Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(3): 773-8, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25568088

RESUMEN

(Macro)autophagy delivers cellular constituents to lysosomes for degradation. Although a cytoplasmic process, autophagy-deficient cells accumulate genomic damage, but an explanation for this effect is currently unclear. We report here that inhibition of autophagy causes elevated proteasomal activity leading to enhanced degradation of checkpoint kinase 1 (Chk1), a pivotal factor for the error-free DNA repair process, homologous recombination (HR). We show that loss of autophagy critically impairs HR and that autophagy-deficient cells accrue micronuclei and sub-G1 DNA, indicators of diminished genomic integrity. Moreover, due to impaired HR, autophagy-deficient cells are hyperdependent on nonhomologous end joining (NHEJ) for repair of DNA double-strand breaks. Consequently, inhibition of NHEJ following DNA damage in the absence of autophagy results in persistence of genomic lesions and rapid cell death. Because autophagy deficiency occurs in several diseases, these findings constitute an important link between autophagy and DNA repair and highlight a synthetic lethal strategy to kill autophagy-deficient cells.


Asunto(s)
Autofagia , Reparación del ADN/genética , Genes Letales , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Recombinación Homóloga , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Cell Commun Signal ; 12: 54, 2014 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-25288205

RESUMEN

BACKGROUND: The myotonic dystrophy kinase-related CDC42-binding kinases MRCKα and MRCKß regulate actin-myosin contractility and have been implicated in cancer metastasis. Along with the related ROCK1 and ROCK2 kinases, the MRCK proteins initiate signalling events that lead to contractile force generation which powers cancer cell motility and invasion. A potential strategy for cancer therapy is to reduce metastasis by blocking MRCK activity, either alone or in combination with ROCK inhibition. However, to date no potent small molecule inhibitors have been developed with selectivity towards MRCK. RESULTS: Screening a kinase-focused small molecule chemical library resulted in the identification of compounds with inhibitory activity towards MRCK. Medicinal chemistry combined with in vitro enzyme profiling led to the discovery of 4-chloro-1-(4-piperidyl)-N-[5-(2-pyridyl)-1H-pyrazol-4-yl]pyrazole-3-carboxamide (BDP00005290; abbreviated as BDP5290) as a potent MRCK inhibitor. X-ray crystallography of the MRCKß kinase domain in complex with BDP5290 revealed how this ligand interacts with the nucleotide binding pocket. BDP5290 demonstrated marked selectivity for MRCKß over ROCK1 or ROCK2 for inhibition of myosin II light chain (MLC) phosphorylation in cells. While BDP5290 was able to block MLC phosphorylation at both cytoplasmic actin stress fibres and peripheral cortical actin bundles, the ROCK selective inhibitor Y27632 primarily reduced MLC phosphorylation on stress fibres. BDP5290 was also more effective at reducing MDA-MB-231 breast cancer cell invasion through Matrigel than Y27632. Finally, the ability of human SCC12 squamous cell carcinoma cells to invade a three-dimensional collagen matrix was strongly inhibited by 2 µM BDP5290 but not the identical concentration of Y27632, despite equivalent inhibition of MLC phosphorylation. CONCLUSIONS: BDP5290 is a potent MRCK inhibitor with activity in cells, resulting in reduced MLC phosphorylation, cell motility and tumour cell invasion. The discovery of this compound will enable further investigations into the biological activities of MRCK proteins and their contributions to cancer progression.


Asunto(s)
Antineoplásicos/farmacología , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Amidas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Proteína Quinasa de Distrofia Miotónica/metabolismo , Invasividad Neoplásica , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
3.
J Cell Biol ; 168(2): 245-55, 2005 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-15657395

RESUMEN

Membrane blebbing during the apoptotic execution phase results from caspase-mediated cleavage and activation of ROCK I. Here, we show that ROCK activity, myosin light chain (MLC) phosphorylation, MLC ATPase activity, and an intact actin cytoskeleton, but not microtubular cytoskeleton, are required for disruption of nuclear integrity during apoptosis. Inhibition of ROCK or MLC ATPase activity, which protect apoptotic nuclear integrity, does not affect caspase-mediated degradation of nuclear proteins such as lamins A, B1, or C. The conditional activation of ROCK I was sufficient to tear apart nuclei in lamin A/C null fibroblasts, but not in wild-type fibroblasts. Thus, apoptotic nuclear disintegration requires actin-myosin contractile force and lamin proteolysis, making apoptosis analogous to, but distinct from, mitosis where nuclear disintegration results from microtubule-based forces and from lamin phosphorylation and depolymerization.


Asunto(s)
Actinas/metabolismo , Apoptosis/fisiología , Núcleo Celular/metabolismo , Laminas/metabolismo , Miosinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Amidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Cicloheximida/farmacología , Citocalasina D/farmacología , Proteínas del Citoesqueleto , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Péptidos y Proteínas de Señalización Intracelular , Laminas/genética , Quinasas Lim , Ratones , Microscopía Electrónica de Transmisión , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mutación/fisiología , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Miosinas/antagonistas & inhibidores , Células 3T3 NIH , Nocodazol/farmacología , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Quinasas Asociadas a rho
4.
Mol Cell Biol ; 26(12): 4612-27, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738326

RESUMEN

The members of the Rho GTPase family are well known for their regulation of actin cytoskeletal structures. In addition, they influence progression through the cell cycle. The RhoA and RhoC proteins regulate numerous effector proteins, with a central and vital signaling role mediated by the ROCK I and ROCK II serine/threonine kinases. The requirement for ROCK function in the proliferation of numerous cell types has been revealed by studies utilizing ROCK-selective inhibitors such as Y-27632. However, the mechanisms by which ROCK signaling promotes cell cycle progression have not been thoroughly characterized. Using a conditionally activated ROCK-estrogen receptor fusion protein, we found that ROCK activation is sufficient to stimulate G1/S cell cycle progression in NIH 3T3 mouse fibroblasts. Further analysis revealed that ROCK acts via independent pathways to alter the levels of cell cycle regulatory proteins: cyclin D1 and p21(Cip1) elevation via Ras and the mitogen-activated protein kinase pathway, increased cyclin A via LIM kinase 2, and reduction of p27(Kip1) protein levels. Therefore, the influence of ROCK on cell cycle regulatory proteins occurs by multiple independent mechanisms.


Asunto(s)
Ciclina A/metabolismo , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Adhesiones Focales , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas , Ratones , Modelos Biológicos , Células 3T3 NIH , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Quinasas Asociadas a rho
5.
Mol Cell Biol ; 26(4): 1480-95, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16449658

RESUMEN

The transcription factor AP-1, which is composed of Fos and Jun family proteins, plays an essential role in tumor cell invasion by altering gene expression. We report here that Krp1, the AP-1 up-regulated protein that has a role in pseudopodial elongation in v-Fos-transformed rat fibroblast cells, forms a novel interaction with the nondifferentially expressed actin binding protein Lasp-1. Krp1 and Lasp-1 colocalize with actin at the tips of pseudopodia, and this localization is maintained by continued AP-1 mediated down-regulation of fibronectin that in turn suppresses integrin and Rho-ROCK signaling and allows pseudopodial protrusion and mesenchyme-like invasion. Mutation analysis of Lasp-1 demonstrates that its SH3 domain is necessary for pseudopodial extension and invasion. The results support the concept of an AP-1-regulated multigenic invasion program in which proteins encoded by differentially expressed genes direct the function, localization, and activity of proteins that are not differentially expressed to enhance the invasiveness of cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Fibronectinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Transformación Celular Neoplásica , Proteínas del Citoesqueleto , Fibronectinas/genética , Genes fos , Péptidos y Proteínas de Señalización Intracelular , Mesodermo/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Invasividad Neoplásica , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Seudópodos/metabolismo , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal , Factor de Transcripción AP-1/genética , Quinasas Asociadas a rho
7.
Cancer Res ; 78(22): 6509-6522, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30279244

RESUMEN

Glioblastoma (GBM) is an aggressive and incurable primary brain tumor that causes severe neurologic, cognitive, and psychologic symptoms. Symptoms are caused and exacerbated by the infiltrative properties of GBM cells, which enable them to pervade the healthy brain and disrupt normal function. Recent research has indicated that although radiotherapy (RT) remains the most effective component of multimodality therapy for patients with GBM, it can provoke a more infiltrative phenotype in GBM cells that survive treatment. Here, we demonstrate an essential role of the actin-myosin regulatory kinase myotonic dystrophy kinase-related CDC42-binding kinase (MRCK) in mediating the proinvasive effects of radiation. MRCK-mediated invasion occurred via downstream signaling to effector molecules MYPT1 and MLC2. MRCK was activated by clinically relevant doses per fraction of radiation, and this activation was concomitant with an increase in GBM cell motility and invasion. Furthermore, ablation of MRCK activity either by RNAi or by inhibition with the novel small-molecule inhibitor BDP-9066 prevented radiation-driven increases in motility both in vitro and in a clinically relevant orthotopic xenograft model of GBM. Crucially, treatment with BDP-9066 in combination with RT significantly increased survival in this model and markedly reduced infiltration of the contralateral cerebral hemisphere.Significance: An effective new strategy for the treatment of glioblastoma uses a novel, anti-invasive chemotherapeutic to prevent infiltration of the normal brain by glioblastoma cells.Cancer Res; 78(22); 6509-22. ©2018 AACR.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Actinas/química , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/radioterapia , Miosinas Cardíacas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Femenino , Glioblastoma/radioterapia , Humanos , Ratones , Ratones Desnudos , Microscopía Fluorescente , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Miosinas/química , Invasividad Neoplásica , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
8.
Cancer Res ; 78(8): 2096-2114, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29382705

RESUMEN

The myotonic dystrophy-related Cdc42-binding kinases MRCKα and MRCKß contribute to the regulation of actin-myosin cytoskeleton organization and dynamics, acting in concert with the Rho-associated coiled-coil kinases ROCK1 and ROCK2. The absence of highly potent and selective MRCK inhibitors has resulted in relatively little knowledge of the potential roles of these kinases in cancer. Here, we report the discovery of the azaindole compounds BDP8900 and BDP9066 as potent and selective MRCK inhibitors that reduce substrate phosphorylation, leading to morphologic changes in cancer cells along with inhibition of their motility and invasive character. In over 750 human cancer cell lines tested, BDP8900 and BDP9066 displayed consistent antiproliferative effects with greatest activity in hematologic cancer cells. Mass spectrometry identified MRCKα S1003 as an autophosphorylation site, enabling development of a phosphorylation-sensitive antibody tool to report on MRCKα status in tumor specimens. In a two-stage chemical carcinogenesis model of murine squamous cell carcinoma, topical treatments reduced MRCKα S1003 autophosphorylation and skin papilloma outgrowth. In parallel work, we validated a phospho-selective antibody with the capability to monitor drug pharmacodynamics. Taken together, our findings establish an important oncogenic role for MRCK in cancer, and they offer an initial preclinical proof of concept for MRCK inhibition as a valid therapeutic strategy.Significance: The development of selective small-molecule inhibitors of the Cdc42-binding MRCK kinases reveals their essential roles in cancer cell viability, migration, and invasive character. Cancer Res; 78(8); 2096-114. ©2018 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Descubrimiento de Drogas , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/enzimología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Neoplasias Cutáneas/enzimología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Curr Biol ; 13(24): 2185-9, 2003 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-14680635

RESUMEN

The epidermis comprises multiple layers of specialized epithelial cells called keratinocytes. As cells are lost from the outermost epidermal layers, they are replaced through terminal differentiation, in which keratinocytes of the basal layer cease proliferating, migrate upwards, and eventually reach the outermost cornified layers. Normal homeostasis of the epidermis requires that the balance between proliferation and differentiation be tightly regulated. The GTP binding protein RhoA plays a fundamental role in the regulation of the actin cytoskeleton and in the adhesion events that are critically important to normal tissue homeostasis. Two central mediators of the signals from RhoA are the ROCK serine/threonine kinases ROCK-I and ROCK-II. We have analyzed ROCK's role in the regulation of epidermal keratinocyte function by using a pharmacological inhibitor and expressing conditionally active or inactive forms of ROCK-II in primary human keratinocytes. We report that blocking ROCK function results in inhibition of keratinocyte terminal differentiation and an increase in cell proliferation. In contrast, activation of ROCK-II in keratinocytes results in cell cycle arrest and an increase in the expression of a number of genes associated with terminal differentiation. Thus, these results indicate that ROCK plays a critical role in regulating the balance between proliferation and differentiation in human keratinocytes.


Asunto(s)
Diferenciación Celular/fisiología , Queratinocitos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Amidas/metabolismo , Western Blotting , Ciclo Celular/fisiología , Electroforesis en Gel de Poliacrilamida , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/metabolismo , Quinasas Asociadas a rho
10.
Methods Enzymol ; 406: 541-53, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16472686

RESUMEN

The Rho-associated kinases ROCK I and ROCK II are serine/threonine kinases that play central and critical roles in regulating the actin cytoskeleton. Activation of ROCK proteins contributes positively to the phosphorylation of myosin II light chains (MLC), myosin ATPase activity, stabilization of filamentous actin, and coupling of the actin-myosin filaments to the plasma membrane, thereby leading to the increased actin-myosin force generation and contractility. We have constructed a conditionally-activated form of ROCK II (called ROCK:ER) by fusing the ROCK II kinase domain to the estrogen receptor hormone-binding domain. In this chapter, we describe the construction and characterization of this regulatable ROCK:ER fusion protein.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Bovinos , Línea Celular , Proteínas Fluorescentes Verdes/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Quinasas Asociadas a rho
11.
Cancer Res ; 64(24): 8994-9001, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15604264

RESUMEN

Progression of tumors to invasive and metastatic forms requires that tumor cells undergo dramatic morphologic changes, a process regulated by Rho GTPases. Elevated expression of RhoA and RhoC, as well as the Rho effector proteins ROCK I and ROCK II, are commonly observed in human cancers and are often associated with more invasive and metastatic phenotypes. To examine how ROCK contributes to the progression of solid tumors, we established a conditionally activated form of ROCK II by fusing the kinase domain to the estrogen receptor hormone-binding domain (ROCK:ER). ROCK:ER-expressing colon carcinoma cells grown as tumors in immunocompromised nude mice organized into discrete clusters surrounding blood vessels. However, ROCK:ER activation resulted in the aggressive dissemination of tumor cells into the surrounding stroma, indicating that increased ROCK signaling is sufficient to promote invasion from solid tumors. In addition, tumors in which ROCK:ER was activated were more highly vascularized, indicating that ROCK contributes to tumor angiogenesis. ROCK:ER activation resulted in changes to epithelial morphology and organization that facilitated motility in vitro, likely by inducing the redistribution of proteins such as ezrin, as well as adherens junction and extracellular matrix-binding proteins. These results suggest that ROCK inhibitors would be useful antimetastatic and antiangiogenic chemotherapeutic agents in tumors associated with elevated RhoA, RhoC, ROCK I, or ROCK II expression.


Asunto(s)
Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Tamoxifeno/análogos & derivados , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Citoesqueleto/enzimología , Citoesqueleto/patología , Activación Enzimática , Células HCT116 , Humanos , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tamoxifeno/farmacología , Quinasas Asociadas a rho
12.
Oncotarget ; 6(36): 38469-86, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26540348

RESUMEN

The actin and microtubule cytoskeletons are critically important for cancer cell proliferation, and drugs that target microtubules are widely-used cancer therapies. However, their utility is compromised by toxicities due to dose and exposure. To overcome these issues, we characterized how inhibition of the actin and microtubule cytoskeleton regulatory LIM kinases could be used in drug combinations to increase efficacy. A previously-described LIMK inhibitor (LIMKi) induced dose-dependent microtubule alterations that resulted in significant mitotic defects, and increased the cytotoxic potency of microtubule polymerization inhibitors. By combining LIMKi with 366 compounds from the GSK Published Kinase Inhibitor Set, effective combinations were identified with kinase inhibitors including EGFR, p38 and Raf. These findings encouraged a drug discovery effort that led to development of CRT0105446 and CRT0105950, which potently block LIMK1 and LIMK2 activity in vitro, and inhibit cofilin phosphorylation and increase αTubulin acetylation in cells. CRT0105446 and CRT0105950 were screened against 656 cancer cell lines, and rhabdomyosarcoma, neuroblastoma and kidney cancer cells were identified as significantly sensitive to both LIMK inhibitors. These large-scale screens have identified effective LIMK inhibitor drug combinations and sensitive cancer types. In addition, the LIMK inhibitory compounds CRT0105446 and CRT0105950 will enable further development of LIMK-targeted cancer therapy.


Asunto(s)
Quinasas Lim/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Células MCF-7 , Microtúbulos/metabolismo , Mitosis/fisiología , Neoplasias/enzimología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/enzimología , Neuroblastoma/patología
13.
Biomaterials ; 33(9): 2701-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200536

RESUMEN

The potential of gene therapy to treat cancer is hampered by the lack of safe and efficacious gene delivery systems able to selectively deliver therapeutic genes to tumors by intravenous administration. With the long-term aim of developing an efficacious cancer-targeted gene medicine, we demonstrated that transferrin-bearing polypropylenimine dendrimer complexed to a plasmid DNA encoding p73 led to an enhanced anti-proliferative activity in vitro, by up to 120-fold in A431 compared to the unmodified dendriplex. In vivo, the intravenous administration of this p73-encoding dendriplex resulted in a rapid and sustained inhibition of tumor growth over one month, with complete tumor suppression for 10% of A431 and B16-F10 tumors and long-term survival of the animals. The treatment was well tolerated by the animals, with no apparent signs of toxicity. These results suggest that the p73-encoding tumor-targeted polypropylenimine dendrimer should be further explored as a therapeutic strategy for cancer therapy.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/uso terapéutico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Melanoma Experimental/patología , Melanoma Experimental/terapia , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapéutico , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/uso terapéutico , Aminobutiratos/química , Animales , Línea Celular Tumoral , Proliferación Celular , ADN/metabolismo , Dendrímeros/administración & dosificación , Dendrímeros/química , Diagnóstico por Imagen , Ensayo de Cambio de Movilidad Electroforética , Femenino , Humanos , Inyecciones Intravenosas , Mediciones Luminiscentes , Ratones , Ratones Endogámicos BALB C , Inducción de Remisión , Transfección , Transferrina/metabolismo , Proteína Tumoral p73 , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Transcription ; 2(5): 211-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22231116

RESUMEN

Signaling through the Rho family of small GTPases regulates a variety of cellular processes via changes in the actin cytoskeleton. Here we discuss recent findings that show the transcription factor p53 regulates the expression of several Rho pathway signaling molecules, and how mutation of p53 in cancer dramatically alters signaling output through this pathway.


Asunto(s)
Transducción de Señal , Transcripción Genética , Proteínas de Unión al GTP rho/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Apoptosis , Citoesqueleto/metabolismo , Daño del ADN , Humanos , Quinasas Lim/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Proteína rhoC de Unión a GTP
15.
Autophagy ; 7(11): 1295-301, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21808150

RESUMEN

(Macro)autophagy is a membrane-trafficking process that serves to sequester cellular constituents in organelles termed autophagosomes, which target their degradation in the lysosome. Autophagy operates at basal levels in all cells where it serves as a homeostatic mechanism to maintain cellular integrity. The levels and cargoes of autophagy can, however, change in response to a variety of stimuli, and perturbations in autophagy are known to be involved in the aetiology of various human diseases. Autophagy must therefore be tightly controlled. We report here that the Drosophila cyclin-dependent kinase PITSLRE is a modulator of autophagy. Loss of the human PITSLRE orthologue, CDK11, initially appears to induce autophagy, but at later time points CDK11 is critically required for autophagic flux and cargo digestion. Since PITSLRE/CDK11 regulates autophagy in both Drosophila and human cells, this kinase represents a novel phylogenetically conserved component of the autophagy machinery.


Asunto(s)
Autofagia , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Animales , Línea Celular Tumoral , Humanos , Lisosomas/metabolismo , Fagosomas/metabolismo , Homología de Secuencia de Aminoácido
16.
Cell Res ; 21(4): 666-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21079653

RESUMEN

The central arbiter of cell fate in response to DNA damage is p53, which regulates the expression of genes involved in cell cycle arrest, survival and apoptosis. Although many responses initiated by DNA damage have been characterized, the role of actin cytoskeleton regulators is largely unknown. We now show that RhoC and LIM kinase 2 (LIMK2) are direct p53 target genes induced by genotoxic agents. Although RhoC and LIMK2 have well-established roles in actin cytoskeleton regulation, our results indicate that activation of LIMK2 also has a pro-survival function following DNA damage. LIMK inhibition by siRNA-mediated knockdown or selective pharmacological blockade sensitized cells to radio- or chemotherapy, such that treatments that were sub-lethal when administered singly resulted in cell death when combined with LIMK inhibition. Our findings suggest that combining LIMK inhibitors with genotoxic therapies could be more efficacious than single-agent administration, and highlight a novel connection between actin cytoskeleton regulators and DNA damage-induced cell survival mechanisms.


Asunto(s)
Actinas/metabolismo , Regulación de la Expresión Génica , Quinasas Lim/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Citoesqueleto , Daño del ADN , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Ratones , Análisis por Micromatrices , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Quinasas Asociadas a rho/metabolismo , Proteína rhoC de Unión a GTP
17.
Cancer Cell ; 19(6): 776-91, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21665151

RESUMEN

Tumors and associated stroma manifest mechanical properties that promote cancer. Mechanosensation of tissue stiffness activates the Rho/ROCK pathway to increase actomyosin-mediated cellular tension to re-establish force equilibrium. To determine how actomyosin tension affects tissue homeostasis and tumor development, we expressed conditionally active ROCK2 in mouse skin. ROCK activation elevated tissue stiffness via increased collagen. ß-catenin, a key element of mechanotranscription pathways, was stabilized by ROCK activation leading to nuclear accumulation, transcriptional activation, and consequent hyperproliferation and skin thickening. Inhibiting actomyosin contractility by blocking LIMK or myosin ATPase attenuated these responses, as did FAK inhibition. Tumor number, growth, and progression were increased by ROCK activation, while ROCK blockade was inhibitory, implicating actomyosin-mediated cellular tension and consequent collagen deposition as significant tumor promoters.


Asunto(s)
Actomiosina/fisiología , Epidermis/patología , Neoplasias Cutáneas/etiología , beta Catenina/fisiología , Animales , Fenómenos Biomecánicos , Proliferación Celular , Células Cultivadas , Humanos , Hiperplasia , Ratones , Papiloma/etiología , Transducción de Señal , Quinasas Asociadas a rho/análisis , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/fisiología
18.
J Cell Biol ; 191(1): 169-85, 2010 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-20876278

RESUMEN

LIM kinases 1 and 2 (LIMK1/2) are centrally positioned regulators of actin cytoskeleton dynamics. Using siRNA-mediated knockdown or a novel small molecule inhibitor, we show LIMK is required for path generation by leading tumor cells and nontumor stromal cells during collective tumor cell invasion. LIMK inhibition lowers cofilin phosphorylation, F-actin levels, serum response factor transcriptional activity and collagen contraction, and reduces invasion in three-dimensional invasion assays. Although motility was unaffected, LIMK inhibition impairs matrix protein degradation and invadopodia formation associated with significantly faster recovery times in FRAP assays indicative of reduced F-actin stability. When LIMK is knocked down in MDA-MB-231 cells, they lose the ability to lead strands of collectively invading cells. Similarly, when LIMK activity is blocked in cancer-associated fibroblasts, they are unable to lead the collective invasion of squamous carcinoma cells in an organotypic skin model. These results show that LIMK is required for matrix remodeling activities for path generation by leading cells in collective invasion.


Asunto(s)
Quinasas Lim/fisiología , Invasividad Neoplásica , Células del Estroma/enzimología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Humanos , Quinasas Lim/antagonistas & inhibidores , Fosforilación , Estabilidad Proteica , Interferencia de ARN
19.
Cancer Cell ; 14(5): 349-51, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18977323

RESUMEN

Switching between elongated and rounded modes of movement allows invasive tumor cells to adapt to varying microenvironments. In a recent issue of Cell, Sanz-Moreno et al. identify DOCK3, NEDD9, WAVE2, and ARHGAP22 as key molecules regulating Rac and Rho signaling that determine the mode of movement driving melanoma cell metastasis.


Asunto(s)
Movimiento Celular/fisiología , Melanoma/patología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/secundario , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA