Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Curr Biol ; 32(9): 1986-2000.e5, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35381185

RESUMO

Tissue morphogenesis arises from the culmination of changes in cell-cell junction length. Mechanochemical signaling in the form of RhoA underlies these ratcheted contractions, which occur asymmetrically. The underlying mechanisms of asymmetry remain unknown. We use optogenetically controlled RhoA in model epithelia together with biophysical modeling to uncover the mechanism lending to asymmetric vertex motion. Using optogenetic and pharmacological approaches, we find that both local and global RhoA activation can drive asymmetric junction contraction in the absence of tissue-scale patterning. We find that standard vertex models with homogeneous junction properties are insufficient to recapitulate the observed junction dynamics. Furthermore, these experiments reveal a local coupling of RhoA activation with E-cadherin accumulation. This motivates a coupling of RhoA-mediated increases in tension and E-cadherin-mediated adhesion strengthening. We then demonstrate that incorporating this force-sensitive adhesion strengthening into a continuum model is successful in capturing the observed junction dynamics. Thus, we find that a force-dependent intercellular "clutch" at tricellular vertices stabilizes vertex motion under increasing tension and is sufficient to generate asymmetries in junction contraction.


Assuntos
Junções Aderentes , Células Epiteliais , Junções Aderentes/fisiologia , Caderinas/genética , Adesão Celular , Epitélio , Morfogênese
3.
J Cell Sci ; 134(17)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34368835

RESUMO

Epithelia migrate as physically coherent populations of cells. Previous studies have revealed that mechanical stress accumulates in these cellular layers as they move. These stresses are characteristically tensile in nature and have often been inferred to arise when moving cells pull upon the cell-cell adhesions that hold them together. We now report that epithelial tension at adherens junctions between migrating cells also increases due to an increase in RhoA-mediated junctional contractility. We found that active RhoA levels were stimulated by p114 RhoGEF (also known as ARHGEF18) at the junctions between migrating MCF-7 monolayers, and this was accompanied by increased levels of actomyosin and mechanical tension. Applying a strategy to restore active RhoA specifically at adherens junctions by manipulating its scaffold, anillin, we found that this junctional RhoA signal was necessary to stabilize junctional E-cadherin (CDH1) during epithelial migration and promoted orderly collective movement. We suggest that stabilization of E-cadherin by RhoA serves to increase cell-cell adhesion to protect against the mechanical stresses of migration. This article has an associated First Person interview with the first author of the paper.


Assuntos
Junções Aderentes , Proteína rhoA de Ligação ao GTP , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Junções Aderentes/metabolismo , Caderinas/genética , Caderinas/metabolismo , Células Epiteliais/metabolismo , Humanos , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(48): 30476-30487, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33214152

RESUMO

None of the current superresolution microscopy techniques can reliably image the changes in endogenous protein nanoclustering dynamics associated with specific conformations in live cells. Single-domain nanobodies have been invaluable tools to isolate defined conformational states of proteins, and we reasoned that expressing these nanobodies coupled to single-molecule imaging-amenable tags could allow superresolution analysis of endogenous proteins in discrete conformational states. Here, we used anti-GFP nanobodies tagged with photoconvertible mEos expressed as intrabodies, as a proof-of-concept to perform single-particle tracking on a range of GFP proteins expressed in live cells, neurons, and small organisms. We next expressed highly specialized nanobodies that target conformation-specific endogenous ß2-adrenoreceptor (ß2-AR) in neurosecretory cells, unveiling real-time mobility behaviors of activated and inactivated endogenous conformers during agonist treatment in living cells. We showed that activated ß2-AR (Nb80) is highly immobile and organized in nanoclusters. The Gαs-GPCR complex detected with Nb37 displayed higher mobility with surprisingly similar nanoclustering dynamics to that of Nb80. Activated conformers are highly sensitive to dynamin inhibition, suggesting selective targeting for endocytosis. Inactivated ß2-AR (Nb60) molecules are also largely immobile but relatively less sensitive to endocytic blockade. Expression of single-domain nanobodies therefore provides a unique opportunity to capture highly transient changes in the dynamic nanoscale organization of endogenous proteins.


Assuntos
Modelos Moleculares , Conformação Proteica , Receptores Adrenérgicos beta 2/química , Imagem Individual de Molécula , Anticorpos de Domínio Único/química , Animais , Linhagem Celular , Endocitose , Imunofluorescência , Expressão Gênica , Genes Reporter , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Ligação Proteica , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Recombinantes de Fusão , Imagem Individual de Molécula/métodos , Anticorpos de Domínio Único/metabolismo , Peixe-Zebra
5.
Mol Biol Cell ; 31(23): 2557-2569, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32903148

RESUMO

Epithelia can eliminate apoptotic cells by apical extrusion. This is a complex morphogenetic event where expulsion of the apoptotic cell is accompanied by rearrangement of its immediate neighbors to form a rosette. A key mechanism for extrusion is constriction of an actomyosin network that neighbor cells form at their interface with the apoptotic cell. Here we report a complementary process of cytoskeletal relaxation that occurs when cortical contractility is down-regulated at the junctions between those neighbor cells themselves. This reflects a mechanosensitive Src family kinase (SFK) signaling pathway that is activated in neighbor cells when the apoptotic cell relaxes shortly after injury. Inhibiting SFK signaling blocks both the expulsion of apoptotic cells and the rosette formation among their neighbor cells. This reveals the complex pattern of spatially distinct contraction and relaxation that must be established in the neighboring epithelium for apoptotic cells to be extruded.


Assuntos
Junções Aderentes/metabolismo , Apoptose/fisiologia , Quinases da Família src/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Junções Aderentes/fisiologia , Células Epiteliais/metabolismo , Epitélio/metabolismo , Epitélio/fisiologia , Humanos , Células MCF-7 , Morfogênese , Transdução de Sinais , Quinases da Família src/fisiologia
6.
Bioessays ; 42(10): e2000055, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32735042

RESUMO

Scaffolds are fundamental to many cellular signaling pathways. In this essay, a novel class of scaffolds are proposed, whose action bears striking resemblance to kinetic proofreading. Commonly, scaffold proteins are thought to work as tethers, bringing different components of a pathway together to improve the likelihood of their interaction. However, recent studies show that the cytoskeletal scaffold, anillin, supports contractile signaling by a novel, non-tethering mechanism that controls the membrane dissociation kinetics of RhoA. More generally, such proof-reading-like scaffolds are distinguished from tethers by a rare type of cooperativity, manifest as a super-linear relationship between scaffold concentration and signaling efficiency. The evidence for this hypothesis is reviewed, its conceptual ramifications are considered, and research questions for the future are discussed.


Assuntos
Proteínas Contráteis , Citocinese , Proteínas Contráteis/metabolismo , Citoesqueleto/metabolismo , Transdução de Sinais
7.
Dev Cell ; 54(1): 75-91.e7, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32485139

RESUMO

Epithelia are active materials where mechanical tension governs morphogenesis and homeostasis. But how that tension is regulated remains incompletely understood. We now report that caveolae control epithelial tension and show that this is necessary for oncogene-transfected cells to be eliminated by apical extrusion. Depletion of caveolin-1 (CAV1) increased steady-state tensile stresses in epithelial monolayers. As a result, loss of CAV1 in the epithelial cells surrounding oncogene-expressing cells prevented their apical extrusion. Epithelial tension in CAV1-depleted monolayers was increased by cortical contractility at adherens junctions. This reflected a signaling pathway, where elevated levels of phosphoinositide-4,5-bisphosphate (PtdIns(4,5)P2) recruited the formin, FMNL2, to promote F-actin bundling. Steady-state monolayer tension and oncogenic extrusion were restored to CAV1-depleted monolayers when tension was corrected by depleting FMNL2, blocking PtdIns(4,5)P2, or disabling the interaction between FMNL2 and PtdIns(4,5)P2. Thus, caveolae can regulate active mechanical tension for epithelial homeostasis by controlling lipid signaling to the actin cytoskeleton.


Assuntos
Cavéolas/metabolismo , Células Epiteliais/metabolismo , Proteínas Oncogênicas/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Células CACO-2 , Caveolina 1/metabolismo , Células Epiteliais/ultraestrutura , Forminas/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Proteínas Oncogênicas/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Estresse Mecânico
8.
J Cell Sci ; 133(13)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32467325

RESUMO

Cell extrusion is a morphogenetic process that is implicated in epithelial homeostasis and elicited by stimuli ranging from apoptosis to oncogenic transformation. To explore whether the morphogenetic transcription factor Snail (SNAI1) induces extrusion, we inducibly expressed a stabilized Snail6SA transgene in confluent MCF-7 monolayers. When expressed in small clusters (less than three cells) within otherwise wild-type confluent monolayers, Snail6SA expression induced apical cell extrusion. In contrast, larger clusters or homogenous cultures of Snail6SA cells did not show enhanced apical extrusion, but eventually displayed sporadic basal delamination. Transcriptomic profiling revealed that Snail6SA did not substantively alter the balance of epithelial and mesenchymal genes. However, we identified a transcriptional network that led to upregulated RhoA signalling and cortical contractility in cells expressing Snail6SA Enhanced contractility was necessary, but not sufficient, to drive extrusion, suggesting that Snail collaborates with other factors. Indeed, we found that the transcriptional downregulation of cell-matrix adhesion cooperates with contractility to mediate basal delamination. This provides a pathway for Snail to influence epithelial morphogenesis independently of classic epithelial-to-mesenchymal transition.


Assuntos
Células Epiteliais , Transição Epitelial-Mesenquimal , Junções Célula-Matriz , Transição Epitelial-Mesenquimal/genética , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição/genética
9.
Proc Natl Acad Sci U S A ; 117(12): 6890-6900, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32152092

RESUMO

Inefficient physiological transitions are known to cause metabolic disorders. Therefore, investigating mechanisms that constitute molecular switches in a central metabolic organ like the liver becomes crucial. Specifically, upstream mechanisms that control temporal engagement of transcription factors, which are essential to mediate physiological fed-fast-refed transitions are less understood. SIRT1, a NAD+-dependent deacetylase, is pivotal in regulating hepatic gene expression and has emerged as a key therapeutic target. Despite this, if/how nutrient inputs regulate SIRT1 interactions, stability, and therefore downstream functions are still unknown. Here, we establish nutrient-dependent O-GlcNAcylation of SIRT1, within its N-terminal domain, as a crucial determinant of hepatic functions. Our findings demonstrate that during a fasted-to-refed transition, glycosylation of SIRT1 modulates its interactions with various transcription factors and a nodal cytosolic kinase involved in insulin signaling. Moreover, sustained glycosylation in the fed state causes nuclear exclusion and cytosolic ubiquitin-mediated degradation of SIRT1. This mechanism exerts spatiotemporal control over SIRT1 functions by constituting a previously unknown molecular relay. Of note, loss of SIRT1 glycosylation discomposed these interactions resulting in aberrant gene expression, mitochondrial dysfunctions, and enhanced hepatic gluconeogenesis. Expression of nonglycosylatable SIRT1 in the liver abrogated metabolic flexibility, resulting in systemic insulin resistance, hyperglycemia, and hepatic inflammation, highlighting the physiological costs associated with its overactivation. Conversely, our study also reveals that hyperglycosylation of SIRT1 is associated with aging and high-fat-induced obesity. Thus, we establish that nutrient-dependent glycosylation of SIRT1 is essential to gate its functions and maintain physiological fitness.


Assuntos
Gluconeogênese , Homeostase , Hiperglicemia/prevenção & controle , Fígado/metabolismo , Processamento de Proteína Pós-Traducional , Sirtuína 1/metabolismo , Acetilglucosamina/metabolismo , Envelhecimento/fisiologia , Animais , Jejum , Glicosilação , Células HEK293 , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Resistência à Insulina , Fígado/imunologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/patologia , Obesidade/prevenção & controle , Fosforilação , Sirtuína 1/química , Análise Espaço-Temporal
10.
Dev Cell ; 49(6): 894-906.e12, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31105010

RESUMO

RhoA stimulates cell contractility by recruiting downstream effectors to the cortical plasma membrane. We now show that direct binding by anillin is required for effective signaling: this antagonizes the otherwise labile membrane association of GTP-RhoA to promote effector recruitment. However, since its binding to RhoA blocks access by other effectors, we demonstrate that anillin must also concentrate membrane phosphoinositide-4,5-P2 (PIP2) to promote signaling. We propose and test a sequential pathway where GTP-RhoA first binds to anillin and then is retained at the membrane by PIP2 after it disengages from anillin. Importantly, re-binding of membrane GTP-RhoA to anillin, regulated by the cortical density of anillin, creates cycles through this pathway. These cycles repeatedly reset the dissociation kinetics of GTP-RhoA, substantially increasing its dwell time to recruit effectors. Thus, anillin regulates RhoA signaling by a paradigm of kinetic scaffolding that may apply to other signals whose efficacy depends on their cortical dwell times.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular/efeitos dos fármacos , Proteínas Contráteis/farmacologia , Citocinese/fisiologia , Guanosina Trifosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Citocinese/efeitos dos fármacos , Feminino , Humanos , Cinética , Células MCF-7 , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/genética
11.
Dev Cell ; 47(4): 439-452.e6, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30318244

RESUMO

Adherens junctions are tensile structures that couple epithelial cells together. Junctional tension can arise from cell-intrinsic application of contractility or from the cell-extrinsic forces of tissue movement. Here, we report a mechanosensitive signaling pathway that activates RhoA at adherens junctions to preserve epithelial integrity in response to acute tensile stress. We identify Myosin VI as the force sensor, whose association with E-cadherin is enhanced when junctional tension is increased by mechanical monolayer stress. Myosin VI promotes recruitment of the heterotrimeric Gα12 protein to E-cadherin, where it signals for p114 RhoGEF to activate RhoA. Despite its potential to stimulate junctional actomyosin and further increase contractility, tension-activated RhoA signaling is necessary to preserve epithelial integrity. This is explained by an increase in tensile strength, especially at the multicellular vertices of junctions, that is due to mDia1-mediated actin assembly.


Assuntos
Junções Aderentes/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Estresse Mecânico , Proteína rhoA de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Caderinas/metabolismo , Humanos , Resistência à Tração
12.
13.
Dev Cell ; 43(4): 381-382, 2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-29161587

RESUMO

α-catenin is a scaffolding molecule that can bind F-actin and other cytoskeletal proteins. It is best known for its contribution to cell-cell adhesion. In this issue of Developmental Cell, Vassilev et al. (2017) identify an extrajunctional pool of α-catenin that regulates RhoA signaling and controls directional migration of single cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Caderinas/metabolismo , Adesão Celular/fisiologia , alfa Catenina/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Humanos , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Nat Commun ; 8(1): 790, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28983097

RESUMO

Contractile adherens junctions support cell-cell adhesion, epithelial integrity, and morphogenesis. Much effort has been devoted to understanding how contractility is established; however, less is known about whether contractility can be actively downregulated at junctions nor what function this might serve. We now identify such an inhibitory pathway that is mediated by the cytoskeletal scaffold, cortactin. Mutations of cortactin that prevent its tyrosine phosphorylation downregulate RhoA signaling and compromise the ability of epithelial cells to generate a contractile zonula adherens. This is mediated by the RhoA antagonist, SRGAP1. We further demonstrate that this mechanism is co-opted by hepatocyte growth factor to promote junctional relaxation and motility in epithelial collectives. Together, our findings identify a novel function of cortactin as a regulator of RhoA signaling that can be utilized by morphogenetic regulators for the active downregulation of junctional contractility.Epithelial cell-cell adhesions are contractile junctions, but whether contractility can be down-regulated is not known. Here the authors report how tyrosine dephosphorylation of the cytoskeletal scaffold, cortactin, recruits the RhoA antagonist SRGAP1 to relax adherens junctions in response to HGF.


Assuntos
Junções Aderentes/metabolismo , Cortactina/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Tirosina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Células CACO-2 , Adesão Celular , Cortactina/genética , Citoesqueleto , Regulação para Baixo , Células Epiteliais/metabolismo , Células HEK293 , Humanos , Mutação , Fosforilação , Transdução de Sinais
15.
PLoS Comput Biol ; 13(3): e1005411, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28273072

RESUMO

Mechanical coherence of cell layers is essential for epithelia to function as tissue barriers and to control active tissue dynamics during morphogenesis. RhoA signaling at adherens junctions plays a key role in this process by coupling cadherin-based cell-cell adhesion together with actomyosin contractility. Here we propose and analyze a mathematical model representing core interactions involved in the spatial localization of junctional RhoA signaling. We demonstrate how the interplay between biochemical signaling through positive feedback, combined with diffusion on the cell membrane and mechanical forces generated in the cortex, can determine the spatial distribution of RhoA signaling at cell-cell junctions. This dynamical mechanism relies on the balance between a propagating bistable signal that is opposed by an advective flow generated by an actomyosin stress gradient. Experimental observations on the behavior of the system when contractility is inhibited are in qualitative agreement with the predictions of the model.


Assuntos
Actomiosina/fisiologia , Junções Aderentes/fisiologia , Células Epiteliais/fisiologia , Mecanotransdução Celular/fisiologia , Contração Muscular/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Actomiosina/química , Junções Aderentes/química , Animais , Simulação por Computador , Células Epiteliais/química , Humanos , Modelos Biológicos , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/fisiologia , Estresse Mecânico , Proteína rhoA de Ligação ao GTP/química
16.
Cell Cycle ; 15(22): 3033-3041, 2016 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-27650961

RESUMO

Non-muscle myosin II (NMII) motor proteins are responsible for generating contractile forces inside eukaryotic cells. There is also a growing interest in the capacity for these motor proteins to influence cell signaling through scaffolding, especially in the context of RhoA GTPase signaling. We previously showed that NMIIA accumulation and stability within specific regions of the cell cortex, such as the zonula adherens (ZA), allows the formation of a stable RhoA signaling zone. Now we demonstrate a key role for Coronin 1B in maintaining this junctional pool of NMIIA, as depletion of Coronin 1B significantly compromised myosin accumulation and stability at junctions. The loss of junctional NMIIA, upon Coronin 1B knockdown, perturbed RhoA signaling due to enhanced junctional recruitment of the RhoA antagonist, p190B Rho GAP. This effect was blocked by the expression of phosphomimetic MRLC-DD, thus reinforcing the central role of NMII in regulating RhoA signaling.


Assuntos
4-Butirolactona/análogos & derivados , Junções Intercelulares/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , 4-Butirolactona/metabolismo , Actomiosina/metabolismo , Junções Aderentes/metabolismo , Células CACO-2 , Caderinas/metabolismo , Células Epiteliais/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Células MCF-7 , Modelos Biológicos , Cadeias Leves de Miosina/metabolismo , Fenótipo , Estabilidade Proteica
17.
J Biol Chem ; 291(23): 12003-13, 2016 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-27059955

RESUMO

Keratins 8/18 (K8/18) are phosphoglycoproteins and form the major intermediate filament network of simple epithelia. The three O-GlcNAcylation (Ser(29), Ser(30), and Ser(48)) and two phosphorylation (Ser(33) and Ser(52)) serine sites on K18 are well characterized. Both of these modifications have been reported to increase K18 solubility and regulate its filament organization. In this report, we investigated the site-specific interplay between these two modifications in regulating the functional properties of K18, like solubility, stability, and filament organization. An immortalized hepatocyte cell line (HHL-17) stably expressing site-specific single, double, and triple O-GlcNAc and phosphomutants of K18 were used to identify the site(s) critical for regulating these functions. Keratin 18 mutants where O-GlcNAcylation at Ser(30) was abolished (K18-S30A) exhibited reduced phosphorylation induced solubility, increased stability, defective filament architecture, and slower migration. Interestingly, K18-S30A mutants also showed loss of phosphorylation at Ser(33), a modification known to regulate the solubility of K18. Further to this, the K18 phosphomutant (K18-S33A) mimicked K18-S30A in its stability, filament organization, and cell migration. These results indicate that O-GlcNAcylation at Ser(30) promotes phosphorylation at Ser(33) to regulate the functional properties of K18 and also impact cellular processes like migration. O-GlcNAcylation and phosphorylation on the same or adjacent sites on most proteins antagonize each other in regulating protein functions. Here we report a novel, positive interplay between O-GlcNAcylation and phosphorylation at adjacent sites on K18 to regulate its fundamental properties.


Assuntos
Acetilglucosamina/metabolismo , Queratina-18/metabolismo , Serina/metabolismo , Acilação , Sítios de Ligação/genética , Linhagem Celular , Movimento Celular/genética , Fibronectinas/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Immunoblotting , Queratina-18/genética , Microscopia Confocal , Mutação de Sentido Incorreto , Fosforilação , Serina/genética
18.
Nat Cell Biol ; 17(10): 1282-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26368311

RESUMO

Actomyosin at the epithelial zonula adherens (ZA) generates junctional tension for tissue integrity and morphogenesis. This requires the RhoA GTPase, which establishes a strikingly stable active zone at the ZA. Mechanisms must then exist to confer robustness on junctional RhoA signalling at the population level. We now identify a feedback network that generates a stable mesoscopic RhoA zone out of dynamic elements. The key is scaffolding of ROCK1 to the ZA by myosin II. ROCK1 protects junctional RhoA by phosphorylating Rnd3 to prevent the cortical recruitment of the Rho suppressor, p190B RhoGAP. Combining predictive modelling and experimentation, we show that this network constitutes a bistable dynamical system that is realized at the population level of the ZA. Thus, stability of the RhoA zone is an emergent consequence of the network of interactions that allow myosin II to feedback to RhoA.


Assuntos
Junções Aderentes/metabolismo , Caderinas/metabolismo , Miosina Tipo II/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Células CACO-2 , Caderinas/genética , Cães , Retroalimentação Fisiológica , Transferência Ressonante de Energia de Fluorescência , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Células HEK293 , Humanos , Immunoblotting , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Células MCF-7 , Células Madin Darby de Rim Canino , Microscopia Confocal , Microscopia de Fluorescência , Miosina Tipo II/genética , Fosforilação , Interferência de RNA , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética
19.
Biochem Biophys Res Commun ; 460(2): 302-7, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25791476

RESUMO

Poly-N-acetyl-lactosamine (polyLacNAc) on N-glycans facilitate lung specific metastasis of melanoma cells by serving as high affinity ligands for galectin-3, expressed in highest amounts in the lungs, on almost all its tissue compartments including on the surface of vascular endothelium. PolyLacNAc not only aids in initial arrest on the organ endothelium but in all the events of extravasation. Inhibition of polyLacNAc synthesis, or competitive inhibition of its interaction with galectin-3 all inhibited these processes and experimental metastasis. Transgenic galectin-3 mice, viz., gal-3(+/+) (wild type), gal-3(+/-) (hemizygous) and gal-3(-/-) (null) have been used to prove that galectin-3/polyLacNAc interactions are indeed critical for lung specific metastasis. Gal-3(+/-) mice which showed <50% expression of galectin-3 on the lungs also showed proportionate decrease in the number of B16F10 melanoma metastatic colonies affirming that galectin-3 and polyLacNAc interactions are indeed key determinants of lung metastasis. However, surprisingly, the number and size of metastatic colonies in gal-3(-/-) mice was very similar as that seen in gal-3(+/+) mice. The levels of lactose binding lectins on the lungs and the transcripts of other galectins (galectin-1, -8 and -9) which are expressed on lungs and have similar sugar binding specificities as galectins-3, remain unchanged in gal-3(+/+) and gal-3(-/-) mice. Further, inhibition of N-glycosylation with Swainsonine (SW) which drastically reduces metastasis of B16F10 cells in gal-3(+/+) mice, did not affect lung metastasis when assessed in gal-3(-/-) mice. Together, these results rule out the possibility of some other galectin taking over the function of galectin-3 in gal-3(-/-) mice. Chimeric mice generated to assess if absence of any effect on metastasis is due to compromised tumor immunity by replacing bone marrow of gal-3(-/-) mice with that from gal-3(+/+) mice, also failed to impact melanoma metastasis. As galectin-3 regulates several immune functions including maturation of different immune cells, compromised tumor immunity could be the major determinant of melanoma metastasis in gal-3(-/-) mice and warrants thorough investigation.


Assuntos
Galectina 3/fisiologia , Metástase Neoplásica , Polissacarídeos/fisiologia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Galectina 3/genética , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase
20.
J Cancer Res Clin Oncol ; 141(9): 1563-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25614122

RESUMO

PURPOSE: Expression of lysosome-associated membrane protein-1 (LAMP1) on the surface correlates with metastatic potential of B16 melanoma cells. Downregulation of their expression in high metastatic (B16F10) cells reduced their surface expression and metastatic potential. Present investigations explore if overexpression of LAMP1 on the surface of low metastatic (B16F1) cells augment their metastatic ability, and if so, how? METHODS: B16F1 cells were transduced with lentiviral vector carrying mutant-LAMP1 (Y386A) (mutLAMP1). Surface expression of LAMP1 and carbohydrates was analyzed by flow cytometry, immunofluorescence and/or immunoprecipitation and Western blotting. Cell spreading and motility were assessed on components of extracellular matrix (ECM) (fibronectin) and basement membrane (BM) (matrigel), and galectin-3-coated coverslips/plates. Metastatic potential was assessed using experimental metastasis assay. RESULTS: Pre-incubation with anti-LAMP1 antibodies significantly reduced lung metastasis of B16F10 cells. Overexpression of mutLAMP1 significantly increased its surface expression on B16F1 cells, resulting in increased cellular spreading and motility on fibronectin and matrigel. LAMP1 is the major carrier of poly-N-acetyllactosamine (polyLacNAc) on B16F10 cells. However, significantly higher expression of mutLAMP1 had no effect on galectin-3 binding on cell surface or on spreading or motility of cells on galectin-3-coated coverslips/plates. These cells also failed to show any gain in metastatic ability. This could be because LAMP1 from these cells carried significantly lower levels of polyLacNAc in comparison with B16F10 cells. CONCLUSIONS: PolyLacNAc on B16F10 cells and galectin-3 on lungs are the major participants in melanoma metastasis. Although surface LAMP1 promotes interactions with organ ECM and BM, carbohydrates on LAMP1 play a decisive role in dictating lung metastasis.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Proteínas de Membrana Lisossomal/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Animais , Metabolismo dos Carboidratos , Movimento Celular/fisiologia , Feminino , Galectina 3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...