Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 201(10): 2873-2878, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30305325

RESUMEN

Sepsis disproportionately affects the very old and the very young. IL-1 signaling is important in innate host defense but may also play a deleterious role in acute inflammatory conditions (including sepsis) by promulgating life-threatening inflammation. IL-1 signaling is mediated by two distinct ligands: IL-1α and IL-1ß, both acting on a common receptor (IL-1R1). IL-1R1 targeting has not reduced adult human sepsis mortality despite biologic plausibility. Because the specific role of IL-1α or IL-1ß in sepsis survival is unknown in any age group and the role of IL-1 signaling remains unknown in neonates, we studied the role of IL-1 signaling, including the impact of IL-1α and IL-1ß, on neonatal murine sepsis survival. IL-1 signaling augments the late plasma inflammatory response to sepsis. IL-1α and not IL-1ß is the critical mediator of sepsis mortality, likely because of paracrine actions within the tissue. These data do not support targeting IL-1 signaling in neonates.


Asunto(s)
Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Receptores Tipo I de Interleucina-1/inmunología , Sepsis/inmunología , Animales , Animales Recién Nacidos , Femenino , Humanos , Recién Nacido , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/mortalidad , Transducción de Señal/inmunología
2.
Am J Respir Cell Mol Biol ; 58(6): 736-744, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29314863

RESUMEN

Loss of secretory IgA is common in the small airways of patients with chronic obstructive pulmonary disease and may contribute to disease pathogenesis. Using mice that lack secretory IgA in the airways due to genetic deficiency of polymeric Ig receptor (pIgR-/- mice), we investigated the role of neutrophils in driving the fibrotic small airway wall remodeling and emphysema that develops spontaneously in these mice. By flow cytometry, we found an increase in the percentage of neutrophils among CD45+ cells in the lungs, as well as an increase in total neutrophils, in pIgR-/- mice compared with wild-type controls. This increase in neutrophils in pIgR-/- mice was associated with elastin degradation in the alveolar compartment and around small airways, along with increased collagen deposition in small airway walls. Neutrophil depletion using anti-Ly6G antibodies or treatment with broad-spectrum antibiotics inhibited development of both emphysema and small airway remodeling, suggesting that airway bacteria provide the stimulus for deleterious neutrophilic inflammation in this model. Exogenous bacterial challenge using lysates prepared from pathogenic and nonpathogenic bacteria worsened neutrophilic inflammation and lung remodeling in pIgR-/- mice. This phenotype was abrogated by antiinflammatory therapy with roflumilast. Together, these studies support the concept that disruption of the mucosal immune barrier in small airways contributes to chronic obstructive pulmonary disease progression by allowing bacteria to stimulate chronic neutrophilic inflammation, which, in turn, drives progressive airway wall fibrosis and emphysematous changes in the lung parenchyma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Neutrófilos/patología , Neumonía Bacteriana/patología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Bacillus/patogenicidad , Benzamidas/farmacología , Ciclopropanos/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Mutantes , Neutrófilos/microbiología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfisema Pulmonar/patología , Receptores de Superficie Celular/genética
3.
Development ; 141(24): 4751-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25395457

RESUMEN

Integrin-dependent interactions between cells and extracellular matrix regulate lung development; however, specific roles for ß1-containing integrins in individual cell types, including epithelial cells, remain incompletely understood. In this study, the functional importance of ß1 integrin in lung epithelium during mouse lung development was investigated by deleting the integrin from E10.5 onwards using surfactant protein C promoter-driven Cre. These mutant mice appeared normal at birth but failed to gain weight appropriately and died by 4 months of age with severe hypoxemia. Defects in airway branching morphogenesis in association with impaired epithelial cell adhesion and migration, as well as alveolarization defects and persistent macrophage-mediated inflammation were identified. Using an inducible system to delete ß1 integrin after completion of airway branching, we showed that alveolarization defects, characterized by disrupted secondary septation, abnormal alveolar epithelial cell differentiation, excessive collagen I and elastin deposition, and hypercellularity of the mesenchyme occurred independently of airway branching defects. By depleting macrophages using liposomal clodronate, we found that alveolarization defects were secondary to persistent alveolar inflammation. ß1 integrin-deficient alveolar epithelial cells produced excessive monocyte chemoattractant protein 1 and reactive oxygen species, suggesting a direct role for ß1 integrin in regulating alveolar homeostasis. Taken together, these studies define distinct functions of epithelial ß1 integrin during both early and late lung development that affect airway branching morphogenesis, epithelial cell differentiation, alveolar septation and regulation of alveolar homeostasis.


Asunto(s)
Células Epiteliales/metabolismo , Integrina beta1/metabolismo , Pulmón/embriología , Organogénesis/fisiología , Alveolos Pulmonares/embriología , Animales , Lavado Broncoalveolar , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quimiocina CCL2/metabolismo , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/metabolismo , Integrasas/metabolismo , Ratones , Microscopía Confocal , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico
4.
Am J Pathol ; 186(7): 1786-1800, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27181406

RESUMEN

The highly orchestrated interactions between the epithelium and mesenchyme required for normal lung development can be disrupted by perinatal inflammation in preterm infants, although the mechanisms are incompletely understood. We used transgenic (inhibitory κB kinase ß transactivated) mice that conditionally express an activator of the NF-κB pathway in airway epithelium to investigate the impact of epithelial-derived inflammation during lung development. Epithelial NF-κB activation selectively impaired saccular stage lung development, with a phenotype comprising rapidly progressive distal airspace dilation, impaired gas exchange, and perinatal lethality. Epithelial-derived inflammation resulted in disrupted elastic fiber organization and down-regulation of elastin assembly components, including fibulins 4 and 5, lysyl oxidase like-1, and fibrillin-1. Fibulin-5 expression by saccular stage lung fibroblasts was consistently inhibited by treatment with bronchoalveolar lavage fluid from inhibitory κB kinase ß transactivated mice, Escherichia coli lipopolysaccharide, or tracheal aspirates from preterm infants exposed to chorioamnionitis. Expression of a dominant NF-κB inhibitor in fibroblasts restored fibulin-5 expression after lipopolysaccharide treatment, whereas reconstitution of fibulin-5 rescued extracellular elastin assembly by saccular stage lung fibroblasts. Elastin organization was disrupted in saccular stage lungs of preterm infants exposed to systemic inflammation. Our study reveals a critical window for elastin assembly during the saccular stage that is disrupted by inflammatory signaling and could be amenable to interventions that restore elastic fiber assembly in the developing lung.


Asunto(s)
Elastina/metabolismo , Epitelio/metabolismo , Inflamación/complicaciones , Pulmón/embriología , Animales , Western Blotting , Desarrollo Fetal , Humanos , Inmunohistoquímica , Recién Nacido , Recien Nacido Prematuro , Inflamación/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Modelos Animales , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
J Cell Physiol ; 227(2): 801-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21503880

RESUMEN

Polyploidy has been linked to tumorigenicity mainly due to the chromosomal aberrations. Elevated reactive oxygen species (ROS) generation, on the other hand, has also been associated with oncogenic transformation in most cancer cells. However, a possible link between ploidy and ROS is largely unexplored. Here we have examined the role of ROS in the tumorigenicity of polyploid cells. We show that polyploid prostate and mammary epithelial cells contain higher levels of ROS due to their higher mitochondrial contents. ROS levels and mitochondrial mass are also higher in dihydrocytochalasin B (DCB)-induced polyploid cells, suggesting that higher levels of ROS observed in polyploid cell can occur due to cytokinesis failure. Interestingly, polyploid cells were more sensitive to the inhibitory effect of the antioxidant, N-Acetyl-L-cysteine (NAC), than control diploid cells. Treatment of polyploid/diploid cells with NAC led to the selective elimination of polyploid cells over time and abrogated the tumorigenicity of polyploid cells. This effect was partially mediated via the Akt signaling pathway. We next explored a possible role for ROS in promoting chromosomal instability by analyzing the effects of ROS on the mitotic stage of the cell cycle. Enhancing ROS levels by treating cells with hydrogen peroxide delayed not only entry into and but also exit from mitosis. Furthermore, increasing ROS levels significantly increased taxol resistance. Our results indicated that increased ROS in polyploid cells can contribute to tumorigenicity and highlight the therapeutic potential of antioxidants by selectively targeting the tumorigenic polyploid cells and by reversing taxol resistance.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Poliploidía , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Línea Celular , Resistencia a Antineoplásicos , Femenino , Humanos , Peróxido de Hidrógeno , Masculino , Glándulas Mamarias Humanas/citología , Mitocondrias/fisiología , Mitosis/efectos de los fármacos , Mitosis/fisiología , Paclitaxel/farmacología , Próstata/citología
7.
J Immunol ; 185(8): 4896-903, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20861353

RESUMEN

Bronchopulmonary dysplasia (BPD) is a frequent complication of preterm birth. This chronic lung disease results from arrested saccular airway development and is most common in infants exposed to inflammatory stimuli. In experimental models, inflammation inhibits expression of fibroblast growth factor-10 (FGF-10) and impairs epithelial-mesenchymal interactions during lung development; however, the mechanisms connecting inflammatory signaling with reduced growth factor expression are not yet understood. In this study we found that soluble inflammatory mediators present in tracheal fluid from preterm infants can prevent saccular airway branching. In addition, LPS treatment led to local production of mediators that inhibited airway branching and FGF-10 expression in LPS-resistant C.C3-Tlr4(Lpsd)/J fetal mouse lung explants. Both direct NF-κB activation and inflammatory cytokines (IL-1ß and TNF-α) that activate NF-κB reduced FGF-10 expression, whereas chemokines that signal via other inflammatory pathways had no effect. Mutational analysis of the FGF-10 promoter failed to identify genetic elements required for direct NF-κB-mediated FGF-10 inhibition. Instead, NF-κB activation appeared to interfere with the normal stimulation of FGF-10 expression by Sp1. Chromatin immunoprecipitation and nuclear coimmunoprecipitation studies demonstrated that the RelA subunit of NF-κB and Sp1 physically interact at the FGF-10 promoter. These findings indicate that inflammatory signaling through NF-κB disrupts the normal expression of FGF-10 in fetal lung mesenchyme by interfering with the transcriptional machinery critical for lung morphogenesis.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/biosíntesis , Pulmón/embriología , FN-kappa B/metabolismo , Proteínas Quinasas/metabolismo , Animales , Corioamnionitis/metabolismo , Inmunoprecipitación de Cromatina , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Inmunoprecipitación , Recién Nacido , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Embarazo , Nacimiento Prematuro , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
8.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33108351

RESUMEN

Emerging evidence indicates that early life events can increase the risk for developing chronic obstructive pulmonary disease (COPD). Using an inducible transgenic mouse model for NF-κB activation in the airway epithelium, we found that a brief period of inflammation during the saccular stage (P3-P5) but not alveolar stage (P10-P12) of lung development disrupted elastic fiber assembly, resulting in permanent reduction in lung function and development of a COPD-like lung phenotype that progressed through 24 months of age. Neutrophil depletion prevented disruption of elastic fiber assembly and restored normal lung development. Mechanistic studies uncovered a role for neutrophil elastase (NE) in downregulating expression of critical elastic fiber assembly components, particularly fibulin-5 and elastin. Further, purified human NE and NE-containing exosomes from tracheal aspirates of premature infants with lung inflammation downregulated elastin and fibulin-5 expression by saccular-stage mouse lung fibroblasts. Together, our studies define a critical developmental window for assembling the elastin scaffold in the distal lung, which is required to support lung structure and function throughout the lifespan. Although neutrophils play a well-recognized role in COPD development in adults, neutrophilic inflammation may also contribute to early-life predisposition to COPD.


Asunto(s)
Elastina/metabolismo , Neutrófilos/metabolismo , Alveolos Pulmonares/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Animales , Elastina/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Ratones , Ratones Transgénicos , Neutrófilos/patología , Alveolos Pulmonares/patología , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología
9.
Carcinogenesis ; 28(10): 2184-92, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17468514

RESUMEN

Little is known about early carcinogen-induced protein alterations in mammary epithelium. Detection of early alterations would enhance our understanding of early-stage carcinogenesis. Here, normal human mammary epithelial cells (HMECs) were exposed to dietary and environmental carcinogens [2-amino-1-methyl-6-phenylimidazo[4,5b]pyridine (PhIP), 4-aminobiphenyl (ABP), benzo[a]pyrene, 2,3,7,8-tetrachlorodibenzo-p-dioxin] individually or in combination. A phage display library of single-chain variable fragment antibodies was used to screen protein targets altered by the treatment. In combination with matrix-assisted laser desorption time of flight, we identified histone H3 as a target antigen. Although histone H3 total protein remained unchanged in control and treated HMEC, the methylation of lysine 4 was altered. A reduction in mono-methyl histone H3 (Lys 4) was observed in treated HMEC compared with control HMEC. This alteration was shown to be dependent on carcinogen concentration and specific for PhIP and ABP. To characterize potential histone demethylation mechanisms, localization and protein expression patterns of lysine-specific demethylase 1 (LSD1) were analyzed. In control HMEC, LSD1 was present at the nuclear periphery. However, following 72 h carcinogen treatment, LSD1 localized within the nucleus. Within 48 h after treatment, mono-methyl histone H3 (Lys 4) was restored and LSD1 localization was reversed. Protein expression levels of LSD1 were also increased in treated HMEC compared with control HMEC. Our data suggest that the induction of a single enzyme, LSD1, represents an early response to carcinogen exposure, which leads to the demethylation of histone H3 (Lys 4), which, in turn, may influence the expression of multiple genes critical in early-stage mammary carcinogenesis.


Asunto(s)
Mama/citología , Mama/fisiología , Carcinógenos/toxicidad , Células Epiteliales/fisiología , Histonas/metabolismo , Compuestos de Aminobifenilo/toxicidad , Benzo(a)pireno/toxicidad , Biotinilación , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Histonas/efectos de los fármacos , Histonas/aislamiento & purificación , Humanos , Imidazoles/toxicidad , Región Variable de Inmunoglobulina/inmunología , Biblioteca de Péptidos , Dibenzodioxinas Policloradas/toxicidad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
Leuk Res ; 29(10): 1181-90, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15936073

RESUMEN

Bifunctional alkylating agents that cross-link DNA are implicated in the pathogenesis of therapy related myelodysplastic syndromes (MDS) and MDS related acute myeloid leukemia (MDR-AML). We exposed HL60 cells to the highest level of bifunctional alkylating nitrogen mustard mechlorethamine (HN2) that was consistent with recovery following suppressed growth. Microarray analyses showed minor changes in transcripts in HN2 treated cells. A moderate up-regulation of S100P mRNA was consistently observed after 1 day of exposure to bifunctional alkylating agents and expression was not induced with monofunctional agents. Elevated S100P protein/antigen was not detected until days later in a subset of non-mitotic G2 cells. Elevated S100P protein persisted over the course of a delayed recovery phase. The results confirm recent reports indicating that S100P is a survival factor. In addition, our results indicate that S100P has a specific role in G2 cell function associated with a prolonged phase of recovery after exposure to bifunctional alkylating agents.


Asunto(s)
Alquilantes/farmacología , Proteínas de Unión al Calcio/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Fase G2/efectos de los fármacos , Mecloretamina/farmacología , Proteínas de Neoplasias/metabolismo , Northern Blotting , Western Blotting , Proteínas de Unión al Calcio/genética , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Perfilación de la Expresión Génica , Células HL-60 , Humanos , Técnicas para Inmunoenzimas , Cariotipificación , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fracciones Subcelulares , Regulación hacia Arriba
11.
J Natl Cancer Inst ; 107(2)2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25505253

RESUMEN

BACKGROUND: PIM1 kinase is coexpressed with c-MYC in human prostate cancers (PCs) and dramatically enhances c-MYC-induced tumorigenicity. Here we examine the effects of a novel oral PIM inhibitor, AZD1208, on prostate tumorigenesis and recurrence. METHODS: A mouse c-MYC/Pim1-transduced tissue recombination PC model, Myc-CaP allografts, and human PC xenografts were treated with AZD1208 (n = 5-11 per group). Androgen-sensitive and castrate-resistant prostate cancer (CRPC) models were studied as well as the effects of hypoxia and radiation. RNA sequencing was used to analyze drug-induced gene expression changes. Results were analyzed with χ(2) test. Student's t test and nonparametric Mann-Whitney rank sum U Test. All statistical tests were two-sided. RESULTS: AZD1208 inhibited tumorigenesis in tissue recombinants, Myc-CaP, and human PC xenograft models. PIM inhibition decreased c-MYC/Pim1 graft growth by 54.3 ± 39% (P < .001), decreased cellular proliferation by 46 ± 14% (P = .016), and increased apoptosis by 326 ± 170% (P = .039). AZD1208 suppressed multiple protumorigenic pathways, including the MYC gene program. However, it also downregulated the p53 pathway. Hypoxia and radiation induced PIM1 in prostate cancer cells, and AZD1208 functioned as a radiation sensitizer. Recurrent tumors postcastration responded transiently to either AZD1208 or radiation treatment, and combination treatment resulted in more sustained inhibition of tumor growth. Cell lines established from recurrent, AZD1208-resistant tumors again revealed downregulation of the p53 pathway. Irradiated AZD1208-treated tumors robustly upregulated p53, providing a possible mechanistic explanation for the effectiveness of combination therapy. Finally, an AZD1208-resistant gene signature was found to be associated with biochemical recurrence in PC patients. CONCLUSIONS: PIM inhibition is a potential treatment for MYC-driven prostate cancers including CRPC, and its effectiveness may be enhanced by activators of the p53 pathway, such as radiation.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Tiazolidinas/farmacología , Administración Oral , Aloinjertos , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/administración & dosificación , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes myc , Humanos , Masculino , Ratones , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Tiazolidinas/administración & dosificación , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Clin Cancer Res ; 20(12): 3211-21, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24771642

RESUMEN

PURPOSE: To identify genes whose depletion is detrimental to Pim1-overexpressing prostate cancer cells and to validate this finding in vitro and in vivo. EXPERIMENTAL DESIGN: RNAi screening was used to identify genes whose depletion is detrimental to Pim1-overexpressing cells. Our finding was validated using shRNA or PLK1-specific inhibitor BI 2536. Xenograft studies were performed using both PLK1-knockdown cells and BI 2536 to investigate the effects of PLK1 inhibition on tumorigenesis in Pim1-overexpressing cells. Finally, PLK1 and PIM1 expression patterns in human prostate tumors were examined by immunohistochemistry using tissue microarrays. RESULTS: We identified the mitotic regulator polo-like kinase (PLK1) as a gene whose depletion is particularly detrimental to the viability of Pim1-overexpressing prostate cancer. Inhibition of PLK1 by shRNA or BI 2536 in Pim1-overexpressing prostate cancer xenograft models resulted in a dramatic inhibition of tumor progression. Notably, Pim1-overexpressing cells were more prone to mitotic arrest followed by apoptosis due to PLK1 inhibition than control cells. Furthermore, inhibition of PLK1 led to the reduction of MYC protein levels both in vitro and in vivo. Our data also suggest that PIM1 and PLK1 physically interact and PIM1 might phosphorylate PLK1. Finally, PLK1 and PIM1 are frequently co-expressed in human prostate tumors, and co-expression of PLK1 and PIM1 was significantly correlated to higher Gleason grades. CONCLUSIONS: Our findings demonstrate that PIM1-overexpressing cancer cells are particularly sensitive to PLK1 inhibition, suggesting that PIM1 might be used as a marker for identifying patients who will benefit from PLK1 inhibitor treatment.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Interferencia de ARN/fisiología , ARN Interferente Pequeño/genética , Animales , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Mitosis/fisiología , Clasificación del Tumor , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/genética , Análisis de Matrices Tisulares , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
13.
J Clin Invest ; 122(5): 1907-19, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22484818

RESUMEN

Cooperativity between oncogenic mutations is recognized as a fundamental feature of malignant transformation, and it may be mediated by synergistic regulation of the expression of pro- and antitumorigenic target genes. However, the mechanisms by which oncogenes and tumor suppressors coregulate downstream targets and pathways remain largely unknown. Here, we used ChIP coupled to massively parallel sequencing (ChIP-seq) and gene expression profiling in mouse prostates to identify direct targets of the tumor suppressor Nkx3.1. Further analysis indicated that a substantial fraction of Nkx3.1 target genes are also direct targets of the oncoprotein Myc. We also showed that Nkx3.1 and Myc bound to and crossregulated shared target genes in mouse and human prostate epithelial cells and that Nkx3.1 could oppose the transcriptional activity of Myc. Furthermore, loss of Nkx3.1 cooperated with concurrent overexpression of Myc to promote prostate cancer in transgenic mice. In human prostate cancer patients, dysregulation of shared NKX3.1/MYC target genes was associated with disease relapse. Our results indicate that NKX3.1 and MYC coregulate prostate tumorigenesis by converging on, and crossregulating, a common set of target genes. We propose that coregulation of target gene expression by oncogenic/tumor suppressor transcription factors may represent a general mechanism underlying the cooperativity of oncogenic mutations during tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/fisiología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
14.
Cancer Cell ; 17(1): 41-52, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20129246

RESUMEN

The sirtuin gene family (SIRT) is hypothesized to regulate the aging process and play a role in cellular repair. This work demonstrates that SIRT3(-/-) mouse embryonic fibroblasts (MEFs) exhibit abnormal mitochondrial physiology as well as increases in stress-induced superoxide levels and genomic instability. Expression of a single oncogene (Myc or Ras) in SIRT3(-/-) MEFs results in in vitro transformation and altered intracellular metabolism. Superoxide dismutase prevents transformation by a single oncogene in SIRT3(-/-) MEFs and reverses the tumor-permissive phenotype as well as stress-induced genomic instability. In addition, SIRT3(-/-) mice develop ER/PR-positive mammary tumors. Finally, human breast and other human cancer specimens exhibit reduced SIRT3 levels. These results identify SIRT3 as a genomically expressed, mitochondria-localized tumor suppressor.


Asunto(s)
Envejecimiento/fisiología , Transformación Celular Neoplásica/genética , Genes Supresores de Tumor , Mitocondrias/metabolismo , Sirtuina 3/genética , Estrés Fisiológico/fisiología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Estrés Oxidativo/fisiología , Sirtuina 3/metabolismo , Superóxidos/metabolismo
15.
PLoS One ; 3(7): e2572, 2008 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-18596907

RESUMEN

BACKGROUND: Polyploidy is a prominent feature of many human cancers, and it has long been hypothesized that polyploidy may contribute to tumorigenesis by promoting genomic instability. In this study, we investigated whether polyploidy per se induced by a relevant oncogene can promote genomic instability and tumorigenicity in human epithelial cells. PRINCIPAL FINDINGS: When the oncogenic serine-threonine kinase Pim-1 is overexpressed in immortalized, non-tumorigenic human prostate and mammary epithelial cells, these cells gradually converted to polyploidy and became tumorigenic. To assess the contribution of polyploidy to tumorigenicity, we obtained sorted, matched populations of diploid and polyploid cells expressing equivalent levels of the Pim-1 protein. Spectral karyotyping revealed evidence of emerging numerical and structural chromosomal abnormalities in polyploid cells, supporting the proposition that polyploidy promotes chromosomal instability. Polyploid cells displayed an intact p53/p21 pathway, indicating that the viability of polyploid cells in this system is not dependent on the inactivation of the p53 signaling pathway. Remarkably, only the sorted polyploid cells were tumorigenic in vitro and in vivo. CONCLUSIONS: Our results support the notion that polyploidy can promote chromosomal instability and the initiation of tumorigenesis in human epithelial cells.


Asunto(s)
Neoplasias de la Mama/genética , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Poliploidía , Próstata/metabolismo , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Animales , Carcinoma/genética , Citometría de Flujo , Inestabilidad Genómica , Humanos , Masculino , Glándulas Mamarias Humanas/patología , Ratones , Proteínas Proto-Oncogénicas c-pim-1/genética , Ratas , Ratas Sprague-Dawley , Telomerasa/metabolismo
16.
J Biol Chem ; 282(35): 25790-800, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17602165

RESUMEN

Transcription factor haploinsufficiency plays a role in the pathogenesis of many diseases, including cancer. In a mouse model of prostate tumor initiation, loss of a single allele of the tumor suppressor Nkx3.1 stochastically inactivates the expression of a class of dosage-sensitive target genes. Here we show that dosage sensitivity is associated with the differential histone H3/H4 acetylation states of Nkx3.1 target genes. When histone acetylation is induced in Nkx3.1+/- mouse prostates with the histone deacetylase inhibitor Trichostatin A, Nkx3.1 can bind to and reactivate the expression of dosage-sensitive target genes. We incorporated our findings into a mathematical model that entails the association of Nkx3.1 with histone acetyltransferase activity. Subsequent experiments indicate that Nkx3.1 associates with and recruits the histone acetyltransferase p300/CREB-binding protein-associated factor to chromatin. Finally, we demonstrate a role for the dosage-sensitive target gene intelectin/omentin in suppressing prostate tumorigenicity. Our results reveal how the interplay between transcription factor dosage and chromatin affects target gene expression in tumor initiation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Ensamble y Desensamble de Cromatina , Dosificación de Gen , Proteínas de Homeodominio/biosíntesis , Pérdida de Heterocigocidad , Modelos Biológicos , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Acetilación , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromatina/genética , Cromatina/metabolismo , Cromatina/patología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Ácidos Hidroxámicos/farmacología , Lectinas/biosíntesis , Lectinas/genética , Pérdida de Heterocigocidad/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Masculino , Ratones , Ratones Mutantes , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
17.
Int J Biol Sci ; 3(5): 281-91, 2007 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-17505536

RESUMEN

To characterize the role of BRCA1 in mammary gland development and tumor suppression, a transgenic mouse model of BRCA1 overexpression was developed. Using the mouse mammary tumor virus (MMTV) promoter/enhancer, transgenic mice expressing human BRCA1 or select mutant controls were generated. Transgenic animals examined during adolescence were shown to express the human transgene in their mammary glands. The mammary glands of 13-week-old virgin homozygous MMTV-BRCA1 mice presented the morphology of moderately increased lobulo-alveolar development. The mammary ductal trees of both hemizygous and homozygous MMTV-BRCA1t340 were similar to those of control non-transgenic littermates. Interestingly, both hemi- and homozygous mice expressing a splice variant of BRCA1 lacking the N-terminal RING finger domain (MMTV-BRCA1sv) exhibited marked mammary lobulo-alveolar development, particularly terminal end bud proliferation. Morphometric analyses of mammary gland whole mount preparations were used to measure epithelial staining indices of ~35% for homozygous MMTV-BRCA1 mice and ~60% for both hemizygous and homozygous MMTV-BRCA1sv mice versus ~25% for non-transgenic mice. Homozygous MMTV-BRCA1 mice showed delayed development of tumors when challenged with 7,12 dimethylbenzanthracene (DMBA), relative to non-transgenic and homozygous BRCA1t340 expressing mice. In contrast, homozygous MMTV-BRCA1sv transgenic animals were sensitized to DMBA treatment and exhibited a very rapid onset of mammary tumor development and accelerated mortality. MMTV-BRCA1 effects on mortality were restricted to DMBA-induced tumors of the mammary gland. These results demonstrate in vivo roles for BRCA1 in both mammary gland development and in tumor suppression against mutagen-induced mammary gland neoplasia.


Asunto(s)
Proteína BRCA1/fisiología , Genes BRCA1 , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinógenos/toxicidad , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Glándulas Mamarias Animales/anatomía & histología , Neoplasias Mamarias Experimentales/inducido químicamente , Virus del Tumor Mamario del Ratón , Ratones , Ratones Endogámicos , Ratones Transgénicos , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA