Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 530
Filtrar
1.
Life Sci Alliance ; 7(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38719752

RESUMO

Septins are cytoskeletal proteins that participate in cell adhesion, migration, and polarity establishment. The septin subunit SEPT9 directly interacts with the single LIM domain of epithelial protein lost in neoplasm (EPLIN), an actin-bundling protein. Using a human SEPT9 KO fibroblast cell line, we show that cell adhesion and migration are regulated by the interplay between both proteins. The low motility of SEPT9-depleted cells could be partly rescued by increased levels of EPLIN. The normal organization of actin-related filopodia and stress fibers was directly dependent on the expression level of SEPT9 and EPLIN. Increased levels of SEPT9 and EPLIN enhanced the size of focal adhesions in cell protrusions, correlating with stabilization of actin bundles. Conversely, decreased levels had the opposite effect. Our work thus establishes the interaction between SEPT9 and EPLIN as an important link between the septin and the actin cytoskeleton, influencing cell adhesion, motility, and migration.


Assuntos
Adesão Celular , Movimento Celular , Fibroblastos , Adesões Focais , Proteínas com Domínio LIM , Septinas , Humanos , Septinas/metabolismo , Septinas/genética , Movimento Celular/genética , Fibroblastos/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas com Domínio LIM/genética , Adesões Focais/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Pseudópodes/metabolismo , Citoesqueleto de Actina/metabolismo , Linhagem Celular , Actinas/metabolismo , Fibras de Estresse/metabolismo
2.
J Biol Chem ; 300(1): 105580, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141763

RESUMO

Cancer cells acquire malignant phenotypes through an epithelial-mesenchymal transition, which is induced by environmental factors or extracellular signaling molecules, including transforming growth factor-ß (TGF-ß). Among epithelial-mesenchymal transition-associated cell responses, cell morphological changes and cell motility are closely associated with remodeling of the actin stress fibers. Here, we examined the TGF-ß signaling pathways leading to these cell responses. Through knockdown experiments in A549 lung adenocarcinoma cells, we found that Smad3-mediated induction of Snail, but not that of Slug, is indispensable for morphological changes, stress fiber formation, and enhanced motility in cells stimulated with TGF-ß. Ectopic expression of Snail in SMAD3-knockout cells rescued the defect in morphological changes and stress fiber formation by TGF-ß, indicating that the role of Smad3 in these responses is to upregulate Snail expression. Mechanistically, Snail is required for TGF-ß-induced upregulation of Wnt5b, which in turn activates RhoA and subsequent stress fiber formation in cooperation with phosphoinositide 3-kinase. However, ectopic expression of Snail in SMAD3-knockout cells failed to rescue the defect in cell motility enhancement by TGF-ß, indicating that activation of the Smad3/Snail/Wnt5b axis is indispensable but not sufficient for enhancing cell motility; a Smad3-dependent but Snail-independent pathway to activate Rac1 is additionally required. Therefore, the Smad3-dependent pathway leading to enhanced cell motility has two branches: a Snail-dependent branch to activate RhoA and a Snail-independent branch to activate Rac1. Coordinated activation of these branches, together with activation of non-Smad signaling pathways, mediates enhanced cell motility induced by TGF-ß.


Assuntos
Transdução de Sinais , Proteína Smad3 , Fatores de Transcrição da Família Snail , Fibras de Estresse , Fator de Crescimento Transformador beta , Proteínas rho de Ligação ao GTP , Humanos , Células A549 , Movimento Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína Smad3/deficiência , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fatores de Transcrição da Família Snail/deficiência , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fibras de Estresse/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ativação Enzimática , Actinas/metabolismo , Mesoderma/metabolismo , Mesoderma/patologia
3.
Respir Res ; 24(1): 318, 2023 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-38105232

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease (ILD) with unknown etiology, characterized by sustained damage repair of epithelial cells and abnormal activation of fibroblasts, the underlying mechanism of the disease remains elusive. METHODS: To evaluate the role of Tuftelin1 (TUFT1) in IPF and elucidate its molecular mechanism. We investigated the level of TUFT1 in the IPF and bleomycin-induced mouse models and explored the influence of TUFT1 deficiency on pulmonary fibrosis. Additionally, we explored the effect of TUFT1 on the cytoskeleton and illustrated the relationship between stress fiber and pulmonary fibrosis. RESULTS: Our results demonstrated a significant upregulation of TUFT1 in IPF and the bleomycin (BLM)-induced fibrosis model. Disruption of TUFT1 exerted inhibitory effects on pulmonary fibrosis in both in vivo and in vitro. TUFT1 facilitated the assembly of microfilaments in A549 and MRC-5 cells, with a pronounced association between TUFT1 and Neuronal Wiskott-Aldrich syndrome protein (N-WASP) observed during microfilament formation. TUFT1 can promote the phosphorylation of tyrosine residue 256 (Y256) of the N-WASP (pY256N-WASP). Furthermore, TUFT1 promoted transforming growth factor-ß1 (TGF-ß1) induced fibroblast activation by increasing nuclear translocation of pY256N-WASP in fibroblasts, while wiskostatin (Wis), an N-WASP inhibitor, suppressed these processes. CONCLUSIONS: Our findings suggested that TUFT1 plays a critical role in pulmonary fibrosis via its influence on stress fiber, and blockade of TUFT1 effectively reduces pro-fibrotic phenotypes. Pharmacological targeting of the TUFT1-N-WASP axis may represent a promising therapeutic approach for pulmonary fibrosis.


Assuntos
Fibrose Pulmonar Idiopática , Doenças Pulmonares Intersticiais , Animais , Camundongos , Bleomicina/toxicidade , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Doenças Pulmonares Intersticiais/metabolismo , Camundongos Endogâmicos C57BL , Fibras de Estresse/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
4.
Sci Rep ; 13(1): 8662, 2023 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-37248294

RESUMO

Stress fibers are actomyosin bundles that regulate cellular mechanosensation and force transduction. Interacting with the extracellular matrix through focal adhesion complexes, stress fibers are highly dynamic structures regulated by myosin motors and crosslinking proteins. Under external mechanical stimuli such as tensile forces, the stress fiber remodels its architecture to adapt to external cues, displaying properties of viscoelastic materials. How the structural remodeling of stress fibers is related to the generation of contractile force is not well understood. In this work, we simulate mechanochemical dynamics and force generation of stress fibers using the molecular simulation platform MEDYAN. We model stress fiber as two connecting bipolar bundles attached at the ends to focal adhesion complexes. The simulated stress fibers generate contractile force that is regulated by myosin motors and [Formula: see text]-actinin crosslinkers. We find that stress fibers enhance contractility by reducing the distance between actin filaments to increase crosslinker binding, and this structural remodeling ability depends on the crosslinker turnover rate. Under tensile pulling force, the stress fiber shows an instantaneous increase of the contractile forces followed by a slow relaxation into a new steady state. While the new steady state contractility after pulling depends only on the overlap between actin bundles, the short-term contractility enhancement is sensitive to the tensile pulling distance. We further show that this mechanical response is also sensitive to the crosslinker turnover rate. Our results provide new insights into the stress fiber mechanics that have significant implications for understanding cellular adaptation to mechanical signaling.


Assuntos
Actinina , Fibras de Estresse , Actinina/metabolismo , Fibras de Estresse/metabolismo , Miosinas/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Citoesqueleto de Actina/metabolismo
5.
Mol Biol Cell ; 34(6): ar62, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36989016

RESUMO

Mammalian cell migration in open spaces requires F-actin polymerization and myosin contraction. While many studies have focused on myosin's coupling to focal adhesion and stress fibers, the indirect effect of myosin contraction on cell migration through actin depolymerization is not well studied. In this work, we quantified how cell velocity and effective power output are influenced by the rate of actin depolymerization, which is affected by myosin contraction. In addition, we derived scaling laws to provide physical insights into cell migration. Model analysis shows that the cell migration velocity displays a biphasic dependence on the rate of actin depolymerization and myosin contraction. Our model further predicts that the effective cell energy output depends not only on the cell velocity but also on myosin contractility. The work has implications on in vivo processes such as immune response and cancer metastasis, where cells overcome barriers imposed by the physical environment.


Assuntos
Actinas , Miosinas , Animais , Actinas/metabolismo , Miosinas/metabolismo , Movimento Celular/fisiologia , Citoesqueleto de Actina/metabolismo , Fibras de Estresse/metabolismo , Mamíferos/metabolismo
6.
Commun Biol ; 6(1): 29, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631535

RESUMO

Signaling through cAMP/protein kinase A (PKA) promotes endothelial barrier function to prevent plasma leakage induced by inflammatory mediators. The discovery of PKA substrates in endothelial cells increases our understanding of the molecular mechanisms involved in vessel maturation. In this study, we evaluate a cAMP inducer, forskolin, and a phospho-PKA substrate antibody to identify ZNF185 as a PKA substrate. ZNF185 interacts with PKA and colocalizes with F-actin in endothelial cells. Both ZNF185 and F-actin accumulate in the plasma membrane region in response to forskolin to stabilize the cortical actin structure. By contrast, ZNF185 knockdown disrupts actin filaments and promotes stress fiber formation without inflammatory mediators. Constitutive activation of RhoA is induced by ZNF185 knockdown, which results in forskolin-resistant endothelial barrier dysfunction. Knockout of mouse Zfp185 which is an orthologous gene of human ZNF185 increases vascular leakage in response to inflammatory stimuli in vivo. Thrombin protease is used as a positive control to assemble stress fibers via RhoA activation. Unexpectedly, ZNF185 is cleaved by thrombin, resulting in an N-terminal actin-targeting domain and a C-terminal PKA-interacting domain. Irreversible dysfunction of ZNF185 protein potentially causes RhoA-dependent stress fiber formation by thrombin.


Assuntos
Actinas , Células Endoteliais , Proteínas com Domínio LIM , Fibras de Estresse , Proteína rhoA de Ligação ao GTP , Animais , Humanos , Camundongos , Actinas/metabolismo , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteínas com Domínio LIM/metabolismo , Camundongos Knockout , Proteína rhoA de Ligação ao GTP/metabolismo , Fibras de Estresse/metabolismo , Trombina/farmacologia , Trombina/metabolismo
7.
Nat Commun ; 13(1): 6032, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229430

RESUMO

Contractile actomyosin bundles are key force-producing and mechanosensing elements in muscle and non-muscle tissues. Whereas the organization of muscle myofibrils and mechanism regulating their contractility are relatively well-established, the principles by which myosin-II activity and force-balance are regulated in non-muscle cells have remained elusive. We show that Caldesmon, an important component of smooth muscle and non-muscle cell actomyosin bundles, is an elongated protein that functions as a dynamic cross-linker between myosin-II and tropomyosin-actin filaments. Depletion of Caldesmon results in aberrant lateral movement of myosin-II filaments along actin bundles, leading to irregular myosin distribution within stress fibers. This manifests as defects in stress fiber network organization and contractility, and accompanied problems in cell morphogenesis, migration, invasion, and mechanosensing. These results identify Caldesmon as critical factor that ensures regular myosin-II spacing within non-muscle cell actomyosin bundles, and reveal how stress fiber networks are controlled through dynamic cross-linking of tropomyosin-actin and myosin filaments.


Assuntos
Fibras de Estresse , Tropomiosina , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Músculo Liso/metabolismo , Miosina Tipo II/metabolismo , Miosinas/metabolismo , Fibras de Estresse/metabolismo , Tropomiosina/metabolismo
8.
Asian Pac J Cancer Prev ; 23(7): 2379-2386, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35901345

RESUMO

OBJECTIVE: Glioblastoma is the most aggressive and lethal brain tumor in adults with highly invasive properties. In this present study, we explored the effects of Phyllanthus taxodiifolius Beille extract on molecules known to be hallmarks of aggressive glioblastoma including N-cadherin and vimentin, mesenchymal markers, as well as paxillin, a major adaptor protein that regulates the linking of focal adhesions to the actin cytoskeleton. METHODS: P. taxodiifolius were air-dried, powdered and percolated with methanol, filtered, concentrated and lyophilized to yield a crude methanol extract. C6 glioblastoma cell line was used in this study. The expression of N-cadherin and vimentin, as well as the activation of paxillin was determined using Western blot analysis. The effect of the extract on focal adhesions and actin cytoskeleton were investigated using immunofluorescence staining and confocal imaging. RESULTS: In the presence of 40 µg/ml Phyllanthus taxodiifolius Beille extract, the expression of N-cadherin and vimentin were significantly decreased (p<0.001 and p<0.05, respectively). Activation of paxillin was also diminished as indicated by a reduction of phosphorylated-paxillin (p<0.01). Consequently, actin stress fibers in glioblastoma cells were abolished as evidenced by the decrease in focal adhesion (p<0.001) and stress fibers numbers (p<0.001). CONCLUSION: Our study demonstrates for the first time that P. taxodiifolius interferes with multiple key molecules related to pathological hallmarks of glioblastoma. These molecules are involved with cell contacts, focal adhesions, and the formation and stabilization of actin stress fibers, which are required for glioblastoma metastatic behavior. These results provide further evidence supporting the potential of P. taxodiifolius and its bioactive compounds as anti-cancer agents.


Assuntos
Glioblastoma , Phyllanthus , Actinas/metabolismo , Caderinas/metabolismo , Adesão Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Glioblastoma/patologia , Humanos , Metanol , Paxilina/metabolismo , Paxilina/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Phyllanthus/metabolismo , Extratos Vegetais/farmacologia , Fibras de Estresse/metabolismo , Fibras de Estresse/patologia , Vimentina
9.
Biochem Biophys Res Commun ; 620: 49-55, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-35777134

RESUMO

The tension in the stress fibers (SFs) of cells plays a pivotal role in determining biological processes such as cell migration, morphological formation, and protein synthesis. Our previous research developed a method to evaluate the cellular contraction force generated in SFs based on photoelasticity-associated retardation of polarized light; however, we employed live cells, which could have caused an increase in retardation and not contraction force. Therefore, the present study aimed to confirm that polarized light retardation increases inherently due to contraction, regardless of cell activity. We also explored the reason why retardation increased with SF contractions. We used SFs physically isolated from vascular smooth muscle cells to stop cell activity. The retardation of SFs was measured after ATP administration, responsible for contracting SFs. The SFs were imaged under optical and electron microscopes to measure SF length, width, and retardation. The retardation of isolated SFs after ATP administration was significantly higher than before. Thus, we confirmed that retardation increased with elevated tension in individual SFs. Furthermore, the SF diameter decreased while the SF length remained almost constant. Thus, we conclude that a contraction force-driven increase in the density of SFs is the main factor for the rise in polarized light retardation.


Assuntos
Miócitos de Músculo Liso , Fibras de Estresse , Trifosfato de Adenosina/metabolismo , Movimento Celular , Miócitos de Músculo Liso/fisiologia , Fibras de Estresse/metabolismo , Estresse Mecânico
10.
J Cell Biol ; 221(5)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35416930

RESUMO

The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.


Assuntos
Actomiosina , Células Epiteliais , Junções Intercelulares , Proteínas dos Microfilamentos , Fibras de Estresse , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Células Epiteliais/metabolismo , Junções Intercelulares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fibras de Estresse/metabolismo
11.
Cells ; 11(2)2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35053376

RESUMO

The transporter protein lipocalin-2 (LCN2) also termed neutrophil-gelatinase-associated lipocalin (NGAL) has pleiotropic effects in tumorigenesis in various cancers. Since the precise role of LCN2 in prostate cancer (PCa) is poorly understood, we aimed to elucidate its functions in PCa in vitro. For this purpose, LCN2 was transiently suppressed or permanently depleted in human PC-3 cells using siRNA or CRISPR/Cas9-mediated knockout. Effects of LCN2 suppression on expression of different tumorigenic markers were investigated by Western blot analysis and RT-qPCR. LCN2 knockout cells were analyzed for cellular changes and their ability to cope endoplasmic stress compared to parenteral PC-3 cells. Reduced LCN2 was accompanied by decreased expression of IL-1ß and Cx43. In PC-3 cells, LCN2 deficiency leads to reduced proliferation, diminished expression of pro-inflammatory cytokines, lower adhesion, and disrupted F-actin distribution. In addition, IL-1ß expression strongly correlated with LCN2 levels. LCN2 knockout cells showed enhanced and sustained activation of unfolded protein response proteins when treated with tunicamycin or cultured under glucose deprivation. Interestingly, an inverse correlation between phosphorylation of eukaryotic initiation factor 2 α subunit (p-eIF2α) and LCN2 expression was observed suggesting that LCN2 triggers protein synthesis under stress conditions. The finding that LCN2 depletion leads to significant phenotypic and cellular changes in PC-3 cells adds LCN2 as a valuable target for the treatment of PCa.


Assuntos
Lipocalina-2/deficiência , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Actinas/metabolismo , Biomarcadores Tumorais/metabolismo , Sistemas CRISPR-Cas/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Adesão Celular , Proliferação de Células , Citoesqueleto/metabolismo , Estresse do Retículo Endoplasmático , Técnicas de Silenciamento de Genes , Humanos , Interleucina-1beta/metabolismo , Lipocalina-2/metabolismo , Masculino , Metástase Neoplásica , Células PC-3 , RNA Interferente Pequeno/metabolismo , Fibras de Estresse/metabolismo , Resposta a Proteínas não Dobradas
12.
J Cell Sci ; 135(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34854883

RESUMO

Septins, a family of GTP-binding proteins that assemble into higher order structures, interface with the membrane, actin filaments and microtubules, and are thus important regulators of cytoarchitecture. Septin 9 (SEPT9), which is frequently overexpressed in tumors and mutated in hereditary neuralgic amyotrophy (HNA), mediates the binding of septins to microtubules, but the molecular determinants of this interaction remained uncertain. We demonstrate that a short microtubule-associated protein (MAP)-like motif unique to SEPT9 isoform 1 (SEPT9_i1) drives septin octamer-microtubule interaction in cells and in vitro reconstitutions. Septin-microtubule association requires polymerizable septin octamers harboring SEPT9_i1. Although outside of the MAP-like motif, HNA mutations abrogate this association, identifying a putative regulatory domain. Removal of this domain from SEPT9_i1 sequesters septins on microtubules, promotes microtubule stability and alters actomyosin fiber distribution and tension. Thus, we identify key molecular determinants and potential regulatory roles of septin-microtubule interaction, paving the way to deciphering the mechanisms underlying septin-associated pathologies. This article has an associated First Person interview with the first author of the paper.


Assuntos
Septinas , Fibras de Estresse , Humanos , Proteínas Associadas aos Microtúbulos , Microtúbulos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Septinas/genética , Septinas/metabolismo , Fibras de Estresse/metabolismo
13.
Sci Rep ; 11(1): 21473, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728615

RESUMO

In obese adipose tissue (AT), hypertrophic expansion of adipocytes is not matched by new vessel formation, leading to AT hypoxia. As a result, hypoxia inducible factor-1⍺ (HIF-1⍺) accumulates in adipocytes inducing a transcriptional program that upregulates profibrotic genes and biosynthetic enzymes such as lysyl oxidase (LOX) synthesis. This excess synthesis and crosslinking of extracellular matrix (ECM) components cause AT fibrosis. Although fibrosis is a hallmark of obese AT, the role of fibroblasts, cells known to regulate fibrosis in other fibrosis-prone tissues, is not well studied. Here we have developed an in vitro model of AT to study adipocyte-fibroblast crosstalk in a hypoxic environment. Further, this in vitro model was used to investigate the effect of hypoxia on adipocyte mechanical properties via ras homolog gene family member A (RhoA)/Rho-associated coiled-coil kinases (ROCK) signaling pathways. We confirmed that hypoxia creates a diseased phenotype by inhibiting adipocyte maturation and inducing actin stress fiber formation facilitated by myocardin-related transcription factor A (MRTF-A/MKL1) nuclear translocation. This work presents new potential therapeutic targets for obesity by improving adipocyte maturation and limiting mechanical stress in obese AT.


Assuntos
Adipócitos/patologia , Fibroblastos/patologia , Fibrose/patologia , Hipóxia/fisiopatologia , Células-Tronco Mesenquimais/patologia , Obesidade/patologia , Fibras de Estresse/patologia , Adipócitos/metabolismo , Fibroblastos/metabolismo , Fibrose/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Obesidade/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Fibras de Estresse/metabolismo , Transativadores/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
14.
FASEB J ; 35(12): e22012, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34724245

RESUMO

Rho-kinase (ROK)-mediated migration of vascular smooth muscle cells plays a crucial role in cardiovascular diseases. Previously we demonstrated Fyn tyrosine kinase as an upstream molecule of ROK to mediate actin stress fiber formation that plays an important role in cell migration, but the molecular mechanism between the two kinases was unclear. To discover a novel signaling molecule that exists between Fyn and ROK, we identified paxillin acting downstream of the active Fyn by combined use of pulldown assay and mass spectrometry. Immunofluorescence staining confirmed co-localization of Fyn and paxillin at the ends of actin stress fibers in human coronary artery smooth muscle cells (CASMCs). Surface plasmon resonance assay demonstrated direct binding between constitutively active Fyn (CA-Fyn) and N-terminus of paxillin (N-pax). The sphingosylphosphorylcholine (SPC)-induced ROK activation, actin stress fiber formation and cell migration were inhibited by paxillin knockdown, which were rescued by full-length paxillin (FL-pax) but not N-pax. N-pax co-localized with CA-Fyn at the cytosol and overexpression of N-pax inhibited the SPC-induced actin stress fiber formation and cell migration, indicating that the direct binding of FL-pax and CA-Fyn at the ends of actin stress fibers is essential for the ROK-mediated actin stress fiber formation and cell migration. Paxillin, as a novel signalling molecule, mediates the SPC-induced actin stress fiber formation and migration in human CASMCs via the Fyn/paxillin/ROK signalling pathway by direct binding of active Fyn.


Assuntos
Actinas/metabolismo , Movimento Celular , Vasos Coronários/patologia , Músculo Liso Vascular/patologia , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Fibras de Estresse/patologia , Vasos Coronários/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Fosforilação , Fibras de Estresse/metabolismo
15.
Cells ; 10(8)2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34440749

RESUMO

The cellular mechanisms of basement membrane (BM) invasion remain poorly understood. We investigated the invasion-promoting mechanisms of actin cytoskeleton reorganization in BM-covered MCF10A breast acini. High-resolution confocal microscopy has characterized actin cell protrusion formation and function in response to tumor-resembling ECM stiffness and soluble EGF stimulation. Traction force microscopy quantified the mechanical BM stresses that invasion-triggered acini exerted on the BM-ECM interface. We demonstrate that acini use non-proteolytic actin microspikes as functional precursors of elongated protrusions to initiate BM penetration and ECM probing. Further, these microspikes mechanically widened the collagen IV pores to anchor within the BM scaffold via force-transmitting focal adhesions. Pre-invasive basal cells located at the BM-ECM interface exhibited predominantly cortical actin networks and actin microspikes. In response to pro-invasive conditions, these microspikes accumulated and converted subsequently into highly contractile stress fibers. The phenotypical switch to stress fiber cells matched spatiotemporally with emerging high BM stresses that were driven by actomyosin II contractility. The activation of proteolytic invadopodia with MT1-MMP occurred at later BM invasion stages and only in cells already disseminating into the ECM. Our study demonstrates that BM pore-widening filopodia bridge mechanical ECM probing function and contractility-driven BM weakening. Finally, these EMT-related cytoskeletal adaptations are critical mechanisms inducing the invasive transition of benign breast acini.


Assuntos
Actinas/metabolismo , Membrana Basal/metabolismo , Miosina Tipo II/metabolismo , Fibras de Estresse/metabolismo , Células Acinares/citologia , Células Acinares/metabolismo , Mama/citologia , Mama/metabolismo , Adesão Celular , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Matriz Extracelular/metabolismo , Feminino , Humanos , Microscopia Confocal , Podossomos/metabolismo , Pseudópodes/metabolismo , Fibras de Estresse/química
16.
Molecules ; 26(16)2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34443489

RESUMO

Hydrogel formulations (masks or patches, without tissue support) represent the new frontier for customizable skin beauty and health. The employment of these materials is becoming popular in wound dressing, to speed up the healing process while protecting the affected area, as well as to provide a moisturizing reservoir, control the inflammatory process and the onset of bacterial development. Most of these hydrogels are acrylic-based at present, not biodegradable and potentially toxic, due to acrylic monomers residues. In this work, we selected a new class of cellulose-derived and biodegradable hydrogel films to incorporate and convey an active compound for dermatological issues. Films were obtained from a combination of different polysaccharides and clays, and berberine hydrochloride, a polyphenolic molecule showing anti-inflammatory, immunomodulatory, antibacterial and antioxidant properties, was chosen and then embedded in the hydrogel films. These innovative hydrogel-based systems were characterized in terms of water uptake profile, in vitro cytocompatibility and skin permeation kinetics by Franz diffusion cell. Berberine permeation fitted well to Korsmeyer-Peppas kinetic model and achieved a release higher than 100 µg/cm2 within 24 h. The latter study, exploiting a reliable skin model membrane, together with the biological assessment, gained insights into the most promising formulation for future investigations.


Assuntos
Berberina/administração & dosagem , Sistemas de Liberação de Medicamentos , Metilgalactosídeos/química , Pele/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Células HaCaT , Humanos , Cinética , Permeabilidade , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo , Difração de Raios X
17.
Cells ; 10(6)2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34207462

RESUMO

The plasma membrane is a key actor of cell migration. For instance, its tension controls persistent cell migration and cell surface caveolae integrity. Then, caveolae constituents such as caveolin-1 can initiate a mechanotransduction loop that involves actin- and focal adhesion-dependent control of the mechanosensor YAP to finely tune cell migration. Tetraspanin CD82 (also named KAI-1) is an integral membrane protein and a metastasis suppressor. Its expression is lost in many cancers including breast cancer. It is a strong inhibitor of cell migration by a little-known mechanism. We demonstrated here that CD82 controls persistent 2D migration of EGF-induced single cells, stress fibers and focal adhesion sizes and dynamics. Mechanistically, we found that CD82 regulates membrane tension, cell surface caveolae abundance and YAP nuclear translocation in a caveolin-1-dependent manner. Altogether, our data show that CD82 controls 2D cell migration using membrane-driven mechanics involving caveolin and the YAP pathway.


Assuntos
Membrana Celular/metabolismo , Movimento Celular/fisiologia , Proteína Kangai-1/metabolismo , Metástase Neoplásica/patologia , Neoplasias/metabolismo , Fibras de Estresse/metabolismo , Tetraspaninas/metabolismo , Caveolina 1/metabolismo , Adesão Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Mecanotransdução Celular/fisiologia , Proteínas de Membrana/metabolismo , Neoplasias/patologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
18.
Am J Physiol Cell Physiol ; 320(6): C1153-C1163, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33881935

RESUMO

Cells adapt to applied cyclic stretch (CS) to circumvent chronic activation of proinflammatory signaling. Currently, the molecular mechanism of the selective disassembly of actin stress fibers (SFs) in the stretch direction, which occurs at the early stage of the cellular response to CS, remains controversial. Here, we suggest that the mechanosensitive behavior of myosin II, a major cross-linker of SFs, primarily contributes to the directional disassembly of the actomyosin complex SFs in bovine vascular smooth muscle cells and human U2OS osteosarcoma cells. First, we identified that CS with a shortening phase that exceeds in speed the inherent contractile rate of individual SFs leads to the disassembly. To understand the biological basis, we investigated the effect of expressing myosin regulatory light-chain mutants and found that SFs with less actomyosin activities disassemble more promptly upon CS. We consequently created a minimal mathematical model that recapitulates the salient features of the direction-selective and threshold-triggered disassembly of SFs to show that disassembly or, more specifically, unbundling of the actomyosin bundle SFs is enhanced with sufficiently fast cell shortening. We further demonstrated that similar disassembly of SFs is inducible in the presence of an active LIM-kinase-1 mutant that deactivates cofilin, suggesting that cofilin is dispensable as opposed to a previously proposed mechanism.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Miosina Tipo II/metabolismo , Fibras de Estresse/metabolismo , Actomiosina/metabolismo , Animais , Bovinos , Linhagem Celular Tumoral , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Humanos , Contração Muscular/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteossarcoma/metabolismo , Estresse Mecânico
19.
Nat Commun ; 12(1): 2226, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33850145

RESUMO

At the basis of cell shape and behavior, the organization of actomyosin and its ability to generate forces are widely studied. However, the precise regulation of this contractile network in space and time is unclear. Here, we study the role of the epithelial-specific protein EpCAM, a contractility modulator, in cell shape and motility. We show that EpCAM is required for stress fiber generation and front-rear polarity acquisition at the single cell level. In fact, EpCAM participates in the remodeling of a transient zone of active RhoA at the cortex of spreading epithelial cells. EpCAM and RhoA route together through the Rab35/EHD1 fast recycling pathway. This endosomal pathway spatially organizes GTP-RhoA to fine tune the activity of actomyosin resulting in polarized cell shape and development of intracellular stiffness and traction forces. Impairment of GTP-RhoA endosomal trafficking either by silencing EpCAM or by expressing Rab35/EHD1 mutants prevents proper myosin-II activity, stress fiber formation and ultimately cell polarization. Collectively, this work shows that the coupling between co-trafficking of EpCAM and RhoA, and actomyosin rearrangement is pivotal for cell spreading, and advances our understanding of how biochemical and mechanical properties promote cell plasticity.


Assuntos
Endossomos/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Células Epiteliais/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Actomiosina/metabolismo , Células CACO-2 , Movimento Celular/fisiologia , Polaridade Celular , Forma Celular , Células HeLa , Humanos , Miosina Tipo II/metabolismo , Fibras de Estresse/metabolismo
20.
Biomed Environ Sci ; 34(2): 139-151, 2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33685573

RESUMO

OBJECTIVE: The underlying mechanism of Ezrin in ovarian cancer (OVCA) is far from being understood. Therefore, this study aimed to assess the role of Ezrin in OVCA cells (SKOV3 and CaOV3) and investigate the associated molecular mechanisms. METHODS: We performed Western blotting, reverse transcription-quantitative polymerase chain reaction, MTT, cell colony, cell wound healing, transwell migration and invasion, RhoA and Rac active pull down assays, and confocal immunofluorescence experiments to evaluate the functions and molecular mechanisms of Ezrin overexpression or knockdown in the proliferation and metastasis of OVCA cells. RESULTS: The ectopic expression of Ezrin significantly increased cell proliferation, invasiveness, and epithelial-mesenchymal transition (EMT) in OVCA cells. By contrast, the knockdown of endogenous Ezrin prevented OVCA cell proliferation, invasiveness, and EMT. Lastly, we observed that Ezrin can positively regulate the active forms of RhoA rather than Rac-1 in OVCA cells, thereby promoting robust stress fiber formation. CONCLUSION: Our results indicated that Ezrin regulates OVCA cell proliferation and invasiveness by modulating EMT and induces actin stress fiber formation by regulating Rho-GTPase activity, which provides novel insights into the treatment of the OVCA.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas do Citoesqueleto/genética , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Fibras de Estresse/genética , Fibras de Estresse/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA