Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(20): 8402-7, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21536877

RESUMEN

Fibroblasts are important participants in inflammation. Although not leukocytes, their capacity to produce cytokines, chemokines, and other inflammatory factors locally in tissues suggests that they can contribute to inflammatory diseases. For example, fibroblasts in a rheumatoid arthritis (RA) joint are a dominant source of IL-6 and RANKL in the synovium, both of which are therapeutic targets for inflammation and bone erosion. Previously, we found that fibroblasts can be targeted by mAb directed against cadherin-11 (cad-11), a mesenchymal cadherin that fibroblasts selectively express. Targeting cad-11 significantly reduced inflammation as assessed by joint swelling and clinical inflammation scores. However, the mechanism by which anti-cad-11 reduced inflammation was not known. Here, we show that cad-11 engagement induces synovial fibroblasts to secret proinflammatory cytokines including IL-6. Cad-11 engagement strongly synergized with TNF-α and IL-1ß in the induction of IL-6. Importantly, cad-11 activated MAP kinases and NF-κB for IL-6 induction. IL-6 levels in ankles of inflamed joints were reduced in cad-11 mutant mice compared to wild-type mice with inflammatory arthritis. Thus, we suggest that cad-11 modulates synovial fibroblasts to evoke inflammatory factors that may contribute to the inflammatory process in RA.


Asunto(s)
Cadherinas/fisiología , Fibroblastos/patología , Inflamación/patología , Animales , Artritis Experimental , Artritis Reumatoide , Células Cultivadas , Citocinas/metabolismo , Humanos , Interleucina-1beta , Interleucina-6 , Ratones , Factor de Necrosis Tumoral alfa
2.
Immunol Rev ; 233(1): 256-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20193004

RESUMEN

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease affecting the joint synovium. The normal synovium consists of a lining layer of fibroblast-like synoviocytes (FLS) and macrophages, one to three cells deep that overlies the loose connective tissue of the synovial sublining. During the course of RA, the synovium is the site of inflammation where immune cells are massively infiltrated, and the lining layer becomes hyperplastic and transforms into a pannus tissue that destroys articular cartilage and bone. FLS play an important role in this RA pathogenesis. In this review, we explain that cadherin-11, an adhesion molecule, is selectively expressed on FLS and required for synovial lining formation. In addition, cadherin-11 on FLS contributes to synovial inflammation and mediates cartilage degradation in a mouse model of inflammatory arthritis. Therefore, we suggest that FLS are critical regulators of synovial inflammation and arthritis pathology via mechanisms that are mediated by cadherin-11.


Asunto(s)
Artritis Reumatoide/inmunología , Cadherinas/inmunología , Fibroblastos/inmunología , Transducción de Señal/inmunología , Membrana Sinovial/inmunología , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/patología , Cartílago Articular/inmunología , Cartílago Articular/patología , Fibroblastos/patología , Humanos , Ratones , Fenotipo , Membrana Sinovial/patología
3.
Cancers (Basel) ; 16(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38893085

RESUMEN

Recent studies highlight the integral role of the interferon gamma receptor (IFNγR) pathway in T cell-mediated cytotoxicity against solid but not liquid tumors. IFNγ not only directly facilitates tumor cell death by T cells but also indirectly promotes cytotoxicity via myeloid phagocytosis in the tumor microenvironment. Meanwhile, full human ex vivo immune checkpoint drug screening remains challenging. We hypothesized that an engineered gamma interferon activation site response element luciferase reporter (GAS-Luc2) can be utilized for immune checkpoint drug screening in diverse ex vivo T cell-solid tumor cell co-culture systems. We comprehensively profiled cell surface proteins in ATCC's extensive collection of human tumor and immune cell lines, identifying those with endogenously high expression of established and novel immune checkpoint molecules and binding ligands. We then engineered three GAS-Luc2 reporter tumor cell lines expressing immune checkpoints PD-L1, CD155, or B7-H3/CD276. Luciferase expression was suppressed upon relevant immune checkpoint-ligand engagement. In the presence of an immune checkpoint inhibitor, T cells released IFNγ, activating the JAK-STAT pathway in GAS-Luc2 cells, and generating a quantifiable bioluminescent signal for inhibitor evaluation. These reporter lines also detected paracrine IFNγ signaling for immune checkpoint-targeted ADCC drug screening. Further development into an artificial antigen-presenting cell line (aAPC) significantly enhanced T cell signaling for superior performance in these ex vivo immune checkpoint drug screening platforms.

4.
J Am Soc Nephrol ; 23(9): 1506-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22859853

RESUMEN

The aquaporin 2 (AQP2) water channel, expressed in kidney collecting ducts, contributes critically to water homeostasis in mammals. Animals lacking or having significantly reduced levels of AQP2, however, have not only urinary concentrating abnormalities but also renal tubular defects that lead to neonatal mortality from renal failure. Here, we show that AQP2 is not only a water channel but also an integrin-binding membrane protein that promotes cell migration and epithelial morphogenesis. AQP2 expression modulates the trafficking and internalization of integrin ß1, facilitating its turnover at focal adhesions. In vitro, disturbing the interaction between AQP2 and integrin ß1 by mutating the RGD motif led to reduced endocytosis, retention of integrin ß1 at the cell surface, and defective cell migration and tubulogenesis. Similarly, in vivo, AQP2-null mice exhibited significant retention of integrin ß1 at the basolateral membrane and had tubular abnormalities. In summary, these data suggest that the water channel AQP2 interacts with integrins to promote renal epithelial cell migration, contributing to the structural and functional integrity of the mammalian kidney.


Asunto(s)
Acuaporina 2/fisiología , Movimiento Celular/fisiología , Células Epiteliales/citología , Riñón/citología , Morfogénesis/fisiología , Animales , Acuaporina 2/deficiencia , Acuaporina 2/genética , Línea Celular , Permeabilidad de la Membrana Celular/fisiología , Perros , Endocitosis/fisiología , Células Epiteliales/fisiología , Técnicas In Vitro , Integrina beta1/fisiología , Riñón/crecimiento & desarrollo , Riñón/fisiología , Ratones , Ratones Noqueados , Modelos Animales , Mutación/genética , Oligopéptidos/genética , Oligopéptidos/fisiología , Porcinos , Transfección
5.
Sci Adv ; 9(2): eabq6480, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36630496

RESUMEN

Cells tune adherens junction dynamics to regulate epithelial integrity in diverse (patho)physiological processes, including cancer metastasis. We hypothesized that the spatially confining architecture of peritumor stroma promotes metastatic cell dissemination by remodeling cell-cell adhesive interactions. By combining microfluidics with live-cell imaging, FLIM/FRET biosensors, and optogenetic tools, we show that confinement induces leader cell dissociation from cohesive ensembles. Cell dissociation is triggered by myosin IIA (MIIA) dismantling of E-cadherin cell-cell junctions, as recapitulated by a mathematical model. Elevated MIIA contractility is controlled by RhoA/ROCK activation, which requires distinct guanine nucleotide exchange factors (GEFs). Confinement activates RhoA via nucleocytoplasmic shuttling of the cytokinesis-regulatory proteins RacGAP1 and Ect2 and increased microtubule dynamics, which results in the release of active GEF-H1. Thus, confining microenvironments are sufficient to induce cell dissemination from primary tumors by remodeling E-cadherin cell junctions via the interplay of microtubules, nuclear trafficking, and RhoA/ROCK/MIIA pathway and not by down-regulating E-cadherin expression.


Asunto(s)
Citocinesis , Uniones Intercelulares , Cadherinas/metabolismo , Citocinesis/fisiología , Uniones Intercelulares/metabolismo , Microtúbulos/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Humanos
6.
Cancers (Basel) ; 14(21)2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36358868

RESUMEN

BRAF V600E mutation drives uncontrolled cell growth in most melanomas. While BRAF V600E tumors are initially responsive to BRAF inhibitors, prolonged treatment results in inhibitor resistance and tumor regrowth. Clinical data have linked the NRAS Q61K, KRAS G13D and MEK1 Q56P mutations to the BRAF inhibitor resistance. However, development of novel therapeutics is hindered by the lack of relevant isogeneic cell models. We employed CRISPR/Cas9 genome engineering to introduce NRAS Q61K, KRAS G13D and MEK1 Q56P mutations into the A375 melanoma cell line with endogenously high expression of BRAF V600E. The resulting isogenic cell lines are resistant to BRAF inhibitors. The A375 MEK1 Q56P isogenic cells are additionally resistant to MEK inhibitors as single agent, but interestingly, these cells become sensitive to MEK/BRAF inhibitor combo. Our results suggest that resistance in the NRAS and MEK isogenic lines is driven by constitutive MEK/ERK signaling, while the resistance in the KRAS isogenic line is driven by EGFR overexpression. Interestingly, the KRAS G13D isogenic line displays elevated PD-L1 expression suggesting the KRAS G13D mutation could be a potential indication for immunotherapy. Overall, these three novel isogenic cell models with endogenous level RAS and MEK1 point mutations provide direct bio-functional evidence demonstrating that acquiring a drug-resistant gene drives tumor cell survival and may simultaneously introduce new indications for combo therapy or immunotherapy in the clinic.

7.
J Exp Med ; 218(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34143182

RESUMEN

Central precocious puberty (CPP), largely caused by germline mutations in the MKRN3 gene, has been epidemiologically linked to cancers. MKRN3 is frequently mutated in non-small cell lung cancers (NSCLCs) with five cohorts. Genomic MKRN3 aberrations are significantly enriched in NSCLC samples harboring oncogenic KRAS mutations. Low MKRN3 expression levels correlate with poor patient survival. Reconstitution of MKRN3 in MKRN3-inactivated NSCLC cells directly abrogates in vitro and in vivo tumor growth and proliferation. MKRN3 knockout mice are susceptible to urethane-induced lung cancer, and lung cell-specific knockout of endogenous MKRN3 accelerates NSCLC tumorigenesis in vivo. A mass spectrometry-based proteomics screen identified PABPC1 as a major substrate for MKRN3. The tumor suppressor function of MKRN3 is dependent on its E3 ligase activity, and MKRN3 missense mutations identified in patients substantially compromise MKRN3-mediated PABPC1 ubiquitination. Furthermore, MKRN3 modulates cell proliferation through PABPC1 nonproteolytic ubiquitination and subsequently, PABPC1-mediated global protein synthesis. Our integrated approaches demonstrate that the CPP-associated gene MKRN3 is a tumor suppressor.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Unión Proteica , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/genética , Reproducibilidad de los Resultados , Análisis de Supervivencia , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Uretano
8.
Eur J Cell Biol ; 85(3-4): 305-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16546574

RESUMEN

Caldesmon is an actin-binding protein that is capable of stabilizing actin filaments against actin-severing proteins, inhibiting actomyosin ATPase activity, and inhibiting Arp2/3-mediated actin polymerization in vitro. Caldesmon is a substrate of cdc2 kinase and Erk1/2 MAPK, and phosphorylation by either of these kinases reverses the inhibitory effects of caldesmon. Cdc2-mediated caldesmon phosphorylation and the resulting dissociation of caldesmon from actin filaments are essential for M-phase progression during mitosis. Cells overexpressing the actin-binding carboxyterminal fragment of caldesmon fail to release the fragment completely from actin filaments during mitosis, resulting in a higher frequency of multinucleated cells. PKC-mediated MEK/Erk/caldesmon phosphorylation is an important signaling cascade in the regulation of smooth muscle contraction. Furthermore, PKC activation has been shown to remodel actin stress fibers into F-actin-enriched podosome columns in cultured vascular smooth muscle cells. Podosomes are cytoskeletal adhesion structures associated with the release of metalloproteases and degradation of extracellular matrix during cell invasion. Interestingly, caldesmon is one of the few actin-binding proteins that is associated with podosomes but excluded from focal adhesions. Caldesmon also inhibits the function of gelsolin and Arp2/3 complex that are essential for the formation of podosomes. Thus, caldesmon appears to be well positioned for playing a modulatory role in the formation of podosomes. Defining the roles of actin filament-stabilizing proteins such as caldesmon and tropomyosin in the formation of podosomes should provide a more complete understanding of molecular systems that regulate the remodeling of the actin cytoskeleton in cell transformation and invasion.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Animales , Ratones , Mitosis , Fosforilación
9.
Cell Rep ; 15(7): 1430-1441, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27160899

RESUMEN

Cells adopt distinct signaling pathways to optimize cell locomotion in different physical microenvironments. However, the underlying mechanism that enables cells to sense and respond to physical confinement is unknown. Using microfabricated devices and substrate-printing methods along with FRET-based biosensors, we report that, as cells transition from unconfined to confined spaces, intracellular Ca(2+) level is increased, leading to phosphodiesterase 1 (PDE1)-dependent suppression of PKA activity. This Ca(2+) elevation requires Piezo1, a stretch-activated cation channel. Moreover, differential regulation of PKA and cell stiffness in unconfined versus confined cells is abrogated by dual, but not individual, inhibition of Piezo1 and myosin II, indicating that these proteins can independently mediate confinement sensing. Signals activated by Piezo1 and myosin II in response to confinement both feed into a signaling circuit that optimizes cell motility. This study provides a mechanism by which confinement-induced signaling enables cells to sense and adapt to different physical microenvironments.


Asunto(s)
Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Canales Iónicos/metabolismo , Miosina Tipo II/metabolismo , Transducción de Señal , Animales , Células CHO , Calcio/metabolismo , Calcio/farmacología , Línea Celular Tumoral , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Espacio Intracelular/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Melanoma/metabolismo , Melanoma/patología
10.
Bioarchitecture ; 4(3): 116-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25029598

RESUMEN

Current knowledge understands the mesenchymal cell invasion in a 3D matrix as a combined process of cell-to-matrix adhesion based cell migration and matrix remodeling. Excluding cell invasion stimulated by cytokines and chemokines, the basal cell invasion itself is a complicated process that can be regulated by matrix ligand type, density, geometry, and stiffness, etc. Understanding such a complicated biological process requires delicate dissections into simplified model studies by altering only one or two elements at a time. Past cell motility studies focusing on matrix stiffness have revealed that a stiffer matrix promotes 2D X-Y axis lateral cell motility. Here, we comment on two recent studies that report, instead of stiffer matrix, a softer matrix promotes matrix proteolysis and the formation of invadosome-like protrusions (ILPs) along the 3D Z axis. These studies also reveal that soft matrix precisely regulates such ILPs formation in the stiffness scale range of 0.1 kilopascal in normal cells. In contrast, malignant cells such as cancer cells can form ILPs in response to a much wider range of matrix stiffness. Further, different cancer cells respond to their own favorable range of matrix stiffness to spontaneously form ILPs. Thus, we hereby propose the idea of utilizing the matrix stiffness to precisely regulate ILP formation as a mechanophenotyping tool for cancer metastasis prediction and pathological diagnosis.


Asunto(s)
Uniones Adherentes/metabolismo , Extensiones de la Superficie Celular/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Fibroblastos/patología , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Mecanotransducción Celular , Oligopéptidos/farmacología , Seudópodos/metabolismo , Animales , Humanos
11.
Curr Opin Cell Biol ; 30: 41-50, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24973724

RESUMEN

Cell migration is a fundamental process underlying diverse (patho)physiological phenomena. The classical understanding of the molecular mechanisms of cell migration has been based on in vitro studies on two-dimensional substrates. More recently, mounting evidence from intravital studies has shown that during metastasis, tumor cells must navigate complex microenvironments in vivo, including narrow, pre-existing microtracks created by anatomical structures. It is becoming apparent that unraveling the mechanisms of confined cell migration in this context requires a multi-disciplinary approach through integration of in vivo and in vitro studies, along with sophisticated bioengineering techniques and mathematical modeling. Here, we highlight such an approach that has led to discovery of a new model for cell migration in confined microenvironments (i.e., the Osmotic Engine Model).


Asunto(s)
Bioingeniería , Movimiento Celular , Animales , Técnicas de Cultivo de Célula , Humanos , Modelos Biológicos , Neoplasias/patología , Microambiente Tumoral
12.
Mol Biol Cell ; 25(4): 457-69, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24336521

RESUMEN

Directional mesenchymal cell invasion in vivo is understood to be a stimulated event and to be regulated by cytokines, chemokines, and types of extracellular matrix (ECM). Instead, by focusing on the cellular response to ECM stiffness, we found that soft ECM (low stiffness) itself is sufficient to prevent stable cell-to-cell adherens junction formation, up-regulate matrix metalloproteinase (MMP) secretion, promote MMP activity, and induce invadosome-like protrusion (ILP) formation. Consistently, similar ILP formation was also detected in a three-dimensional directional invasion assay in soft matrix. Primary human fibroblasts spontaneously form ILPs in a very narrow range of ECM stiffness (0.1-0.4 kPa), and such ILP formation is Src family kinase dependent. In contrast, spontaneous ILP formation in malignant cancer cells and fibrosarcoma cells occurs across a much wider range of ECM stiffness, and these tumor cell ILPs are also more prominent at lower stiffness. These findings suggest that ECM softness is a natural stimulator for cellular invasiveness.


Asunto(s)
Uniones Adherentes/metabolismo , Extensiones de la Superficie Celular/metabolismo , Matriz Extracelular/química , Fibroblastos/patología , Mecanotransducción Celular , Uniones Adherentes/ultraestructura , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Extensiones de la Superficie Celular/ultraestructura , Elasticidad , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Dureza , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Invasividad Neoplásica , Cultivo Primario de Células , Propiedades de Superficie , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
13.
Nat Genet ; 46(6): 601-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24793134

RESUMEN

Many common human mesenchymal tumors, including gastrointestinal stromal tumor (GIST), rhabdomyosarcoma (RMS) and leiomyosarcoma (LMS), feature myogenic differentiation. Here we report that intragenic deletion of the dystrophin-encoding and muscular dystrophy-associated DMD gene is a frequent mechanism by which myogenic tumors progress to high-grade, lethal sarcomas. Dystrophin is expressed in the non-neoplastic and benign counterparts of GIST, RMS and LMS tumors, and DMD deletions inactivate larger dystrophin isoforms, including 427-kDa dystrophin, while preserving the expression of an essential 71-kDa isoform. Dystrophin inhibits myogenic sarcoma cell migration, invasion, anchorage independence and invadopodia formation, and dystrophin inactivation was found in 96%, 100% and 62% of metastatic GIST, embryonal RMS and LMS samples, respectively. These findings validate dystrophin as a tumor suppressor and likely anti-metastatic factor, suggesting that therapies in development for muscular dystrophies may also have relevance in the treatment of cancer.


Asunto(s)
Distrofina/genética , Distrofina/fisiología , Distrofias Musculares/genética , Sarcoma/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Femenino , Tumores del Estroma Gastrointestinal/genética , Eliminación de Gen , Genes Supresores de Tumor , Humanos , Hibridación Fluorescente in Situ , Células Intersticiales de Cajal/patología , Leiomiosarcoma/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Desarrollo de Músculos/genética , Músculo Esquelético/patología , Distrofias Musculares/patología , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Rabdomiosarcoma/genética
14.
Vascul Pharmacol ; 58(1-2): 87-97, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22940282

RESUMEN

Cigarette smoking is a significant risk factor for atherosclerosis, which involves the invasion of vascular smooth muscle cells (VSMCs) from the media to intima. A hallmark of many invasive cells is actin cytoskeletal remodeling in the form of podosomes, accompanied by extracellular matrix (ECM) degradation. A7r5 VSMCs form podosomes in response to PKC activation. In this study, we found that cigarette smoke extract, nicotine, and the cholinergic agonist, carbachol, were similarly effective in inducing the formation of podosome rosettes in A7r5 VSMCs. α-Bungarotoxin and atropine experiments confirmed the involvement of nicotinic acetylcholine receptors (nAChRs). Western blotting and immunofluorescence experiments revealed the aggregation of nAChRs at podosome rosettes. Cycloheximide experiments and media exchange experiments suggested that autocrine factor(s) and intracellular phenotypic modulation are putative mechanisms. In situ zymography experiments indicated that, in response to PKC activation, nicotine-treated cells degraded ECM near podosome rosettes, and possibly endocytose ECM fragments to intracellular compartments. Invasion assay of human aortic smooth muscle cells indicated that nicotine and PKC activation individually and synergistically enhanced cell invasion through ECM. Results from this study suggest that nicotine enhances the ability of VSMCs to degrade and invade ECM. nAChR activation, actin cytoskeletal remodeling and phenotypic modulation are possible mechanisms.


Asunto(s)
Músculo Liso Vascular/efectos de los fármacos , Nicotina/toxicidad , Receptores Nicotínicos/metabolismo , Humo/efectos adversos , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Carbacol/farmacología , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Fumar/efectos adversos , Nicotiana/química
15.
J Cell Biol ; 193(1): 61-70, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21464228

RESUMEN

During cell migration, integrins are redistributed from focal adhesions undergoing disassembly at the cell's trailing edges to new focal adhesions assembling at leading edges. The initial step of integrin redistribution is thought to require clathrin-mediated endocytosis. However, whether clathrin-mediated endocytosis functions in different contexts, such as basal versus stimulated migration, has not been determined. In this paper, we examine the spatial and temporal redistribution of integrins from focal adhesions upon stimulation by growth factors. Four-dimensional confocal live-cell imaging along with functional analysis reveals that surface integrins do not undergo significant endocytosis at ventral focal adhesions upon cell stimulation with the platelet-derived growth factor. Rather, they abruptly redistribute to dorsal circular ruffles, where they are internalized through macropinocytosis. The internalized integrins then transit through recycling endosomal compartments to repopulate newly formed focal adhesions on the ventral surface. These findings explain why integrins have long been observed to redistribute through both surface-based and internal routes and identify a new function for macropinocytosis during growth factor-induced cell migration.


Asunto(s)
Estructuras de la Membrana Celular/fisiología , Integrinas/metabolismo , Pinocitosis/fisiología , Animales , Movimiento Celular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Exp Cell Res ; 313(5): 849-66, 2007 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-17239373

RESUMEN

We tested the hypothesis that the MEK/Erk/caldesmon phosphorylation cascade regulates PKC-mediated podosome dynamics in A7r5 cells. We observed the phosphorylation of MEK, Erk and caldesmon, and their translocation to the podosomes upon phorbol dibutyrate (PDBu) stimulation, together with the nuclear translocation of phospho-MEK and phospho-Erk. After MEK inhibition by U0126, Erk translocated to the interconnected actin-rich columns but failed to translocate to the nucleus, suggesting that podosomes served as a site for Erk phosphorylation. The interconnected actin-rich columns in U0126-treated, PDBu-stimulated cells contained alpha-actinin, caldesmon, vinculin, and metalloproteinase-2. Caldesmon and vinculin became integrated with F-actin at the columns, in contrast to their typical location at the ring of podosomes. Live-imaging experiments suggested the growth of these columns from podosomes that were slow to disassemble. The observed modulation of podosome size and life time in A7r5 cells overexpressing wild-type and phosphorylation-deficient caldesmon-GFP mutants in comparison to untransfected cells suggests that caldesmon and caldesmon phosphorylation modulate podosome dynamics in A7r5 cells. These results suggest that Erk1/2 and caldesmon differentially modulate PKC-mediated formation and/or dynamics of podosomes in A7r5 vascular smooth muscle cells.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Extensiones de la Superficie Celular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Actinas/metabolismo , Animales , Butadienos/farmacología , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/fisiología , Carcinógenos , Línea Celular , Inhibidores Enzimáticos , Regulación de la Expresión Génica , Guanosina Trifosfato/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Músculo Liso Vascular/citología , Nitrilos/farmacología , Forbol 12,13-Dibutirato/farmacología , Fosforilación , Transporte de Proteínas , Ratas , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA