Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
FASEB J ; 37(3): e22786, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36786724

RESUMEN

Adherens junctions (AJs) are a defining feature of all epithelial cells. They regulate epithelial tissue architecture and integrity, and their dysregulation is a key step in tumor metastasis. AJ remodeling is crucial for cancer progression, and it plays a key role in tumor cell survival, growth, and dissemination. Few studies have examined AJ remodeling in cancer cells consequently, it remains poorly understood and unleveraged in the treatment of metastatic carcinomas. Fascin1 is an actin-bundling protein that is absent from the normal epithelium but its expression in colon cancer is linked to metastasis and increased mortality. Here, we provide the molecular mechanism of AJ remodeling in colon cancer cells and identify for the first time, fascin1's function in AJ remodeling. We show that in colon cancer cells fascin1 remodels junctional actin and actomyosin contractility which makes AJs less stable but more dynamic. By remodeling AJs fascin1 drives mechanoactivation of WNT/ß-catenin signaling and generates "collective plasticity" which influences the behavior of cells during cell migration. The impact of mechanical inputs on WNT/ß-catenin activation in cancer cells remains poorly understood. Our findings highlight the role of AJ remodeling and mechanosensitive WNT/ß-catenin signaling in the growth and dissemination of colorectal carcinomas.


Asunto(s)
Uniones Adherentes , Neoplasias del Colon , Humanos , Uniones Adherentes/metabolismo , Actinas/metabolismo , beta Catenina/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias del Colon/metabolismo , Cadherinas/metabolismo
2.
Mol Biol Cell ; 31(17): 1857-1866, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32520642

RESUMEN

Villin is a major actin-bundling protein that assembles the brush border of intestinal and renal epithelial cells. The villin "headpiece" domain and the actin-binding residues within it regulate its actin-bundling function. Substantial experimental and theoretical information about the three-dimensional structure of the isolated villin headpiece, including a description of the actin-binding residues within the headpiece, is available. Despite that, the actin-bundling site in the full-length (FL) villin protein remains unidentified. We used this existing villin headpiece nuclear magnetic resonance data and performed mutational analysis and functional assays to identify the actin-bundling site in FL human villin protein. By careful evaluation of these conserved actin-binding residues in human advillin protein, we demonstrate their functional significance in the over 30 proteins that contain a villin-type headpiece domain. Our study is the first that combines the available structural data on villin headpiece with functional assays to identify the actin-binding residues in FL villin that regulate its filament-bundling activity. Our findings could have wider implications for other actin-bundling proteins that contain a villin-type headpiece domain.


Asunto(s)
Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Sitios de Unión/genética , Proteínas Portadoras/metabolismo , Citoesqueleto/metabolismo , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Proteínas de Microfilamentos/fisiología , Unión Proteica/genética , Dominios Proteicos/genética , Estructura Terciaria de Proteína
3.
Sci Rep ; 10(1): 8877, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483224

RESUMEN

Tuft (or brush) cells are solitary chemosensory cells scattered throughout the epithelia of the respiratory and alimentary tract. The actin-binding protein villin (Vil1) is used as a marker of tuft cells and the villin promoter is frequently used to drive expression of the Cre recombinase in tuft cells. While there is widespread agreement about the expression of villin in tuft cells there are several disagreements related to tuft cell lineage commitment and function. We now show that many of these inconsistencies could be resolved by our surprising finding that intestinal tuft cells, in fact, do not express villin protein. Furthermore, we show that a related actin-binding protein, advillin which shares 75% homology with villin, has a tuft cell restricted expression in the gastrointestinal epithelium. Our study identifies advillin as a marker of tuft cells and provides a mechanism for driving gene expression in tuft cells but not in other epithelial cells of the gastrointestinal tract. Our findings fundamentally change the way we identify and study intestinal tuft cells.


Asunto(s)
Mucosa Intestinal/citología , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Animales , Células Cultivadas , Técnicas de Inactivación de Genes , Mucosa Intestinal/metabolismo , Ratones , Especificidad de Órganos , Análisis de Secuencia de ARN
4.
Gastroenterology ; 154(5): 1405-1420.e2, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29274870

RESUMEN

BACKGROUND & AIMS: Cell stress signaling pathways result in phosphorylation of the eukaryotic translation initiation factor 2 subunit alpha (EIF2S1 or EIF2A), which affects regulation of protein translation. Translation reprogramming mitigates stress by activating pathways that result in autophagy and cell death, to eliminate damaged cells. Actin is modified during stress and EIF2A is dephosphorylated to restore homeostasis. It is not clear how actin affects EIF2A signaling. We studied the actin-binding proteins villin 1 (VIL1) and gelsolin (GSN) in intestinal epithelial cells (IECs) to determine whether they respond to cell stress response and affect signaling pathways. METHODS: We performed studies with mice with disruptions in Vil1 and Gsn (double-knockout mice). Wild-type (WT) mice either were or were not (controls) exposed to cell stressors such as tumor necrosis factor and adherent-invasive Escherichia coli. Distal ileum tissues were collected from mice; IECs and enteroids were cultured and analyzed by histology, immunoblots, phalloidin staining, immunohistochemistry, electron microscopy, and flow cytometry. HT-29 cells were incubated with cell stressors such as DTT, IFN, and adherent-invasive E coli or control agents; cells were analyzed by immunoblots and quantitative polymerase chain reaction. Green fluorescent protein and green fluorescent protein tagged mutant EIF2A were expressed from a lentiviral vector. The mouse immunity-related GTPase (IRGM1) was overexpressed in embryonic fibroblasts from dynamin1 like (DNM1L) protein-knockout mice or their WT littermates. IRGM1 was overexpressed in embryonic fibroblasts from receptor interacting serine/threonine kinase 1-knockout mice or their WT littermates. Human IRGM was overexpressed in human epithelial cell lines incubated with the DNM1L-specific inhibitor Mdivi-1. Mitochondria were analyzed by semi-quantitative confocal imaging. We performed immunohistochemical analyses of distal ileum tissues from 6-8 patients with Crohn's disease (CD) and 6-8 individuals without CD (controls). RESULTS: In IECs exposed to cell stressors, EIF2A signaling reduced expression of VIL1 and GSN. However, VIL1 and GSN were required for dephosphorylation of EIF2A and recovery from cell stress. In mouse and human IECs, prolonged, unresolved stress was accompanied by continued down-regulation of VIL1 and GSN, resulting in constitutive phosphorylation of EIF2A and overexpression of IRGM1 (or IRGM), which regulates autophagy. Overexpression of IRGM1 (or IRGM) induced cell death by necroptosis, accompanied by release of damage-associated molecular patterns (DAMPs). In double-knockout mice, constitutive phosphorylation of EIF2A and over-expression of IRGM1 resulted in spontaneous ileitis that resembled human CD in symptoms and histology. Distal ileum tissues from patients with CD had lower levels of VIL1 and GSN, increased phosphorylation of EIF2A, increased levels of IRGM and necroptosis, and increased release of nuclear DAMPs compared with controls. CONCLUSIONS: In studies of intestinal epithelial tissues from patients with CD and embryonic fibroblasts from mice, along with enteroids and human IEC lines, we found that induction of cell stress alters the cytoskeleton in IECs via changes in the actin-binding proteins VIL1 and GSN. Acute changes in actin dynamics increase IEC survival, whereas long-term changes in actin dynamics lead to IEC death and intestinal inflammation. IRGM regulates necroptosis and release of DAMPs to induce gastrointestinal inflammation, linking IRGM activity with CD.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Enfermedad de Crohn/metabolismo , Células Epiteliales/metabolismo , Gelsolina/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Microfilamentos/metabolismo , Transducción de Señal , Estrés Fisiológico , Citoesqueleto de Actina/patología , Alarminas/metabolismo , Animales , Muerte Celular , Supervivencia Celular , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Modelos Animales de Enfermedad , Células Epiteliales/patología , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Gelsolina/deficiencia , Gelsolina/genética , Células HT29 , Células HeLa , Humanos , Íleon/patología , Mucosa Intestinal/patología , Ratones Noqueados , Proteínas de Microfilamentos/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Fosforilación , Interferencia de ARN , Factores de Tiempo , Transfección
5.
J Clin Invest ; 127(12): 4257-4269, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29058690

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of chronic kidney disease. Here, we identified recessive mutations in the gene encoding the actin-binding protein advillin (AVIL) in 3 unrelated families with SRNS. While all AVIL mutations resulted in a marked loss of its actin-bundling ability, truncation of AVIL also disrupted colocalization with F-actin, thereby leading to impaired actin binding and severing. Additionally, AVIL colocalized and interacted with the phospholipase enzyme PLCE1 and with the ARP2/3 actin-modulating complex. Knockdown of AVIL in human podocytes reduced actin stress fibers at the cell periphery, prevented recruitment of PLCE1 to the ARP3-rich lamellipodia, blocked EGF-induced generation of diacylglycerol (DAG) by PLCE1, and attenuated the podocyte migration rate (PMR). These effects were reversed by overexpression of WT AVIL but not by overexpression of any of the 3 patient-derived AVIL mutants. The PMR was increased by overexpression of WT Avil or PLCE1, or by EGF stimulation; however, this increased PMR was ameliorated by inhibition of the ARP2/3 complex, indicating that ARP-dependent lamellipodia formation occurs downstream of AVIL and PLCE1 function. Together, these results delineate a comprehensive pathogenic axis of SRNS that integrates loss of AVIL function with alterations in the action of PLCE1, an established SRNS protein.


Asunto(s)
Proteínas de Microfilamentos , Mutación , Síndrome Nefrótico/congénito , Fosfoinositido Fosfolipasa C , Podocitos , Seudópodos , Complejo 2-3 Proteico Relacionado con la Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Movimiento Celular/genética , Diglicéridos/genética , Diglicéridos/metabolismo , Femenino , Humanos , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Fosfoinositido Fosfolipasa C/genética , Fosfoinositido Fosfolipasa C/metabolismo , Podocitos/metabolismo , Podocitos/patología , Seudópodos/genética , Seudópodos/metabolismo
6.
Sci Rep ; 6: 35491, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27765954

RESUMEN

In the small intestine, epithelial cells are derived from stem cells in the crypts, migrate up the villus as they differentiate and are ultimately shed from the villus tips. This process of proliferation and shedding is tightly regulated to maintain the intestinal architecture and tissue homeostasis. Apoptosis regulates both the number of stem cells in the crypts as well as the sloughing of cells from the villus tips. Previously, we have shown that villin, an epithelial cell-specific actin-binding protein functions as an anti-apoptotic protein in the gastrointestinal epithelium. The expression of villin is highest in the apoptosis-resistant villus cells and lowest in the apoptosis-sensitive crypts. In this study we report that villin is cleaved in the intestinal mucosa to generate a pro-apoptotic fragment that is spatially restricted to the villus tips. This cleaved villin fragment severs actin in an unregulated fashion to initiate the extrusion and subsequent apoptosis of effete cells from the villus tips. Using villin knockout mice, we validate the physiological role of villin in apoptosis and cell extrusion from the gastrointestinal epithelium. Our study also highlights the potential role of villin's pro-apoptotic function in the pathogenesis of inflammatory bowel disease, ischemia-reperfusion injury, enteroinvasive bacterial and parasitic infections.


Asunto(s)
Apoptosis , Homeostasis , Intestinos/citología , Proteínas de Microfilamentos/metabolismo , Animales , Movimiento Celular , Perros , Epitelio/metabolismo , Intestinos/ultraestructura , Células de Riñón Canino Madin Darby , Ratones Noqueados , Modelos Biológicos
7.
Mol Biol Cell ; 27(3): 535-48, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26658611

RESUMEN

Villin is a tissue-specific, actin-binding protein involved in the assembly and maintenance of microvilli in polarized epithelial cells. Conversely, villin is also linked with the loss of epithelial polarity and gain of the mesenchymal phenotype in migrating, invasive cells. In this study, we describe for the first time how villin can switch between these disparate functions to change tissue architecture by moonlighting in the nucleus. Our study reveals that the moonlighting function of villin in the nucleus may play an important role in tissue homeostasis and disease. Villin accumulates in the nucleus during wound repair, and altering the cellular microenvironment by inducing hypoxia increases the nuclear accumulation of villin. Nuclear villin is also associated with mouse models of tumorigenesis, and a systematic analysis of a large cohort of colorectal cancer specimens confirmed the nuclear distribution of villin in a subset of tumors. Our study demonstrates that nuclear villin regulates epithelial-mesenchymal transition (EMT). Altering the nuclear localization of villin affects the expression and activity of Slug, a key transcriptional regulator of EMT. In addition, we find that villin directly interacts with a transcriptional corepressor and ligand of the Slug promoter, ZBRK1. The outcome of this study underscores the role of nuclear villin and its binding partner ZBRK1 in the regulation of EMT and as potential new therapeutic targets to inhibit tumorigenesis.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas de Microfilamentos/fisiología , Proteínas Represoras/metabolismo , Transporte Activo de Núcleo Celular , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Perros , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Ratones SCID , Trasplante de Neoplasias , Señales de Localización Nuclear , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
J Cell Sci ; 126(Pt 1): 312-26, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23132923

RESUMEN

During embryonic development and in metastatic cancers, cells detach from the epithelium and migrate with persistent directionality. Directional cell migration is also crucial for the regeneration and maintenance of the epithelium and impaired directional migration is linked to chronic inflammatory diseases. Despite its significance, the mechanisms controlling epithelial cell migration remain poorly understood. Villin is an epithelial-cell-specific actin modifying protein that regulates epithelial cell plasticity and motility. In motile cells villin is associated with the highly branched and the unbranched actin filaments of lamellipodia and filopodia, respectively. In this study we demonstrate for the first time that villin regulates directionally persistent epithelial cell migration. Functional characterization of wild-type and mutant villin proteins revealed that the ability of villin to self-associate and bundle actin as well as its direct interaction with phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] regulates villin-induced filopodial assembly and directional cell migration. Our findings suggest that convergence of different signaling cascades could spatially restrict villin activity to areas of high PtdIns(4,5)P(2) and F-actin concentration to assemble filopodia. Furthermore, our data reveal the ability of villin to undergo actin- and PtdIns(4,5)P(2)-induced self-association, which may be particularly suited to coalesce and reorganize actin bundles within the filopodia.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/fisiología , Animales , Perros , Células HT29 , Humanos , Células de Riñón Canino Madin Darby , Proteínas de Microfilamentos/metabolismo , Microscopía Fluorescente , Microtúbulos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Seudópodos/metabolismo
9.
Cell Adh Migr ; 5(5): 409-20, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21975550

RESUMEN

The goal of this review is to highlight how emerging new models of filopodia assembly, which include tissue specific actin-bundling proteins, could provide more comprehensive representations of filopodia assembly that would describe more adequately and effectively the complexity and plasticity of epithelial cells.  This review also describes how the true diversity of actin bundling proteins must be considered to predict the far-reaching significance and versatile functions of filopodia in epithelial cells.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Microvellosidades/química , Seudópodos/química , Seudópodos/metabolismo , Actinas/química , Actinas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Movimiento Celular , Humanos , Proteínas de Microfilamentos/genética , Microvellosidades/metabolismo , Modelos Moleculares , Metástasis de la Neoplasia , Seudópodos/genética
10.
J Biol Chem ; 284(51): 35278-82, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19808673

RESUMEN

We have previously reported that the epithelial cell-specific actin-binding protein villin directly associates with phosphatidylinositol 4,5-bisphosphate (PIP(2)) through three binding sites that overlap with actin-binding sites in villin. As a result, association of villin with PIP(2) inhibits actin depolymerization and enhances actin cross-linking by villin. In this study, we demonstrate that these three PIP(2)-binding sites also bind the more hydrophilic phospholipid, lysophosphatidic acid (LPA) but with a higher affinity than PIP(2) (dissociation constant (K(d)) of 22 mum versus 39.5 mum for PIP(2)). More interestingly, unlike PIP(2), the association of villin with LPA inhibits all actin regulatory functions of villin. In addition, unlike PIP(2), LPA dramatically stimulates the tyrosine phosphorylation of villin by c-Src kinase. These studies suggest that in cells, selective interaction of villin with either PIP(2) or LPA could have dramatically different outcomes on actin reorganization as well as phospholipid-regulated cell signaling. These studies provide a novel regulatory mechanism for phospholipid-induced changes in the microfilament structure and cell function and suggest that LPA could be an intracellular regulator of the actin cytoskeleton.


Asunto(s)
Actinas/química , Lisofosfolípidos/química , Proteínas de Microfilamentos/química , Fosfatidilinositol 4,5-Difosfato/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Sitios de Unión/fisiología , Proteína Tirosina Quinasa CSK , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lisofosfolípidos/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosforilación/fisiología , Unión Proteica/fisiología , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas
11.
J Biol Chem ; 283(33): 22709-22, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18482983

RESUMEN

The ubiquitously expressed Src tyrosine kinases (c-Src, c-Yes, and c-Fyn) regulate intestinal cell growth and differentiation. Src activity is also elevated in the majority of malignant and premalignant tumors of the colon. The development of fibroblasts with the three ubiquitously expressed kinases deleted (SYF cells) has identified the role of Src proteins in the regulation of actin dynamics associated with increased cell migration and invasion. Despite this, unexpectedly nothing is known about the role of the individual Src kinases on intestinal cell cytoskeleton and/or cell migration. We have previously reported that villin, an epithelial cell-specific actin-modifying protein that regulates actin reorganization, cell morphology, cell migration, cell invasion, and apoptosis, is tyrosine-phosphorylated. In this report using the SYF cells reconstituted individually with c-Src, c-Yes, c-Fyn, and wild type or phosphorylation site mutants of villin, we demonstrate for the first time the absolute requirement for c-Src in villin-induced regulation of cell migration. The other major finding of our study is that contrary to previous reports, the nonreceptor tyrosine kinase, Jak3 (Janus kinase 3), does not regulate phosphorylation of villin or villin-induced cell migration and is, in fact, not expressed in intestinal epithelial cells. Further, we identify SHP-2 and PTP-PEST (protein-tyrosine phosphatase proline-, glutamate-, serine-, and threonine-rich sequence) as negative regulators of c-Src kinase and demonstrate a new function for these phosphatases in intestinal cell migration. Together, these data suggest that in colorectal carcinogenesis, elevation of c-Src or down-regulation of SHP-2 and/or PTP-PEST may promote cancer metastases and invasion by regulating villin-induced cell migration and cell invasion.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Tirosina Quinasas/metabolismo , Proteína Tirosina Quinasa CSK , Células CACO-2/fisiología , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Células HeLa , Humanos , Janus Quinasa 3/metabolismo , Invasividad Neoplásica , Fosforilación , Familia-src Quinasas
12.
FEBS Lett ; 582(14): 2128-39, 2008 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-18307996

RESUMEN

Villin is a tissue-specific actin modifying protein that is associated with actin filaments in the microvilli and terminal web of epithelial cells. It belongs to a large family of actin-binding proteins which includes actin-capping, -nucleating and/or -severing proteins such as gelsolin, severin, fragmin, adseverin/scinderin and actin crosslinking proteins such as dematin and supervillin. Studies done in epithelial cell lines and villin knock-out mice have demonstrated the function of villin in regulating actin dynamics, cell morphology, epithelial-to-mesenchymal transition, cell migration and cell survival. In addition, the ligand-binding properties of villin (F-actin, G-actin, calcium, phospholipids and phospholipase C-gamma1) are mechanistically important for the crosstalk between signaling pathways and actin reorganization in epithelial cells.


Asunto(s)
Actinas/metabolismo , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Epiteliales/fisiología , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Movimiento Celular , Supervivencia Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Estructura Terciaria de Proteína
13.
J Biol Chem ; 283(14): 9454-64, 2008 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-18198174

RESUMEN

Apoptosis is a key regulator for the normal turnover of the intestinal mucosa, and abnormalities associated with this function have been linked to inflammatory bowel disease and colorectal cancer. Despite this, little is known about the mechanism(s) mediating intestinal epithelial cell apoptosis. Villin is an actin regulatory protein that is expressed in every cell of the intestinal epithelium as well as in exocrine glands associated with the gastrointestinal tract. In this study we demonstrate for the first time that villin is an epithelial cell-specific anti-apoptotic protein. Absence of villin predisposes mice to dextran sodium sulfate-induced colitis by promoting apoptosis. To better understand the cellular and molecular mechanisms of the anti-apoptotic function of villin, we overexpressed villin in the Madin-Darby canine kidney Tet-Off epithelial cell line to demonstrate that expression of villin protects cells from apoptosis by maintaining mitochondrial integrity thus inhibiting the activation of caspase-9 and caspase-3. Furthermore, we report that the anti-apoptotic response of villin depends on activation of the pro-survival proteins, phosphatidylinositol 3-kinase and phosphorylated Akt. The results of our studies shed new light on the previously unrecognized function of villin in the regulation of apoptosis in the gastrointestinal epithelium.


Asunto(s)
Apoptosis , Células Epiteliales/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Proteínas de Microfilamentos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran/toxicidad , Perros , Células Epiteliales/patología , Células HeLa , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt
14.
Exp Cell Res ; 314(3): 530-42, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18054784

RESUMEN

Autotaxin (ATX) is a potent tumor cell motogen that can produce lysophosphatidic acid (LPA) from lysophosphatidylcholine. LPA is a lipid mediator that has also been shown to modulate tumor cell invasion. Autotaxin mRNA is expressed at significant levels in the intestine. Likewise, LPA2 receptor levels have been shown to be elevated in colon cancers. The molecular mechanism of ATX/LPA-induced increase in intestinal cell migration however, remains poorly understood. Villin is an intestinal and renal epithelial cell specific actin regulatory protein that modifies epithelial cell migration. In this study we demonstrate that both Caco-2 (endogenous villin) and MDCK (exogenous villin) cells, which express primarily LPA2 receptors, show enhanced cell migration in response to ATX/LPA. ATX and LPA treatment results in the rapid formation of lamellipodia and redistribution of villin to these cell surface structures, suggesting a role for villin in regulating this initial event of cell locomotion. The LPA-induced increase in cell migration required activation of c-src kinase and downstream tyrosine phosphorylation of villin by c-src kinase. LPA stimulated cell motility was determined to be insensitive to pertussis toxin, but was regulated by activation of PLC-gamma 1. Together, our results show that in epithelial cells ATX and LPA act as strong stimulators of cell migration by recruiting PLC-gamma 1 and villin, both of which participate in the initiation of protrusion.


Asunto(s)
Movimiento Celular/fisiología , Mucosa Intestinal/metabolismo , Lisofosfolípidos/fisiología , Proteínas de Microfilamentos/metabolismo , Complejos Multienzimáticos/fisiología , Fosfodiesterasa I/fisiología , Seudópodos/metabolismo , Pirofosfatasas/fisiología , Actinas/metabolismo , Animales , Células CACO-2 , Carcinoma/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Perros , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Neoplasias Intestinales/metabolismo , Lisofosfolípidos/farmacología , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/farmacología , Complejos Multienzimáticos/farmacología , Fosfodiesterasa I/farmacología , Fosfolipasa C gamma/efectos de los fármacos , Fosfolipasa C gamma/metabolismo , Hidrolasas Diéster Fosfóricas , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Seudópodos/efectos de los fármacos , Seudópodos/ultraestructura , Pirofosfatasas/farmacología , Receptores del Ácido Lisofosfatídico/efectos de los fármacos , Receptores del Ácido Lisofosfatídico/metabolismo , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo
15.
J Biol Chem ; 282(36): 26528-41, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17606613

RESUMEN

Villin is a major actin-bundling protein in the brush border of epithelial cells. In this study we demonstrate for the first time that villin can bundle actin filaments using a single F-actin binding site, because it has the ability to self-associate. Using fluorescence resonance energy transfer, we demonstrate villin self-association in living cells in microvilli and in growth factor-stimulated cells in membrane ruffles and lamellipodia. Using sucrose density gradient, size-exclusion chromatography, and matrix-assisted laser desorption ionization time-of-flight, the majority of villin was identified as a monomer or dimer. Villin dimers were also identified in Caco-2 cells, which endogenously express villin and Madin-Darby canine kidney cells that ectopically express villin. Using truncation mutants of villin, site-directed mutagenesis, and fluorescence resonance energy transfer, an amino-terminal dimerization site was identified that regulated villin self-association in parallel conformation as well as actin bundling by villin. This detailed analysis describes for the first time microvillus assembly by villin, redefines the actin-bundling function of villin, and provides a molecular mechanism for actin bundling by villin, which could have wider implications for other actin cross-linking proteins that share a villin-like headpiece domain. Our study also provides a molecular basis to separate the morphologically distinct actin-severing and actin-bundling properties of villin.


Asunto(s)
Actinas/metabolismo , Células Epiteliales/metabolismo , Proteínas de Microfilamentos/metabolismo , Seudópodos/metabolismo , Eliminación de Secuencia , Actinas/química , Actinas/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células CACO-2 , Línea Celular , Dimerización , Perros , Células Epiteliales/química , Células Epiteliales/ultraestructura , Transferencia Resonante de Energía de Fluorescencia , Humanos , Proteínas de Microfilamentos/genética , Microvellosidades/química , Microvellosidades/genética , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Seudópodos/química , Seudópodos/genética , Seudópodos/ultraestructura , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
Am J Physiol Cell Physiol ; 292(5): C1775-86, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17229814

RESUMEN

While there is circumstantial evidence to suggest a requirement for phospholipase C-gamma(1) (PLC-gamma(1)) in actin reorganization and cell migration, few studies have examined the direct mechanisms that link regulators of the actin cytoskeleton with this crucial signaling molecule. This study was aimed to examine the role that villin, an epithelial cell-specific actin-binding protein, and its ligand PLC-gamma(1) play in migration in intestinal and renal epithelial cell lines that endogenously or ectopically express human villin. Basal as well as epidermal growth factor (EGF)-stimulated cell migration was accompanied by tyrosine phosphorylation of villin and its association with PLC-gamma(1). Inhibition of villin phosphorylation prevented villin-PLC-gamma(1) complex formation as well as villin-induced cell migration. The absolute requirement for PLC-gamma(1) in villin-induced cell migration was demonstrated by measuring cell motility in PLC-gamma(1)(-/-) cells and by downregulation of endogenous PLC-gamma(1). EGF-stimulated direct interaction of villin with the Src homology domain 2 domain of PLC-gamma(1) at the plasma membrane was demonstrated in living cells by using fluorescence resonance energy transfer. These results demonstrate that villin provides an important link between the activation of phosphoinositide signal transduction pathway and epithelial cell migration.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Células Epiteliales/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfolipasa C gamma/metabolismo , Transducción de Señal , Cicatrización de Heridas , Animales , Células CACO-2 , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Perros , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Estrenos/farmacología , Transferencia Resonante de Energía de Fluorescencia , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Riñón/citología , Riñón/metabolismo , Ratones , Proteínas de Microfilamentos/genética , Mutación , Fosfolipasa C gamma/antagonistas & inhibidores , Fosfolipasa C gamma/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirrolidinonas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Tirosina/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Dominios Homologos src , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
17.
J Biol Chem ; 277(20): 17978-86, 2002 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-11844793

RESUMEN

The first report of slow-tight inhibition of xylanase by a bifunctional inhibitor alkalo-thermophilic Bacillus inhibitor (ATBI), from an extremophilic Bacillus sp. is described. ATBI inhibits aspartic protease (Dash, C., and Rao, M. (2001) J. Biol. Chem., 276, 2487-2493) and xylanase (Xyl I) from a Thermomonospora sp. The steady-state kinetics revealed time-dependent competitive inhibition of Xyl I by ATBI, consistent with two-step inhibition mechanism. The inhibition followed a rapid equilibrium step to form a reversible enzyme-inhibitor complex (EI), which isomerizes to the second enzyme-inhibitor complex (EI*), which dissociated at a very slow rate. The rate constants determined for the isomerization of EI to EI*, and the dissociation of EI* were 13 +/- 1 x 10(-6) s(-1) and 5 +/- 0.5 x 10(-8) s(-1), respectively. The K(i) value for the formation of EI complex was 2.5 +/- 0.5 microm, whereas the overall inhibition constant K(i)* was 7 +/- 1 nm. The conformational changes induced in Xyl I by ATBI were monitored by fluorescence spectroscopy and the rate constants derived were in agreement with the kinetic data. Thus, the conformational alterations were correlated to the isomerization of EI to EI*. ATBI binds to the active site of the enzyme and disturbs the native interaction between the histidine and lysine, as demonstrated by the abolished isoindole fluorescence of o-phthalaldehyde (OPTA)-labeled Xyl I. Our results revealed that the inactivation of Xyl I is due to the disruption of the hydrogen-bonding network between the essential histidine and other residues involved in catalysis and a model depicting the probable interaction between ATBI or OPTA with Xyl I has been proposed.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Proteínas Bacterianas/farmacología , Inhibidores Enzimáticos/farmacología , Xilosidasas/metabolismo , Actinomycetales , Bacillus , Isomerismo , Cinética , Modelos Químicos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Espectrometría de Fluorescencia , Relación Estructura-Actividad , Especificidad por Sustrato , Xilano Endo-1,3-beta-Xilosidasa , o-Ftalaldehído
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA