Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 291(4): 1605-1618, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26589794

RESUMEN

Cancer cells can invade in three-dimensional collagen as single cells or as a cohesive group of cells that require coordination of cell-cell junctions and the actin cytoskeleton. To examine the role of Gα13, a G12 family heterotrimeric G protein, in regulating cellular invasion in three-dimensional collagen, we established a novel method to track cell invasion by membrane type 1 matrix metalloproteinase-expressing cancer cells. We show that knockdown of Gα13 decreased membrane type 1 matrix metalloproteinase-driven proteolytic invasion in three-dimensional collagen and enhanced E-cadherin-mediated cell-cell adhesion. E-cadherin knockdown reversed Gα13 siRNA-induced cell-cell adhesion but failed to reverse the effect of Gα13 siRNA on proteolytic invasion. Instead, concurrent knockdown of E-cadherin and Gα13 led to an increased number of single cells rather than groups of cells. Significantly, knockdown of discoidin domain receptor 1 (DDR1), a collagen-binding protein that also co-localizes to cell-cell junctions, reversed the effects of Gα13 knockdown on cell-cell adhesion and proteolytic invasion in three-dimensional collagen. Knockdown of the polarity protein Par3, which can function downstream of DDR1, also reversed the effects of Gα13 knockdown on cell-cell adhesion and proteolytic invasion in three-dimensional collagen. Overall, we show that Gα13 and DDR1-Par3 differentially regulate cell-cell junctions and the actin cytoskeleton to mediate invasion in three-dimensional collagen.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Colágeno/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Cadherinas/metabolismo , Adhesión Celular , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular , Receptor con Dominio Discoidina 1 , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Invasividad Neoplásica , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/fisiopatología , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal
2.
Infect Immun ; 80(7): 2382-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22526672

RESUMEN

The Staphylococcus aureus global regulator CodY responds to nutrient availability by controlling the expression of target genes. In vitro, CodY represses the transcription of virulence genes, but it is not known if CodY also represses virulence in vivo. The dominant community-associated methicillin-resistant S. aureus (CA-MRSA) clone, USA300, is hypervirulent and has increased transcription of global regulators and virulence genes; these features are reminiscent of a strain defective in CodY. Sequence analysis revealed, however, that the codY genes of USA300 and other sequenced S. aureus isolates are not significantly different from the codY genes in strains known to have active CodY. codY was expressed in USA300, as well as in other pulsotypes assessed. Deletion of codY from a USA300 clinical isolate resulted in modestly increased expression of the global regulators agr and saeRS, as well as the gene encoding the toxin alpha-hemolysin (hla). A substantial increase (>30-fold) in expression of the lukF-PV gene, encoding part of the Panton-Valentine leukocidin (PVL), was observed in the codY mutant. All of these expression differences were reversed by complementation with a functional codY gene. Moreover, purified CodY protein bound upstream of the lukSF-PV operon, indicating that CodY directly represses expression of lukSF-PV. Deletion of codY increased the virulence of USA300 in necrotizing pneumonia and skin infection. Interestingly, deletion of lukSF-PV from the codY mutant did not attenuate virulence, indicating that the hypervirulence of the codY mutant was not explained by overexpression of PVL. These results demonstrate that CodY is active in USA300 and that CodY-mediated repression restrains the virulence of USA300.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/patogenicidad , Proteínas Represoras/deficiencia , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Toxinas Bacterianas/biosíntesis , ADN Bacteriano/metabolismo , Exotoxinas/biosíntesis , Eliminación de Gen , Prueba de Complementación Genética , Proteínas Hemolisinas/biosíntesis , Leucocidinas/biosíntesis , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteínas Quinasas/biosíntesis , Proteínas Represoras/genética , Transactivadores/biosíntesis , Factores de Transcripción , Virulencia , Factores de Virulencia/biosíntesis
3.
Dev Biol ; 314(1): 224-35, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18177852

RESUMEN

In the early stages of lung development, the endoderm undergoes extensive and stereotypic branching morphogenesis. During this process, a simple epithelial bud develops into a complex tree-like system of tubes specialized for the transport and exchange of gas with blood. The endodermal cells in the distal tips of the developing lung express a special set of genes, have a higher proliferation rate than proximal part, undergo shape change and initiate branching morphogenesis. In this study, we found that of the four p38 genes, only p38 alpha mRNA is localized specifically to the distal endoderm suggesting a role in the regulation of budding morphogenesis. Chemical inhibitors specific for the p38 alpha and p38 beta isoforms suppress budding of embryonic mouse lung explants and isolated endoderm in vitro. Specific knockdown of p38 alpha in cultured lung endoderm using shRNA also inhibited budding morphogenesis, consistent with the chemical inhibition of the p38 signaling pathway. Disruption of p38 alpha did not affect proliferation or expression of the distal cell markers, Sox9 and Erm. However, the amount of E-cadherin protein increased significantly and ectopic expression of E-cadherin also impaired budding of endoderm in vitro. These results suggest that p38 alpha modulates epithelial cell-cell interactions and possibly cell rearrangement during branching morphogenesis. This study provides the first evidence that p38 alpha is involved in the morphogenesis of an epithelial organ.


Asunto(s)
Pulmón/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Adhesión Celular/fisiología , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Endodermo/embriología , Epitelio/embriología , Epitelio/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Pulmón/embriología , Ratones , Ratones Endogámicos ICR , Morfogénesis , Isoformas de Proteínas/fisiología , Factor de Transcripción SOX9 , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
4.
Methods Mol Biol ; 1882: 161-169, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30378053

RESUMEN

Since many tumors are associated with a pronounced collagen-rich stromal reaction, there is increasing interest in understanding mechanisms by which cancer cells invade through the collagen barrier. Here we describe a quantitative method to track cell invasion in 3D collagen I gels. We analyze invasion by quantifying proteolytic tracks generated by invading cancer cells through a 3D collagen microenvironment. We provide a detailed protocol for this quantitative assay, which can be used to characterize signaling pathways that regulate invasion in the 3D microenvironment.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Técnicas de Cultivo de Célula/métodos , Colágeno Tipo I/metabolismo , Neoplasias Pancreáticas/patología , Técnicas de Cultivo de Célula/instrumentación , Línea Celular Tumoral , Movimiento Celular , Matriz Extracelular/metabolismo , Geles , Humanos , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Invasividad Neoplásica/patología , Proteolisis , ARN Interferente Pequeño/metabolismo
5.
Mol Cancer Ther ; 18(2): 235-244, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30446586

RESUMEN

BET inhibitors (BETi), which target transcription of key oncogenic genes, are currently being evaluated in early-phase clinical trials. However, because BETis show limited single-agent activity, there is increasing interest in identifying signaling pathways to enhance the efficacy of BETis. Here, we demonstrate increased MNK kinase-dependent eIF4E phosphorylation following treatment with BETis, indicating activation of a prosurvival feedback mechanism in response to BETis. BET PROTACs, which promote degradation of BET proteins, also induced eIF4E phosphorylation in cancer cells. Mechanistically, we show that the effect of BETis on MNK-eIF4E phosphorylation was mediated by p38 MAPKs. We also show that BETis suppressed RacGAP1 to induce Rac signaling-mediated eIF4E phosphorylation. Significantly, MNK inhibitors and MNK1/2 knockdown enhanced the efficacy of BETis in suppressing proliferation of cancer cells in vitro and in a syngeneic mouse model. Together, these results demonstrate a novel prosurvival feedback signaling induced by BETis, providing a mechanistic rationale for combination therapy with BET and MNK inhibitors for synergistic inhibition of cancer cells.


Asunto(s)
Acetanilidas/administración & dosificación , Compuestos de Anilina/administración & dosificación , Azepinas/administración & dosificación , Factor 4E Eucariótico de Iniciación/metabolismo , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Proteínas Serina-Treonina Quinasas/metabolismo , Purinas/administración & dosificación , Neoplasias de la Tiroides/tratamiento farmacológico , Triazoles/administración & dosificación , Acetanilidas/farmacología , Compuestos de Anilina/farmacología , Animales , Azepinas/farmacología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Factor 4E Eucariótico de Iniciación/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Ratones , Fosforilación/efectos de los fármacos , Purinas/farmacología , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cell Biol ; 38(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29866656

RESUMEN

Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Interferón Tipo I/metabolismo , FN-kappa B/metabolismo , Animales , Sitios de Unión/genética , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Fosfoproteínas Fosfatasas/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal
7.
Sci Rep ; 8(1): 13225, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185888

RESUMEN

The fibrotic reaction is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of fibrosis in vivo. While there is increasing interest in the regulation of PD-L1 expression in cancer and immune cells, the expression and regulation of PD-L1 in other stromal cells, such as PSCs, has not been fully evaluated. Here we show that PSCs in vitro express higher PD-L1 mRNA and protein levels compared to the levels present in PDAC cells. We show that inhibitors targeting bromodomain and extra-terminal (BET) proteins and BRD4 knockdown decrease interferon-γ (IFN-γ)-induced PD-L1 expression in PSCs. We also show that c-MYC, one of the well-established targets of BET inhibitors, does not mediate IFN-γ-regulated PD-L1 expression in PSCs. Instead we show that interferon regulatory factor 1 (IRF1) mediates IFN-γ-induced PD-L1 expression in PSCs. Finally, while we show that BET inhibitors do not regulate IFN-γ-induced IRF1 expression in PSCs, BET inhibitors decrease binding of IRF1 and BRD4 to the PD-L1 promoter. Together, these results demonstrate the interplay between IRF1 and BRD4 in the regulation of PD-L1 in PSCs.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Factores de Transcripción/metabolismo , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Factores de Transcripción/genética , Neoplasias Pancreáticas
8.
JCI Insight ; 2(3): e88032, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28194432

RESUMEN

The fibrotic reaction, which can account for over 70%-80% of the tumor mass, is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of collagen I production and fibrosis in vivo. In this report, we show that members of the bromodomain and extraterminal (BET) family of proteins are expressed in primary PSCs isolated from human PDAC tumors, with BRD4 positively regulating, and BRD2 and BRD3 negatively regulating, collagen I expression in primary cancer-associated PSCs. We show that the inhibitory effect of pan-BET inhibitors on collagen I expression in primary cancer-associated PSCs is through blocking of BRD4 function. Importantly, we show that FOSL1 is repressed by BRD4 in primary cancer-associated PSCs and negatively regulates collagen I expression. While BET inhibitors do not affect viability or induce PSC apoptosis or senescence, BET inhibitors induce primary cancer-associated PSCs to become quiescent. Finally, we show that BET inhibitors attenuate stellate cell activation, fibrosis, and collagen I production in the EL-KrasG12D transgenic mouse model of pancreatic tumorigenesis. Our results demonstrate that BET inhibitors regulate fibrosis by modulating the activation and function of cancer-associated PSCs.


Asunto(s)
Azepinas/administración & dosificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Colágeno Tipo I/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Células Estrelladas Pancreáticas/efectos de los fármacos , Factores de Transcripción/metabolismo , Triazoles/administración & dosificación , Animales , Azepinas/farmacología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Colágeno Tipo I/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/citología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
9.
Sci Rep ; 6: 28260, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27301426

RESUMEN

Although the translational function of tRNA has long been established, extra translational functions of tRNA are still being discovered. We previously developed a computational method to systematically predict new tRNA-protein complexes and experimentally validated six candidate proteins, including the mitogen-activated protein kinase kinase 2 (MEK2), that interact with tRNA in HEK293T cells. However, consequences of the interaction between tRNA and these proteins remain to be elucidated. Here we tested the consequence of the interaction between tRNA and MEK2 in pancreatic cancer cell lines. We also generated disease and drug resistance-derived MEK2 mutants (Q60P, P128Q, S154F, E207K) to evaluate the function of the tRNA-MEK2 interaction. Our results demonstrate that tRNA interacts with the wild-type and mutant MEK2 in pancreatic cancer cells; furthermore, the MEK2 inhibitor U0126 significantly reduces the tRNA-MEK2 interaction. In addition, tRNA affects the catalytic activity of the wild type and mutant MEK2 proteins in different ways. Overall, our findings demonstrate the interaction of tRNA with MEK2 in pancreatic cancer cells and suggest that tRNA may impact MEK2 activity in cancer cells.


Asunto(s)
MAP Quinasa Quinasa 2/metabolismo , Neoplasias Pancreáticas/metabolismo , ARN de Transferencia/metabolismo , Butadienos/farmacología , Línea Celular Tumoral , Células HEK293 , Humanos , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , Mutación , Nitrilos/farmacología , Neoplasias Pancreáticas/patología
10.
Mol Cancer Res ; 14(2): 216-27, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26609108

RESUMEN

UNLABELLED: Human pancreatic ductal adenocarcinoma (PDAC) tumors are associated with dysregulation of mRNA translation. In this report, it is demonstrated that PDAC cells grown in collagen exhibit increased activation of the MAPK-interacting protein kinases (MNK) that mediate eIF4E phosphorylation. Pharmacologic and genetic targeting of MNKs reverse epithelial-mesenchymal transition (EMT), decrease cell migration, and reduce protein expression of the EMT-regulator ZEB1 without affecting ZEB1 mRNA levels. Paradoxically, targeting eIF4E, the best-characterized effector of MNKs, increases ZEB1 mRNA expression through repression of ZEB1-targeting miRNAs, miR-200c and miR-141. In contrast, targeting the MNK effector hnRNPA1, which can function as a translational repressor, increases ZEB1 protein without increasing ZEB1 mRNA levels. Importantly, treatment with MNK inhibitors blocks growth of chemoresistant PDAC cells in collagen and decreases the number of aldehyde dehydrogenase activity-positive (Aldefluor+) cells. Significantly, MNK inhibitors increase E-cadherin mRNA levels and decrease vimentin mRNA levels in human PDAC organoids without affecting ZEB1 mRNA levels. Importantly, MNK inhibitors also decrease growth of human PDAC organoids. IMPLICATIONS: These results demonstrate differential regulation of ZEB1 and EMT by MNKs and eIF4E, and identify MNKs as potential targets in pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Transición Epitelial-Mesenquimal , Factor 4E Eucariótico de Iniciación/metabolismo , Proteínas de Homeodominio/genética , Neoplasias Pancreáticas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Carcinoma Ductal Pancreático/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Neoplasias Pancreáticas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
11.
Sci Rep ; 6: 29133, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27364947

RESUMEN

Cells in the pancreas that have undergone acinar-ductal metaplasia (ADM) can transform into premalignant cells that can eventually become cancerous. Although the epithelial-mesenchymal transition regulator Snail (Snai1) can cooperate with Kras in acinar cells to enhance ADM development, the contribution of Snail-related protein Slug (Snai2) to ADM development is not known. Thus, transgenic mice expressing Slug and Kras in acinar cells were generated. Surprisingly, Slug attenuated Kras-induced ADM development, ERK1/2 phosphorylation and proliferation. Co-expression of Slug with Kras also attenuated chronic pancreatitis-induced changes in ADM development and fibrosis. In addition, Slug attenuated TGF-α-induced acinar cell metaplasia to ductal structures and TGF-α-induced expression of ductal markers in ex vivo acinar explant cultures. Significantly, blocking the Rho-associated protein kinase ROCK1/2 in the ex vivo cultures induced expression of ductal markers and reversed the effects of Slug by inducing ductal structures. In addition, blocking ROCK1/2 activity in Slug-expressing Kras mice reversed the inhibitory effects of Slug on ADM, ERK1/2 phosphorylation, proliferation and fibrosis. Overall, these results increase our understanding of the role of Slug in ADM, an early event that can eventually lead to pancreatic cancer development.


Asunto(s)
Metaplasia/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción de la Familia Snail/genética , Células Acinares/patología , Animales , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Humanos , Metaplasia/patología , Ratones , Ratones Transgénicos , Conductos Pancreáticos/metabolismo , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Pancreatitis Crónica/genética , Pancreatitis Crónica/patología , Transducción de Señal/genética
12.
Sci Rep ; 5: 9489, 2015 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-25807524

RESUMEN

JQ1 and I-BET151 are selective inhibitors of BET bromodomain proteins that have efficacy against a number of different cancers. Since the effectiveness of targeted therapies is often limited by development of resistance, we examined whether it was possible for cancer cells to develop resistance to the BET inhibitor JQ1. Here we show that pancreatic cancer cells developing resistance to JQ1 demonstrate cross-resistance to I-BET151 and insensitivity to BRD4 downregulation. The resistant cells maintain expression of c-MYC, increase expression of JQ1-target genes FOSL1 and HMGA2, and demonstrate evidence of epithelial-mesenchymal transition (EMT). However, reverting EMT fails to sensitize the resistant cells to JQ1 treatment. Importantly, the JQ1-resistant cells remain dependent on c-MYC that now becomes co-regulated by high levels of GLI2. Furthermore, downregulating GLI2 re-sensitizes the resistant cells to JQ1. Overall, these results identify a mechanism by which cancer cells develop resistance to BET inhibitors.


Asunto(s)
Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Apoptosis/efectos de los fármacos , Azepinas/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Factores de Transcripción de Tipo Kruppel/biosíntesis , Proteínas Nucleares/biosíntesis , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Triazoles/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Gli2 con Dedos de Zinc
13.
Mol Cancer Ther ; 13(7): 1907-17, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807963

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is associated with pronounced fibrosis that contributes to chemoresistance, in part, through increased histone acetylation. Because bromodomain (BRD) and extra terminal domain (BET) proteins are "readers" of histone acetylation marks, we targeted BET proteins in PDAC cells grown in three-dimensional collagen. We show that treatment with BET inhibitors decreases growth of PDAC cells (AsPC1, CD18, and Panc1) in collagen. Transfection with siRNA against BRD4, which is increased in human PDAC tumors, also decreases growth of PDAC cells. BET inhibitors additionally decrease growth in collagen of PDAC cells that have undergone epithelial-to-mesenchymal transition or have become resistant to chemotherapy. Although BET inhibitors and BRD4 siRNA repress c-MYC only in AsPC1 and CD18 cells, downregulating c-MYC decreases growth of all three PDAC cell lines in collagen. FOSL1, which is also targeted by BET inhibitors and BRD4 siRNA in AsPC1, CD18, and Panc1 cells, additionally regulates growth of all three PDAC cell lines in collagen. BET inhibitors and BRD4 siRNA repress HMGA2, an architectural protein that modulates chromatin state and also contributes to chemoresistance, in PDAC cells grown in collagen. Importantly, we show that there is a statistically significant correlation between BRD4 and HMGA2 in human PDAC tumors. Significantly, overexpression of HMGA2 partially mitigates the effect of BET inhibitors on growth and c-MYC and/or FOSL1 expression in collagen. Overall, these results demonstrate that BET inhibitors block growth of PDAC cells in collagen and that BET proteins may be potential targets for the treatment of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Factores de Transcripción/antagonistas & inhibidores , Carcinoma Ductal Pancreático/genética , Proteínas de Ciclo Celular , Colágeno , Transición Epitelial-Mesenquimal , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Factores de Transcripción/genética , Transfección , Células Tumorales Cultivadas
14.
Mol Cancer Res ; 12(10): 1440-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24944064

RESUMEN

UNLABELLED: Pancreatic ductal adenocarcinoma (PDAC) is associated with a pronounced fibro-inflammatory stromal reaction that contributes to tumor progression. A critical step in invasion and metastasis is the epithelial-to-mesenchymal transition (EMT), which can be regulated by the Snail family of transcription factors. Overexpression of Snail (Snai1) and mutant Kras(G12D) in the pancreas of transgenic mice, using an elastase (EL) promoter, resulted in fibrosis. To identify how Snail modulates inflammation in the pancreas, we examined the effect of expressing Snail in EL-Kras(G12D) mice (Kras(G12D)/Snail) on mast cell infiltration, which has been linked to PDAC progression. Using this animal model system, it was demonstrated that there are increased numbers of mast cells in the pancreas of Kras(G12D)/Snail mice compared with control Kras(G12D) mice. In addition, it was revealed that human primary PDAC tumors with increased Snail expression are associated with increased mast cell infiltration, and that Snail expression in these clinical specimens positively correlated with the expression of stem cell factor (SCF/KITLG), a cytokine known to regulate mast cell migration. Concomitantly, SCF levels are increased in the Kras(G12D)/Snail mice than in control mice. Moreover, overexpression of Snail in PDAC cells increased SCF levels, and the media conditioned by Snail-expressing PDAC cells promoted mast cell migration. Finally, inhibition of SCF using a neutralizing antibody significantly attenuated Snail-induced migration of mast cells. IMPLICATIONS: Together, these results elucidate how the EMT regulator Snail contributes to inflammation associated with PDAC tumors.


Asunto(s)
Mastocitos/patología , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Células Madre/metabolismo , Factores de Transcripción/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Movimiento Celular , Humanos , Inflamación/patología , Ratones , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción de la Familia Snail , Triptasas/metabolismo
15.
PLoS One ; 8(5): e64566, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696899

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is associated with a pronounced collagen-rich stromal reaction that has been shown to contribute to chemo-resistance. We have previously shown that PDAC cells are resistant to gemcitabine chemotherapy in the collagen microenvironment because of increased expression of the chromatin remodeling protein high mobility group A2 (HMGA2). We have now found that human PDAC tumors display higher levels of histone H3K9 and H3K27 acetylation in fibrotic regions. We show that relative to cells grown on tissue culture plastic, PDAC cells grown in three-dimensional collagen gels demonstrate increased histone H3K9 and H3K27 acetylation, along with increased expression of p300, PCAF and GCN5 histone acetyltransferases (HATs). Knocking down HMGA2 attenuates the effect of collagen on histone H3K9 and H3K27 acetylation and on collagen-induced p300, PCAF and GCN5 expression. We also show that human PDAC tumors with HMGA2 demonstrate increased histone H3K9 and H3K27 acetylation. Additionally, we show that cells in three-dimensional collagen gels demonstrate increased protection against gemcitabine. Significantly, down-regulation of HMGA2 or p300, PCAF and GCN5 HATs sensitizes the cells to gemcitabine in three-dimensional collagen. Overall, our results increase our understanding of how the collagen microenvironment contributes to chemo-resistance in vitro and identify HATs as potential therapeutic targets against this deadly cancer.


Asunto(s)
Colágeno Tipo I/farmacología , Desoxicitidina/análogos & derivados , Proteína HMGA2/metabolismo , Histona Acetiltransferasas/metabolismo , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/metabolismo , Línea Celular Tumoral , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Proteína HMGA2/genética , Histona Acetiltransferasas/genética , Humanos , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo , Gemcitabina
16.
Mol Cancer Res ; 11(9): 1078-87, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23761168

RESUMEN

UNLABELLED: Patients with pancreatic cancer, which is characterized by an extensive collagen-rich fibrotic reaction, often present with metastases. A critical step in cancer metastasis is epithelial-to-mesenchymal transition (EMT), which can be orchestrated by the Snail family of transcription factors. To understand the role of Snail (SNAI1) in pancreatic cancer development, we generated transgenic mice expressing Snail in the pancreas. Because chronic pancreatitis can contribute to pancreatic cancer development, Snail-expressing mice were treated with cerulein to induce pancreatitis. Although significant tissue injury was observed, a minimal difference in pancreatitis was seen between control and Snail-expressing mice. However, because Kras mutation is necessary for tumor development in mouse models of pancreatic cancer, we generated mice expressing both mutant Kras(G12D) and Snail (Kras(+)/Snail(+)). Compared with control mice (Kras(+)/Snai(-)), Kras(+)/Snail(+) mice developed acinar ectasia and more advanced acinar-to-ductal metaplasia. The Kras(+)/Snail(+) mice exhibited increased fibrosis, increased phosphorylated Smad2, increased TGF-ß2 expression, and activation of pancreatic stellate cells. To further understand the mechanism by which Snail promoted fibrosis, we established an in vitro model to examine the effect of Snail expression in pancreatic cancer cells on stellate cell collagen production. Snail expression in pancreatic cancer cells increased TGF-ß2 levels, and conditioned media from Snail-expressing pancreatic cancer cells increased collagen production by stellate cells. Additionally, inhibiting TGF-ß signaling in stellate cells attenuated the conditioned media-induced collagen production by stellate cells. Together, these results suggest that Snail contributes to pancreatic tumor development by promoting fibrotic reaction through increased TGF-ß signaling. IMPLICATIONS: Expression of the EMT regulator Snail in the context of mutant Kras provides new insight into pancreatic cancer progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Ceruletida , Colágeno/metabolismo , Fibrosis/metabolismo , Ratones , Ratones Transgénicos , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Pancreatitis Crónica/inducido químicamente , Pancreatitis Crónica/metabolismo , Pancreatitis Crónica/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Proteína Smad2/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta2/metabolismo , Células Tumorales Cultivadas
17.
EMBO J ; 25(12): 2867-77, 2006 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-16710293

RESUMEN

Phosphorylation of neural proteins in response to a diverse array of external stimuli is one of the main mechanisms underlying dynamic changes in neural circuitry. The NR2B subunit of the NMDA receptor is tyrosine-phosphorylated in the brain, with Tyr-1472 its major phosphorylation site. Here, we generate mice with a knockin mutation of the Tyr-1472 site to phenylalanine (Y1472F) and show that Tyr-1472 phosphorylation is essential for fear learning and amygdaloid synaptic plasticity. The knockin mice show impaired fear-related learning and reduced amygdaloid long-term potentiation. NMDA receptor-mediated CaMKII signaling is impaired in YF/YF mice. Electron microscopic analyses reveal that the Y1472F mutant of the NR2B subunit shows improper localization at synapses in the amygdala. We thus identify Tyr-1472 phosphorylation as a key mediator of fear learning and amygdaloid synaptic plasticity.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico , Miedo/fisiología , Plasticidad Neuronal , Fosfotirosina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/ultraestructura , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Aprendizaje/fisiología , Ratones , Mutación/genética , Fosforilación , Transporte de Proteínas , Receptores de N-Metil-D-Aspartato/ultraestructura , Transmisión Sináptica , Tetania
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA