Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Toxicol Appl Pharmacol ; 274(2): 200-8, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24269878

RESUMEN

Sarin is an organophosphate nerve agent that is among the most lethal chemical toxins known to mankind. Because of its vaporization properties and ease and low cost of production, sarin is the nerve agent with a strong potential for use by terrorists and rouge nations. The primary route of sarin exposure is through inhalation and, depending on the dose, sarin leads to acute respiratory failure and death. The mechanism(s) of sarin-induced respiratory failure is poorly understood. Sarin irreversibly inhibits acetylcholine esterase, leading to excessive synaptic levels of acetylcholine and, we have previously shown that sarin causes marked ventilatory changes including weakened response to hypoxia. We now show that LD50 sarin inhalation causes severe bronchoconstriction in rats, leading to airway resistance, increased hypoxia-induced factor-1α, and severe lung epithelium injury. Transferring animals into 60% oxygen chambers after sarin exposure improved the survival from about 50% to 75% at 24h; however, many animals died within hours after removal from the oxygen chambers. On the other hand, if LD50 sarin-exposed animals were administered the bronchodilator epinephrine, >90% of the animals survived. Moreover, while both epinephrine and oxygen treatments moderated cardiorespiratory parameters, the proinflammatory cytokine surge, and elevated expression of hypoxia-induced factor-1α, only epinephrine consistently reduced the sarin-induced bronchoconstriction. These data suggest that severe bronchoconstriction is a critical factor in the mortality induced by LD50 sarin inhalation, and epinephrine may limit the ventilatory, inflammatory, and lethal effects of sarin.


Asunto(s)
Broncoconstricción/efectos de los fármacos , Sustancias para la Guerra Química/toxicidad , Epinefrina/farmacología , Enfermedades Pulmonares/tratamiento farmacológico , Oxígeno/farmacología , Sarín/toxicidad , Enfermedad Aguda , Administración por Inhalación , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Inhibidores de la Colinesterasa/toxicidad , Relación Dosis-Respuesta a Droga , Precursores Enzimáticos/metabolismo , Gelatinasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Dosificación Letal Mediana , Pulmón/efectos de los fármacos , Pulmón/patología , Enfermedades Pulmonares/inducido químicamente , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Endogámicas F344 , Sarín/administración & dosificación
2.
Inhal Toxicol ; 24(13): 907-17, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23121300

RESUMEN

Tire and road wear particles (TRWP) are a component of ambient particulate matter (PM) produced from the interaction of tires with the roadway. Inhalation of PM has been associated with cardiopulmonary morbidities and mortalities thought to stem from pulmonary inflammation. To determine whether TRWP may contribute to these events, the effects of subacute inhalation of TRWP were evaluated in rats. TRWP were collected at a road simulator laboratory, aerosolized, and used to expose male and female Sprague-Dawley rats (n = 10/treatment group) at ~10, 40, or 100 µg/m³ TRWP via nose-only inhalation for 6 h/day for 28 days. Particle size distribution of the aerosolized TRWP was found to be within the respirable range for rats. Toxicity was assessed following OECD guidelines (TG 412). No TRWP-related effects were observed on survival, clinical observations, body or organ weights, gross pathology, food consumption, immune system endpoints, serum chemistry, or biochemical markers of inflammation or cytotoxicity. Rare to few focal areas of subacute inflammatory cell infiltration associated with TWRP exposure were observed in the lungs of one mid and four high exposure animals, but not the low-exposure animals. These alterations were minimal, widely scattered and considered insufficient in extent or severity to have an impact on pulmonary function. Furthermore, it is expected that these focal lesions would remain limited and may undergo resolution without long-term or progressive pulmonary alterations. Therefore, from this study we identified a no-observable-adverse-effect-level (NOAEL) of 112 µg/m³ of TRWP in rats for future use in risk assessment of TRWP.


Asunto(s)
Contaminantes Ambientales/toxicidad , Pulmón/efectos de los fármacos , Vehículos a Motor , Material Particulado/toxicidad , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Citocinas/metabolismo , Contaminantes Ambientales/química , Pulmón/metabolismo , Pulmón/patología , Nivel sin Efectos Adversos Observados , Tamaño de la Partícula , Material Particulado/química , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Pruebas de Toxicidad Subaguda
3.
Nat Med ; 8(10): 1171-4, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12219086

RESUMEN

With the advent of HIV and the widespread emergence of drug-resistant strains of Mycobacterium tuberculosis, newer control strategies in the form of a better vaccine could decrease the global incidence of tuberculosis. A desirable trait in an effective live attenuated vaccine strain is an ability to persist within the host in a limited fashion in order to produce important protective antigens in vivo. Attenuated M. tuberculosis vaccine candidates have been constructed by deleting genes required for growth in mice. These candidate vaccines did not elicit adequate protective immunity in animal models, due to their inability to persist sufficiently long within the host tissues. Here we report that an auxotrophic mutant of M. tuberculosis defective in the de novo biosynthesis of pantothenic acid (vitamin B5) is highly attenuated in immunocompromised SCID mice and in immunocompetent BALB/c mice. SCID mice infected with the pantothenate auxotroph survived significantly longer (250 days) than mice infected with either bacille Calmette-Guerin (BCG) vaccine or virulent M. tuberculosis (77 and 35 days, respectively). Subcutaneous immunization with this auxotroph conferred protection in C57BL/6J mice against an aerosol challenge with virulent M. tuberculosis, which was comparable with that afforded by BCG vaccination. Our findings highlight the importance of de novo pantothenate biosynthesis in limiting the intracellular survival and pathogenesis of M. tuberculosis without reducing its immunogenicity in vaccinated mice.


Asunto(s)
Mycobacterium tuberculosis/inmunología , Ácido Pantoténico/metabolismo , Vacunas contra la Tuberculosis , Tuberculosis/prevención & control , Animales , Genes Bacterianos , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Tasa de Supervivencia , Tuberculosis/inmunología , Tuberculosis/patología , Vacunas Atenuadas
4.
Mol Cell Biol ; 26(19): 7116-29, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16980615

RESUMEN

Obstacles to the expansion of cells with proliferative potential include the induction of cell death, telomere-based senescence, and the pRb and p53 tumor suppressors. Not infrequently, the molecular pathways regulating oncogenesis recapitulate aberrations of processes governing embryogenesis. The genetic network, consisting of the dachshund (dac), eyes absent (eya), eyeless, and sine oculis (so) genes, regulates cell fate determination in metazoans, with dac serving as a cointegrator through a So DNA-binding factor. Here, DACH1 inhibited oncogene-mediated breast oncogenesis, blocking breast cancer epithelial cell DNA synthesis, colony formation, growth in Matrigel, and tumor growth in mice. Genetic deletion studies demonstrated a requirement for cyclin D1 in DACH1-mediated inhibition of DNA synthesis. DACH1 repressed cyclin D1 through a novel mechanism via a c-Jun DNA-binding partner, requiring the DACH1 alpha-helical DS domain which recruits corepressors to the local chromatin. Analysis of over 2,000 patients demonstrated increased nuclear DACH1 expression correlated inversely with cellular mitosis and predicted improved breast cancer patient survival. The cell fate determination factor, DACH1, arrests breast tumor proliferation and growth in vivo providing a new mechanistic and potential therapeutic insight into this common disease.


Asunto(s)
Neoplasias de la Mama/patología , Ciclina D1/metabolismo , Proteínas del Ojo/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Ciclina D1/genética , ADN/biosíntesis , ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Fenotipo , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Transcripción AP-1/metabolismo , Ensayo de Tumor de Célula Madre , Proteínas ras/metabolismo
5.
Int J Toxicol ; 28(2): 99-112, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19482834

RESUMEN

An oral sulfate salt solution (OSS), under development as a bowel cleansing agent for colonoscopy in humans, is studied in rats and dogs. In rats, amaximumpractical oral OSS dose (5 g/kg/d) is compared with an oral sodium phosphate (OSP) solution, both at about 7 times the clinical dose. OSS induces the intended effects of loose stools and diarrhea. In rats, higher urine sodium and potassium accompany higher clearance rates, considered adaptive to the osmotic load of OSS. OSS for 28 days is well tolerated in rats and dogs. In contrast, OSP causes increased mortality, reduced body weight and food consumption, severe kidney tubular degeneration, and calcium phosphate deposition in rats. These are accompanied by mineralization in the stomach and aorta, along with cardiac and hepatic degeneration and necrosis. The greater safety margin of OSS over OSP at similarmultiples of the clinical dose indicates its suitability for human use.


Asunto(s)
Colonoscopía , Riñón/efectos de los fármacos , Fosfatos/toxicidad , Sulfatos/toxicidad , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Calcio/sangre , Creatinina/metabolismo , Perros , Femenino , Riñón/patología , Masculino , Potasio/orina , Ratas , Sodio/sangre , Sodio/orina
6.
J Clin Invest ; 115(5): 1163-76, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15841211

RESUMEN

The interactions of transformed cells with the surrounding stromal cells are of importance for tumor progression and metastasis. The relevance of adipocyte-derived factors to breast cancer cell survival and growth is well established. However, it remains unknown which specific adipocyte-derived factors are most critical in this process. Collagen VI is abundantly expressed in adipocytes. Collagen(-/-) mice in the background of the mouse mammary tumor virus/polyoma virus middle T oncogene (MMTV-PyMT) mammary cancer model demonstrate dramatically reduced rates of early hyperplasia and primary tumor growth. Collagen VI promotes its growth-stimulatory and pro-survival effects in part by signaling through the NG2/chondroitin sulfate proteoglycan receptor expressed on the surface of malignant ductal epithelial cells to sequentially activate Akt and beta-catenin and stabilize cyclin D1. Levels of the carboxyterminal domain of collagen VIalpha3, a proteolytic product of the full-length molecule, are dramatically upregulated in murine and human breast cancer lesions. The same fragment exerts potent growth-stimulatory effects on MCF-7 cells in vitro. Therefore, adipocytes play a vital role in defining the ECM environment for normal and tumor-derived ductal epithelial cells and contribute significantly to tumor growth at early stages through secretion and processing of collagen VI.


Asunto(s)
Adipocitos/metabolismo , Colágeno Tipo VI/metabolismo , Neoplasias Mamarias Animales/metabolismo , Animales , Colágeno Tipo VI/deficiencia , Colágeno Tipo VI/genética , Ciclina D1/metabolismo , Proteínas del Citoesqueleto/metabolismo , Femenino , Inmunohistoquímica , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Poliomavirus/metabolismo , Transactivadores/metabolismo , beta Catenina
7.
Mol Cell Biol ; 25(9): 3563-74, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15831462

RESUMEN

The inhibitor of differentiation Id2 is a target of the retinoblastoma (Rb) protein during mouse embryogenesis. In Rb(+/-) mice, LOH at the wild-type Rb allele initiates pituitary adenocarcinoma, a tumor derived from embryonic melanotropes. Here we identify a critical role for Id2 in initiation, growth, and angiogenesis of pituitary tumors from Rb(+/-) mice. We show that proliferation and differentiation are intimately coupled in Rb(+/-) pituitary cells before tumor initiation. In Id2-null pituitaries, premature activation of basic helix-loop-helix-mediated transcription and expression of the cdk inhibitor p27(Kip1) impairs the proliferation of melanotropes and tumor initiation. Without Id2, Rb(+/-) mice have fewer early tumor lesions and a markedly decreased proliferation rate of the tumor foci. Expression of Id2 by pituitary tumor cells promotes growth and angiogenesis by functioning as a master regulator of vascular endothelial growth factor (VEGF). In human neuroblastoma, the N-Myc-driven expression of Id2 is sufficient and necessary for expression of VEGF. These results establish that aberrant Id2 activity directs initiation and progression of embryonal cancer.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neovascularización Patológica/genética , Neuroblastoma/genética , Neoplasias Hipofisarias/genética , Proteínas Represoras/fisiología , Proteína de Retinoblastoma/genética , Factores de Transcripción/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Ciclina G , Ciclina G1 , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Proteínas de Unión al ADN/genética , Proteína 2 Inhibidora de la Diferenciación , Ratones , Ratones Mutantes , Mutación/genética , Hipófisis/patología , Neoplasias Hipofisarias/irrigación sanguínea , Proteínas Represoras/genética , Proteína de Retinoblastoma/fisiología , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/metabolismo
8.
Mol Biol Cell ; 13(10): 3416-30, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12388746

RESUMEN

It is well established that mammary gland development and lactation are tightly controlled by prolactin signaling. Binding of prolactin to its cognate receptor (Prl-R) leads to activation of the Jak-2 tyrosine kinase and the recruitment/tyrosine phosphorylation of STAT5a. However, the mechanisms for attenuating the Prl-R/Jak-2/STAT5a signaling cascade are just now being elucidated. Here, we present evidence that caveolin-1 functions as a novel suppressor of cytokine signaling in the mammary gland, akin to the SOCS family of proteins. Specifically, we show that caveolin-1 expression blocks prolactin-induced activation of a STAT5a-responsive luciferase reporter in mammary epithelial cells. Furthermore, caveolin-1 expression inhibited prolactin-induced STAT5a tyrosine phosphorylation and DNA binding activity, suggesting that caveolin-1 may negatively regulate the Jak-2 tyrosine kinase. Because the caveolin-scaffolding domain bears a striking resemblance to the SOCS pseudosubstrate domain, we examined whether Jak-2 associates with caveolin-1. In accordance with this homology, we demonstrate that Jak-2 cofractionates and coimmunoprecipitates with caveolin-1. We next tested the in vivo relevance of these findings using female Cav-1 (-/-) null mice. If caveolin-1 normally functions as a suppressor of cytokine signaling in the mammary gland, then Cav-1 null mice should show premature development of the lobuloalveolar compartment because of hyperactivation of the prolactin signaling cascade via disinhibition of Jak-2. In accordance with this prediction, Cav-1 null mice show accelerated development of the lobuloalveolar compartment, premature milk production, and hyperphosphorylation of STAT5a (pY694) at its Jak-2 phosphorylation site. In addition, the Ras-p42/44 MAPK cascade is hyper-activated. Because a similar premature lactation phenotype is observed in SOCS1 (-/-) null mice, we conclude that caveolin-1 is a novel suppressor of cytokine signaling.


Asunto(s)
Caveolinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Trastornos de la Lactancia , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Represoras , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factores de Transcripción , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Caveolina 1 , Caveolinas/genética , Línea Celular , Membrana Celular/metabolismo , Regulación hacia Abajo/fisiología , Activación Enzimática , Células Epiteliales/metabolismo , Estrógenos/metabolismo , Femenino , Genes Reporteros , Janus Quinasa 2 , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Leche/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Embarazo , Progesterona/metabolismo , Prolactina/metabolismo , Proteínas/genética , Factor de Transcripción STAT5 , Alineación de Secuencia , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
9.
Cancer Res ; 65(9): 3950-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867396

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) represents a ligand-dependent nuclear receptor family that regulates multiple metabolic processes associated with fatty acid beta-oxidation, glucose utilization, and cholesterol transport. These and other receptor-mediated actions pertain to their role in hypolipidemic and antidiabetic therapies and as potential targets for cancer chemopreventive agents. The present study evaluated the chemopreventive activity of two highly potent and selective PPARgamma and PPARdelta agonists in a progestin- and carcinogen-induced mouse mammary tumorigenesis model. Animals treated with the PPARgamma agonist GW7845 exhibited a moderate delay in tumor formation. In contrast, animals treated with the PPARdelta agonist GW501516 showed accelerated tumor formation. Significantly, tumors from GW7845-treated mice were predominantly ductal adenocarcinomas, whereas tumors from GW501516-treated animals were adenosquamous and squamous cell carcinomas. Gene expression analysis of tumors arising from GW7845- and GW501516-treated mice identified expression profiles that were distinct from each other and from untreated control tumors of the same histopathology. Only tumors from mice treated with the PPARgamma agonist expressed estrogen receptor-alpha in luminal transit cells, suggesting increased ductal progenitor cell expansion. Tumors from mice treated with the PPARdelta agonist exhibited increased PPARdelta levels and activated 3-phosphoinositide-dependent protein kinase-1 (PDK1), which co-associated, suggesting a link between the known oncogenic activity of PDK1 in mammary epithelium and PPARdelta activation. These results indicate that PPARdelta and PPARgamma agonists produce diverse, yet profound effects on mammary tumorigenesis that give rise to distinctive histopathologic patterns of tumor differentiation and tumor development.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma Ductal/prevención & control , Neoplasias Mamarias Experimentales/prevención & control , Oxazoles/farmacología , PPAR delta/agonistas , PPAR gamma/agonistas , Tiazoles/farmacología , Tirosina/análogos & derivados , Tirosina/farmacología , Animales , Carcinoma Adenoescamoso/inducido químicamente , Carcinoma Adenoescamoso/tratamiento farmacológico , Carcinoma Adenoescamoso/patología , Carcinoma Adenoescamoso/prevención & control , Carcinoma Ductal/inducido químicamente , Carcinoma Ductal/tratamiento farmacológico , Carcinoma Ductal/patología , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/prevención & control , Diferenciación Celular/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones
10.
Cancer Res ; 64(20): 7220-5, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15492237

RESUMEN

Oncogenic signals elevate expression of Id2 in multiple tumor types. When deregulated, Id2 inactivates the tumor suppressor proteins retinoblastoma, p107, and p130. Here, we report a novel and unexpected tumor inhibitory function of Id2 in the intestinal epithelium. First, genetic ablation of Id2 in the mouse prevents differentiation and cell cycle arrest of enterocytes at the time of formation of the crypt-villus unit. Later, these developmental abnormalities evolve toward neoplastic transformation with complete penetrance. Id2-null tumors contain severe dysplastic and metaplastic lesions and express aberrant amounts of beta-catenin. Thus, our data are the first to establish a direct requirement of basic helix-loop-helix inhibitors in driving differentiation and define an unexpected role for the retinoblastoma-binding protein Id2 in preventing tumor formation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Mucosa Intestinal/patología , Neoplasias Intestinales/patología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Femenino , Proteína 2 Inhibidora de la Diferenciación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
11.
Cancer Res ; 64(12): 4122-30, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15205322

RESUMEN

The Ink4a/Arf locus encodes two structurally unrelated tumor suppressor proteins, p16(INK4a) and p14(ARF) (murine p19(ARF)). Invariant inactivation of either the p16(INK4a)-cyclin D/CDK-pRb pathway and/or p53-p14(ARF) pathway occurs in most human tumors. Cyclin D1 is frequently overexpressed in breast cancer cells contributing an alternate mechanism inactivating the p16(INK4a)/pRb pathway. Targeted overexpression of cyclin D1 to the mammary gland is sufficient for tumorigenesis, and cyclin D1-/- mice are resistant to Ras-induced mammary tumors. Recent studies suggest cyclin D1 and p16(INK4a) expression are reciprocal in human breast cancers. Herein, reciprocal regulation of cyclin D1 and p16(INK4a) was observed in tissues of mice mutant for the Ink4a/Arf locus. p16(INK4a) and p19(ARF) inhibited DNA synthesis in MCF7 cells. p16(INK4a) repressed cyclin D1 expression and transcription. Repression of cyclin D1 by p16(INK4a) occurred independently of the p16(INK4a)-cdk4-binding function and required a cAMP-response element/activating transcription factor-2-binding site. p19(ARF) repressed cyclin D1 through a novel distal cis-element at -1137, which bound p53 in chromatin-immunoprecipitation assays. Transcriptional repression of the cyclin D1 gene through distinct DNA sequences may contribute to the tumor suppressor function of the Ink4a/Arf locus.


Asunto(s)
Ciclina D1/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteína p14ARF Supresora de Tumor/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclina D1/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/biosíntesis , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transcripción Genética , Transfección , Proteína p14ARF Supresora de Tumor/biosíntesis
12.
Cancer Res ; 63(12): 3395-402, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810676

RESUMEN

Most human tumors display inactivation of the p53 and the p16(INK4)/pRb pathway. The Ink4a/alternative reading frame (ARF) locus encodes the p16(INK4a) and p14(ARF) (murine p19(ARF)) proteins. p16(INK4a) is deleted in 40-60% of breast cancer cell lines, and p16(INK4a) inactivation by DNA methylation occurs in < or =30% of human breast cancers. In mice genetically heterozygous for p16(INK4a) or Ink4a/Arf, predisposition to specific tumor types is enhanced. Ink4a/Arf(+/-) mice have increased E micro -Myc-induced lymphomagenesis and epidermal growth factor receptor-induced gliomagenesis. ErbB2 (epidermal growth factor receptor-related protein B2) is frequently overexpressed in human breast cancer and is sufficient for mammary tumorigenesis in vivo. We determined the role of heterozygosity at the Ink4a/Arf locus in ErbB2-induced mammary tumorigenesis. Compared with mouse mammary tumor virus-ErbB2 Ink4a/Arf(+/-) mice, mouse mammary tumor virus-ErbB2 Ink4a/Arf(wt) mammary tumors showed increased p16(INK4a), reduced Ki-67 expression, and reduced cyclin D1 protein but increased mammary tumor apoptosis with no significant change in the risk of developing mammary tumors. These studies demonstrate the contribution of Ink4a/Arf heterozygosity to tumor progression is tissue specific in vivo. In view of the important role of Ink4a/Arf in response to chemotherapy, these transgenic mice may provide a useful model for testing breast tumor therapies.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Genes erbB-2 , Genes p16 , Neoplasias Mamarias Experimentales/genética , Proteína p14ARF Supresora de Tumor/fisiología , Adenocarcinoma/patología , Aneuploidia , Animales , Apoptosis , Neoplasias de la Mama/patología , Ciclo Celular , Transformación Celular Viral/genética , Cruzamientos Genéticos , Ciclina D1/biosíntesis , Ciclina D1/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Antígeno Ki-67/biosíntesis , Antígeno Ki-67/genética , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos , Transfección
13.
Mol Cancer Ther ; 1(13): 1191-200, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12479700

RESUMEN

A number of cancer chemotherapeutic drugs designed to have cytotoxic actions on tumor cells have recently been shown to also have antiangiogenic activities. Endothelial cell migration and proliferation are key components of tumor angiogenesis, and agents that target the microtubule cytoskeleton can interfere with these processes. In this study, the effect on endothelial cell functions of the microtubule-stabilizing drugs Taxotere and Taxol were evaluated in three in vitro assays: a chemokinetic migration assay, an angiogenesis factor-mediated chemotactic migration assay, and a three-dimensional Matrigel tubule formation assay, using rat fat pad endothelial cells (RFPECs) and/or human umbilical vein endothelial cells (HUVECs). Taxotere was active in all three assays at concentrations that were not cytotoxic and did not inhibit endothelial cell proliferation. In the RFPEC chemokinetic migration and in vitro tubule formation assays, the IC50 values were approximately 10(-9) M for both Taxotere and Taxol. HUVEC migration, however, was more sensitive to Taxotere, with an observed IC50 of 10(-12) M in a chemokinetic assay. In a Boyden chamber assay, HUVEC chemotaxis stimulated by either of two angiogenic factors, thymidine phosphorylase or vascular endothelial growth factor, was inhibited by Taxotere with an IC50 of 10(-11) M and was ablated at 10(-9) M. Taxotere was also up to 1000-fold more potent than Taxol in inhibiting either chemokinetic or chemotactic migration. When the microtubule cytoskeleton was visualized using immunofluorescence staining of alpha-tubulin, there were no gross morphological changes observed in HUVECs or RFPECs treated with Taxotere at concentrations that inhibited endothelial cell migration but not proliferation. The effects of Taxotere on migration were associated with a reduction in the reorientation of the cell's centrosome, at concentrations that did not affect gross microtubule morphology or proliferation. Reorientation of the centrosome, which acts as the microtubule organizing center, in the intended direction of movement is a critical early step in the stabilization of directed cell migration. These data indicate that endothelial cell migration correlates more closely with changes in microtubule plasticity than with microtubule gross structure. The antiangiogenic activity of Taxotere in vivo was assessed in a Matrigel plug assay. In this assay, the angiogenic response to fibroblast growth factor 2 was inhibited in vivo by Taxotere with an ID50 of 5.4 mg/kg when injected twice weekly over a 14-day period, and angiogenesis was completely blocked in mice that received 10 mg/kg Taxotere. The in vivo data further suggested that Taxotere had selectivity for endothelial cell migration and/or microvessel formation because infiltration of inflammatory cells into the Matrigel plug was much less sensitive to inhibition by Taxotere. In conclusion, Taxotere is a potent and potentially specific inhibitor of endothelial cell migration in vitro and angiogenesis in vitro and in vivo.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Endotelio Vascular/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Neovascularización Patológica/prevención & control , Paclitaxel/análogos & derivados , Paclitaxel/farmacología , Taxoides , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Centrosoma , Colágeno , Docetaxel , Combinación de Medicamentos , Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Laminina , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Proteoglicanos , Ratas , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/trasplante , Venas Umbilicales/citología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Endocrinology ; 145(1): 367-83, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14576179

RESUMEN

Adiponectin is a plasma protein expressed exclusively in adipose tissue. Adiponectin levels are linked to insulin sensitivity, but a direct effect of chronically elevated adiponectin on improved insulin sensitivity has not yet been demonstrated. We identified a dominant mutation in the collagenous domain of adiponectin that elevated circulating adiponectin values in mice by 3-fold. Adiponectinemia raised lipid clearance and lipoprotein lipase activity, and suppressed insulin-mediated endogenous glucose production. The induction of adiponectin during puberty and the sexual dimorphism in adult adiponectin values were preserved in these transgenic animals. As a result of elevated adiponectin, serum PRL values and brown adipose mass both increased. The effects on carbohydrate and lipid metabolism were associated with elevated phosphorylation of 5'-AMP-activated protein kinase in liver and elevated expression of peroxisomal proliferator-activated receptor gamma2, caveolin-1, and mitochondrial markers in white adipose tissue. These studies strongly suggest that increasing endogenous adiponectin levels has direct effects on insulin sensitivity and may induce similar physiological responses as prolonged treatment with peroxisomal proliferator-activated receptor gamma agonists.


Asunto(s)
Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intercelular , Proteínas/genética , Proteínas/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adiponectina , Tejido Adiposo/anatomía & histología , Tejido Adiposo/metabolismo , Animales , Composición Corporal , Calorimetría Indirecta , Colágeno/genética , Ingestión de Alimentos , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Técnica de Clampeo de la Glucosa , Intolerancia a la Glucosa/metabolismo , Ratones , Ratones Transgénicos , Prolactina/sangre , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/agonistas , Factores de Transcripción/agonistas , Transcripción Genética
15.
Biochem Pharmacol ; 64(5-6): 827-36, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213576

RESUMEN

The neu (c-erbB-2, HER2) proto-oncogene encodes a receptor tyrosine kinase that is a member of an important growth factor receptor family which includes the epidermal growth factor receptor (EGFR, ErbB1), ErbB3 and ErbB4. The neu is found over-expressed in 20-30% of human breast tumors. The c-erbB-2 is sufficient for the induction of mammary tumorigenesis in transgenic mice and the pathology of these mammary tumors strongly resembles human breast cancer. Murine transgenic models engineered to recapitulate human breast cancer provide an excellent and straightforward approach to dissect the molecular mechanisms governing the onset and progression of this disease. The molecular mechanisms by which ErbB-2 transforms cells involves direct effects on components of the cell-cycle regulatory apparatus. Recent studies have demonstrated a key role for components of the cell-cycle, in particular cyclin D1 and p27Kip1 (p27) in the onset and progression of ErbB-2-induced murine mammary tumorigenesis. Such studies have provided further impetus to therapeutics targeting these cell-cycle proteins.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Ciclina D1/fisiología , Genes Supresores de Tumor/fisiología , Neoplasias Mamarias Animales/fisiopatología , Receptor ErbB-2/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Pruebas de Carcinogenicidad , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Modelos Animales de Enfermedad , Femenino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Proto-Oncogenes Mas
16.
Proc Natl Acad Sci U S A ; 104(18): 7438-43, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17460049

RESUMEN

The serine threonine kinase Akt1 has been implicated in the control of cellular metabolism, survival and growth. Here, disruption of the ubiquitously expressed member of the Akt family of genes, Akt1, in the mouse demonstrates a requirement for Akt1 in ErbB2-induced mammary tumorigenesis. Akt1 deficiency delayed tumor growth and reduced lung metastases, correlating with a reduction in phosphorylation of the Akt1 target, tuberous sclerosis 2 (TSC2) at Ser-939. Akt1-deficient mammary epithelial tumor cells (MEC) were reduced in size and proliferative capacity, with reduced cyclin D1 and p27(KIP1) abundance. Akt1 deficiency abrogated the oncogene-induced changes in polarization of MEC in three-dimensional culture and reverted oncogene-induced relocalization of the phosphorylated ezrin-radixin-moesin proteins. Akt1 increased MEC migration across an endothelial cell barrier, enhancing the persistence of migratory directionality. An unbiased proteomic analysis demonstrated Akt1 mediated MEC migration through paracrine signaling via induction of expression and secretion of CXCL16 and MIP1gamma. Akt1 governs MEC polarity, migratory directionality and breast cancer onset induced by ErbB2 in vivo.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Neoplasias de la Mama/genética , Aumento de la Célula , Movimiento Celular , Polaridad Celular , Proliferación Celular , Transformación Celular Neoplásica/patología , Células Cultivadas , Progresión de la Enfermedad , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
17.
Immunology ; 120(2): 192-206, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17076705

RESUMEN

The global epidemic of tuberculosis, fuelled by acquired immune-deficiency syndrome, necessitates the development of a safe and effective vaccine. We have constructed a DeltaRD1DeltapanCD mutant of Mycobacterium tuberculosis (mc(2)6030) that undergoes limited replication and is severely attenuated in immunocompromised mice, yet induces significant protection against tuberculosis in wild-type mice and even in mice that completely lack CD4(+) T cells as a result of targeted disruption of their CD4 genes (CD4(-/-) mice). Ex vivo studies of T cells from mc(2)6030-immunized mice showed that these immune cells responded to protein antigens of M. tuberculosis in a major histocompatibility complex (MHC) class II-restricted manner. Antibody depletion experiments showed that antituberculosis protective responses in the lung were not diminished by removal of CD8(+), T-cell receptor gammadelta (TCR-gammadelta(+)) and NK1.1(+) T cells from vaccinated CD4(-/-) mice before challenge, implying that the observed recall and immune effector functions resulting from vaccination of CD4(-/-) mice with mc(2)6030 were attributable to a population of CD4(-) CD8(-) (double-negative) TCR-alphabeta(+), TCR-gammadelta(-), NK1.1(-) T cells. Transfer of highly enriched double-negative TCR-alphabeta(+) T cells from mc(2)6030-immunized CD4(-/-) mice into naive CD4(-/-) mice resulted in significant protection against an aerosol tuberculosis challenge. Enriched pulmonary double-negative T cells transcribed significantly more interferon-gamma and interleukin-2 mRNA than double-negative T cells from naive mice after a tuberculous challenge. These results confirmed previous findings on the potential for a subset of MHC class II-restricted T cells to develop and function without expression of CD4 and suggest novel vaccination strategies to assist in the control of tuberculosis in human immunodeficiency virus-infected humans who have chronic depletion of their CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Huésped Inmunocomprometido , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control , Traslado Adoptivo , Animales , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Inmunidad Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-2/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos , ARN Mensajero/genética , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Vacunación/métodos , Vacunas Atenuadas/inmunología
18.
Toxicol Pathol ; 34(6): 699-707, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17074738

RESUMEN

We present a status report from the NCI Mouse Models of Human Cancers Consortium (MMHCC) Precancers Workshop held November 8 and 9, 2004. An expert panel, the Mouse Models Group (MMG) evaluated the status of mouse models of precancer emphasizing genetically engineered mouse models, especially of lining epithelium and their utilitarian value to human carcinogenesis. An outline of the background for the panel's considerations is provided with examples of past and current precancerous lesions in mice. The experimental use of oncogenic viruses and chemical carcinogens in mice led to operational definitions of initiation, promotion, and preneoplasia Preneoplastic and precancerous lesions are found in these models. In this precancer concept, most preneoplastic lesions are considered as potentially precancerous or at least an earlier stage in cancer development than typical pre-invasive epithelial lesions, which are often seen in these mouse models. Genetically engineered mice, used to test the oncogenicity of individual genes, develop precancers that are initiated by defined molecular and histopathologic changes. The mouse can be used to isolate and study precancers in detail, thereby providing a level of biological understanding not readily available in clinical disease. These studies suggest that genetically engineered mice are very useful preclinical models for chemoprevention and therapy.


Asunto(s)
Carcinógenos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/genética , Infecciones Tumorales por Virus/genética , Animales , Antineoplásicos/uso terapéutico , Carcinoma in Situ/inducido químicamente , Carcinoma in Situ/genética , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Ensayos de Selección de Medicamentos Antitumorales , Células Epiteliales/patología , Humanos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Oncogenes/genética , Virus Oncogénicos/genética , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/prevención & control , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Infecciones Tumorales por Virus/patología
19.
Mol Carcinog ; 44(1): 42-50, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15937957

RESUMEN

Mouse mammary tumors arising during medroxyprogesterone-DMBA-mediated mammary carcinogenesis comprised three distinct phenotypes: adenocarcinoma, squamous cell carcinoma, and myoepithelial carcinoma. The molecular signature for each of the three tumor subsets was characterized by gene microarray analysis, and three distinct sets of gene expression profiles were obtained that were corroborated in part by quantitative RT-PCR and immunohistochemistry. These results suggest that this carcinogenesis and gene expression model will be useful for rapidly assessing the histopathological differences arising in mammary carcinogenesis and the effects of tumor promoting or chemoprevention agents.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/genética , Medroxiprogesterona/farmacología , Animales , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Inmunohistoquímica , Neoplasias Mamarias Experimentales/clasificación , Neoplasias Mamarias Experimentales/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Urology ; 66(5): 1127-33, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16286152

RESUMEN

OBJECTIVES: To perform a comparison to determine which of two methods of partial urethral ligation produces the most consistent outcome and fewest side effects. Such a study has not been previously reported. Partial urethral ligation is a means of causing reproducible bladder outlet obstruction. In the male rat model, partial urethral obstruction can be performed either by perineal incision and bulbous urethral ligation or retropubic incision and midprostatic obstruction. METHODS: Fifteen male Sprague-Dawley rats were studied. Five were selected for bulbous urethral obstruction through a perineal incision, five for midprostatic obstruction using a retropubic approach, and five for a sham operation through a perineal incision. RESULTS: The operative time was shorter and morbidity lower with the perineal approach compared with the retropubic approach. Inflammation or infection, or both, were seen in the prostate, bladder, proximal urethra, ureters, and kidneys in the rats in which a midprostatic obstruction was performed. The proximal urethra and prostate were mildly inflamed in those rats that underwent bulbous obstruction. Sham-operated rats exhibited mild prostatitis only. CONCLUSIONS: The perineal approach to the bulbous urethra is the method of choice for creating a partial urethral obstruction model of bladder outlet obstruction in the male rat.


Asunto(s)
Modelos Animales de Enfermedad , Obstrucción Uretral , Obstrucción del Cuello de la Vejiga Urinaria , Animales , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA