Miométrio/ultraestrutura , NF-kappa B/metabolismo , Trabalho de Parto Prematuro , Ocitócicos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C beta/metabolismo , Gravidez , Proteína Quinase C/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/antagonistas & inibidores
14.
Nano Lett ; 20(4): 2230-2245, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32142297

RESUMO

Cellular mechanics play a crucial role in tissue homeostasis and are often misregulated in disease. Traction force microscopy is one of the key methods that has enabled researchers to study fundamental aspects of mechanobiology; however, traction force microscopy is limited by poor resolution. Here, we propose a simplified protocol and imaging strategy that enhances the output of traction force microscopy by increasing i) achievable bead density and ii) the accuracy of bead tracking. Our approach relies on super-resolution microscopy, enabled by fluorescence fluctuation analysis. Our pipeline can be used on spinning-disk confocal or widefield microscopes and is compatible with available analysis software. In addition, we demonstrate that our workflow can be used to gain biologically relevant information and is suitable for fast long-term live measurement of traction forces even in light-sensitive cells. Finally, using fluctuation-based traction force microscopy, we observe that filopodia align to the force field generated by focal adhesions.


Assuntos
Microscopia de Força Atômica/métodos , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Adesões Focais/ultraestrutura , Humanos , Microscopia de Força Atômica/instrumentação , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Imagem Óptica/instrumentação , Imagem Óptica/métodos , Pseudópodes/ultraestrutura
15.
Biomolecules ; 10(2)2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31991559

RESUMO

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase with key roles in the regulation of cell adhesion migration, proliferation and survival. In cancer FAK is a major driver of invasion and metastasis and its upregulation is associated with poor patient prognosis. FAK is autoinhibited in the cytosol, but activated upon localisation into a protein complex, known as focal adhesion complex. This complex forms upon cell adhesion to the extracellular matrix (ECM) at the cytoplasmic side of the plasma membrane at sites of ECM attachment. FAK is anchored to the complex via multiple sites, including direct interactions with specific membrane lipids and connector proteins that attach focal adhesions to the actin cytoskeleton. In migrating cells, the contraction of actomyosin stress fibres attached to the focal adhesion complex apply a force to the complex, which is likely transmitted to the FAK protein, causing stretching of the FAK molecule. In this review we discuss the current knowledge of the FAK structure and how specific structural features are involved in the regulation of FAK signalling. We focus on two major regulatory mechanisms known to contribute to FAK activation, namely interactions with membrane lipids and stretching forces applied to FAK, and discuss how they might induce structural changes that facilitate FAK activation.


Assuntos
Adesão Celular/genética , Quinase 1 de Adesão Focal/genética , Adesões Focais/genética , Mecanotransdução Celular/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/ultraestrutura , Movimento Celular , Quinase 1 de Adesão Focal/ultraestrutura , Adesões Focais/ultraestrutura , Humanos , Membranas/ultraestrutura , Fosforilação , Transdução de Sinais/genética
16.
PLoS One ; 15(1): e0220019, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31945053

RESUMO

The migration of cancer cells is highly regulated by the biomechanical properties of their local microenvironment. Using 3D scaffolds of simple composition, several aspects of cancer cell mechanosensing (signal transduction, EMC remodeling, traction forces) have been separately analyzed in the context of cell migration. However, a combined study of these factors in 3D scaffolds that more closely resemble the complex microenvironment of the cancer ECM is still missing. Here, we present a comprehensive, quantitative analysis of the role of cell-ECM interactions in cancer cell migration within a highly physiological environment consisting of mixed Matrigel-collagen hydrogel scaffolds of increasing complexity that mimic the tumor microenvironment at the leading edge of cancer invasion. We quantitatively show that the presence of Matrigel increases hydrogel stiffness, which promotes ß1 integrin expression and metalloproteinase activity in H1299 lung cancer cells. Then, we show that ECM remodeling activity causes matrix alignment and compaction that favors higher tractions exerted by the cells. However, these traction forces do not linearly translate into increased motility due to a biphasic role of cell adhesions in cell migration: at low concentration Matrigel promotes migration-effective tractions exerted through a high number of small sized focal adhesions. However, at high Matrigel concentration, traction forces are exerted through fewer, but larger focal adhesions that favor attachment yielding lower cell motility.


Assuntos
Colágeno/farmacologia , Células Epiteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Laminina/farmacologia , Mecanotransdução Celular , Proteoglicanas/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Colágeno/química , Combinação de Medicamentos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Adesões Focais/ultraestrutura , Expressão Gênica , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Laminina/química , Modelos Biológicos , Proteoglicanas/química , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Propriedades de Superfície , Microambiente Tumoral/efeitos dos fármacos
17.
Nat Cell Biol ; 21(11): 1357-1369, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31659275

RESUMO

αMß2 integrin (complement receptor 3) is a major receptor for phagocytosis in macrophages. In other contexts, integrins' activities and functions are mechanically linked to actin dynamics through focal adhesions. We asked whether mechanical coupling of αMß2 integrin to the actin cytoskeleton mediates phagocytosis. We found that particle internalization was driven by formation of Arp2/3 and formin-dependent actin protrusions that wrapped around the particle. Focal complex-like adhesions formed in the phagocytic cup that contained ß2 integrins, focal adhesion proteins and tyrosine kinases. Perturbation of talin and Syk demonstrated that a talin-dependent link between integrin and actin and Syk-mediated recruitment of vinculin enable force transmission to target particles and promote phagocytosis. Altering target mechanical properties demonstrated more efficient phagocytosis of stiffer targets. Thus, macrophages use tyrosine kinase signalling to build a mechanosensitive, talin- and vinculin-mediated, focal adhesion-like molecular clutch, which couples integrins to cytoskeletal forces to drive particle engulfment.


Assuntos
Macrófagos/imunologia , Mecanotransdução Celular , Fagocitose/imunologia , Quinase Syk/genética , Talina/genética , Vinculina/genética , Citoesqueleto de Actina/imunologia , Citoesqueleto de Actina/ultraestrutura , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/imunologia , Actinas/genética , Actinas/imunologia , Animais , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Adesões Focais/imunologia , Adesões Focais/ultraestrutura , Forminas/genética , Forminas/imunologia , Regulação da Expressão Gênica , Humanos , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Fagossomos/imunologia , Fagossomos/ultraestrutura , Poliestirenos , Cultura Primária de Células , Células RAW 264.7 , Quinase Syk/imunologia , Células THP-1 , Talina/imunologia , Vinculina/imunologia
18.
J Cell Biol ; 218(8): 2481-2491, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31315944

RESUMO

Cell sensing of externally applied mechanical strain through integrin-mediated adhesions is critical in development and physiology of muscle, lung, tendon, and arteries, among others. We examined the effects of strain on force transmission through the essential cytoskeletal linker talin. Using a fluorescence-based talin tension sensor (TS), we found that uniaxial stretch of cells on elastic substrates increased tension on talin, which was unexpectedly independent of the orientation of the focal adhesions relative to the direction of strain. High-resolution electron microscopy of the actin cytoskeleton revealed that stress fibers (SFs) are integrated into an isotropic network of cortical actin filaments in which filamin A (FlnA) localizes preferentially to points of intersection between SFs and cortical actin. Knockdown (KD) of FlnA resulted in more isolated, less integrated SFs. After FlnA KD, tension on talin was polarized in the direction of stretch, while FlnA reexpression restored tensional symmetry. These data demonstrate that a FlnA-dependent cortical actin network distributes applied forces over the entire cytoskeleton-matrix interface.


Assuntos
Actinas/metabolismo , Filaminas/metabolismo , Estresse Mecânico , Animais , Fenômenos Biomecânicos , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Células NIH 3T3 , Fibras de Estresse/metabolismo , Fibras de Estresse/ultraestrutura , Talina/metabolismo
19.
FASEB J ; 33(10): 10618-10632, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31225977

RESUMO

Biomechanical cues within tissue microenvironments are critical for maintaining homeostasis, and their disruption can contribute to malignant transformation and metastasis. Once transformed, metastatic cancer cells can migrate persistently by adapting (plasticity) to changes in the local fibrous extracellular matrix, and current strategies to recapitulate persistent migration rely exclusively on the use of aligned geometries. Here, the controlled interfiber spacing in suspended crosshatch networks of nanofibers induces cells to exhibit plasticity in migratory behavior (persistent and random) and the associated cytoskeletal arrangement. At dense spacing (3 and 6 µm), unexpectedly, elongated cells migrate persistently (in 1 dimension) at high speeds in 3-dimensional shapes with thick nuclei, and short focal adhesion cluster (FAC) lengths. With increased spacing (18 and 36 µm), cells attain 2-dimensional morphologies, have flattened nuclei and longer FACs, and migrate randomly by rapidly detaching their trailing edges that strain the nuclei by ∼35%. At 54-µm spacing, kite-shaped cells become near stationary. Poorly developed filamentous actin stress fibers are found only in cells on 3-µm networks. Gene-expression profiling shows a decrease in transcriptional potential and a differential up-regulation of metabolic pathways. The consistency in observed phenotypes across cell lines supports using this platform to dissect hallmarks of plasticity in migration in vitro.-Jana, A., Nookaew, I., Singh, J., Behkam, B., Franco, A. T., Nain, A. S. Crosshatch nanofiber networks of tunable interfiber spacing induce plasticity in cell migration and cytoskeletal response.


Assuntos
Movimento Celular/fisiologia , Citoesqueleto/fisiologia , Citoesqueleto de Actina/fisiologia , Citoesqueleto de Actina/ultraestrutura , Animais , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Movimento Celular/genética , Núcleo Celular/fisiologia , Núcleo Celular/ultraestrutura , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/ultraestrutura , Microambiente Celular/genética , Microambiente Celular/fisiologia , Citoesqueleto/ultraestrutura , Matriz Extracelular/fisiologia , Matriz Extracelular/ultraestrutura , Adesões Focais/fisiologia , Adesões Focais/ultraestrutura , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Modelos Biológicos , Nanofibras/ultraestrutura
20.
PLoS Genet ; 15(5): e1008083, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31116733

RESUMO

How biochemical and mechanical information are integrated during tissue development is a central question in morphogenesis. In many biological systems, the PIX-GIT complex localises to focal adhesions and integrates both physical and chemical information. We used Drosophila melanogaster egg chamber formation to study the function of PIX and GIT orthologues (dPix and Git, respectively), and discovered a central role for this complex in controlling myosin activity and epithelial monolayering. We found that Git's focal adhesion targeting domain mediates basal localisation of this complex to filament structures and the leading edge of migrating cells. In the absence of dpix and git, tissue disruption is driven by contractile forces, as reduction of myosin activators restores egg production and morphology. Further, dpix and git mutant eggs closely phenocopy defects previously reported in pak mutant epithelia. Together, these results indicate that the dPix-Git complex controls egg chamber morphogenesis by controlling myosin contractility and Pak kinase downstream of focal adhesions.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Ativadoras de GTPase/genética , Morfogênese/genética , Miosinas/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Proteínas Ativadoras de GTPase/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular , Miosinas/metabolismo , Zigoto/citologia , Zigoto/crescimento & desenvolvimento , Zigoto/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA