Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37548995

RESUMO

Cell-generated forces play a major role in coordinating the large-scale behavior of cell assemblies, in particular during development, wound healing, and cancer. Mechanical signals propagate faster than biochemical signals, but can have similar effects, especially in epithelial tissues with strong cell-cell adhesion. However, a quantitative description of the transmission chain from force generation in a sender cell, force propagation across cell-cell boundaries, and the concomitant response of receiver cells is missing. For a quantitative analysis of this important situation, here we propose a minimal model system of two epithelial cells on an H-pattern ('cell doublet'). After optogenetically activating RhoA, a major regulator of cell contractility, in the sender cell, we measure the mechanical response of the receiver cell by traction force and monolayer stress microscopies. In general, we find that the receiver cells show an active response so that the cell doublet forms a coherent unit. However, force propagation and response of the receiver cell also strongly depend on the mechano-structural polarization in the cell assembly, which is controlled by cell-matrix adhesion to the adhesive micropattern. We find that the response of the receiver cell is stronger when the mechano-structural polarization axis is oriented perpendicular to the direction of force propagation, reminiscent of the Poisson effect in passive materials. We finally show that the same effects are at work in small tissues. Our work demonstrates that cellular organization and active mechanical response of a tissue are key to maintain signal strength and lead to the emergence of elasticity, which means that signals are not dissipated like in a viscous system, but can propagate over large distances.


Assuntos
Células Epiteliais , Fenômenos Mecânicos , Células Epiteliais/fisiologia , Epitélio , Adesão Celular/fisiologia , Elasticidade , Estresse Mecânico
2.
Nat Commun ; 14(1): 2740, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37217519

RESUMO

Cell migration is crucial for cancer dissemination. We find that AMP-activated protein kinase (AMPK) controls cell migration by acting as an adhesion sensing molecular hub. In 3-dimensional matrices, fast-migrating amoeboid cancer cells exert low adhesion/low traction linked to low ATP/AMP, leading to AMPK activation. In turn, AMPK plays a dual role controlling mitochondrial dynamics and cytoskeletal remodelling. High AMPK activity in low adhering migratory cells, induces mitochondrial fission, resulting in lower oxidative phosphorylation and lower mitochondrial ATP. Concurrently, AMPK inactivates Myosin Phosphatase, increasing Myosin II-dependent amoeboid migration. Reducing adhesion or mitochondrial fusion or activating AMPK induces efficient rounded-amoeboid migration. AMPK inhibition suppresses metastatic potential of amoeboid cancer cells in vivo, while a mitochondrial/AMPK-driven switch is observed in regions of human tumours where amoeboid cells are disseminating. We unveil how mitochondrial dynamics control cell migration and suggest that AMPK is a mechano-metabolic sensor linking energetics and the cytoskeleton.


Assuntos
Proteínas Quinases Ativadas por AMP , Dinâmica Mitocondrial , Neoplasias , Humanos , Trifosfato de Adenosina/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Adesão Celular , Movimento Celular/fisiologia , Miosina Tipo II/metabolismo , Fosforilação Oxidativa , Fosforilação
3.
Proc Natl Acad Sci U S A ; 119(49): e2201600119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36454762

RESUMO

The direction in which a cell divides is set by the orientation of its mitotic spindle and is important for determining cell fate, controlling tissue shape, and maintaining tissue architecture. Divisions parallel to the epithelial plane sustain tissue expansion. By contrast, divisions perpendicular to the plane promote tissue stratification and lead to the loss of epithelial cells from the tissue-an event that has been suggested to promote metastasis. Much is known about the molecular machinery involved in orienting the spindle, but less is known about the contribution of mechanical factors, such as tissue tension, in ensuring spindle orientation in the plane of the epithelium. This is important as epithelia are continuously subjected to mechanical stresses. To explore this further, we subjected suspended epithelial monolayers devoid of extracellular matrix to varying levels of tissue tension to study the orientation of cell divisions relative to the tissue plane. This analysis revealed that lowering tissue tension by compressing epithelial monolayers or by inhibiting myosin contractility increased the frequency of out-of-plane divisions. Reciprocally, increasing tissue tension by elevating cell contractility or by tissue stretching restored accurate in-plane cell divisions. Moreover, a characterization of the geometry of cells within these epithelia suggested that spindles can sense tissue tension through its impact on tension at subcellular surfaces, independently of their shape. Overall, these data suggest that accurate spindle orientation in the plane of the epithelium relies on a threshold level of tension at intercellular junctions.


Assuntos
Células Epiteliais , Junções Intercelulares , Epitélio , Divisão Celular , Matriz Extracelular
4.
Nat Commun ; 12(1): 6511, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764258

RESUMO

In animal cells, shape is mostly determined by the actomyosin cortex, a thin cytoskeletal network underlying the plasma membrane. Myosin motors generate tension in the cortex, and tension gradients result in cellular deformations. As such, many cell morphogenesis studies have focused on the mechanisms controlling myosin activity and recruitment to the cortex. Here, we demonstrate using super-resolution microscopy that myosin does not always overlap with actin at the cortex, but remains restricted towards the cytoplasm in cells with low cortex tension. We propose that this restricted penetration results from steric hindrance, as myosin minifilaments are considerably larger than the cortical actin meshsize. We identify myosin activity and actin network architecture as key regulators of myosin penetration into the cortex, and show that increasing myosin penetration increases cortical tension. Our study reveals that the spatial coordination of myosin and actin at the cortex regulates cell surface mechanics, and unveils an important mechanism whereby myosin size controls its action by limiting minifilament penetration into the cortical actin network. More generally, our findings suggest that protein size could regulate function in dense cytoskeletal structures.


Assuntos
Miosinas/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Membrana Celular/metabolismo
5.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445693

RESUMO

Mechanical forces acting on cell-cell adhesion modulate the barrier function of endothelial cells. The actively remodeled actin cytoskeleton impinges on cell-cell adhesion to counteract external forces. We applied stress on endothelial monolayers by mechanical stretch to uncover the role of BRAF in the stress-induced response. Control cells responded to external forces by organizing and stabilizing actin cables in the stretched cell junctions. This was accompanied by an increase in intercellular gap formation, which was prevented in BRAF knockdown monolayers. In the absence of BRAF, there was excess stress fiber formation due to the enhanced reorganization of actin fibers. Our findings suggest that stretch-induced intercellular gap formation, leading to a decrease in barrier function of blood vessels, can be reverted by BRAF RNAi. This is important when the endothelium experiences changes in external stresses caused by high blood pressure, leading to edema, or by immune or cancer cells in inflammation or metastasis.


Assuntos
Células Endoteliais/metabolismo , Junções Comunicantes/fisiologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Actinas/fisiologia , Adesão Celular/fisiologia , Células Cultivadas , Citoesqueleto/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Humanos , Junções Intercelulares/fisiologia , Fenômenos Mecânicos , Proteínas Proto-Oncogênicas B-raf/fisiologia
6.
Elife ; 102021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-34014166

RESUMO

How cells with different genetic makeups compete in tissues is an outstanding question in developmental biology and cancer research. Studies in recent years have revealed that cell competition can either be driven by short-range biochemical signalling or by long-range mechanical stresses in the tissue. To date, cell competition has generally been characterised at the population scale, leaving the single-cell-level mechanisms of competition elusive. Here, we use high time-resolution experimental data to construct a multi-scale agent-based model for epithelial cell competition and use it to gain a conceptual understanding of the cellular factors that governs competition in cell populations within tissues. We find that a key determinant of mechanical competition is the difference in homeostatic density between winners and losers, while differences in growth rates and tissue organisation do not affect competition end result. In contrast, the outcome and kinetics of biochemical competition is strongly influenced by local tissue organisation. Indeed, when loser cells are homogenously mixed with winners at the onset of competition, they are eradicated; however, when they are spatially separated, winner and loser cells coexist for long times. These findings suggest distinct biophysical origins for mechanical and biochemical modes of cell competition.


Assuntos
Competição entre as Células , Células Epiteliais/fisiologia , Mecanotransdução Celular , Modelos Biológicos , Animais , Apoptose , Fenômenos Biomecânicos , Comunicação Celular , Proliferação de Células , Simulação por Computador , Cães , Genótipo , Cinética , Células Madin Darby de Rim Canino , Fenótipo , Análise de Célula Única , Estresse Mecânico
7.
Nat Cell Biol ; 22(7): 803-814, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32572169

RESUMO

Cell shape is controlled by the submembranous cortex, an actomyosin network mainly generated by two actin nucleators: the Arp2/3 complex and the formin mDia1. Changes in relative nucleator activity may alter cortical organization, mechanics and cell shape. Here we investigate how nucleation-promoting factors mediate interactions between nucleators. In vitro, the nucleation-promoting factor SPIN90 promotes formation of unbranched filaments by Arp2/3, a process thought to provide the initial filament for generation of dendritic networks. Paradoxically, in cells, SPIN90 appears to favour a formin-dominated cortex. Our in vitro experiments reveal that this feature stems mainly from two mechanisms: efficient recruitment of mDia1 to SPIN90-Arp2/3 nucleated filaments and formation of a ternary SPIN90-Arp2/3-mDia1 complex that greatly enhances filament nucleation. Both mechanisms yield rapidly elongating filaments with mDia1 at their barbed ends and SPIN90-Arp2/3 at their pointed ends. Thus, in networks, SPIN90 lowers branching densities and increases the proportion of long filaments elongated by mDia1.


Assuntos
Citoesqueleto de Actina/fisiologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Forminas/metabolismo , Melanoma/patologia , Proteínas Musculares/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Blástula/citologia , Blástula/metabolismo , Forma Celular , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Forminas/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteínas Musculares/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
8.
Semin Cancer Biol ; 63: 60-68, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31108201

RESUMO

Cell competition is a quality control mechanism in tissues that results in the elimination of less fit cells. Over the past decade, the phenomenon of cell competition has been identified in many physiological and pathological contexts, driven either by biochemical signaling or by mechanical forces within the tissue. In both cases, competition has generally been characterized based on the elimination of loser cells at the population level, but significantly less attention has been focused on determining how single-cell dynamics and interactions regulate population-wide changes. In this review, we describe quantitative strategies and outline the outstanding challenges in understanding the single cell rules governing tissue-scale competition dynamics. We propose quantitative metrics to characterize single cell behaviors in competition and use them to distinguish the types and outcomes of competition. We describe how such metrics can be measured experimentally using a novel combination of high-throughput imaging and machine learning algorithms. We outline the experimental challenges to quantify cell fate dynamics with high-statistical precision, and describe the utility of computational modeling in testing hypotheses not easily accessible in experiments. In particular, cell-based modeling approaches that combine mechanical interaction of cells with decision-making rules for cell fate choices provide a powerful framework to understand and reverse-engineer the diverse rules of cell competition.


Assuntos
Aprendizado de Máquina , Imagem Molecular/métodos , Neoplasias/patologia , Análise de Célula Única/métodos , Animais , Comunicação Celular/fisiologia , Simulação por Computador , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/etiologia , Neoplasias/metabolismo , Transdução de Sinais
9.
Nat Mater ; 19(1): 109-117, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31451778

RESUMO

Throughout embryonic development and adult life, epithelia are subjected to compressive deformations. While these have been shown to trigger mechanosensitive responses such as cell extrusion and differentiation, which span tens of minutes, little is known about how epithelia adapt to compression over shorter timescales. Here, using suspended epithelia, we uncover the immediate response of epithelial tissues to the application of in-plane compressive strains (5-80%). We show that fast compression induces tissue buckling followed by actomyosin-dependent tissue flattening that erases the buckle within tens of seconds, in both mono- and multi-layered epithelia. Strikingly, we identify a well-defined limit to this response, so that stable folds form in the tissue when compressive strains exceed a 'buckling threshold' of ~35%. A combination of experiment and modelling shows that this behaviour is orchestrated by adaptation of the actomyosin cytoskeleton as it re-establishes tissue tension following compression. Thus, tissue pre-tension allows epithelia to both buffer against deformation and sets their ability to form and retain folds during morphogenesis.


Assuntos
Actomiosina/química , Epitélio/fisiologia , Animais , Caderinas/fisiologia , Força Compressiva , Citoesqueleto , Cães , Elasticidade , Células Epiteliais/citologia , Epitélio/embriologia , Proteínas de Fluorescência Verde , Células Madin Darby de Rim Canino , Microscopia Confocal , Modelos Biológicos , Morfogênese , Estresse Mecânico , Viscosidade
10.
Dev Cell ; 45(5): 565-579.e3, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29870718

RESUMO

Contact inhibition of locomotion is defined as the behavior of cells to cease migrating in their former direction after colliding with another cell. It has been implicated in multiple developmental processes and its absence has been linked to cancer invasion. Cellular forces are thought to govern this process; however, the exact role of traction through cell-matrix adhesions and tension through cell-cell adhesions during contact inhibition of locomotion remains unknown. Here we use neural crest cells to address this and show that cell-matrix adhesions are rapidly disassembled at the contact between two cells upon collision. This disassembly is dependent upon the formation of N-cadherin-based cell-cell adhesions and driven by Src and FAK activity. We demonstrate that the loss of cell-matrix adhesions near the contact leads to a buildup of tension across the cell-cell contact, a step that is essential to drive cell-cell separation after collision.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Embrião não Mamífero/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Crista Neural/fisiologia , Xenopus laevis/fisiologia , Quinases da Família src/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Embrião não Mamífero/citologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Crista Neural/citologia , Fosforilação , Xenopus laevis/embriologia , Quinases da Família src/genética
11.
Methods Mol Biol ; 1749: 375-386, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29526011

RESUMO

Cell migration plays a key role in many physiological and pathological conditions during which cells migrate primarily in the 3D environments formed by tissues. Microfluidics enables the design of simple devices that can mimic in a highly controlled manner the geometry and dimensions of the interstices encountered by cells in the body. Here we describe the design, fabrication, and implementation of an array of channels with a range of cross sections to investigate migration of cells and cell clusters through confined spaces. By combining this assay with a motorized microscope stage, image data can be acquired with high throughput to determine the physical limits of migration in confined environments and their biological origin.


Assuntos
Movimento Celular/fisiologia , Microfluídica/métodos , Linhagem Celular Tumoral , Feminino , Humanos
12.
Nature ; 554(7693): 523-527, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29443958

RESUMO

Collective cell migration is essential for morphogenesis, tissue remodelling and cancer invasion. In vivo, groups of cells move in an orchestrated way through tissues. This movement involves mechanical as well as molecular interactions between cells and their environment. While the role of molecular signals in collective cell migration is comparatively well understood, how tissue mechanics influence collective cell migration in vivo remains unknown. Here we investigated the importance of mechanical cues in the collective migration of the Xenopus laevis neural crest cells, an embryonic cell population whose migratory behaviour has been likened to cancer invasion. We found that, during morphogenesis, the head mesoderm underlying the cephalic neural crest stiffens. This stiffening initiates an epithelial-to-mesenchymal transition in neural crest cells and triggers their collective migration. To detect changes in their mechanical environment, neural crest cells use mechanosensation mediated by the integrin-vinculin-talin complex. By performing mechanical and molecular manipulations, we show that mesoderm stiffening is necessary and sufficient to trigger neural crest migration. Finally, we demonstrate that convergent extension of the mesoderm, which starts during gastrulation, leads to increased mesoderm stiffness by increasing the cell density underneath the neural crest. These results show that convergent extension of the mesoderm has a role as a mechanical coordinator of morphogenesis, and reveal a link between two apparently unconnected processes-gastrulation and neural crest migration-via changes in tissue mechanics. Overall, we demonstrate that changes in substrate stiffness can trigger collective cell migration by promoting epithelial-to-mesenchymal transition in vivo. More broadly, our results raise the idea that tissue mechanics combines with molecular effectors to coordinate morphogenesis.


Assuntos
Movimento Celular , Mecanotransdução Celular , Mesoderma/fisiologia , Morfogênese , Crista Neural/citologia , Xenopus laevis/embriologia , Animais , Transição Epitelial-Mesenquimal , Matriz Extracelular , Feminino , Gastrulação , Dureza , Integrinas/metabolismo , Mesoderma/citologia , Mesoderma/embriologia
13.
J Cell Biol ; 217(1): 195-209, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29133484

RESUMO

Centrosome amplification is a common feature of human tumors. To survive, cancer cells cluster extra centrosomes during mitosis, avoiding the detrimental effects of multipolar divisions. However, it is unclear whether clustering requires adaptation or is inherent to all cells. Here, we show that cells have varied abilities to cluster extra centrosomes. Epithelial cells are innately inefficient at clustering even in the presence of HSET/KIFC1, which is essential but not sufficient to promote clustering. The presence of E-cadherin decreases cortical contractility during mitosis through a signaling cascade leading to multipolar divisions, and its knockout promotes clustering and survival of cells with multiple centrosomes. Cortical contractility restricts centrosome movement at a minimal distance required for HSET/KIFC1 to exert its function, highlighting a biphasic model for centrosome clustering. In breast cancer cell lines, increased levels of centrosome amplification are accompanied by efficient clustering and loss of E-cadherin, indicating that this is an important adaptation mechanism to centrosome amplification in cancer.


Assuntos
Neoplasias da Mama/patologia , Caderinas/genética , Centrossomo/metabolismo , Receptor com Domínio Discoidina 1/genética , Células Epiteliais/patologia , Comunicação Celular/genética , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Cinesinas/metabolismo , Mitose/genética
14.
Mol Biol Cell ; 28(23): 3215-3228, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-28931601

RESUMO

Cell competition is a quality-control mechanism through which tissues eliminate unfit cells. Cell competition can result from short-range biochemical inductions or long-range mechanical cues. However, little is known about how cell-scale interactions give rise to population shifts in tissues, due to the lack of experimental and computational tools to efficiently characterize interactions at the single-cell level. Here, we address these challenges by combining long-term automated microscopy with deep-learning image analysis to decipher how single-cell behavior determines tissue makeup during competition. Using our high-throughput analysis pipeline, we show that competitive interactions between MDCK wild-type cells and cells depleted of the polarity protein scribble are governed by differential sensitivity to local density and the cell type of each cell's neighbors. We find that local density has a dramatic effect on the rate of division and apoptosis under competitive conditions. Strikingly, our analysis reveals that proliferation of the winner cells is up-regulated in neighborhoods mostly populated by loser cells. These data suggest that tissue-scale population shifts are strongly affected by cellular-scale tissue organization. We present a quantitative mathematical model that demonstrates the effect of neighbor cell-type dependence of apoptosis and division in determining the fitness of competing cell lines.


Assuntos
Proteínas de Drosophila/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Proteínas de Membrana/metabolismo , Microscopia/métodos , Animais , Apoptose , Comunicação Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Cães , Drosophila melanogaster/metabolismo , Processamento de Imagem Assistida por Computador/estatística & dados numéricos , Células Madin Darby de Rim Canino , Ativação Transcricional , Proteínas Supressoras de Tumor
15.
Nat Commun ; 8: 14396, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28186127

RESUMO

Contractile forces are the end effectors of cell migration, division, morphogenesis, wound healing and cancer invasion. Here we report optogenetic tools to upregulate and downregulate such forces with high spatiotemporal accuracy. The technology relies on controlling the subcellular activation of RhoA using the CRY2/CIBN light-gated dimerizer system. We fused the catalytic domain (DHPH domain) of the RhoA activator ARHGEF11 to CRY2-mCherry (optoGEF-RhoA) and engineered its binding partner CIBN to bind either to the plasma membrane or to the mitochondrial membrane. Translocation of optoGEF-RhoA to the plasma membrane causes a rapid and local increase in cellular traction, intercellular tension and tissue compaction. By contrast, translocation of optoGEF-RhoA to mitochondria results in opposite changes in these physical properties. Cellular changes in contractility are paralleled by modifications in the nuclear localization of the transcriptional regulator YAP, thus showing the ability of our approach to control mechanotransductory signalling pathways in time and space.


Assuntos
Movimento Celular/fisiologia , Mecanotransdução Celular/fisiologia , Optogenética/métodos , Transdução de Sinais/fisiologia , Animais , Membrana Celular/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Cães , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Células Madin Darby de Rim Canino , Membranas Mitocondriais/metabolismo , Ligação Proteica , Transporte Proteico , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína Vermelha Fluorescente
16.
Dev Cell ; 38(4): 371-83, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27554857

RESUMO

Fascin is an F-actin-bundling protein shown to stabilize filopodia and regulate adhesion dynamics in migrating cells, and its expression is correlated with poor prognosis and increased metastatic potential in a number of cancers. Here, we identified the nuclear envelope protein nesprin-2 as a binding partner for fascin in a range of cell types in vitro and in vivo. Nesprin-2 interacts with fascin through a direct, F-actin-independent interaction, and this binding is distinct and separable from a role for fascin within filopodia at the cell periphery. Moreover, disrupting the interaction between fascin and nesprin-2 C-terminal domain leads to specific defects in F-actin coupling to the nuclear envelope, nuclear movement, and the ability of cells to deform their nucleus to invade through confined spaces. Together, our results uncover a role for fascin that operates independently of filopodia assembly to promote efficient cell migration and invasion.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Pseudópodes/fisiologia , Células 3T3 , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Drosophila , Células HeLa , Humanos , Camundongos , Complexos Multiproteicos/metabolismo , Invasividade Neoplásica/patologia , Neoplasias/patologia , Membrana Nuclear/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína
17.
Curr Opin Cell Biol ; 42: 113-120, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27371889

RESUMO

Cellularised materials are composed of cells interfaced through specialised intercellular junctions that link the cytoskeleton of one cell to that of its neighbours allowing for transmission of forces. Cellularised materials are common in early development and adult tissues where they can be found in the form of cell sheets, cysts, or amorphous aggregates and in pathophysiological conditions such as cancerous tumours. Given the growing realisation that forces can regulate cell physiology and developmental processes, understanding how cellularised materials deform under mechanical stress or dissipate stress appear as key biological questions. In this review, we will discuss the dynamic mechanical properties of cellularised materials devoid of extracellular matrix.


Assuntos
Células/metabolismo , Animais , Fenômenos Biomecânicos , Agregação Celular , Humanos , Modelos Biológicos , Morfogênese , Reologia
18.
Development ; 143(13): 2305-10, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226324

RESUMO

Coordinating cell differentiation with cell growth and division is crucial for the successful development, homeostasis and regeneration of multicellular tissues. Here, we use bristle patterning in the fly notum as a model system to explore the regulatory and functional coupling of cell cycle progression and cell fate decision-making. The pattern of bristles and intervening epithelial cells (ECs) becomes established through Notch-mediated lateral inhibition during G2 phase of the cell cycle, as neighbouring cells physically interact with each other via lateral contacts and/or basal protrusions. Since Notch signalling controls cell division timing downstream of Cdc25, ECs in lateral contact with a Delta-expressing cell experience higher levels of Notch signalling and divide first, followed by more distant neighbours, and lastly Delta-expressing cells. Conversely, mitotic entry and cell division makes ECs refractory to lateral inhibition signalling, fixing their fate. Using a combination of experiments and computational modelling, we show that this reciprocal relationship between Notch signalling and cell cycle progression acts like a developmental clock, providing a delimited window of time during which cells decide their fate, ensuring efficient and orderly bristle patterning.


Assuntos
Padronização Corporal , Ciclo Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Drosophila melanogaster/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Órgãos dos Sentidos/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo , Vibrissas/citologia , Vibrissas/embriologia
19.
J Leukoc Biol ; 99(5): 699-710, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26590149

RESUMO

The immunological synapse is a highly structured and molecularly dynamic interface between communicating immune cells. Although the immunological synapse promotes T cell activation by dendritic cells, the specific organization of the immunological synapse on the dendritic cell side in response to T cell engagement is largely unknown. In this study, confocal and electron microscopy techniques were used to investigate the role of dendritic cell actin regulation in immunological synapse formation, stabilization, and function. In the dendritic cell-restricted absence of the Wiskott-Aldrich syndrome protein, an important regulator of the actin cytoskeleton in hematopoietic cells, the immunological synapse contact with T cells occupied a significantly reduced surface area. At a molecular level, the actin network localized to the immunological synapse exhibited reduced stability, in particular, of the actin-related protein-2/3-dependent, short-filament network. This was associated with decreased polarization of dendritic cell-associated ICAM-1 and MHC class II, which was partially dependent on Wiskott-Aldrich syndrome protein phosphorylation. With the use of supported planar lipid bilayers incorporating anti-ICAM-1 and anti-MHC class II antibodies, the dendritic cell actin cytoskeleton organized into recognizable synaptic structures but interestingly, formed Wiskott-Aldrich syndrome protein-dependent podosomes within this area. These findings demonstrate that intrinsic dendritic cell cytoskeletal remodeling is a key regulatory component of normal immunological synapse formation, likely through consolidation of adhesive interaction and modulation of immunological synapse stability.


Assuntos
Citoesqueleto de Actina/metabolismo , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Sinapses Imunológicas/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Animais , Recuperação de Fluorescência Após Fotodegradação , Molécula 1 de Adesão Intercelular/metabolismo , Bicamadas Lipídicas/metabolismo , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Podossomos/metabolismo
20.
Cell Rep ; 13(12): 2699-714, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26711338

RESUMO

Cancer-associated fibroblasts (CAFs) are non-cancerous cells found in solid tumors that remodel the tumor matrix and promote cancer invasion and angiogenesis. Here, we demonstrate that Cdc42EP3/BORG2 is required for the matrix remodeling, invasion, angiogenesis, and tumor-growth-promoting abilities of CAFs. Cdc42EP3 functions by coordinating the actin and septin networks. Furthermore, depletion of SEPT2 has similar effects to those of loss of Cdc42EP3, indicating a role for the septin network in the tumor stroma. Cdc42EP3 is upregulated early in fibroblast activation and precedes the emergence of the highly contractile phenotype characteristic of CAFs. Depletion of Cdc42EP3 in normal fibroblasts prevents their activation by cancer cells. We propose that Cdc42EP3 sensitizes fibroblasts to further cues-in particular, those activating actomyosin contractility-and thereby enables the generation of the pathological activated fibroblast state.


Assuntos
Fibroblastos/metabolismo , Fibroblastos/patologia , Reguladores de Proteínas de Ligação ao GTP/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Septinas/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA