Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 7791, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38057326

RESUMEN

Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Ratones , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Páncreas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Fibroblastos/metabolismo , Carcinogénesis/patología , Microambiente Tumoral
2.
bioRxiv ; 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37745372

RESUMEN

Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.

3.
Nat Commun ; 14(1): 5195, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37673892

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy in need of new therapeutic options. Using unbiased analyses of super-enhancers (SEs) as sentinels of core genes involved in cell-specific function, here we uncover a druggable SE-mediated RNA-binding protein (RBP) cascade that supports PDAC growth through enhanced mRNA translation. This cascade is driven by a SE associated with the RBP heterogeneous nuclear ribonucleoprotein F, which stabilizes protein arginine methyltransferase 1 (PRMT1) to, in turn, control the translational mediator ubiquitin-associated protein 2-like. All three of these genes and the regulatory SE are essential for PDAC growth and coordinately regulated by the Myc oncogene. In line with this, modulation of the RBP network by PRMT1 inhibition reveals a unique vulnerability in Myc-high PDAC patient organoids and markedly reduces tumor growth in male mice. Our study highlights a functional link between epigenetic regulation and mRNA translation and identifies components that comprise unexpected therapeutic targets for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Masculino , Animales , Ratones , ARN , Epigénesis Genética , Secuencias Reguladoras de Ácidos Nucleicos , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Metiltransferasas , Proteínas de Unión al ARN/genética
4.
Proc Natl Acad Sci U S A ; 119(51): e2213041119, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36508655

RESUMEN

The pleiotropic actions of the Farnesoid X Receptor (FXR) are required for gut health, and reciprocally, reduced intestinal FXR signaling is seen in inflammatory bowel diseases (IBDs). Here, we show that activation of FXR selectively in the intestine is protective in inflammation-driven models of IBD. Prophylactic activation of FXR restored homeostatic levels of pro-inflammatory cytokines, most notably IL17. Importantly, these changes were attributed to FXR regulation of innate lymphoid cells (ILCs), with both the inflammation-driven increases in ILCs, and ILC3s in particular, and the induction of Il17a and Il17f in ILC3s blocked by FXR activation. Moreover, a population of ILC precursor-like cells increased with treatment, implicating FXR in the maturation/differentiation of ILC precursors. These findings identify FXR as an intrinsic regulator of intestinal ILCs and a potential therapeutic target in inflammatory intestinal diseases.


Asunto(s)
Inmunidad Innata , Enfermedades Inflamatorias del Intestino , Humanos , Linfocitos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Citocinas , Inflamación
5.
J Addict Nurs ; 28(3): 124-130, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28863053

RESUMEN

Stigma and insufficient training for addressing alcohol or drug problems in clinical settings are two significant barriers to earlier identification and access to care for patients with alcohol or drug (AOD)-related problems. Nurses are crucial players in this strategy, yet nurses often report poor motivation and insufficient training for working with AOD-affected patients. This article describes and reports preliminary evaluation results of an educational experience that integrated AOD curriculum into a community/public health nursing clinical practicum. The purpose of the intervention was to meet the required objectives for our university's community/public health nursing clinical course, improve students' attitudes and therapeutic commitment for working with AOD-affected patients, and prepare students for providing high-quality nursing care for individuals with AOD-related problems.


Asunto(s)
Actitud del Personal de Salud , Curriculum , Estudiantes de Enfermería , Trastornos Relacionados con Sustancias/enfermería , Adolescente , Adulto , Bachillerato en Enfermería/métodos , Femenino , Humanos , Masculino , Evaluación de Programas y Proyectos de Salud , Encuestas y Cuestionarios , Texas , Adulto Joven
6.
Nat Rev Cancer ; 17(5): 332, 2017 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-28436470
7.
Proc Natl Acad Sci U S A ; 114(5): 1129-1134, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096419

RESUMEN

A fibroinflammatory stromal reaction cooperates with oncogenic signaling to influence pancreatic ductal adenocarcinoma (PDAC) initiation, progression, and therapeutic outcome, yet the mechanistic underpinning of this crosstalk remains poorly understood. Here we show that stromal cues elicit an adaptive response in the cancer cell including the rapid mobilization of a transcriptional network implicated in accelerated growth, along with anabolic changes of an altered metabolome. The close overlap of stroma-induced changes in vitro with those previously shown to be regulated by oncogenic Kras in vivo suggests that oncogenic Kras signaling-a hallmark and key driver of PDAC-is contingent on stromal inputs. Mechanistically, stroma-activated cancer cells show widespread increases in histone acetylation at transcriptionally enhanced genes, implicating the PDAC epigenome as a presumptive point of convergence between these pathways and a potential therapeutic target. Notably, inhibition of the bromodomain and extraterminal (BET) family of epigenetic readers, and of Bromodomain-containing protein 2 (BRD2) in particular, blocks stroma-inducible transcriptional regulation in vitro and tumor progression in vivo. Our work suggests the existence of a molecular "AND-gate" such that tumor activation is the consequence of mutant Kras and stromal cues, providing insight into the role of the tumor microenvironment in the origin and treatment of Ras-driven tumors.


Asunto(s)
Carcinoma Ductal Pancreático/fisiopatología , Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Código de Histonas , Metaboloma , Neoplasias Pancreáticas/fisiopatología , Células del Estroma/fisiología , Microambiente Tumoral/fisiología , Acetilación , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Citocinas/metabolismo , Metabolismo Energético , Elementos de Facilitación Genéticos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/fisiología , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción , Células Tumorales Cultivadas
8.
Sci Signal ; 9(430): ra57, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27245614

RESUMEN

Rapamycin has been used as a clinical immunosuppressant for many years; however, the molecular basis for its selective effects on lymphocytes remains unclear. We investigated the role of two canonical effectors of the mammalian target of rapamycin (mTOR): ribosomal S6 kinases (S6Ks) and eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs). S6Ks are thought to regulate cell growth (increase in cell size), and 4E-BPs are thought to control proliferation (increase in cell number), with mTORC1 signaling serving to integrate these processes. However, we found that the 4E-BP-eIF4E signaling axis controlled both the growth and proliferation of lymphocytes, processes for which the S6Ks were dispensable. Furthermore, rapamycin disrupted eIF4E function selectively in lymphocytes, which was due to the increased abundance of 4E-BP2 relative to that of 4E-BP1 in these cells and the greater sensitivity of 4E-BP2 to rapamycin. Together, our findings suggest that the 4E-BP-eIF4E axis is uniquely rapamycin-sensitive in lymphocytes and that this axis promotes clonal expansion of these cells by coordinating growth and proliferation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Linfocitos/efectos de los fármacos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Animales , Proteínas de Ciclo Celular , Aumento de la Célula , Membrana Celular/metabolismo , Proliferación Celular , Cruzamientos Genéticos , Factores Eucarióticos de Iniciación , Femenino , Regulación de la Expresión Génica , Guanosina Trifosfato , Inmunosupresores/farmacología , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Sirolimus/farmacología
9.
Nat Rev Cancer ; 16(5): 288-304, 2016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27112207

RESUMEN

The past several years have seen dramatic leaps in our understanding of how gene expression is rewired at the translation level during tumorigenesis to support the transformed phenotype. This work has been driven by an explosion in technological advances and is revealing previously unimagined regulatory mechanisms that dictate functional expression of the cancer genome. In this Review we discuss emerging trends and exciting new discoveries that reveal how this translational circuitry contributes to specific aspects of tumorigenesis and cancer cell function, with a particular focus on recent insights into the role of translational control in the adaptive response to oncogenic stress conditions.


Asunto(s)
Genoma , Neoplasias/genética , Biosíntesis de Proteínas , Humanos , Oncogenes , ARN Neoplásico/genética
10.
Cancer Discov ; 6(3): 270-85, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26715645

RESUMEN

UNLABELLED: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.


Asunto(s)
Comunicación Celular/inmunología , Sistema Inmunológico/citología , Sistema Inmunológico/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias Pancreáticas/genética , Receptores de IgG/metabolismo , Transducción de Señal
11.
Cell ; 162(1): 59-71, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26095252

RESUMEN

eIF4E, the major cap-binding protein, has long been considered limiting for translating the mammalian genome. However, the eIF4E dose requirement at an organismal level remains unexplored. By generating an Eif4e haploinsufficient mouse, we found that a 50% reduction in eIF4E expression, while compatible with normal development and global protein synthesis, significantly impeded cellular transformation. Genome-wide translational profiling uncovered a translational program induced by oncogenic transformation and revealed a critical role for the dose of eIF4E, specifically in translating a network of mRNAs enriched for a unique 5' UTR signature. In particular, we demonstrate that the dose of eIF4E is essential for translating mRNAs that regulate reactive oxygen species, fueling transformation and cancer cell survival in vivo. Our findings indicate eIF4E is maintained at levels in excess for normal development that are hijacked by cancer cells to drive a translational program supporting tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Embrión de Mamíferos/metabolismo , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Dosificación de Gen , Regiones no Traducidas 5' , Animales , Carcinogénesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Biosíntesis de Proteínas , Especies Reactivas de Oxígeno/metabolismo
12.
Cancer Res ; 75(14): 2851-62, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25977330

RESUMEN

Oncogenic K-Ras mutation occurs frequently in several types of cancers, including pancreatic and lung cancers. Tumors with K-Ras mutation are resistant to chemotherapeutic drugs as well as molecular targeting agents. Although numerous approaches are ongoing to find effective ways to treat these tumors, there are still no effective therapies for K-Ras mutant cancer patients. Here we report that K-Ras mutant cancers are more dependent on K-Ras in anchorage-independent culture conditions than in monolayer culture conditions. In seeking to determine mechanisms that contribute to the K-Ras dependency in anchorage-independent culture conditions, we discovered the involvement of Met in K-Ras-dependent, anchorage-independent cell growth. The Met signaling pathway is enhanced and plays an indispensable role in anchorage-independent growth even in cells in which Met is not amplified. Indeed, Met expression is elevated under anchorage-independent growth conditions and is regulated by K-Ras in a MAPK/ERK kinase (MEK)-dependent manner. Remarkably, in spite of a global downregulation of mRNA translation during anchorage-independent growth, we find that Met mRNA translation is specifically enhanced under these conditions. Importantly, ectopic expression of an active Met mutant rescues K-Ras ablation-derived growth suppression, indicating that K-Ras-mediated Met expression drives "K-Ras addiction" in anchorage-independent conditions. Our results indicate that enhanced Met expression and signaling is essential for anchorage-independent growth of K-Ras mutant cancer cells and suggests that pharmacological inhibitors of Met could be effective for K-Ras mutant tumor patients.


Asunto(s)
Proliferación Celular/genética , Genes ras/fisiología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Adhesión Celular/genética , Células Cultivadas , Células HCT116 , Células HEK293 , Humanos , Ratones , Biosíntesis de Proteínas , Transducción de Señal/fisiología , Técnicas de Cultivo de Tejidos
13.
Proc Natl Acad Sci U S A ; 110(29): 11988-93, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23803853

RESUMEN

Myc is one of the most commonly deregulated oncogenes in human cancer, yet therapies directly targeting Myc hyperactivation are not presently available in the clinic. The evolutionarily conserved function of Myc in modulating protein synthesis control is critical to the Myc oncogenic program. Indeed, enhancing the protein synthesis capacity of cancer cells directly contributes to their survival, proliferation, and genome instability. Therefore, inhibiting enhanced protein synthesis may represent a highly relevant strategy for the treatment of Myc-dependent human cancers. However, components of the translation machinery that can be exploited as therapeutic targets for Myc-driven cancers remain poorly defined. Here, we uncover a surprising and important functional link between Myc and mammalian target of rapamycin (mTOR)-dependent phosphorylation of eukaryotic translation initiation factor 4E binding protein-1 (4EBP1), a master regulator of protein synthesis control. Using a pharmacogenetic approach, we find that mTOR-dependent phosphorylation of 4EBP1 is required for cancer cell survival in Myc-dependent tumor initiation and maintenance. We further show that a clinical mTOR active site inhibitor, which is capable of blocking mTOR-dependent 4EBP1 phosphorylation, has remarkable therapeutic efficacy in Myc-driven hematological cancers. Additionally, we demonstrate the clinical implications of these results by delineating a significant link between Myc and mTOR-dependent phosphorylation of 4EBP1 and therapeutic response in human lymphomas. Together, these findings reveal that an important mTOR substrate is found hyperactivated downstream of Myc oncogenic activity to promote tumor survival and confers synthetic lethality, thereby revealing a unique therapeutic approach to render Myc druggable in the clinic.


Asunto(s)
Linfocitos B/fisiología , Benzoxazoles/farmacología , Proteínas Portadoras/metabolismo , Transformación Celular Neoplásica/metabolismo , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Benzoxazoles/administración & dosificación , Western Blotting , Proteínas de Ciclo Celular , Factores Eucarióticos de Iniciación , Everolimus , Humanos , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Fosforilación , Pirimidinas/administración & dosificación , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
14.
Nature ; 493(7432): 371-7, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23172145

RESUMEN

Hyperconnectivity of neuronal circuits due to increased synaptic protein synthesis is thought to cause autism spectrum disorders (ASDs). The mammalian target of rapamycin (mTOR) is strongly implicated in ASDs by means of upstream signalling; however, downstream regulatory mechanisms are ill-defined. Here we show that knockout of the eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2)-an eIF4E repressor downstream of mTOR-or eIF4E overexpression leads to increased translation of neuroligins, which are postsynaptic proteins that are causally linked to ASDs. Mice that have the gene encoding 4E-BP2 (Eif4ebp2) knocked out exhibit an increased ratio of excitatory to inhibitory synaptic inputs and autistic-like behaviours (that is, social interaction deficits, altered communication and repetitive/stereotyped behaviours). Pharmacological inhibition of eIF4E activity or normalization of neuroligin 1, but not neuroligin 2, protein levels restores the normal excitation/inhibition ratio and rectifies the social behaviour deficits. Thus, translational control by eIF4E regulates the synthesis of neuroligins, maintaining the excitation-to-inhibition balance, and its dysregulation engenders ASD-like phenotypes.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Factor 4E Eucariótico de Iniciación/metabolismo , Biosíntesis de Proteínas , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Factor 4E Eucariótico de Iniciación/antagonistas & inhibidores , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Masculino , Ratones , Ratones Noqueados , Fenotipo , Sinapsis/metabolismo
15.
Science ; 338(6108): 818-22, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23042294

RESUMEN

The endoplasmic reticulum (ER) is the primary organelle for folding and maturation of secretory and transmembrane proteins. Inability to meet protein-folding demand leads to "ER stress," and activates IRE1α, an ER transmembrane kinase-endoribonuclease (RNase). IRE1α promotes adaptation through splicing Xbp1 mRNA or apoptosis through incompletely understood mechanisms. Here, we found that sustained IRE1α RNase activation caused rapid decay of select microRNAs (miRs -17, -34a, -96, and -125b) that normally repress translation of Caspase-2 mRNA, and thus sharply elevates protein levels of this initiator protease of the mitochondrial apoptotic pathway. In cell-free systems, recombinant IRE1α endonucleolytically cleaved microRNA precursors at sites distinct from DICER. Thus, IRE1α regulates translation of a proapoptotic protein through terminating microRNA biogenesis, and noncoding RNAs are part of the ER stress response.


Asunto(s)
Caspasa 2/genética , Caspasa 2/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Regiones no Traducidas 3' , Animales , Apoptosis , Brefeldino A/farmacología , Sistema Libre de Células , Células Cultivadas , Regulación hacia Abajo , Retículo Endoplásmico/metabolismo , Endorribonucleasas/química , Endorribonucleasas/genética , Activación Enzimática , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteínas Mutantes , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
16.
Nat Med ; 17(4): 500-3, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21460848

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease. Overall survival is typically 6 months from diagnosis. Numerous phase 3 trials of agents effective in other malignancies have failed to benefit unselected PDA populations, although patients do occasionally respond. Studies in other solid tumors have shown that heterogeneity in response is determined, in part, by molecular differences between tumors. Furthermore, treatment outcomes are improved by targeting drugs to tumor subtypes in which they are selectively effective, with breast and lung cancers providing recent examples. Identification of PDA molecular subtypes has been frustrated by a paucity of tumor specimens available for study. We have overcome this problem by combined analysis of transcriptional profiles of primary PDA samples from several studies, along with human and mouse PDA cell lines. We define three PDA subtypes: classical, quasimesenchymal and exocrine-like, and we present evidence for clinical outcome and therapeutic response differences between them. We further define gene signatures for these subtypes that may have utility in stratifying patients for treatment and present preclinical model systems that may be used to identify new subtype specific therapies.


Asunto(s)
Carcinoma Ductal Pancreático/clasificación , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/clasificación , Neoplasias Pancreáticas/genética , Animales , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Clorhidrato de Erlotinib , Femenino , Factor de Transcripción GATA6/genética , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Farmacogenética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Quinazolinas/farmacología , Proteínas ras/genética , Gemcitabina
17.
Genes Cancer ; 1(2): 125-41, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20975924

RESUMEN

Factors associated with tumor sensitivity to epidermal growth factor receptor (EGFR) inhibitors in the context of wild-type EGFR remain elusive. This study investigates the mechanistic basis of responsiveness to EGFR inhibitors in the RIP1-Tag2 (RT2) mouse model of pancreatic neuroendocrine tumorigenesis (PNET). Upon treatment of RT2 mice with EGFR inhibitors, PNET tumors harboring wild-type, nonamplified alleles of Egfr grow at a markedly reduced rate and display a significant increase in tumor cell apoptosis, as well as reduced neovascularization. The authors identify Tgf-α and Hb-egf as key limiting mediators of separable pathological functions of Egfr in neuroendocrine tumor progression: Tgf-α mutant tumors present with an elevated apoptotic index, whereas Hb-egf mutant lesions exhibit decreased angiogenic switching and neovascularization. This study not only associates Tgf-α and Hb-egf expression with wild-type Egfr oncogenicity but also ascribes the proangiogenic activity of Egfr in this tumor model to a novel mesenchymal Hb-egf/Egfr signaling axis, whereby endothelial and pericyte-derived Hb-egf activates Egfr specifically in tumor-associated perivascular cells, leading to increased pericyte coverage of the tumor endothelium and enhanced angiogenesis.

18.
Cancer Cell ; 17(2): 135-47, 2010 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-20138012

RESUMEN

Cancer-associated fibroblasts (CAFs) support tumorigenesis by stimulating angiogenesis, cancer cell proliferation, and invasion. We demonstrate that CAFs also mediate tumor-enhancing inflammation. Using a mouse model of squamous skin carcinogenesis, we found a proinflammatory gene signature in CAFs isolated from dysplastic skin. This signature was maintained in CAFs from subsequent skin carcinomas and was evident in mammary and pancreatic tumors in mice and in cognate human cancers. The inflammatory signature was already activated in CAFs isolated from the initial hyperplastic stage in multistep skin tumorigenesis. CAFs from this pathway promoted macrophage recruitment, neovascularization, and tumor growth, activities that are abolished when NF-kappaB signaling was inhibited. Additionally, we show that normal dermal fibroblasts can be "educated" by carcinoma cells to express proinflammatory genes.


Asunto(s)
Fibroblastos/fisiología , Mediadores de Inflamación/metabolismo , FN-kappa B/fisiología , Neoplasias/genética , Animales , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Femenino , Fibroblastos/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Patológica , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias del Cuello Uterino/irrigación sanguínea , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología
19.
Genes Dev ; 23(1): 24-36, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19136624

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular , Supervivencia Celular , Células Cultivadas , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Receptor Smoothened , Factor de Crecimiento Transformador beta/metabolismo , Proteína con Dedos de Zinc GLI1
20.
Dis Model Mech ; 1(2-3): 155-67; discussion 165, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19048079

RESUMEN

The tumor microenvironment consists of stromal cells and extracellular factors that evolve in parallel with carcinoma cells. To gain insights into the activities of stromal cell populations, we developed and applied multicolor imaging techniques to analyze the behavior of these cells within different tumor microenvironments in the same live mouse. We found that regulatory T-lymphocytes (Tregs) migrated in proximity to blood vessels. Dendritic-like cells, myeloid cells and carcinoma-associated fibroblasts all exhibited higher motility in the microenvironment at the tumor periphery than within the tumor mass. Since oxygen levels differ between tumor microenvironments, we tested if acute hypoxia could account for the differences in cell migration. Direct visualization revealed that Tregs ceased migration under acute systemic hypoxia, whereas myeloid cells continued migrating. In the same mouse and microenvironment, we experimentally subdivided the myeloid cell population and revealed that uptake of fluorescent dextran defined a low-motility subpopulation expressing markers of tumor-promoting, alternatively activated macrophages. In contrast, fluorescent anti-Gr1 antibodies marked myeloid cells patrolling inside tumor vessels and in the stroma. Our techniques allow real-time combinatorial analysis of cell populations based on spatial location, gene expression, behavior and cell surface molecules within intact tumors. The techniques are not limited to investigations in cancer, but could give new insights into cell behavior more broadly in development and disease.


Asunto(s)
Microscopía Confocal/métodos , Neoplasias/patología , Células del Estroma/patología , Hipoxia de la Célula , Movimiento Celular , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...