Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38801098

RESUMEN

Focal adhesions serve as structural and signaling hubs, facilitating bidirectional communication at the cell-extracellular matrix interface. Paxillin and the related Hic-5 (TGFß1i1) are adaptor/scaffold proteins that recruit numerous structural and regulatory proteins to focal adhesions, where they perform both overlapping and discrete functions. In this study, paxillin and Hic-5 were expressed in U2OS osteosarcoma cells as biotin ligase (BioID2) fusion proteins and used as bait proteins for proximity-dependent biotinylation in order to directly compare their respective interactomes. The fusion proteins localized to both focal adhesions and the centrosome, resulting in biotinylation of components of each of these structures. Biotinylated proteins were purified and analyzed by mass spectrometry. The list of proximity interactors for paxillin and Hic-5 comprised numerous shared core focal adhesion proteins that likely contribute to their similar functions in cell adhesion and migration, as well as proteins unique to paxillin and Hic-5 that have been previously localized to focal adhesions, the centrosome, or the nucleus. Western blotting confirmed biotinylation and enrichment of FAK and vinculin, known interactors of Hic-5 and paxillin, as well as several potentially unique proximity interactors of Hic-5 and paxillin, including septin 7 and ponsin, respectively. Further investigation into the functional relationship between the unique interactors and Hic-5 or paxillin may yield novel insights into their distinct roles in cell migration.

2.
Exp Cell Res ; 435(2): 113930, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38237846

RESUMEN

The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/patología , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral/genética
3.
Mol Biol Cell ; 34(7): ar65, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37043310

RESUMEN

Rab GTPase-mediated vesicle trafficking of cell surface proteins, including integrins, through endocytic and recycling pathways is important in controlling cell-extracellular matrix interactions during cell migration. The focal adhesion adaptor protein, paxillin, plays a central role in regulating adhesion dynamics and was previously shown to promote anterograde vesicle trafficking through modulation of microtubule acetylation via its inhibition of the deacetylase HDAC6. The role of paxillin in retrograde trafficking is unknown. Herein, we identified a role for paxillin in the modulation of the Rab5 GTPase, which is necessary for regulating early endosome dynamics and focal adhesion turnover. Using MDA-MB-231 breast cancer cells and paxillin (-/-) fibroblasts, paxillin was shown to impact Rab5-associated vesicle size and distribution, as well as Rab5 GTPase activity, through its modulation of HDAC6. Using a combination of real-time imaging and particle tracking analysis, paxillin was shown to promote Rab5-associated vesicle motility through inhibition of HDAC6-mediated micro-tubule deacetylation, along with the localization of active integrin to focal adhesions.


Asunto(s)
Adhesiones Focales , Procesamiento Proteico-Postraduccional , Humanos , Paxillin/metabolismo , Acetilación , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Microtúbulos/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Adhesión Celular/fisiología
4.
Mol Biol Cell ; 33(2): ar14, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34851720

RESUMEN

Distant organ metastasis is linked to poor prognosis during cancer progression. The expression level of the focal adhesion adapter protein paxillin varies among different human cancers, but its role in tumor progression is unclear. Herein we utilize a newly generated PyMT mammary tumor mouse model with conditional paxillin ablation in breast tumor epithelial cells, combined with in vitro three-dimensional (3D) tumor organoids invasion analysis and 2D calcium switch assays, to assess the roles for paxillin in breast tumor cell invasion. Paxillin had little effect on primary tumor initiation and growth but is critical for the formation of distant lung metastasis. In paxillin-depleted 3D tumor organoids, collective cell invasion was substantially perturbed. The 2D cell culture revealed paxillin-dependent stabilization of adherens junctions (AJ). Mechanistically, paxillin is required for AJ assembly through facilitating E-cadherin endocytosis and recycling and HDAC6-mediated microtubule acetylation. Furthermore, Rho GTPase activity analysis and rescue experiments with a RhoA activator or Rac1 inhibitor suggest paxillin is potentially regulating the E-cadherin-dependent junction integrity and contractility through control of the balance of RhoA and Rac1 activities. Together, these data highlight new roles for paxillin in the regulation of cell-cell adhesion and collective tumor cell migration to promote the formation of distance organ metastases.


Asunto(s)
Uniones Adherentes/fisiología , Invasividad Neoplásica/genética , Paxillin/metabolismo , Uniones Adherentes/genética , Animales , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/metabolismo , Femenino , Adhesiones Focales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Paxillin/genética , Proteínas de Unión al GTP rho/metabolismo
5.
Int Rev Cell Mol Biol ; 355: 1-52, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32859368

RESUMEN

The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.


Asunto(s)
Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Invasividad Neoplásica , Neoplasias/metabolismo , Paxillin/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Neoplasias/patología , Paxillin/fisiología , Transducción de Señal
6.
Mol Biol Cell ; 30(25): 3037-3056, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31644368

RESUMEN

Focal adhesion (FA)-stimulated reorganization of the F-actin cytoskeleton regulates cellular size, shape, and mechanical properties. However, FA cross-talk with the intermediate filament cytoskeleton is poorly understood. Genetic ablation of the FA-associated scaffold protein Hic-5 in mouse cancer-associated fibroblasts (CAFs) promoted a dramatic collapse of the vimentin network, which was rescued following EGFP-Hic-5 expression. Vimentin collapse correlated with a loss of detergent-soluble vimentin filament precursors and decreased vimentin S72/S82 phosphorylation. Additionally, fluorescence recovery after photobleaching analysis indicated impaired vimentin dynamics. Microtubule (MT)-associated EB1 tracking and Western blotting of MT posttranslational modifications indicated no change in MT dynamics that could explain the vimentin collapse. However, pharmacological inhibition of the RhoGTPase Cdc42 in Hic-5 knockout CAFs rescued the vimentin collapse, while pan-formin inhibition with SMIFH2 promoted vimentin collapse in Hic-5 heterozygous CAFs. Our results reveal novel regulation of vimentin organization/dynamics by the FA scaffold protein Hic-5 via modulation of RhoGTPases and downstream formin activity.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Adhesiones Focales/metabolismo , Proteínas con Dominio LIM/metabolismo , Vimentina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/genética , Células Cultivadas , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Fosforilación , Factores de Transcripción/metabolismo
7.
Development ; 146(9)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30967426

RESUMEN

Establishing apical-basal epithelial cell polarity is fundamental for mammary gland duct morphogenesis during mammalian development. While the focal adhesion adapter protein paxillin is a well-characterized regulator of mesenchymal cell adhesion signaling, F-actin cytoskeleton remodeling and single cell migration, its role in epithelial tissue organization and mammary gland morphogenesis in vivo has not been investigated. Here, using a newly developed paxillin conditional knockout mouse model with targeted ablation in the mammary epithelium, in combination with ex vivo three-dimensional organoid and acini cultures, we identify new roles for paxillin in the establishment of apical-basal epithelial cell polarity and lumen formation, as well as mammary gland duct diameter and branching. Paxillin is shown to be required for the integrity and apical positioning of the Golgi network, Par complex and the Rab11/MyoVb trafficking machinery. Paxillin depletion also resulted in reduced levels of apical acetylated microtubules, and rescue experiments with the HDAC6 inhibitor tubacin highlight the central role for paxillin-dependent regulation of HDAC6 activity and associated microtubule acetylation in controlling epithelial cell apical-basal polarity and tissue branching morphogenesis.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Paxillin/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Polaridad Celular/genética , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Ratones , Microtúbulos/metabolismo , Morfogénesis/genética , Morfogénesis/fisiología , Paxillin/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
8.
Mol Biol Cell ; 30(11): 1298-1313, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30893012

RESUMEN

Fibroblasts transformed by the proto-oncogene Src form individual invadopodia that can spontaneously self-organize into large matrix-degrading superstructures called rosettes. However, the mechanisms by which the invadopodia can spatiotemporally reorganize their architecture is not well understood. Here, we show that Hic-5, a close relative of the scaffold protein paxillin, is essential for the formation and organization of rosettes in active Src-transfected NIH3T3 fibroblasts and cancer-associated fibroblasts. Live cell imaging, combined with domain-mapping analysis of Hic-5, identified critical motifs as well as phosphorylation sites that are required for the formation and dynamics of rosettes. Using pharmacological inhibition and mutant expression, we show that FAK kinase activity, along with its proximity to and potential interaction with the LD2,3 motifs of Hic-5, is necessary for rosette formation. Invadopodia dynamics and their coalescence into rosettes were also dependent on Rac1, formin, and myosin II activity. Superresolution microscopy revealed the presence of formin FHOD1 and INF2-mediated unbranched radial F-actin fibers emanating from invadopodia and rosettes, which may facilitate rosette formation. Collectively, our data highlight a novel role for Hic-5 in orchestrating the organization of invadopodia into higher-order rosettes, which may promote the localized matrix degradation necessary for tumor cell invasion.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas con Dominio LIM/metabolismo , Podosomas/metabolismo , Procesamiento Proteico-Postraduccional , Familia-src Quinasas/genética , Actinas/metabolismo , Actinas/fisiología , Animales , Línea Celular Transformada , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Fetales/metabolismo , Proteínas Fetales/fisiología , Fibroblastos/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Forminas/metabolismo , Forminas/fisiología , Proteínas con Dominio LIM/fisiología , Ratones , Miosina Tipo II/metabolismo , Miosina Tipo II/fisiología , Células 3T3 NIH , Neuropéptidos/metabolismo , Neuropéptidos/fisiología , Fosforilación , Podosomas/fisiología , Formación de Roseta , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/fisiología
9.
PLoS One ; 14(2): e0211408, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30759123

RESUMEN

Cell motility is critical to biological processes from wound healing to cancer metastasis to embryonic development. The involvement of organelles in cell motility is well established, but the role of organelle positional reorganization in cell motility remains poorly understood. Here we present an automated image analysis technique for tracking the shape and motion of Golgi bodies and cell nuclei. We quantify the relationship between nuclear orientation and the orientation of the Golgi body relative to the nucleus before, during, and after exposure of mouse fibroblasts to a controlled change in cell substrate topography, from flat to wrinkles, designed to trigger polarized motility. We find that the cells alter their mean nuclei orientation, in terms of the nuclear major axis, to increasingly align with the wrinkle direction once the wrinkles form on the substrate surface. This change in alignment occurs within 8 hours of completion of the topographical transition. In contrast, the position of the Golgi body relative to the nucleus remains aligned with the pre-programmed wrinkle direction, regardless of whether it has been fully established. These findings indicate that intracellular positioning of the Golgi body precedes nuclear reorientation during mouse fibroblast directed migration on patterned substrates. We further show that both processes are Rho-associated kinase (ROCK) mediated as they are abolished by pharmacologic ROCK inhibition whereas mouse fibroblast motility is unaffected. The automated image analysis technique introduced could be broadly employed in the study of polarization and other cellular processes in diverse cell types and micro-environments. In addition, having found that the nuclei Golgi vector may be a more sensitive indicator of substrate features than the nuclei orientation, we anticipate the nuclei Golgi vector to be a useful metric for researchers studying the dynamics of cell polarity in response to different micro-environments.


Asunto(s)
Movimiento Celular , Núcleo Celular/ultraestructura , Aparato de Golgi/ultraestructura , Imagen de Lapso de Tiempo/métodos , Animales , Polaridad Celular , Células Cultivadas , Fibroblastos , Ratones
10.
Mol Biol Cell ; 29(13): 1704-1717, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29771639

RESUMEN

The focal adhesion proteins Hic-5 and paxillin have been previously identified as key regulators of MDA-MB-231 breast cancer cell migration and morphologic mesenchymal-amoeboid plasticity in three-dimensional (3D) extracellular matrices (ECMs). However, their respective roles in other cancer cell types have not been evaluated. Herein, utilizing 3D cell-derived matrices and fibronectin-coated one-dimensional substrates, we show that across a variety of cancer cell lines, the level of Hic-5 expression serves as the major indicator of the cells primary morphology, plasticity, and in vitro invasiveness. Domain mapping studies reveal sites critical to the functions of both Hic-5 and paxillin in regulating phenotype, while ectopic expression of Hic-5 in cell lines with low endogenous levels of the protein is sufficient to induce a Rac1-dependent mesenchymal phenotype and, in turn, increase amoeboid-mesenchymal plasticity and invasion. We show that the activity of vinculin, when coupled to the expression of Hic-5 is required for the mesenchymal morphology in the 3D ECM. Taken together, our results identify Hic-5 as a critical modulator of tumor cell phenotype that could be utilized in predicting tumor cell migratory and invasive behavior in vivo.


Asunto(s)
Movimiento Celular , Plasticidad de la Célula , Forma de la Célula , Matriz Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Neoplasias/patología , Secuencias de Aminoácidos , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas con Dominio LIM/química , Mesodermo/patología , Invasividad Neoplásica , Paxillin/metabolismo , Fenotipo , Fosforilación , Unión Proteica , Dominios Proteicos , Vinculina/metabolismo
11.
Oncogene ; 37(13): 1699-1713, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29348458

RESUMEN

The linearization of the stromal extracellular matrix (ECM) by cancer-associated fibroblasts (CAFs) facilitates tumor cell growth and metastasis. However, the mechanism by which the ECM is remodeled is not fully understood. Hic-5 (TGFß1i1), a focal adhesion scaffold protein, has previously been reported to be crucial for stromal ECM deposition and remodeling in vivo. Herein we show that CAFs lacking Hic-5 exhibit a significant reduction in the ability to form fibrillar adhesions, a specialized form of focal adhesion that promote fibronectin fibrillogenesis. Hic-5 was found to promote fibrillar adhesion formation through a newly characterized interaction with tensin1. Furthermore, Src-dependent phosphorylation of Hic-5 facilitated the interaction with tensin1 to prevent ß1 integrin internalization and trafficking to the lysosome. The interaction between Hic-5 and tensin1 was mechanosensitive, promoting fibrillar adhesion formation and fibronectin fibrillogenesis in a rigidity-dependent fashion. Importantly, this Src-dependent mechanism was conserved in three-dimensional (3D) ECM environments. Immunohistochemistry of tensin1 showed enrichment in CAFs in vivo, which was abrogated upon deletion of Hic-5. Interestingly, elevated Hic-5 expression correlates with reduced distant metastasis-free survival in patients with basal-like, HER2+ and grade 3 tumors. Thus, we have identified Hic-5 as a crucial regulator of ECM remodeling in CAFs by promoting fibrillar adhesion formation through a novel interaction with tensin1.


Asunto(s)
Adhesión Celular , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Neoplasias/metabolismo , Tensinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Adhesión Celular/genética , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/patología , Adhesiones Focales/genética , Técnicas de Silenciamiento del Gen , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neoplasias/genética , Neoplasias/patología , Unión Proteica
12.
Mol Biol Cell ; 28(26): 3815-3831, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29046398

RESUMEN

Cell polarization and directed migration play pivotal roles in diverse physiological and pathological processes. Herein, we identify new roles for paxillin-mediated HDAC6 inhibition in regulating key aspects of cell polarization in both two-dimensional and one-dimensional matrix environments. Paxillin, by modulating microtubule acetylation through HDAC6 regulation, was shown to control centrosome and Golgi reorientation toward the leading edge, a hallmark of cell polarization to ensure directed trafficking of promigratory factors. Paxillin was also required for pericentrosomal Golgi localization and centrosome cohesion, independent of its localization to, and role in, focal adhesion signaling. In addition, we provide evidence of an accumulation of paxillin at the centrosome that is dependent on focal adhesion kinase (FAK) and identify an important collaboration between paxillin and FAK signaling in the modulation of microtubule acetylation, as well as centrosome and Golgi organization and polarization. Finally, paxillin was also shown to be required for optimal anterograde vesicular trafficking to the plasma membrane.


Asunto(s)
Movimiento Celular/fisiología , Histona Desacetilasa 6/metabolismo , Paxillin/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Polaridad Celular/fisiología , Centrosoma/fisiología , Proteínas del Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Aparato de Golgi , Histona Desacetilasa 6/antagonistas & inhibidores , Humanos , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/fisiología , Transducción de Señal
13.
Development ; 144(21): 4002-4014, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935710

RESUMEN

Paxillin and Hic-5 are homologous focal adhesion adaptor proteins that coordinate cytoskeletal rearrangements in response to integrin signaling, but their role(s) in cortical development are unknown. Here, we find that Hic-5-deficient mice are postnatal viable with normal cortical layering. Mice with a neural-specific deletion of paxillin are also postnatal viable, but show evidence of a cortical neuron migration delay that is evident pre- and perinatally, but is not detected at postnatal day 35 (P35). This phenotype is not modified by Hic-5 deficiency (double knockout). Specific deletion of paxillin in postmitotic neurons using Nex-Cre-mediated recombination as well as in utero electroporation of a Cre-expression construct identified a cell-autonomous requirement for paxillin in migrating neurons. Paxillin-deficient neurons have shorter leading processes that exhibited multiple swellings in comparison with control. Multiphoton imaging revealed that paxillin-deficient neurons migrate ∼30% slower than control neurons. This phenotype is similar to that produced by deletion of focal adhesion kinase (FAK), a signaling partner of paxillin, and suggests that paxillin and FAK function cell-autonomously to control migrating neuron morphology and speed during cortical development.


Asunto(s)
Movimiento Celular , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Adhesiones Focales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Paxillin/metabolismo , Alelos , Animales , Movimiento Celular/genética , Forma de la Célula , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Integrasas/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones Noqueados , Células-Madre Neurales/metabolismo , Especificidad de Órganos , Paxillin/deficiencia , Paxillin/genética
14.
Biomaterials ; 140: 150-161, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28649015

RESUMEN

In vitro biomaterial models have enabled advances in understanding the role of extracellular matrix (ECM) architecture in the control of cell motility and polarity. Most models are, however, static and cannot mimic dynamic aspects of in vivo ECM remodeling and function. To address this limitation, we present an electrospun shape memory polymer scaffold that can change fiber alignment on command under cytocompatible conditions. Cellular response was studied using the human fibrosarcoma cell line HT-1080 and the murine mesenchymal stem cell line C3H/10T1/2. The results demonstrate successful on-command on/off switching of cell polarized motility and alignment. Decrease in fiber alignment causes a change from polarized motility along the direction of fiber alignment to non-polarized motility and from aligned to unaligned morphology, while increase in fiber alignment causes a change from non-polarized to polarized motility along the direction of fiber alignment and from unaligned to aligned morphology. In addition, the findings are consistent with the hypothesis that increased fiber alignment causes increased cell velocity, while decreased fiber alignment causes decreased cell velocity. On-command on/off switching of cell polarized motility and alignment is anticipated to enable new study of directed cell motility in tumor metastasis, in cell homing, and in tissue engineering.


Asunto(s)
Materiales Biocompatibles/química , Movimiento Celular , Polímeros/química , Células Madre/citología , Andamios del Tejido/química , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/ultraestructura , Polaridad Celular , Forma de la Célula , Matriz Extracelular/química , Humanos , Ratones
15.
Oncotarget ; 8(22): 35902-35918, 2017 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-28415794

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy characterized by resistance to currently employed chemotherapeutic approaches. Members of the mir-17~92 cluster of microRNAs (miRNAs) are upregulated in PDAC, but the precise roles of these miRNAs in PDAC are unknown. Using genetically engineered mouse models, we show that loss of mir-17~92 reduces ERK pathway activation downstream of mutant KRAS and promotes the regression of KRASG12D-driven precursor pancreatic intraepithelial neoplasias (PanINs) and their replacement by normal exocrine tissue. In a PDAC model driven by concomitant KRASG12D expression and Trp53 heterozygosity, mir-17~92 deficiency extended the survival of mice that lacked distant metastasis. Moreover, mir-17~92-deficient PDAC cell lines display reduced invasion activity in transwell assays, form fewer invadopodia rosettes than mir-17~92-competent cell lines and are less able to degrade extracellular matrix. Specific inhibition of miR-19 family miRNAs with antagomirs recapitulates these phenotypes, suggesting that miR-19 family miRNAs are important mediators of PDAC cell invasion. Together these data demonstrate an oncogenic role for mir-17~92 at multiple stages of pancreatic tumorigenesis and progression; specifically, they link this miRNA cluster to ERK pathway activation and precursor lesion maintenance in vivo and identify a novel role for miR-19 family miRNAs in promoting cancer cell invasion.


Asunto(s)
Transformación Celular Neoplásica/genética , MicroARNs/genética , Familia de Multigenes , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Edad , Alelos , Animales , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Neoplasias Pancreáticas/metabolismo , Fenotipo
16.
Dev Biol ; 425(1): 70-84, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28315297

RESUMEN

Paxillin (Pxn) is a key adapter protein and signaling regulator at sites of cell-extracellular matrix (ECM) adhesion. Here, we investigated the role of Pxn during vertebrate development using the zebrafish embryo as a model system. We have characterized two Pxn genes, pxna and pxnb, in zebrafish that are maternally supplied and expressed in multiple tissues. Gene editing and antisense gene knockdown approaches were used to uncover Pxn functions during zebrafish development. While mutation of either pxna or pxnb alone did not cause gross embryonic phenotypes, double mutants lacking maternally supplied pxna or pxnb displayed defects in cardiovascular, axial, and skeletal muscle development. Transient knockdown of Pxn proteins resulted in similar defects. Irregular myotome shape and ECM composition were observed, suggesting an "inside-out" signaling role for Paxillin genes in the development of myotendinous junctions. Inhibiting non-muscle Myosin-II during somitogenesis altered the subcellular localization of Pxn protein and phenocopied pxn gene loss-of-function. This indicates that Paxillin genes are effectors of actomyosin contractility-driven morphogenesis of trunk musculature in zebrafish. Together, these results reveal new functions for Pxn during muscle development and provide novel genetic models to elucidate Pxn functions.


Asunto(s)
Actomiosina/metabolismo , Morfogénesis , Músculo Esquelético/metabolismo , Paxillin/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Western Blotting , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Técnicas de Silenciamiento del Gen , Microscopía Confocal , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Mutación , Paxillin/genética , Isoformas de Proteínas/genética , Homología de Secuencia de Ácido Nucleico , Somitos/embriología , Somitos/metabolismo , Imagen de Lapso de Tiempo/métodos , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
17.
PLoS One ; 11(11): e0165266, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27806088

RESUMEN

BACKGROUND: Paxillin family proteins regulate intracellular signaling downstream of extracellular matrix adhesion. Tissue expression patterns and cellular functions of Paxillin proteins during embryo development remain poorly understood. Additionally, the evolution of this gene family has not been thoroughly investigated. RESULTS: This report characterizes the evolution and expression of a novel Paxillin gene, called Paxillin-b, in Teleosts. Alignments indicate that Teleost Paxillin-a and Paxillin-b proteins are highly homologous to each other and to human Paxillin. Phylogenetic and synteny analyses suggest that these genes originated from the duplication of an ancestral Paxillin gene that was in a common ancestor of Teleosts and Tetrapods. Analysis of the spatiotemporal expression profiles of Paxillin-a and Paxillin-b using zebrafish revealed both overlapping and distinct domains for Paxillin-a and Paxillin-b during embryo development. Localization of zebrafish Paxillin orthologs expressed in mammalian cells demonstrated that both proteins localize to focal adhesions, similar to mammalian Paxillin. This suggests these proteins regulate adhesion-dependent processes in their endogenous tissues. CONCLUSION: Paxillin-a and Paxillin-b were generated by duplication in Teleosts. These genes likely play similar roles as Paxillin genes in other organisms. This work provides a framework for functional investigation of Paxillin family members during development using the zebrafish as an in vivo model system.


Asunto(s)
Peces/embriología , Adhesiones Focales/metabolismo , Paxillin/genética , Paxillin/metabolismo , Animales , Evolución Molecular , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Peces/genética , Peces/metabolismo , Duplicación de Gen , Regulación del Desarrollo de la Expresión Génica , Humanos , Filogenia , Sintenía , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
18.
Oncotarget ; 7(29): 46419-46432, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27329840

RESUMEN

Despite advancing therapies, thousands of women die every year of breast cancer. Myosins, actin-dependent molecular motors, are likely to contribute to tumor formation and metastasis via their effects on cell adhesion and migration and may provide promising new targets for cancer therapies. Using the MMTV-PyMT murine model of breast cancer, we identified Myosin 1e (MYO1E) as a novel tumor promoter. Tumor latency in mice lacking MYO1E was significantly increased, and tumors formed in the absence of MYO1E displayed unusual papillary morphology, with well-differentiated layers of epithelial cells covering fibrovascular cores, rather than solid sheets of tumor cells typically observed in this cancer model. These tumors were reminiscent of papillary breast cancer in humans that is typically non-invasive and often cured by tumor excision. MYO1E-null tumors exhibited decreased expression of the markers of cell proliferation, which was recapitulated in primary tumor cells derived from MYO1E-null mice. In agreement with our findings, meta-analysis of patient survival data indicated that MYO1E expression level was associated with reduced recurrence-free survival in basal-like breast cancer. Overall, our data suggests that MYO1E contributes to breast tumor malignancy and regulates the differentiation and proliferation state of breast tumor cells.


Asunto(s)
Neoplasias de la Mama/patología , Desdiferenciación Celular/fisiología , Miosinas/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Proliferación Celular/fisiología , Femenino , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miosina Tipo I , Pronóstico
19.
J Vis Exp ; (102): e52949, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26381826

RESUMEN

The composition and mechanical properties of the extracellular matrix are highly variable between tissue types. This connective tissue stroma diversity greatly impacts cell behavior to regulate normal and pathologic processes including cell proliferation, differentiation, adhesion signaling and directional migration. In this regard, the innate ability of certain cell types to migrate towards a stiffer, or less compliant matrix substrate is referred to as durotaxis. This phenomenon plays an important role during embryonic development, wound repair and cancer cell invasion. Here, we describe a straightforward assay to study durotaxis, in vitro, using polydimethylsiloxane (PDMS) substrates. Preparation of the described durotaxis chambers creates a rigidity interface between the relatively soft PDMS gel and a rigid glass coverslip. In the example provided, we have used these durotaxis chambers to demonstrate a role for the cdc42/Rac1 GTPase activating protein, cdGAP, in mechanosensing and durotaxis regulation in human U2OS osteosarcoma cells. This assay is readily adaptable to other cell types and/or knockdown of other proteins of interests to explore their respective roles in mechanosignaling and durotaxis.


Asunto(s)
Movimiento Celular/fisiología , Técnicas Citológicas/métodos , Dimetilpolisiloxanos/química , Fenómenos Biomecánicos , Neoplasias Óseas/patología , Línea Celular Tumoral , Matriz Extracelular/fisiología , Humanos , Técnicas In Vitro , Osteosarcoma/patología , Transducción de Señal
20.
J Cell Biol ; 206(3): 395-413, 2014 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-25070956

RESUMEN

Polarized cell migration is essential for normal organism development and is also a critical component of cancer cell invasion and disease progression. Directional cell motility requires the coordination of dynamic cell-extracellular matrix interactions as well as repositioning of the Golgi apparatus, both of which can be controlled by the microtubule (MT) cytoskeleton. In this paper, we have identified a new and conserved role for the focal adhesion scaffold protein paxillin in regulating the posttranslational modification of the MT cytoskeleton through an inhibitory interaction with the α-tubulin deacetylase HDAC6. We also determined that through HDAC6-dependent regulation of the MT cytoskeleton, paxillin regulates both Golgi organelle integrity and polarized cell invasion and migration in both three-dimensional and two-dimensional matrix microenvironments. Importantly, these data reveal a fundamental role for paxillin in coordinating MT acetylation-dependent cell polarization and migration in both normal and transformed cells.


Asunto(s)
Movimiento Celular , Matriz Extracelular/metabolismo , Aparato de Golgi/ultraestructura , Histona Desacetilasas/metabolismo , Microtúbulos/metabolismo , Paxillin/fisiología , Procesamiento Proteico-Postraduccional , Acetilación , Animales , Línea Celular Tumoral , Polaridad Celular , Aparato de Golgi/metabolismo , Histona Desacetilasa 6 , Humanos , Ratones , Células 3T3 NIH
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA