Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 989
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(1): 188-200.e16, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28867286

RESUMEN

Actin filaments polymerizing against membranes power endocytosis, vesicular traffic, and cell motility. In vitro reconstitution studies suggest that the structure and the dynamics of actin networks respond to mechanical forces. We demonstrate that lamellipodial actin of migrating cells responds to mechanical load when membrane tension is modulated. In a steady state, migrating cell filaments assume the canonical dendritic geometry, defined by Arp2/3-generated 70° branch points. Increased tension triggers a dense network with a broadened range of angles, whereas decreased tension causes a shift to a sparse configuration dominated by filaments growing perpendicularly to the plasma membrane. We show that these responses emerge from the geometry of branched actin: when load per filament decreases, elongation speed increases and perpendicular filaments gradually outcompete others because they polymerize the shortest distance to the membrane, where they are protected from capping. This network-intrinsic geometrical adaptation mechanism tunes protrusive force in response to mechanical load.


Asunto(s)
Citoesqueleto de Actina/química , Citoesqueleto de Actina/ultraestructura , Queratinocitos/ultraestructura , Seudópodos/química , Seudópodos/ultraestructura , Animales , Membrana Celular/química , Queratinocitos/química , Microscopía Electrónica , Pez Cebra
2.
PLoS Biol ; 18(8): e3000774, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32745097

RESUMEN

The Scar/WAVE complex is the principal catalyst of pseudopod and lamellipod formation. Here we show that Scar/WAVE's proline-rich domain is polyphosphorylated after the complex is activated. Blocking Scar/WAVE activation stops phosphorylation in both Dictyostelium and mammalian cells, implying that phosphorylation modulates pseudopods after they have been formed, rather than controlling whether they are initiated. Unexpectedly, phosphorylation is not promoted by chemotactic signaling but is greatly stimulated by cell:substrate adhesion and diminished when cells deadhere. Phosphorylation-deficient or phosphomimetic Scar/WAVE mutants are both normally functional and rescue the phenotype of knockout cells, demonstrating that phosphorylation is dispensable for activation and actin regulation. However, pseudopods and patches of phosphorylation-deficient Scar/WAVE last substantially longer in mutants, altering the dynamics and size of pseudopods and lamellipods and thus changing migration speed. Scar/WAVE phosphorylation does not require ERK2 in Dictyostelium or mammalian cells. However, the MAPKKK homologue SepA contributes substantially-sepA mutants have less steady-state phosphorylation, which does not increase in response to adhesion. The mutants also behave similarly to cells expressing phosphorylation-deficient Scar, with longer-lived pseudopods and patches of Scar recruitment. We conclude that pseudopod engagement with substratum is more important than extracellular signals at regulating Scar/WAVE's activity and that phosphorylation acts as a pseudopod timer by promoting Scar/WAVE turnover.


Asunto(s)
Dictyostelium/genética , MAP Quinasa Quinasa Quinasa 3/genética , Proteínas Protozoarias/genética , Seudópodos/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Animales , Sistemas CRISPR-Cas , Adhesión Celular , Línea Celular Tumoral , Quimiotaxis/genética , Dictyostelium/metabolismo , Dictyostelium/ultraestructura , Edición Génica/métodos , Regulación de la Expresión Génica , MAP Quinasa Quinasa Quinasa 3/metabolismo , Melanocitos/metabolismo , Melanocitos/ultraestructura , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Mutación , Células 3T3 NIH , Fenotipo , Fosforilación , Ploidias , Proteínas Protozoarias/metabolismo , Seudópodos/genética , Seudópodos/ultraestructura , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
3.
Nucleic Acids Res ; 49(1): 1-14, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33275144

RESUMEN

Nucleic acid therapeutics (NATs) have proven useful in promoting the degradation of specific transcripts, modifying gene expression, and regulating mRNA splicing. In each situation, efficient delivery of nucleic acids to cells, tissues and intracellular compartments is crucial-both for optimizing efficacy and reducing side effects. Despite successes in NATs, our understanding of their cellular uptake and distribution in tissues is limited. Current methods have yielded insights into distribution of NATs within cells and tissues, but the sensitivity and resolution of these approaches are limited. Here, we show that nanoscale secondary ion mass spectrometry (NanoSIMS) imaging can be used to define the distribution of 5-bromo-2'-deoxythymidine (5-BrdT) modified antisense oligonucleotides (ASO) in cells and tissues with high sensitivity and spatial resolution. This approach makes it possible to define ASO uptake and distribution in different subcellular compartments and to quantify the impact of targeting ligands designed to promote ASO uptake by cells. Our studies showed that phosphorothioate ASOs are associated with filopodia and the inner nuclear membrane in cultured cells, and also revealed substantial cellular and subcellular heterogeneity of ASO uptake in mouse tissues. NanoSIMS imaging represents a significant advance in visualizing uptake and distribution of NATs; this approach will be useful in optimizing efficacy and delivery of NATs for treating human disease.


Asunto(s)
Oligonucleótidos Antisentido/análisis , Oligonucleótidos Fosforotioatos/análisis , Espectrometría de Masa de Ion Secundario/métodos , Células 3T3-L1 , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/análisis , Animales , Receptor de Asialoglicoproteína/análisis , Cesio , Células HEK293 , Células HeLa , Humanos , Riñón/química , Riñón/ultraestructura , Hígado/química , Hígado/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Miocardio/química , Miocardio/ultraestructura , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Fosforotioatos/farmacocinética , Seudópodos/química , Seudópodos/ultraestructura , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , Fracciones Subcelulares/química , Azufre/análisis , Isótopos de Azufre/análisis , Distribución Tisular
4.
Biochem Biophys Res Commun ; 587: 146-152, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34875533

RESUMEN

Lysyl hydroxylase 2 (LH2) regulates intermolecular cross-linking of collagen molecules. Accumulation of LH2-modified collagen, which is highly stable and resistant to collagenase cleavage, is one cause of fibrosis. We previously demonstrated that conventional LH2 knockout mice showed embryonic lethality. Here we established LH2 conditional knockout mice using a tamoxifen-inducible Cre system. Morphological analysis of LH2-deficient fibroblasts by microscopy showed a dramatic increase in the number of filopodia, the finger-like cell surface projections that enable cell movement. The tips and leading edges of these filopodia exhibited up-regulated expression of Myosin-X (Myo10), a regulator of filopodial integrity. Wound healing assays demonstrated that migration of LH2-deficient cells was significantly faster than that of control cells. Gene expression profiling data also supported this phenotype. Together these findings indicate that LH2 deficiency may prevent fibrosis through decreased accumulation of LH2-cross-linked collagen, and that fibroblasts with faster migration contribute to enhanced wound healing activity. In conclusion, our cellular models provide evidence that LH2 deficiency plays a critical role in cell migration mediated through filopodia formation. Understanding the precise role of this phenotype in LH2-deficient cells may be helpful to define the pathogenesis of fibrosis. As such, detailed analyses of fibrosis and wound healing using LH2-deficient mouse models are needed.


Asunto(s)
Fibroblastos/enzimología , Miosinas/genética , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Seudópodos/enzimología , Animales , Movimiento Celular , Colágeno/genética , Colágeno/metabolismo , Fibroblastos/citología , Fibrosis , Regulación de la Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Miosinas/metabolismo , Fenotipo , Cultivo Primario de Células , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/deficiencia , Seudópodos/ultraestructura , Cicatrización de Heridas/genética
5.
FASEB J ; 35(5): e21582, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33835502

RESUMEN

The rapid development of advanced microscopy techniques over recent decades has significantly increased the quality of imaging and our understanding of subcellular structures, such as the organization of the filaments of the cytoskeleton using fluorescence and electron microscopy. However, these recent improvements in imaging techniques have not been matched by similar development of techniques for computational analysis of the images of filament networks that can now be obtained. Hence, for a wide range of applications, reliable computational analysis of such two-dimensional methods remains challenging. Here, we present a new algorithm for tracing of filament networks. This software can extract many important parameters from grayscale images of filament networks, including the mesh hole size, and filament length and connectivity (also known as Coordination Number). In addition, the method allows sub-networks to be distinguished in two-dimensional images using intensity thresholding. We show that the algorithm can be used to analyze images of cytoskeleton networks obtained using different advanced microscopy methods. We have thus developed a new improved method for computational analysis of two-dimensional images of filamentous networks that has wide applications for existing imaging techniques. The algorithm is available as open-source software.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Algoritmos , Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Electrónica de Rastreo/métodos , Microtúbulos/metabolismo , Seudópodos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Citoesqueleto de Actina/ultraestructura , Células Cultivadas , Humanos , Microtúbulos/ultraestructura , Seudópodos/ultraestructura , Epitelio Pigmentado de la Retina/ultraestructura
6.
PLoS Biol ; 17(10): e3000457, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31600188

RESUMEN

Migratory cells use distinct motility modes to navigate different microenvironments, but it is unclear whether these modes rely on the same core set of polarity components. To investigate this, we disrupted actin-related protein 2/3 (Arp2/3) and the WASP-family verprolin homologous protein (WAVE) complex, which assemble branched actin networks that are essential for neutrophil polarity and motility in standard adherent conditions. Surprisingly, confinement rescues polarity and movement of neutrophils lacking these components, revealing a processive bleb-based protrusion program that is mechanistically distinct from the branched actin-based protrusion program but shares some of the same core components and underlying molecular logic. We further find that the restriction of protrusion growth to one site does not always respond to membrane tension directly, as previously thought, but may rely on closely linked properties such as local membrane curvature. Our work reveals a hidden circuit for neutrophil polarity and indicates that cells have distinct molecular mechanisms for polarization that dominate in different microenvironments.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/genética , Actinas/genética , Polaridad Celular/genética , Quimiotaxis/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Fenómenos Biomecánicos , Sistemas CRISPR-Cas , Adhesión Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Polaridad Celular/efectos de los fármacos , Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Edición Génica , Regulación de la Expresión Génica , Células HEK293 , Células HL-60 , Humanos , Microscopía de Fuerza Atómica , N-Formilmetionina Leucil-Fenilalanina/farmacología , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Seudópodos/ultraestructura , Transducción de Señal , Propiedades de Superficie , Familia de Proteínas del Síndrome de Wiskott-Aldrich/deficiencia
7.
J Am Soc Nephrol ; 32(3): 563-579, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33514561

RESUMEN

BACKGROUND: Previous research demonstrated that small Rho GTPases, modulators of the actin cytoskeleton, are drivers of podocyte foot-process effacement in glomerular diseases, such as FSGS. However, a comprehensive understanding of the regulatory networks of small Rho GTPases in podocytes is lacking. METHODS: We conducted an analysis of podocyte transcriptome and proteome datasets for Rho GTPases; mapped in vivo, podocyte-specific Rho GTPase affinity networks; and examined conditional knockout mice and murine disease models targeting Srgap1. To evaluate podocyte foot-process morphology, we used super-resolution microscopy and electron microscopy; in situ proximity ligation assays were used to determine the subcellular localization of the small GTPase-activating protein SRGAP1. We performed functional analysis of CRISPR/Cas9-generated SRGAP1 knockout podocytes in two-dimensional and three-dimensional cultures and quantitative interaction proteomics. RESULTS: We demonstrated SRGAP1 localization to podocyte foot processes in vivo and to cellular protrusions in vitro. Srgap1fl/fl*Six2Cre but not Srgap1fl/fl*hNPHS2Cre knockout mice developed an FSGS-like phenotype at adulthood. Podocyte-specific deletion of Srgap1 by hNPHS2Cre resulted in increased susceptibility to doxorubicin-induced nephropathy. Detailed analysis demonstrated significant effacement of podocyte foot processes. Furthermore, SRGAP1-knockout podocytes showed excessive protrusion formation and disinhibition of the small Rho GTPase machinery in vitro. Evaluation of a SRGAP1-dependent interactome revealed the involvement of SRGAP1 with protrusive and contractile actin networks. Analysis of glomerular biopsy specimens translated these findings toward human disease by displaying a pronounced redistribution of SRGAP1 in FSGS. CONCLUSIONS: SRGAP1, a podocyte-specific RhoGAP, controls podocyte foot-process architecture by limiting the activity of protrusive, branched actin networks. Therefore, elucidating the complex regulatory small Rho GTPase affinity network points to novel targets for potentially precise intervention in glomerular diseases.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Podocitos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actomiosina/metabolismo , Animales , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Síndrome Nefrótico/etiología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Podocitos/ultraestructura , Mapeo de Interacción de Proteínas , Proteoma , Seudópodos/metabolismo , Seudópodos/ultraestructura , Transcriptoma
8.
J Struct Biol ; 213(4): 107808, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34742832

RESUMEN

A precise quantitative description of the ultrastructural characteristics underlying biological mechanisms is often key to their understanding. This is particularly true for dynamic extra- and intracellular filamentous assemblies, playing a role in cell motility, cell integrity, cytokinesis, tissue formation and maintenance. For example, genetic manipulation or modulation of actin regulatory proteins frequently manifests in changes of the morphology, dynamics, and ultrastructural architecture of actin filament-rich cell peripheral structures, such as lamellipodia or filopodia. However, the observed ultrastructural effects often remain subtle and require sufficiently large datasets for appropriate quantitative analysis. The acquisition of such large datasets has been enabled by recent advances in high-throughput cryo-electron tomography (cryo-ET) methods. This also necessitates the development of complementary approaches to maximize the extraction of relevant biological information. We have developed a computational toolbox for the semi-automatic quantification of segmented and vectorized filamentous networks from pre-processed cryo-electron tomograms, facilitating the analysis and cross-comparison of multiple experimental conditions. GUI-based components simplify the processing of data and allow users to obtain a large number of ultrastructural parameters describing filamentous assemblies. We demonstrate the feasibility of this workflow by analyzing cryo-ET data of untreated and chemically perturbed branched actin filament networks and that of parallel actin filament arrays. In principle, the computational toolbox presented here is applicable for data analysis comprising any type of filaments in regular (i.e. parallel) or random arrangement. We show that it can ease the identification of key differences between experimental groups and facilitate the in-depth analysis of ultrastructural data in a time-efficient manner.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Biología Computacional/métodos , Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Citoesqueleto de Actina/metabolismo , Animales , Línea Celular Tumoral , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Aprendizaje Profundo , Ratones , Seudópodos/metabolismo , Seudópodos/ultraestructura , Reproducibilidad de los Resultados
9.
Development ; 145(3)2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29386244

RESUMEN

The three-dimensional architecture of the pancreatic islet is integral to beta cell function, but the process of islet formation remains poorly understood due to the difficulties of imaging internal organs with cellular resolution. Within transparent zebrafish larvae, the developing pancreas is relatively superficial and thus amenable to live imaging approaches. We performed in vivo time-lapse and longitudinal imaging studies to follow islet development, visualizing both naturally occurring islet cells and cells arising with an accelerated timecourse following an induction approach. These studies revealed previously unappreciated fine dynamic protrusions projecting between neighboring and distant endocrine cells. Using pharmacological compound and toxin interference approaches, and single-cell analysis of morphology and cell dynamics, we determined that endocrine cell motility is regulated by phosphoinositide 3-kinase (PI3K) and G-protein-coupled receptor (GPCR) signaling. Linking cell dynamics to islet formation, perturbation of protrusion formation disrupted endocrine cell coalescence, and correlated with decreased islet cell differentiation. These studies identified novel cell behaviors contributing to islet morphogenesis, and suggest a model in which dynamic exploratory filopodia establish cell-cell contacts that subsequently promote cell clustering.


Asunto(s)
Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Animales Modificados Genéticamente , Agregación Celular , Movimiento Celular , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Islotes Pancreáticos/citología , Queratina-18/genética , Queratina-18/metabolismo , Organogénesis , Inhibidores de las Quinasa Fosfoinosítidos-3 , Seudópodos/metabolismo , Seudópodos/ultraestructura , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
10.
J Immunol ; 202(1): 239-248, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30510066

RESUMEN

The movement of neutrophils between blood and tissues appears to be regulated by chemoattractants and chemorepellents. Compared with neutrophil chemoattractants, relatively little is known about neutrophil chemorepellents. Slit proteins are endogenously cleaved into a variety of N- and C-terminal fragments, and these fragments are neuronal chemorepellents and inhibit chemoattraction of many cell types, including neutrophils. In this report, we show that the ∼140-kDa N-terminal Slit2 fragment (Slit2-N) is a chemoattractant and the ∼110-kDa N-terminal Slit2 fragment (Slit2-S) is a chemorepellent for human neutrophils. The effects of both Slit2 fragments were blocked by Abs to the Slit2 receptor Roundabout homolog 1 or the Slit2 coreceptor Syndecan-4. Slit2-N did not appear to activate Ras but increased phosphatidylinositol 3,4,5-triphosphate levels. Slit2-N-induced chemoattraction was unaffected by Ras inhibitors, reversed by PI3K inhibitors, and blocked by Cdc42 and Rac inhibitors. In contrast, Slit2-S activated Ras but did not increase phosphatidylinositol 3,4,5-triphosphate levels. Slit2-S-induced chemorepulsion was blocked by Ras and Rac inhibitors, not affected by PI3K inhibitors, and reversed by Cdc42 inhibitors. Slit2-N, but not Slit2-S, increased neutrophil adhesion, myosin L chain 2 phosphorylation, and polarized actin formation and single pseudopods at the leading edge of cells. Slit2-S induced multiple pseudopods. These data suggest that Slit2 isoforms use similar receptors but different intracellular signaling pathways and have different effects on the cytoskeleton and pseudopods to induce neutrophil chemoattraction or chemorepulsion.


Asunto(s)
Citoesqueleto de Actina/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neutrófilos/fisiología , Isoformas de Proteínas/metabolismo , Seudópodos/metabolismo , Anticuerpos Bloqueadores/metabolismo , Orientación del Axón , Adhesión Celular , Células Cultivadas , Quimiotaxis/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Seudópodos/ultraestructura , Transducción de Señal , Sindecano-4/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas ras/metabolismo
11.
Nano Lett ; 20(4): 2230-2245, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32142297

RESUMEN

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


Asunto(s)
Microscopía de Fuerza Atómica/métodos , Fenómenos Biomecánicos , Línea Celular Tumoral , Adhesiones Focales/ultraestructura , Humanos , Microscopía de Fuerza Atómica/instrumentación , Microscopía Confocal/instrumentación , Microscopía Confocal/métodos , Imagen Óptica/instrumentación , Imagen Óptica/métodos , Seudópodos/ultraestructura
12.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32393510

RESUMEN

Entamoeba histolytica and its reptilian counterpart and encystation model Entamoeba invadens formed a polarized monopodial morphology when treated with pentoxifylline. This morphology was propelled by retrograde flow of the cell surface resulting from a cyclic sol-gel conversion of cytoplasm and a stable bleb at the leading edge. Pentoxifylline treatment switched the unpolarized, adherent trophozoites to the nonadherent, stable bleb-driven form and altered the motility pattern from slow and random to fast, directionally persistent, and highly chemotactic. Interestingly, exogenously added adenosine produced multiple protrusions and random motility, an opposite phenotype to that of pentoxifylline. Thus, pentoxifylline, an adenosine antagonist, may be inducing the monopodial morphology by preventing lateral protrusions and restricting the leading edge to one site. The polarized form of E. invadens was aggregation competent, and time-lapse microscopy of encystation revealed its appearance during early hours, mediating the cell aggregation by directional cell migration. The addition of purine nucleotides to in vitro encystation culture prevented the formation of polarized morphology and inhibited the cell aggregation and, thus, the encystation, which further showed the importance of the polarized form in the Entamoeba life cycle. Cell polarity and motility are essential in the pathogenesis of Entamoeba parasites, and the stable bleb-driven polarized morphology of Entamoeba may also be important in invasive amoebiasis.


Asunto(s)
Adenosina/farmacología , Entamoeba histolytica/efectos de los fármacos , Entamoeba/efectos de los fármacos , Estadios del Ciclo de Vida/efectos de los fármacos , Pentoxifilina/farmacología , Seudópodos/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Entamoeba/fisiología , Entamoeba/ultraestructura , Entamoeba histolytica/fisiología , Entamoeba histolytica/ultraestructura , Depuradores de Radicales Libres/farmacología , Estadios del Ciclo de Vida/fisiología , Movimiento/efectos de los fármacos , Movimiento/fisiología , Pentoxifilina/antagonistas & inhibidores , Transición de Fase , Seudópodos/fisiología , Seudópodos/ultraestructura , Imagen de Lapso de Tiempo
13.
Genes Cells ; 24(3): 202-213, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30664308

RESUMEN

Fascin, an actin-bundling protein, is present in the filopodia and lamellipodia of growth cones. However, few studies have examined lamellipodial fascin because it is difficult to observe. In this study, we evaluated lamellipodial fascin. We visualized the actin meshwork of lamellipodia in live growth cones by super-resolution microscopy. Fascin was colocalized with the actin meshwork in lamellipodia. Ser39 of fascin is a well-known phosphorylation site that controls the binding of fascin to actin filaments. Fluorescence recovery after photobleaching experiments with confocal microscopy showed that binding of fascin was controlled by phosphorylation of Ser39 in lamellipodia. Moreover, TPA, an agonist of protein kinase C, induced phosphorylation of fascin and dissociation from actin filaments in lamellipodia. Time series images showed that dissociation of fascin from the actin meshwork was induced by TPA. As fascin dissociated from actin filaments, the orientation of the actin filaments became parallel to the leading edge. The angle of actin filaments against the leading edge was changed from 73° to 15°. This decreased the elasticity of the lamellipodia by 40%, as measured by atomic force microscopy. These data suggest that actin bundles made by fascin contribute to elasticity of the growth cone.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Microfilamentos/metabolismo , Seudópodos/metabolismo , Citoesqueleto de Actina/ultraestructura , Animales , Proteínas Portadoras/química , Línea Celular , Elasticidad , Recuperación de Fluorescencia tras Fotoblanqueo , Ratones , Proteínas de Microfilamentos/química , Fosforilación , Seudópodos/ultraestructura
14.
J Microsc ; 278(2): 59-75, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32141623

RESUMEN

In fluorescence microscopy imaging, the segmentation of adjacent cell membranes within cell aggregates, multicellular samples, tissue, organs, or whole organisms remains a challenging task. The lipid bilayer is a very thin membrane when compared to the wavelength of photons in the visual spectra. Fluorescent molecules or proteins used for labelling membranes provide a limited signal intensity, and light scattering in combination with sample dynamics during in vivo imaging lead to poor or ambivalent signal patterns that hinder precise localisation of the membrane sheets. In the proximity of cells, membranes approach and distance each other. Here, the presence of membrane protrusions such as blebs; filopodia and lamellipodia; microvilli; or membrane vesicle trafficking, lead to a plurality of signal patterns, and the accurate localisation of two adjacent membranes becomes difficult. Several computational methods for membrane segmentation have been introduced. However, few of them specifically consider the accurate detection of adjacent membranes. In this article we present ALPACA (ALgorithm for Piecewise Adjacent Contour Adjustment), a novel method based on 2D piecewise parametric active contours that allows: (i) a definition of proximity for adjacent contours, (ii) a precise detection of adjacent, nonadjacent, and overlapping contour sections, (iii) the definition of a polyline for an optimised shared contour within adjacent sections and (iv) a solution for connecting adjacent and nonadjacent sections under the constraint of preserving the inherent cell morphology. We show that ALPACA leads to a precise quantification of adjacent and nonadjacent membrane zones in regular hexagons and live image sequences of cells of the parapineal organ during zebrafish embryo development. The algorithm detects and corrects adjacent, nonadjacent, and overlapping contour sections within a selected adjacency distance d, calculates shared contour sections for neighbouring cells with minimum alterations of the contour characteristics, and presents piecewise active contour solutions, preserving the contour shape and the overall cell morphology. ALPACA quantifies adjacent contours and can improve the meshing of 3D surfaces, the determination of forces, or tracking of contours in combination with previously published algorithms. We discuss pitfalls, strengths, and limits of our approach, and present a guideline to take the best decision for varying experimental conditions for in vivo microscopy.


Asunto(s)
Membrana Celular/ultraestructura , Extensiones de la Superficie Celular/ultraestructura , Procesamiento de Imagen Asistido por Computador/métodos , Imagenología Tridimensional/métodos , Microscopía Fluorescente/métodos , Algoritmos , Animales , Animales Modificados Genéticamente , Vesículas Citoplasmáticas/ultraestructura , Embrión no Mamífero , Humanos , Microvellosidades/ultraestructura , Seudópodos/ultraestructura , Pez Cebra/embriología
15.
PLoS Comput Biol ; 15(4): e1006684, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30958816

RESUMEN

The mechanical properties of the extracellular matrix (ECM)-a complex, 3D, fibrillar scaffold of cells in physiological environments-modulate cell behavior and can drive tissue morphogenesis, regeneration, and disease progression. For simplicity, it is often convenient to assume these properties to be time-invariant. In living systems, however, cells dynamically remodel the ECM and create time-dependent local microenvironments. Here, we show how cell-generated contractile forces produce substantial irreversible changes to the density and architecture of physiologically relevant ECMs-collagen I and fibrin-in a matter of minutes. We measure the 3D deformation profiles of the ECM surrounding cancer and endothelial cells during stages when force generation is active or inactive. We further correlate these ECM measurements to both discrete fiber simulations that incorporate fiber crosslink unbinding kinetics and continuum-scale simulations that account for viscoplastic and damage features. Our findings further confirm that plasticity, as a mechanical law to capture remodeling in these networks, is fundamentally tied to material damage via force-driven unbinding of fiber crosslinks. These results characterize in a multiscale manner the dynamic nature of the mechanical environment of physiologically mimicking cell-in-gel systems.


Asunto(s)
Matriz Extracelular/fisiología , Seudópodos/fisiología , Fenómenos Biomecánicos , Biopolímeros/química , Biopolímeros/fisiología , Línea Celular , Microambiente Celular/fisiología , Biología Computacional , Simulación por Computador , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Células Endoteliales de la Vena Umbilical Humana , Humanos , Imagenología Tridimensional , Cinética , Modelos Biológicos , Seudópodos/química , Seudópodos/ultraestructura
16.
Dev Dyn ; 248(10): 997-1008, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31390119

RESUMEN

BACKGROUND: During zebrafish epiboly, the embryonic cell mass, or blastoderm, spreads to enclose the yolk cell. The blastoderm consists of an outer epithelial sheet, the enveloping layer (EVL), and the underlying deep cell layer (DEL). Studies have provided insights into the mechanisms of EVL and deep cell epiboly, but little is known about the interactions between the two cell layers and what role they may play during epiboly. RESULTS: We used live imaging to examine EVL basal protrusions. We identified them as filopodia based on f-actin content and localization of fluorescently tagged filopodial markers. A spatiotemporal analysis revealed that the largest number of EVL filopodia were present during early epiboly at the animal pole. In functional studies, expression of a constitutively active actin-bundling protein resulted in increased filopodial length and delayed gastrulation. CONCLUSIONS: We identified protrusions on the basal surface of EVL cells as filopodia and showed that they are present throughout the EVL during epiboly. The largest number of filopodia was at the animal pole during early epiboly, which is when and where deep cell radial intercalations occur to the greatest extent. These findings suggest that EVL filopodia may function during epiboly to promote deep cell rearrangements during epiboly initiation.


Asunto(s)
Epitelio/ultraestructura , Seudópodos/ultraestructura , Análisis Espacio-Temporal , Actinas/metabolismo , Animales , Blastodermo/citología , Embrión no Mamífero , Gastrulación , Pez Cebra
17.
Biophys J ; 116(1): 142-150, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30558885

RESUMEN

Fluorescent markers that bind endogenous target proteins are frequently employed for quantitative live-cell imaging. To visualize the actin cytoskeleton in live cells, several actin-binding probes have been widely used. Among them, Lifeact is the most popular probe with ideal properties, including fast exchangeable binding kinetics. Because of its fast kinetics, Lifeact is generally believed to distribute evenly throughout cellular actin structures. In this study, however, we demonstrate misdistribution of Lifeact toward the rear of lamellipodia where actin filaments continuously move inward along the retrograde flow. Similarly, phalloidin showed biased misdistribution toward the rear of lamellipodia in live cells. We show evidence of convection-induced misdistribution of actin probes by both experimental data and physical models. Our findings warn about the potential error arising from the use of target-binding probes in quantitative live imaging.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/metabolismo , Convección , Colorantes Fluorescentes/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Células Cultivadas , Carpa Dorada , Microscopía Fluorescente/métodos , Unión Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , Xenopus laevis
18.
J Biol Chem ; 293(30): 11674-11686, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29880641

RESUMEN

The evolution of cell-adhesion mechanisms in animals facilitated the assembly of organized multicellular tissues. Studies in traditional animal models have revealed two predominant adhesion structures, the adherens junction (AJ) and focal adhesions (FAs), which are involved in the attachment of neighboring cells to each other and to the secreted extracellular matrix (ECM), respectively. The AJ (containing cadherins and catenins) and FAs (comprising integrins, talin, and paxillin) differ in protein composition, but both junctions contain the actin-binding protein vinculin. The near ubiquity of these structures in animals suggests that AJ and FAs evolved early, possibly coincident with multicellularity. However, a challenge to this perspective is that previous studies of sponges-a divergent animal lineage-indicate that their tissues are organized primarily by an alternative, sponge-specific cell-adhesion mechanism called "aggregation factor." In this study, we examined the structure, biochemical properties, and tissue localization of a vinculin ortholog in the sponge Oscarella pearsei (Op). Our results indicate that Op vinculin localizes to both cell-cell and cell-ECM contacts and has biochemical and structural properties similar to those of vertebrate vinculin. We propose that Op vinculin played a role in cell adhesion and tissue organization in the last common ancestor of sponges and other animals. These findings provide compelling evidence that sponge tissues are indeed organized like epithelia in other animals and support the notion that AJ- and FA-like structures extend to the earliest periods of animal evolution.


Asunto(s)
Poríferos/citología , Vinculina/metabolismo , Actinas/análisis , Actinas/metabolismo , Animales , Adhesión Celular , Adhesiones Focales/metabolismo , Modelos Moleculares , Poríferos/metabolismo , Poríferos/ultraestructura , Unión Proteica , Conformación Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , Talina/análisis , Talina/metabolismo , Vinculina/análisis
19.
J Cell Sci ; 130(20): 3427-3435, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-29032357

RESUMEN

The actin cytoskeleton and associated motor proteins provide the driving forces for establishing the astonishing morphological diversity and dynamics of mammalian cells. Aside from functions in protruding and contracting cell membranes for motility, differentiation or cell division, the actin cytoskeleton provides forces to shape and move intracellular membranes of organelles and vesicles. To establish the many different actin assembly functions required in time and space, actin nucleators are targeted to specific subcellular compartments, thereby restricting the generation of specific actin filament structures to those sites. Recent research has revealed that targeting and activation of actin filament nucleators, elongators and myosin motors are tightly coordinated by conserved protein complexes to orchestrate force generation. In this Cell Science at a Glance article and the accompanying poster, we summarize and discuss the current knowledge on the corresponding protein complexes and their modes of action in actin nucleation, elongation and force generation.


Asunto(s)
Citoesqueleto de Actina/fisiología , Seudópodos/fisiología , Citoesqueleto de Actina/ultraestructura , Actinas/fisiología , Actinas/ultraestructura , Animales , Fenómenos Fisiológicos Celulares , Células Cultivadas , Humanos , Multimerización de Proteína , Seudópodos/ultraestructura
20.
J Cell Sci ; 130(11): 1965-1978, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28446539

RESUMEN

Cancer cell invasion is influenced by various biomechanical forces found within the microenvironment. We have previously found that invasion is enhanced in fibrosarcoma cells when transient mechanical stimulation is applied within an in vitro mechano-invasion assay. This enhancement of invasion is dependent on cofilin (CFL1), a known regulator of invadopodia maturation. Invadopodia are actin-rich structures present in invasive cancer cells that are enzymatically active and degrade the surrounding extracellular matrix to facilitate invasion. In this study, we examine changes in gene expression in response to tugging on matrix fibers. Interestingly, we find that integrin ß3 expression is downregulated and leads to an increase in cofilin activity, as evidenced by a reduction in its Ser3 phosphorylation levels. As a result, invadopodia lengthen and have increased enzymatic activity, indicating that transient mechanical stimulation promotes the maturation of invadopodia leading to increased levels of cell invasion. Our results are unique in defining an invasive mechanism specific to the invasive process of cancer cells that is triggered by tugging forces in the microenvironment, as opposed to rigidity, compression or stretch forces.


Asunto(s)
Cofilina 1/genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Integrina beta3/genética , Mecanotransducción Celular , Actinas/genética , Actinas/metabolismo , Fenómenos Biomecánicos , Línea Celular Tumoral , Movimiento Celular , Cofilina 1/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Fibroblastos/ultraestructura , Humanos , Integrina beta3/metabolismo , Invasividad Neoplásica , Fosforilación , Proteolisis , Seudópodos/metabolismo , Seudópodos/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA