Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(1): 125-130, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27999180

RESUMEN

Kabuki syndrome is a Mendelian intellectual disability syndrome caused by mutations in either of two genes (KMT2D and KDM6A) involved in chromatin accessibility. We previously showed that an agent that promotes chromatin opening, the histone deacetylase inhibitor (HDACi) AR-42, ameliorates the deficiency of adult neurogenesis in the granule cell layer of the dentate gyrus and rescues hippocampal memory defects in a mouse model of Kabuki syndrome (Kmt2d+/ßGeo). Unlike a drug, a dietary intervention could be quickly transitioned to the clinic. Therefore, we have explored whether treatment with a ketogenic diet could lead to a similar rescue through increased amounts of beta-hydroxybutyrate, an endogenous HDACi. Here, we report that a ketogenic diet in Kmt2d+/ßGeo mice modulates H3ac and H3K4me3 in the granule cell layer, with concomitant rescue of both the neurogenesis defect and hippocampal memory abnormalities seen in Kmt2d+/ßGeo mice; similar effects on neurogenesis were observed on exogenous administration of beta-hydroxybutyrate. These data suggest that dietary modulation of epigenetic modifications through elevation of beta-hydroxybutyrate may provide a feasible strategy to treat the intellectual disability seen in Kabuki syndrome and related disorders.


Asunto(s)
Anomalías Múltiples/dietoterapia , Dieta Cetogénica/métodos , Cara/anomalías , Enfermedades Hematológicas/dietoterapia , Hipocampo/metabolismo , Histonas/biosíntesis , Discapacidad Intelectual/dietoterapia , Neurogénesis/fisiología , Enfermedades Vestibulares/dietoterapia , Ácido 3-Hidroxibutírico/metabolismo , Anomalías Múltiples/genética , Animales , Modelos Animales de Enfermedad , Enfermedades Hematológicas/genética , Hipocampo/citología , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína de la Leucemia Mieloide-Linfoide/genética , Neurogénesis/genética , Enfermedades Vestibulares/genética
2.
Nature ; 503(7474): 126-30, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24107997

RESUMEN

In systemic sclerosis (SSc), a common and aetiologically mysterious form of scleroderma (defined as pathological fibrosis of the skin), previously healthy adults acquire fibrosis of the skin and viscera in association with autoantibodies. Familial recurrence is extremely rare and causal genes have not been identified. Although the onset of fibrosis in SSc typically correlates with the production of autoantibodies, whether they contribute to disease pathogenesis or simply serve as a marker of disease remains controversial and the mechanism for their induction is largely unknown. The study of SSc is hindered by a lack of animal models that recapitulate the aetiology of this complex disease. To gain a foothold in the pathogenesis of pathological skin fibrosis, we studied stiff skin syndrome (SSS), a rare but tractable Mendelian disorder leading to childhood onset of diffuse skin fibrosis with autosomal dominant inheritance and complete penetrance. We showed previously that SSS is caused by heterozygous missense mutations in the gene (FBN1) encoding fibrillin-1, the main constituent of extracellular microfibrils. SSS mutations all localize to the only domain in fibrillin-1 that harbours an Arg-Gly-Asp (RGD) motif needed to mediate cell-matrix interactions by binding to cell-surface integrins. Here we show that mouse lines harbouring analogous amino acid substitutions in fibrillin-1 recapitulate aggressive skin fibrosis that is prevented by integrin-modulating therapies and reversed by antagonism of the pro-fibrotic cytokine transforming growth factor ß (TGF-ß). Mutant mice show skin infiltration of pro-inflammatory immune cells including plasmacytoid dendritic cells, T helper cells and plasma cells, and also autoantibody production; these findings are normalized by integrin-modulating therapies or TGF-ß antagonism. These results show that alterations in cell-matrix interactions are sufficient to initiate and sustain inflammatory and pro-fibrotic programmes and highlight new therapeutic strategies.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Contractura/tratamiento farmacológico , Contractura/patología , Integrinas/efectos de los fármacos , Integrinas/metabolismo , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Enfermedades Cutáneas Genéticas/tratamiento farmacológico , Enfermedades Cutáneas Genéticas/patología , Secuencias de Aminoácidos/genética , Sustitución de Aminoácidos/genética , Animales , Anticuerpos Antinucleares/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Autoinmunidad/inmunología , Contractura/inmunología , Contractura/prevención & control , Células Dendríticas/efectos de los fármacos , Femenino , Fibrilina-1 , Fibrilinas , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Fibrosis/prevención & control , Masculino , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Mutación Missense/genética , Células Plasmáticas/efectos de los fármacos , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/prevención & control , Enfermedades Cutáneas Genéticas/inmunología , Enfermedades Cutáneas Genéticas/prevención & control , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/inmunología
3.
J Infect Dis ; 214(1): 122-9, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908749

RESUMEN

BACKGROUND: Chronic inflammation and composition of the colon microbiota have been associated with colorectal cancer in humans. The human commensal enterotoxigenic Bacteroides fragilis (ETBF) is linked to both inflammatory bowel disease and colorectal cancer and, in our murine model, causes interleukin 17A (IL-17A)-dependent colon tumors. In these studies, we hypothesized that persistent colonization by ETBF is required for tumorigenesis. METHODS: We established a method for clearing ETBF in mice, using the antibiotic cefoxitin. Multiple intestinal neoplasia mice were colonized with ETBF for the experiment duration or were cleared of infection after 5 or 14 days. Gross tumors and/or microadenomas were then evaluated. In parallel, IL-17A expression was evaluated in wild-type littermates. RESULTS: Cefoxitin treatment resulted in complete and durable clearance of ETBF colonization. We observed a stepwise increase in median colon tumor numbers as the duration of ETBF colonization increased before cefoxitin treatment. ETBF eradication also significantly decreased mucosal IL-17A expression. CONCLUSIONS: The timing of ETBF clearance profoundly influences colon adenoma formation, defining a period during which the colon is susceptible to IL-17A-dependent tumorigenesis in this murine model. This model system can be used to study the microbiota-dependent and molecular mechanisms contributing to IL-17A-dependent colon tumor initiation.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Cefoxitina/efectos adversos , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/complicaciones , Neoplasias del Colon/tratamiento farmacológico , Enterotoxinas/efectos adversos , Enterotoxinas/uso terapéutico , Animales , Bacteroides fragilis/química , Colon/microbiología , Neoplasias del Colon/microbiología , Humanos , Ratones
4.
Kidney Int ; 90(1): 90-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27165822

RESUMEN

Abnormal proliferation of cyst-lining epithelium and increased intracystic fluid secretion via the cystic fibrosis transmembrane conductance regulator (CFTR) are thought to contribute to cyst growth in autosomal dominant polycystic kidney disease (ADPKD). Histone deacetylase 6 (HDAC6) expression and activity are increased in certain cancers, neurodegenerative diseases, and in Pkd1-mutant renal epithelial cells. Inhibition of HDAC6 activity with specific inhibitors slows cancer growth. Here we studied the effect of tubacin, a specific HDAC6 inhibitor, on cyst growth in polycystic kidney disease. Treatment with tubacin prevented cyst formation in MDCK cells, an in vitro model of cystogenesis. Cyclic AMP stimulates cell proliferation and activates intracystic CFTR-mediated chloride secretion in ADPKD. Treatment with tubacin downregulated cyclic AMP levels, inhibited cell proliferation, and inhibited cyclic AMP-activated CFTR chloride currents in MDCK cells. We also found that tubacin reduced cyst growth by inhibiting proliferation of cyst-lining epithelial cells, downregulated cyclic AMP levels, and improved renal function in a Pkd1-conditional mouse model of ADPKD. Thus, HDAC6 could play a role in cyst formation and could serve as a potential therapeutic target in ADPKD.


Asunto(s)
Anilidas/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Riñón/efectos de los fármacos , Riñón Poliquístico Autosómico Dominante/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Cloruros/sangre , Cloruros/metabolismo , AMP Cíclico/sangre , Modelos Animales de Enfermedad , Perros , Regulación hacia Abajo , Células Epiteliales/metabolismo , Femenino , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Riñón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética
5.
Gynecol Oncol ; 141(3): 580-587, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27001612

RESUMEN

OBJECTIVES: Although uterine cancer is the fourth most common cause for cancer death in women worldwide, the molecular underpinnings of tumor progression remain poorly understood. The High Mobility Group A1 (HMGA1) gene is overexpressed in aggressive cancers and high levels portend adverse outcomes in diverse tumors. We previously reported that Hmga1a transgenic mice develop uterine tumors with complete penetrance. Because HMGA1 drives tumor progression by inducing MatrixMetalloproteinase (MMP) and other genes involved in invasion, we explored the HMGA1-MMP-2 pathway in uterine cancer. METHODS: To investigate MMP-2 in uterine tumors driven by HMGA1, we used a genetic approach with mouse models. Next, we assessed HMGA1 and MMP-2 expression in primary human uterine tumors, including low-grade carcinomas (endometrial endometrioid) and more aggressive tumors (endometrial serous carcinomas, uterine carcinosarcomas/malignant mesodermal mixed tumors). RESULTS: Here, we report for the first time that uterine tumor growth is impaired in Hmga1a transgenic mice crossed on to an Mmp-2 deficient background. In human tumors, we discovered that HMGA1 is highest in aggressive carcinosarcomas and serous carcinomas, with lower levels in the more indolent endometrioid carcinomas. Moreover, HMGA1 and MMP-2 were positively correlated, but only in a subset of carcinosarcomas. HMGA1 also occupies the MMP-2 promoter in human carcinosarcoma cells. CONCLUSIONS: Together, our studies define a novel HMGA1-MMP-2 pathway involved in a subset of human carcinosarcomas and tumor progression in murine models. Our work also suggests that targeting HMGA1 could be effective adjuvant therapy for more aggressive uterine cancers and provides compelling data for further preclinical studies.


Asunto(s)
Carcinosarcoma/genética , Cistadenocarcinoma Seroso/genética , Proteína HMGA1a/genética , Metaloproteinasa 2 de la Matriz/genética , Neoplasias Uterinas/genética , Animales , Carcinosarcoma/metabolismo , Inmunoprecipitación de Cromatina , Cistadenocarcinoma Seroso/metabolismo , Femenino , Silenciador del Gen , Proteína HMGA1a/biosíntesis , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Ratones Transgénicos , Regiones Promotoras Genéticas , Regulación hacia Arriba , Neoplasias Uterinas/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(52): 21113-8, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24255108

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) is mutated in approximately one third of acute myeloid leukemia cases. The most common FLT3 mutations in acute myeloid leukemia are internal tandem duplication (ITD) mutations in the juxtamembrane domain (23%) and point mutations in the tyrosine kinase domain (10%). The mutation substituting the aspartic acid at position 838 (equivalent to the human aspartic acid residue at position 835) with a tyrosine (referred to as FLT3/D835Y hereafter) is the most frequent kinase domain mutation, converting aspartic acid to tyrosine. Although both of these mutations constitutively activate FLT3, patients with an ITD mutation have a significantly poorer prognosis. To elucidate the mechanisms behind this prognostic difference, we have generated a knock-in mouse model with a D838Y point mutation in FLT3 that corresponds to the FLT3/D835Y mutation described in humans. Compared with FLT3/ITD knock-in mice, the FLT3/D835Y knock-in mice survive significantly longer. The majority of these mice develop myeloproliferative neoplasms with a less-aggressive phenotype. In addition, FLT3/D835Y mice have distinct hematopoietic development patterns. Unlike the tremendous depletion of the hematopoietic stem cell compartment we have observed in FLT3/ITD mice, FLT3/D835Y mutant mice are not depleted in hematopoietic stem cells. Further comparisons of these FLT3/D835Y knock-in mice with FLT3/ITD mice should provide an ideal platform for dissecting the molecular mechanisms that underlie the prognostic differences between the two different types of FLT3 mutations.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Modelos Animales , Tirosina Quinasa 3 Similar a fms/genética , Animales , Inmunohistoquímica , Ratones , Mutación Missense/genética , Pronóstico , Duplicaciones Segmentarias en el Genoma/genética
7.
Proc Natl Acad Sci U S A ; 110(36): 14717-22, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959886

RESUMEN

Nanoparticle gene therapy holds great promise for the treatment of malignant disease in light of the large number of potent, tumor-specific therapeutic payloads potentially available for delivery. To be effective, gene therapy vehicles must be able to deliver their therapeutic payloads to metastatic lesions after systemic administration. Here we describe nanoparticles comprised of a core of high molecular weight linear polyethylenimine (LPEI) complexed with DNA and surrounded by a shell of polyethyleneglycol-modified (PEGylated) low molecular weight LPEI. Compared with a state-of-the-art commercially available in vivo gene delivery formulation, i.v. delivery of the core/PEGylated shell (CPS) nanoparticles provided more than a 16,000-fold increase in the ratio of tumor to nontumor transfection. The vast majority of examined liver and lung metastases derived from a colorectal cancer cell line showed transgene expression after i.v. CPS injection in an animal model of metastasis. Histological examination of tissues from transfected mice revealed that the CPS nanoparticles selectively transfected neoplastic cells rather than stromal cells within primary and metastatic tumors. However, only a small fraction of neoplastic cells (<1%) expressed the transgene, and the extent of delivery varied with the tumor cell line, tumor site, and host mouse strain used. Our results demonstrate that these CPS nanoparticles offer substantial advantages over previously described formulations for in vivo nanoparticle gene therapeutics. At the same time, they illustrate that major increases in the effectiveness of such approaches are needed for utility in patients with metastatic cancer.


Asunto(s)
Terapia Genética/métodos , Proteínas Luminiscentes/genética , Nanopartículas/administración & dosificación , Neoplasias/terapia , Transfección/métodos , Animales , Línea Celular , Línea Celular Tumoral , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Microscopía Confocal , Microscopía Electrónica de Transmisión , Nanopartículas/química , Nanopartículas/ultraestructura , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Polietilenglicoles/química , Polietileneimina/química , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Am J Physiol Renal Physiol ; 307(11): F1179-86, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25234309

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is a progressive, proliferative renal disease. Kidneys from ADPKD patients are characterized by the presence of cysts that are marked by enhanced proliferation and apoptosis of renal tubular epithelial cells. Current treatment of this disease is supportive, as there are few if any clinically validated targeted therapeutics. Given the parallels between cystic disease and cancer, and in light of our findings of the efficacy of the nuclear transport inhibitors in kidney cancer, which has similarities to ADPKD, we asked whether such inhibitors show utility in ADPKD. In this study, we tested selective inhibitors of nuclear export (SINE) in two human ADPKD cell lines and in an in vivo mouse model of ADPKD. After effective downregulation of a nuclear exporter, exportin 1 (XPO1), with KPT-330, both cell lines showed dose-dependent inhibition of cell proliferation through G0/G1 arrest associated with downregulation of CDK4, with minimal apoptosis. To analyze mechanisms of CDK4 decrease by XPO1 inhibition, localization of various XPO1 target proteins was examined, and C/EBPß was found to be localized in the nucleus by XPO1 inhibition, resulting in an increase of C/EBPα, which activates degradation of CDK4. Furthermore, inhibition of XPO1 with the parallel inhibitor KPT-335 attenuated cyst growth in vivo in the PKD1 mutant mouse model Pkd1(v/v). Thus, inhibition of nuclear export by KPT-330, which has shown no adverse effects in renal serum chemistries and urinalyses in animal models, and which is already in phase 1 trials for cancers, will be rapidly translatable to human ADPKD.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quistes/patología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/patología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Ratones , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Triazoles/farmacología , Proteína Exportina 1
9.
Blood ; 120(7): 1516-27, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22740442

RESUMEN

Blood vessel networks form in a 2-step process of sprouting angiogenesis followed by selective branch regression and stabilization of remaining vessels. Pericytes are known to function in stabilizing blood vessels, but their role in vascular sprouting and selective vessel regression is poorly understood. The endosialin (CD248) receptor is expressed by pericytes associated with newly forming but not stable quiescent vessels. In the present study, we used the Endosialin(-/-) mouse as a means to uncover novel roles for pericytes during the process of vascular network formation. We demonstrate in a postnatal retina model that Endosialin(-/-) mice have normal vascular sprouting but are defective in selective vessel regression, leading to increased vessel density. Examination of the Endosialin(-/-) mouse tumor vasculature revealed an equivalent phenotype, indicating that pericytes perform a hitherto unidentified function to promote vessel destabilization and regression in vivo in both physiologic and pathologic angiogenesis. Mechanistically, Endosialin(-/-) mice have no defect in pericyte recruitment. Rather, endosialin binding to an endothelial associated, but not a pericyte associated, basement membrane component induces endothelial cell apoptosis and detachment. The results of the present study advance our understanding of pericyte biology and pericyte/endothelial cell cooperation during vascular patterning and have implications for the design of both pro- and antiangiogenic therapies.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/patología , Tipificación del Cuerpo , Neovascularización Fisiológica , Pericitos/patología , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Aorta/crecimiento & desarrollo , Aorta/patología , Apoptosis , Membrana Basal/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo , Pericitos/metabolismo , Ratas , Retina/metabolismo , Retina/patología , Vasos Retinianos/crecimiento & desarrollo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(37): 15354-9, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876161

RESUMEN

It is estimated that the etiology of 20-30% of epithelial cancers is directly associated with inflammation, although the direct molecular events linking inflammation and carcinogenesis are poorly defined. In the context of gastrointestinal disease, the bacterium enterotoxigenic Bacteroides fragilis (ETBF) is a significant source of chronic inflammation and has been implicated as a risk factor for colorectal cancer. Spermine oxidase (SMO) is a polyamine catabolic enzyme that is highly inducible by inflammatory stimuli resulting in increased reactive oxygen species (ROS) and DNA damage. We now demonstrate that purified B. fragilis toxin (BFT) up-regulates SMO in HT29/c1 and T84 colonic epithelial cells, resulting in SMO-dependent generation of ROS and induction of γ-H2A.x, a marker of DNA damage. Further, ETBF-induced colitis in C57BL/6 mice is associated with increased SMO expression and treatment of mice with an inhibitor of polyamine catabolism, N(1),N(4)-bis(2,3-butandienyl)-1,4-butanediamine (MDL 72527), significantly reduces ETBF-induced chronic inflammation and proliferation. Most importantly, in the multiple intestinal neoplasia (Min) mouse model, treatment with MDL 72527 reduces ETBF-induced colon tumorigenesis by 69% (P < 0.001). The results of these studies indicate that SMO is a source of bacteria-induced ROS directly associated with tumorigenesis and could serve as a unique target for chemoprevention.


Asunto(s)
Bacteroides fragilis/fisiología , Neoplasias del Colon/microbiología , Poliaminas/metabolismo , Lesiones Precancerosas/microbiología , Acetiltransferasas/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Bacteroides fragilis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Colitis/patología , Neoplasias del Colon/complicaciones , Neoplasias del Colon/patología , Daño del ADN , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Inflamación/complicaciones , Inflamación/patología , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Lesiones Precancerosas/patología , Putrescina/análogos & derivados , Putrescina/farmacología , Proteínas Recombinantes/toxicidad , Poliamino Oxidasa
11.
BMC Cancer ; 13: 157, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23530816

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of death from cancer in the United States. Colorectal cancers have a prolonged latency following initiation that may span decades providing ample time for implementing a chemoprevention strategy that could block or reverse the progression to CRC. Cdk4 pathway alterations have been linked to a number of cancers including CRC. In these experiments we focused on the Cdk4 pathway and its role in intestinal tumorigenesis as a possible target in chemoprevention strategies. METHODS: We evaluated the effect of Cdk4 blockade on the prevention of intestinal tumor formation by crossing Cdk4(-/-) mice to Apc(-/+) mice. In addition, we tested the effect of the dietary compound silibinin on the Cdk4 pathway in Apc(-/+) mice and HT-29 colon cancer cells in culture. RESULTS: Cdk4(-/-) mice backcrossed to Apc(-/+) mice reduced intestinal adenoma formation compared to Apc(-/+) controls. Silibinin effectively targeted the Cdk4 pathway causing hypophosphorylation of the retinoblastoma protein, inhibited cell growth, and induced apoptosis. As a result silibinin blocked the development of intestinal adenomas by 52% in this genetic model (Apc(-/+) mice) of early events in colorectal cancer formation. No toxic abnormalities were detected in mice which received silibinin. CONCLUSIONS: Modification of the Cdk4 pathway using a natural plant-derived compound such as silibinin may be a useful chemopreventive strategy for colorectal carcinomas.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Genes APC , Transducción de Señal/efectos de los fármacos , Silimarina/farmacología , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Adenoma/prevención & control , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioprevención , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Antígeno Ki-67/metabolismo , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/metabolismo , Silibina , Silimarina/uso terapéutico
12.
FASEB J ; 26(6): 2648-56, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22415308

RESUMEN

After separating from a primary tumor, metastasizing cells enter the circulatory system and interact with host cells before lodging in secondary organs. Previous studies have implicated the surface glycoproteins CD44 and carcinoembryonic antigen (CEA) in adhesion, migration, and invasion, suggesting that they may influence metastatic progression. To elucidate the role of these multifunctional molecules while avoiding the potential drawbacks of ectopic expression or monoclonal antibody treatments, we silenced the expression of CD44 and/or CEA in LS174T colon carcinoma cells and analyzed their ability to metastasize in 2 independent mouse models. Quantitative PCR revealed that CD44 knockdown increased lung and liver metastasis >10-fold, while metastasis was decreased by >50% following CEA knockdown. These findings were corroborated by in vitro experiments assessing the metastatic potential of LS174T cells. Cell migration was decreased as a result of silencing CEA but was enhanced in CD44-knockdown cells. In addition, CD44 silencing promoted homotypic aggregation of LS147T cells, a phenotype that was reversed by additional CEA knockdown. Finally, CD44-knockdown cells exhibited greater mechanical compliance than control cells, a property that correlates with increased metastatic potential. Collectively, these data indicate that CEA, but not CD44, is a viable target for therapeutics aimed at curbing colon carcinoma metastasis.


Asunto(s)
Antígeno Carcinoembrionario/fisiología , Neoplasias del Colon/patología , Receptores de Hialuranos/fisiología , Metástasis de la Neoplasia/fisiopatología , Animales , Movimiento Celular/fisiología , Neoplasias del Colon/fisiopatología , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Ratones , Células Tumorales Cultivadas
13.
Arthritis Rheum ; 64(10): 3334-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22674221

RESUMEN

OBJECTIVE: CD248 (tumor endothelial marker 1/endosialin) is found on stromal cells and is highly expressed during malignancy and inflammation. Studies have shown a reduction in inflammatory arthritis in CD248-knockout (CD248(-/-) ) mice. The aim of the present study was to investigate the functional effect of genetic deletion of CD248 on bone mass. METHODS: Western blotting, polymerase chain reaction, and immunofluorescence were used to investigate the expression of CD248 in humans and mice. Micro-computed tomography and the 3-point bending test were used to measure bone parameters and mechanical properties of the tibiae of 10-week-old wild-type (WT) or CD248(-/-) mice. Human and mouse primary osteoblasts were cultured in medium containing 10 mM ß-glycerophosphate and 50 µg/ml ascorbic acid to induce mineralization, and then treated with platelet-derived growth factor BB (PDGF-BB). The mineral apposition rate in vivo was calculated by identifying newly formed bone via calcein labeling. RESULTS: Expression of CD248 was seen in human and mouse osteoblasts, but not osteoclasts. CD248(-/-) mouse tibiae had higher bone mass and superior mechanical properties (increased load required to cause fracture) compared to WT mice. Primary osteoblasts from CD248(-/-) mice induced increased mineralization in vitro and produced increased bone over 7 days in vivo. There was no decrease in bone mineralization and no increase in proliferation of osteoblasts in response to stimulation with PDGF-BB, which could be attributed to a defect in PDGF signal transduction in the CD248(-/-) mice. CONCLUSION: There is an unmet clinical need to address rheumatoid arthritis-associated bone loss. Genetic deletion of CD248 in mice results in high bone mass due to increased osteoblast-mediated bone formation, suggesting that targeting CD248 in rheumatoid arthritis may have the effect of increasing bone mass in addition to the previously reported effect of reducing inflammation.


Asunto(s)
Antígenos CD/metabolismo , Huesos/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/metabolismo , Osteogénesis/fisiología , Animales , Antígenos CD/genética , Becaplermina , Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología
14.
Cancer Cell ; 7(6): 561-73, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15950905

RESUMEN

PIK3CA is mutated in diverse human cancers, but the functional effects of these mutations have not been defined. To evaluate the consequences of PIK3CA alterations, the two most common mutations were inactivated by gene targeting in colorectal cancer (CRC) cells. Biochemical analyses of these cells showed that mutant PIK3CA selectively regulated the phosphorylation of AKT and the forkhead transcription factors FKHR and FKHRL1. PIK3CA mutations had little effect on growth under standard conditions, but reduced cellular dependence on growth factors. PIK3CA mutations resulted in attenuation of apoptosis and facilitated tumor invasion. Treatment with the PI3K inhibitor LY294002 abrogated PIK3CA signaling and preferentially inhibited growth of PIK3CA mutant cells. These data have important implications for therapy of cancers harboring PIK3CA alterations.


Asunto(s)
Proliferación Celular , Invasividad Neoplásica/patología , Fosfatidilinositol 3-Quinasas/genética , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase I , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Marcación de Gen , Sustancias de Crecimiento/deficiencia , Humanos , Insulina/deficiencia , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Desnudos , Morfolinas/farmacología , Mutación , Invasividad Neoplásica/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Proc Natl Acad Sci U S A ; 107(6): 2598-603, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133737

RESUMEN

Phosphotidylinositol-3-kinase (PI3K) signaling is altered in the majority of human cancers. To gain insight into the roles of members of this pathway in growth regulation, we inactivated AKT1, AKT2, or PDPK1 genes by targeted homologous recombination in human colon cancer cell lines. Knockout of either AKT1 or AKT2 had minimum effects on cell growth or downstream signaling. In contrast, knockout of both AKT1 and AKT2 resulted in markedly reduced proliferation in vitro when growth factors were limiting and severely affected experimental metastasis in mice. Unexpectedly, AKT1 and AKT2 appeared to regulate growth through FOXO proteins, but not through either GSK3beta or mTOR. In contrast, inactivation of PDPK1 affected GSK3beta and mTOR activation. These findings show that the PI3K signaling pathway is wired differently in human cancer cells than in other cell types or organisms, which has important implications for the design and testing of drugs that target this pathway.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Apoptosis , Western Blotting , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Silenciador del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HCT116 , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR , Trasplante Heterólogo
16.
Carcinogenesis ; 33(11): 2242-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22831956

RESUMEN

Multiple lines of evidence support a role for curcumin in cancer chemoprevention. Nonetheless, despite its reported efficacy and safety profile, clinical translation of curcumin has been hampered by low oral bioavailability, requiring infeasible 'mega' doses for achieving detectable tissue levels. We have engineered a polymeric nanoparticle encapsulated formulation of curcumin (NanoCurc) to harness its full therapeutic potential. In the current study, we assessed the chemoprevention efficacy of NanoCurc administered via direct intraductal (i.duc) injection in a chemical carcinogen-induced rodent mammary cancer model. Specifically, Sprague-Dawley rats exposed to systemic N-methyl-N-nitrosourea were randomized to receive either oral free curcumin at a previously reported 'mega' dose (200mg/kg) or by direct i.duc injection of free curcumin or NanoCurc, respectively, each delivering 168 µg equivalent of curcumin per rodent teat (a ~20-fold lower dose per animal compared to oral administration). All three chemoprevention modalities resulted in significantly lower mammary tumor incidence compared with control rats; however, there was no significant difference in cancer incidence between the oral dosing and either i.duc arms. On the other hand, mean tumor size, was significantly smaller in the i.duc NanoCurc cohort compared with i.duc free curcumin (P < 0.0001), suggesting the possibility of better resectability for 'breakthrough' cancers. Reduction in cancer incidence was associated with significant decrease in nuclear factor -κB activation in the NanoCurc treated mammary epithelium explants, compared to either control or oral curcumin-administered rats. Our studies confirm the potential for i.duc NanoCurc as an alternative to the oral route for breast cancer chemoprevention in high-risk cohorts.


Asunto(s)
Antineoplásicos/administración & dosificación , Curcumina/administración & dosificación , Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/prevención & control , Nanopartículas , Polímeros/química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Química Farmacéutica , Curcumina/farmacocinética , Curcumina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Técnicas para Inmunoenzimas , Incidencia , Neoplasias Mamarias Experimentales/epidemiología , Neoplasias Mamarias Experimentales/mortalidad , Metilnitrosourea/toxicidad , Ratas , Ratas Sprague-Dawley , Tasa de Supervivencia , Distribución Tisular
17.
Breast Cancer Res Treat ; 135(1): 201-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22752247

RESUMEN

Previously, we have shown that the intraductal (i.duc) administration of pegylated liposomal doxorubicin (PLD) to Her2/neu transgenic mice is associated with mammary tumor regression and prevention. Exploring the mechanism underlying the protection afforded by PLD, we studied: the effects of i.duc PLD-treatment with a subsequent pregnancy on outgrowth of tumors in Her2/neu mice; whether the i.duc PLD antitumor effect was mediated partially through changes in normal mammary stem cells (MaSCs); and the long-term safety of i.duc PLD into the normal mouse mammary gland. Her2/neu mice were treated with two i.duc injections of PLD given four weeks apart; pregnancy was induced and mice were followed up for changes in physiology, and tumor formation. We found that all pups born to i.duc PLD-treated Her2/neu mice died without weight gain within 7 days after birth. Despite an additional pregnancy, compared to vehicle control PLD-treated Her2/neu mice had a significantly longer latency and lower frequency of tumor development. Mammary epithelial cells isolated from untreated and i.duc PLD-treated 6-8 months-old multiparous FVB/N mice were analyzed for their repopulating ability in mammary fat pads of naïve recipients. Mice were also monitored for abnormalities in mammary gland morphology and function, including tumor formation. PLD-treated FVB/N mice displayed histomorphologic changes and a significant reduction in the outgrowth potential of cells from the mammary glands. Thus, i.duc PLD administration altered the mammary gland structurally and functionally by reducing the MaSC population, which could compromise milk production. Followed long term, i.duc PLD-treated FVB/N mice developed malignant mammary tumors, confirming similar published findings on doxorubicin injected into the mammary gland of rats. Unless there are fundamental species differences in PLD metabolism in rodents and humans, this finding seriously limits the consideration of i.duc PLD use in the clinic for treatment or prevention of breast cancer.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/análogos & derivados , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Células Madre/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/efectos adversos , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Lactancia/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inducido químicamente , Ratones , Ratones Transgénicos , Polietilenglicoles/administración & dosificación , Polietilenglicoles/efectos adversos , Embarazo , Células Madre/fisiología
18.
Nat Cell Biol ; 7(7): 706-11, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15965466

RESUMEN

Telomeres protect chromosome ends from fusion, degradation and recombination. Loss of telomere function has opposite effects on tumorigenesis: apoptosis, which inhibits tumour growth, and genomic instability, which accelerates tumour formation. Here we describe a new mechanism by which short telomeres inhibit tumorigenesis through interference with oncogenic translocations. In mice that are null for both ataxia-telangiectasia-mutated (Atm) and telomerase RNA (mTR), the first generation (G1) Atm-/- mTR-/- mice have a lower rate of tumour formation than Atm-/- mTR+/+ mice. These Atm-/- mTR-/- G1 tumours show no increase in either apoptosis or overall genomic instability. Strikingly, the tumours show a high fraction of translocations containing telomere signals at the translocation junctions. Translocations of the T-cell receptors on chromosome 14, which initiate tumorigenesis, were interrupted by fusion with telomeres. Telomere repeats were also detected at the translocation junctions in pre-malignant thymocytes. We propose that telomere fusion to DNA double-strand breaks competes with the generation of oncogenic translocations and thus reduces tumour formation.


Asunto(s)
Rotura Cromosómica/genética , Neoplasias Experimentales/genética , Telómero/metabolismo , Translocación Genética/genética , Factores de Edad , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/química , Linfocitos B/metabolismo , Peso Corporal/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Femenino , Reordenamiento Génico de Linfocito T/genética , Genes Codificadores de los Receptores de Linfocitos T/genética , Inestabilidad Genómica/genética , Genotipo , Hibridación Fluorescente in Situ , Linfoma de Células T/genética , Linfoma de Células T/patología , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Modelos Genéticos , Trasplante de Neoplasias , Neoplasias Experimentales/patología , Proteínas Serina-Treonina Quinasas/genética , Cariotipificación Espectral , Análisis de Supervivencia , Telomerasa/genética , Telómero/genética , Proteínas Supresoras de Tumor/genética
19.
Pancreatology ; 12(4): 372-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22898640

RESUMEN

CONTEXT: Although pancreatic cancer is a common, highly lethal malignancy, the molecular events that enable precursor lesions to become invasive carcinoma remain unclear. We previously reported that the high-mobility group A1 (HMGA1) protein is overexpressed in >90% of primary pancreatic cancers, with absent or low levels in early precursor lesions. METHODS: Here, we investigate the role of HMGA1 in reprogramming pancreatic epithelium into invasive cancer cells. We assessed oncogenic properties induced by HMGA1 in non-transformed pancreatic epithelial cells expressing activated K-RAS. We also explored the HMGA1-cyclooxygenase (COX-2) pathway in human pancreatic cancer cells and the therapeutic effects of COX-2 inhibitors in xenograft tumorigenesis. RESULTS: HMGA1 cooperates with activated K-RAS to induce migration, invasion, and anchorage-independent cell growth in a cell line derived from normal human pancreatic epithelium. Moreover, HMGA1 and COX-2 expression are positively correlated in pancreatic cancer cell lines (r(2) = 0.93; p < 0.001). HMGA1 binds directly to the COX-2 promoter at an AT-rich region in vivo in three pancreatic cancer cell lines. In addition, HMGA1 induces COX-2 expression in pancreatic epithelial cells, while knock-down of HMGA1 results in repression of COX-2 in pancreatic cancer cells. Strikingly, we also discovered that Sulindac (a COX-1/COX-2 inhibitor) or Celecoxib (a more specific COX-2 inhibitor) block xenograft tumorigenesis from pancreatic cancer cells expressing high levels of HMGA1. CONCLUSIONS: Our studies identify for the first time an important role for the HMGA1-COX-2 pathway in pancreatic cancer and suggest that targeting this pathway could be effective to treat, or even prevent, pancreatic cancer.


Asunto(s)
Adenocarcinoma/genética , Ciclooxigenasa 2/genética , Proteína HMGA1a/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/prevención & control , Animales , Celecoxib , División Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Ciclooxigenasa 2/fisiología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Expresión Génica , Proteína HMGA1a/fisiología , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/prevención & control , Pirazoles/administración & dosificación , Sulfonamidas/administración & dosificación , Sulindac/administración & dosificación , Trasplante Heterólogo , Proteínas ras/fisiología
20.
Cancer Cell ; 4(4): 291-9, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14585356

RESUMEN

Id genes regulate tumor angiogenesis and loss of Id1 inhibits tumor xenograft growth in mice. Here we evaluate the role of Id1 in a more clinically relevant tumor model system using a two-step chemical carcinogenesis protocol. Remarkably, we find that Id1-/- mice are more susceptible to skin tumorigenesis compared to their wild-type counterparts. Cutaneous neoplasms in Id1-/- mice show increased proliferation without alterations in tumor angiogenesis; however, Id1-/- mice possess 50% fewer cutaneous gammadelta T cells than their wild-type counterparts due to an intrinsic migration defect associated with loss of expression of the chemokine receptor, CXCR4. We suggest that there are important differences between the mechanisms of angiogenesis in transplanted and autochthonous tumors and that these findings will have significant implications for the potential utility of antiangiogenic therapies in cancer.


Asunto(s)
Melanoma/metabolismo , Neovascularización Patológica/fisiopatología , Proteínas Represoras , Neoplasias Cutáneas/fisiopatología , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Animales , Carcinógenos/toxicidad , Endotelio Vascular/fisiopatología , Citometría de Flujo , Proteína 1 Inhibidora de la Diferenciación , Melanoma/inducido químicamente , Ratones , Ratones Noqueados , Microscopía Fluorescente , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/fisiopatología , Receptores CXCR4/metabolismo , Neoplasias Cutáneas/inducido químicamente , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA