Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Cancer ; 15(1): 78, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27919264

RESUMEN

BACKGROUND: Blocking the mechanistic target of rapamycin complex-1 (mTORC1) with chemical inhibitors such as rapamycin has shown limited clinical efficacy in cancer. The tumor microenvironment is characterized by an acidic pH which interferes with cancer therapies. The consequences of acidity on the anti-cancer efficacy of mTORC1 inhibitors have not been characterized and are thus the focus of our study. METHODS: Cancer cell lines were treated with rapamycin in acidic or physiological conditions and cell proliferation was investigated. The effect of acidity on mTORC1 activity was determined by Western blot. The anticancer efficacy of rapamycin in combination with sodium bicarbonate to increase the intratumoral pH was tested in two different mouse models and compared to rapamycin treatment alone. Histological analysis was performed on tumor samples to evaluate proliferation, apoptosis and necrosis. RESULTS: Exposing cancer cells to acidic pH in vitro significantly reduced the anti-proliferative effect of rapamycin. At the molecular level, acidity significantly decreased mTORC1 activity, suggesting that cancer cell proliferation is independent of mTORC1 in acidic conditions. In contrast, the activation of mitogen-activated protein kinase (MAPK) or AKT were not affected by acidity, and blocking MAPK or AKT with a chemical inhibitor maintained an anti-proliferative effect at low pH. In tumor mouse models, the use of sodium bicarbonate increased mTORC1 activity in cancer cells and potentiated the anti-cancer efficacy of rapamycin. Combining sodium bicarbonate with rapamycin resulted in increased tumor necrosis, increased cancer cell apoptosis and decreased cancer cell proliferation as compared to single treatment. CONCLUSIONS: Taken together, these results emphasize the inefficacy of mTORC1 inhibitors in acidic conditions. They further highlight the potential of combining sodium bicarbonate with mTORC1 inhibitors to improve their anti-tumoral efficacy.


Asunto(s)
Ácidos/efectos adversos , Neoplasias Colorrectales/tratamiento farmacológico , Complejos Multiproteicos/metabolismo , Sirolimus/administración & dosificación , Bicarbonato de Sodio/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Quimioterapia Combinada , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/antagonistas & inhibidores , Sirolimus/farmacología , Bicarbonato de Sodio/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Oncotarget ; 7(24): 36666-36680, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27153561

RESUMEN

The inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) by chemical inhibitors, such as rapamycin, has demonstrated anti-cancer activity in preclinical and clinical trials. Their efficacy is, however, limited and tumors eventually relapse through resistance formation. In this study, using two different cancer mouse models, we identify tumor hypoxia as a novel mechanism of resistance of cancer cells against mTORC1 inhibitors. Indeed, we show that the activity of mTORC1 is mainly restricted to the non-hypoxic tumor compartment, as evidenced by a mutually exclusive staining pattern of the mTORC1 activity marker pS6 and the hypoxia marker pimonidazole. Consequently, whereas rapamycin reduces cancer cell proliferation in non-hypoxic regions, it has no effect in hypoxic areas, suggesting that cancer cells proliferate independently of mTORC1 under hypoxia. Targeting the hypoxic tumor compartment by knockdown of carbonic anhydrase IX (CAIX) using short hairpin RNA or by chemical inhibition of CAIX with acetazolamide potentiates the anti-cancer activity of rapamycin. Taken together, these data emphasize that hypoxia impairs the anti-cancer efficacy of rapalogs. Therapeutic strategies targeting the hypoxic tumor compartment, such as the inhibition of CAIX, potentiate the efficacy of rapamycin and warrant further clinical evaluation.


Asunto(s)
Acetazolamida/farmacología , Anhidrasa Carbónica IX/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacología , Anhidrasa Carbónica IX/genética , Anhidrasa Carbónica IX/metabolismo , Inhibidores de Anhidrasa Carbónica/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Sinergismo Farmacológico , Femenino , Células HT29 , Humanos , Hipoxia , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Interferencia de ARN , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Microbes Infect ; 17(11-12): 755-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26340890

RESUMEN

Parachlamydia acanthamoebae is an obligate intracellular bacterium naturally infecting free-living amoebae. The role of this bacterium as an agent of pneumonia is suggested by sero-epidemiological studies and molecular surveys. Furthermore, P. acanthamoebae may escape macrophages microbicidal effectors. Recently, we demonstrated that intratracheal inoculation of P. acanthamoebae induced pneumonia in 100% of infected mice. However, the intratracheal route of infection is not the natural way of infection and we therefore developed an intranasal murine model. Mice inoculated with P. acanthamoebae by intranasal inoculation lost 18% of their weight up to 8 days post-inoculation. All mice presented histological signs of pneumonia at day 2, 4, 7, and 10 post-inoculation, whereas no control mice harboured signs of pneumonia. A 5-fold increase in bacterial load was observed from day 0 to day 4 post-inoculation. Lungs of inoculated mice were positive by Parachlamydia-specific immunohistochemistry 4 days post-inoculation, and P. acanthamoebae were localized within macrophages. Thus, we demonstrated that P. acanthamoebae induce a severe pneumonia in mice. This animal model (i) further supports the role of P. acanthamoebae as an agent of pneumonia, confirming the third Koch postulate, and (ii) identified alveolar macrophages as one of the initial cells where P. acanthamoebae is localized following infection.


Asunto(s)
Chlamydiales/patogenicidad , Pulmón/microbiología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Administración Intranasal , Animales , Carga Bacteriana , Chlamydiales/inmunología , Modelos Animales de Enfermedad , Pulmón/patología , Macrófagos/inmunología , Ratones , Neumonía Bacteriana/inmunología
4.
J Pharmacol Exp Ther ; 346(2): 281-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23697346

RESUMEN

Chronic renal failure (CRF) is associated with the development of secondary hyperparathyroidism and vascular calcifications. We evaluated the efficacy of PA21, a new iron-based noncalcium phosphate binder, in controlling phosphocalcic disorders and preventing vascular calcifications in uremic rats. Rats with adenine-diet-induced CRF were randomized to receive either PA21 0.5, 1.5, or 5% or CaCO3 3% in the diet for 4 weeks, and were compared with uremic and nonuremic control groups. After 4 weeks of phosphate binder treatment, serum calcium, creatinine, and body weight were similar between all CRF groups. Serum phosphorus was reduced with CaCO3 3% (2.06 mM; P ≤ 0.001), PA21 1.5% (2.29 mM; P < 0.05), and PA21 5% (2.21 mM; P ≤ 0.001) versus CRF controls (2.91 mM). Intact parathyroid hormone was strongly reduced in the PA21 5% and CaCO3 3% CRF groups to a similar extent (1138 and 1299 pg/ml, respectively) versus CRF controls (3261 pg/ml; both P ≤ 0.001). A lower serum fibroblast growth factor 23 concentration was observed in the PA21 5%, compared with CaCO3 3% and CRF, control groups. PA21 5% CRF rats had a lower vascular calcification score compared with CaCO3 3% CRF rats and CRF controls. In conclusion, PA21 was as effective as CaCO3 at controlling phosphocalcic disorders but superior in preventing the development of vascular calcifications in uremic rats. Thus, PA21 represents a possible alternative to calcium-based phosphate binders in CRF patients.


Asunto(s)
Compuestos Férricos/uso terapéutico , Fallo Renal Crónico/tratamiento farmacológico , Calcificación Vascular/prevención & control , Adenina , Animales , Aorta/efectos de los fármacos , Aorta/patología , Presión Sanguínea/efectos de los fármacos , Calcio/sangre , Carbonato de Calcio/uso terapéutico , Factores de Crecimiento de Fibroblastos/sangre , Frecuencia Cardíaca/efectos de los fármacos , Fallo Renal Crónico/inducido químicamente , Fallo Renal Crónico/patología , Masculino , Hormona Paratiroidea/sangre , Fósforo/sangre , Ratas , Ratas Wistar , Calcificación Vascular/patología
5.
PLoS One ; 8(2): e55796, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23405214

RESUMEN

The serine protease CAP1/Prss8 is crucial for skin barrier function, lung alveolar fluid clearance and has been unveiled as diagnostic marker for specific cancer types. Here, we show that a constitutive knockout of CAP1/Prss8 leads to embryonic lethality. These embryos presented no specific defects, but it is during this period, and in particular at E13.5, that wildtype placentas show an increased expression of CAP1/Prss8, thus suggesting a placental defect in the knockout situation. The placentas of knockout embryos exhibited significantly reduced vascular development and incomplete cellular maturation. In contrary, epiblast-specific deletion of CAP1/Prss8 allowed development until birth. These CAP1/Prss8-deficient newborns presented abnormal epidermis, and died soon after birth due to impaired skin function. We thus conclude that a late placental insufficiency might be the primary cause of embryonic lethality in CAP1/Prss8 knockouts. This study highlights a novel and crucial role for CAP1/Prss8 in placental development and function.


Asunto(s)
Oído Interno/crecimiento & desarrollo , Pérdida del Embrión/genética , Embrión de Mamíferos/citología , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica , Placentación/fisiología , Serina Endopeptidasas/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular , Embrión de Mamíferos/enzimología , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Clin Cancer Res ; 18(16): 4365-74, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711708

RESUMEN

PURPOSE: Local breast cancer relapse after breast-saving surgery and radiotherapy is associated with increased risk of distant metastasis formation. The mechanisms involved remain largely elusive. We used the well-characterized 4T1 syngeneic, orthotopic breast cancer model to identify novel mechanisms of postradiation metastasis. EXPERIMENTAL DESIGN: 4T1 cells were injected in 20 Gy preirradiated mammary tissue to mimic postradiation relapses, or in nonirradiated mammary tissue, as control, of immunocompetent BALB/c mice. Molecular, biochemical, cellular, histologic analyses, adoptive cell transfer, genetic, and pharmacologic interventions were carried out. RESULTS: Tumors growing in preirradiated mammary tissue had reduced angiogenesis and were more hypoxic, invasive, and metastatic to lung and lymph nodes compared with control tumors. Increased metastasis involved the mobilization of CD11b(+)c-Kit(+)Ly6G(high)Ly6C(low)(Gr1(+)) myeloid cells through the HIF1-dependent expression of Kit ligand (KitL) by hypoxic tumor cells. KitL-mobilized myeloid cells homed to primary tumors and premetastatic lungs, to give rise to CD11b(+)c-Kit(-) cells. Pharmacologic inhibition of HIF1, silencing of KitL expression in tumor cells, and inhibition of c-Kit with an anti-c-Kit-blocking antibody or with a tyrosine kinase inhibitor prevented the mobilization of CD11b(+)c-Kit(+) cells and attenuated metastasis. C-Kit inhibition was also effective in reducing mobilization of CD11b(+)c-Kit(+) cells and inhibiting lung metastasis after irradiation of established tumors. CONCLUSIONS: Our work defines KitL/c-Kit as a previously unidentified axis critically involved in promoting metastasis of 4T1 tumors growing in preirradiated mammary tissue. Pharmacologic inhibition of this axis represents a potential therapeutic strategy to prevent metastasis in breast cancer patients with local relapses after radiotherapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoxia , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/radioterapia , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Carga Tumoral
7.
Mol Endocrinol ; 26(6): 1000-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22556341

RESUMEN

The glucocorticoid-induced leucine zipper (Tsc22d3-2) is a widely expressed dexamethasone-induced transcript that has been proposed to be important in immunity, adipogenesis, and renal sodium handling based on in vitro studies. To address its function in vivo, we have used Cre/loxP technology to generate mice deficient for Tsc22d3-2. Male knockout mice were viable but surprisingly did not show any major deficiencies in immunological processes or inflammatory responses. Tsc22d3-2 knockout mice adapted to a sodium-deprived diet and to water deprivation conditions but developed a subtle deficiency in renal sodium and water handling. Moreover, the affected animals developed a mild metabolic phenotype evident by a reduction in weight from 6 months of age, mild hyperinsulinemia, and resistance to a high-fat diet. Tsc22d3-2-deficient males were infertile and exhibited severe testis dysplasia from postnatal d 10 onward with increases in apoptotic cells within seminiferous tubules, an increased number of Leydig cells, and significantly elevated FSH and testosterone levels. Thus, our analysis of the Tsc22d3-2-deficient mice demonstrated a previously uncharacterized function of glucocorticoid-induced leucine zipper protein in testis development.


Asunto(s)
Infertilidad Masculina/genética , Factores de Transcripción/genética , Adipogénesis , Animales , Peso Corporal , Recuento de Células , Células Cultivadas , Citocinas/metabolismo , Dexametasona/farmacología , Femenino , Fibroblastos/fisiología , Sitios Genéticos , Hiperinsulinismo/genética , Sistema Inmunológico/crecimiento & desarrollo , Factores Inmunológicos/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Isoformas de Proteínas/genética , Bazo/patología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testículo/patología , Timo/patología , Factores de Transcripción/deficiencia
8.
J Clin Invest ; 121(7): 2794-807, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21646719

RESUMEN

Metastasis depends on the ability of tumor cells to establish a relationship with the newly seeded tissue that is conducive to their survival and proliferation. However, the factors that render tissues permissive for metastatic tumor growth have yet to be fully elucidated. Breast tumors arising during pregnancy display early metastatic proclivity, raising the possibility that pregnancy may constitute a physiological condition of permissiveness for tumor dissemination. Here we have shown that during murine gestation, metastasis is enhanced regardless of tumor type, and that decreased NK cell activity is responsible for the observed increase in experimental metastasis. Gene expression changes in pregnant mouse lung and liver were shown to be similar to those detected in premetastatic sites and indicative of myeloid cell infiltration. Indeed, myeloid-derived suppressor cells (MDSCs) accumulated in pregnant mice and exerted an inhibitory effect on NK cell activity, providing a candidate mechanism for the enhanced metastatic tumor growth observed in gestant mice. Although the functions of MDSCs are not yet understood in the context of pregnancy, our observations suggest that they may represent a shared mechanism of immune suppression occurring during gestation and tumor growth.


Asunto(s)
Tolerancia Inmunológica/fisiología , Terapia de Inmunosupresión , Células Mieloides/fisiología , Metástasis de la Neoplasia/inmunología , Complicaciones Neoplásicas del Embarazo/inmunología , Complicaciones Neoplásicas del Embarazo/patología , Animales , Línea Celular Tumoral , Femenino , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Células Mieloides/citología , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Embarazo
9.
PLoS One ; 6(5): e18640, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21611158

RESUMEN

Primary tumor growth induces host tissue responses that are believed to support and promote tumor progression. Identification of the molecular characteristics of the tumor microenvironment and elucidation of its crosstalk with tumor cells may therefore be crucial for improving our understanding of the processes implicated in cancer progression, identifying potential therapeutic targets, and uncovering stromal gene expression signatures that may predict clinical outcome. A key issue to resolve, therefore, is whether the stromal response to tumor growth is largely a generic phenomenon, irrespective of the tumor type or whether the response reflects tumor-specific properties. To address similarity or distinction of stromal gene expression changes during cancer progression, oligonucleotide-based Affymetrix microarray technology was used to compare the transcriptomes of laser-microdissected stromal cells derived from invasive human breast and prostate carcinoma. Invasive breast and prostate cancer-associated stroma was observed to display distinct transcriptomes, with a limited number of shared genes. Interestingly, both breast and prostate tumor-specific dysregulated stromal genes were observed to cluster breast and prostate cancer patients, respectively, into two distinct groups with statistically different clinical outcomes. By contrast, a gene signature that was common to the reactive stroma of both tumor types did not have survival predictive value. Univariate Cox analysis identified genes whose expression level was most strongly associated with patient survival. Taken together, these observations suggest that the tumor microenvironment displays distinct features according to the tumor type that provides survival-predictive value.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Humanos , Estimación de Kaplan-Meier , Masculino , Especificidad de Órganos/genética , Pronóstico , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Microambiente Tumoral , Regulación hacia Arriba/genética
10.
Int J Cancer ; 128(9): 2105-13, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20635385

RESUMEN

Cervical cancer results from infection with high-risk type human papillomaviruses (HPV). Therapeutic vaccines aiming at controlling existing genital HPV infections and associated lesions are usually tested in mice with HPV-expressing tumor cells subcutaneously implanted into their flank. However, effective vaccine-induced regression of these ectopic tumors strongly contrasts with the poor clinical results of these vaccines produced in patients with HPV-associated genital neoplasia. To assess HPV therapeutic vaccines in a more relevant setting, we have, here, established an orthotopic mouse model where tumors in the genital mucosa (GM) develop after an intravaginal instillation of HPV16 E6/E7-expressing tumor cells transduced with a luciferase-encoding lentiviral vector for in vivo imaging of tumor growth. Tumor take was 80-90% after nonoxynol-9 induced damage of the epithelium. Tumors remained localized in the genital tract, and histological analysis showed that most tumors grew within the squamous epithelium of the vaginal wall. Those tumors induced (i) E7-specific CD8 T cells restricted to the GM and draining lymph nodes, in agreement with their mucosal location and (ii) high Foxp3+ CD4+ infiltrates, similarly to those found in natural non-regressing HPV lesions. This novel genital HPV-tumor model by requiring GM homing of vaccine-induced immune responses able to overcome local immuno-suppression may be more representative of the situation occurring in patients upon therapeutic vaccination.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Modelos Animales de Enfermedad , Infecciones por Papillomavirus/complicaciones , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Animales , Carcinoma de Células Escamosas/virología , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias/inmunología , Trasplante de Neoplasias/métodos , Trasplante de Neoplasias/patología , Trasplante Heterólogo/inmunología , Trasplante Heterólogo/métodos , Trasplante Heterólogo/patología , Neoplasias del Cuello Uterino/virología
11.
J Am Soc Nephrol ; 22(2): 253-61, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051735

RESUMEN

Lithium-induced nephrogenic diabetes insipidus (NDI) is accompanied by polyuria, downregulation of aquaporin 2 (AQP2), and cellular remodeling of the collecting duct (CD). The amiloride-sensitive epithelial sodium channel (ENaC) is a likely candidate for lithium entry. Here, we subjected transgenic mice lacking αENaC specifically in the CD (knockout [KO] mice) and littermate controls to chronic lithium treatment. In contrast to control mice, KO mice did not markedly increase their water intake. Furthermore, KO mice did not demonstrate the polyuria and reduction in urine osmolality induced by lithium treatment in the control mice. Lithium treatment reduced AQP2 protein levels in the cortex/outer medulla and inner medulla (IM) of control mice but only partially reduced AQP2 levels in the IM of KO mice. Furthermore, lithium induced expression of H(+)-ATPase in the IM of control mice but not KO mice. In conclusion, the absence of functional ENaC in the CD protects mice from lithium-induced NDI. These data support the hypothesis that ENaC-mediated lithium entry into the CD principal cells contributes to the pathogenesis of lithium-induced NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/inducido químicamente , Canales Epiteliales de Sodio/fisiología , Cloruro de Litio/toxicidad , Absorción , Animales , Acuaporina 2/análisis , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Cloruro de Litio/farmacocinética , Ratones , Ratones Noqueados , ATPasas de Translocación de Protón/análisis
12.
Cancer Res ; 70(16): 6619-28, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20682801

RESUMEN

Canonical Wnt signaling plays a critical role in stem cell maintenance in epithelial homeostasis and carcinogenesis. Here, we show that in the mouse this role is critically mediated by Bcl9/Bcl9l, the mammalian homologues of Legless, which in Drosophila is required for Armadillo/beta-catenin signaling. Conditional ablation of Bcl9/Bcl9l in the intestinal epithelium, where the essential role of Wnt signaling in epithelial homeostasis and stem cell maintenance is well documented, resulted in decreased expression of intestinal stem cell markers and impaired regeneration of ulcerated colon epithelium. Adenocarcinomas with aberrant Wnt signaling arose with similar incidence in wild-type and mutant mice. However, transcriptional profiles were vastly different: Whereas wild-type tumors displayed characteristics of epithelial-mesenchymal transition (EMT) and stem cell-like properties, these properties were largely abrogated in mutant tumors. These findings reveal an essential role for Bcl9/Bcl9l in regulating a subset of Wnt target genes involved in controlling EMT and stem cell-related features and suggest that targeting the Bcl9/Bcl9l arm of Wnt signaling in Wnt-activated cancers might attenuate these traits, which are associated with tumor invasion, metastasis, and resistance to therapy.


Asunto(s)
Adenocarcinoma/patología , Neoplasias del Colon/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Neoplásicas/patología , Proteínas Wnt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/fisiología , Factores de Transcripción , Proteínas Wnt/biosíntesis , Proteínas Wnt/genética
13.
Genes Dev ; 24(9): 916-32, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20382729

RESUMEN

Cancer stem cells (CSCs) display plasticity and self-renewal properties reminiscent of normal tissue stem cells, but the events responsible for their emergence remain obscure. We recently identified CSCs in Ewing sarcoma family tumors (ESFTs) and showed that they retain mesenchymal stem cell (MSC) plasticity. In the present study, we addressed the mechanisms that underlie ESFT CSC development. We show that the EWS-FLI-1 fusion gene, associated with 85%-90% of ESFTs and believed to initiate their pathogenesis, induces expression of the embryonic stem cell (ESC) genes OCT4, SOX2, and NANOG in human pediatric MSCs (hpMSCs) but not in their adult counterparts. Moreover, under appropriate culture conditions, hpMSCs expressing EWS-FLI-1 generate a cell subpopulation displaying ESFT CSC features in vitro. We further demonstrate that induction of the ESFT CSC phenotype is the result of the combined effect of EWS-FLI-1 on its target gene expression and repression of microRNA-145 (miRNA145) promoter activity. Finally, we provide evidence that EWS-FLI-1 and miRNA-145 function in a mutually repressive feedback loop and identify their common target gene, SOX2, in addition to miRNA145 itself, as key players in ESFT cell differentiation and tumorigenicity. Our observations provide insight for the first time into the mechanisms whereby a single oncogene can reprogram primary cells to display a CSC phenotype.


Asunto(s)
Reprogramación Celular , Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/citología , MicroARNs/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Factores de Transcripción SOXB1/metabolismo , Adolescente , Adulto , Diferenciación Celular , Línea Celular Tumoral , Niño , Proteínas de Homeodominio/metabolismo , Humanos , Proteína Homeótica Nanog , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Sarcoma de Ewing/fisiopatología , Células Tumorales Cultivadas
14.
Cancer Res ; 69(24): 9211-8, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19934320

RESUMEN

Overexpression of the polycomb group protein enhancer of zeste homologue 2 (EZH2) occurs in diverse malignancies, including prostate cancer, breast cancer, and glioblastoma multiforme (GBM). Based on its ability to modulate transcription of key genes implicated in cell cycle control, DNA repair, and cell differentiation, EZH2 is believed to play a crucial role in tissue-specific stem cell maintenance and tumor development. Here, we show that targeted pharmacologic disruption of EZH2 by the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep), or its specific downregulation by short hairpin RNA (shRNA), strongly impairs GBM cancer stem cell (CSC) self-renewal in vitro and tumor-initiating capacity in vivo. Using genome-wide expression analysis of DZNep-treated GBM CSCs, we found the expression of c-myc, recently reported to be essential for GBM CSCs, to be strongly repressed upon EZH2 depletion. Specific shRNA-mediated downregulation of EZH2 in combination with chromatin immunoprecipitation experiments revealed that c-myc is a direct target of EZH2 in GBM CSCs. Taken together, our observations provide evidence that direct transcriptional regulation of c-myc by EZH2 may constitute a novel mechanism underlying GBM CSC maintenance and suggest that EZH2 may be a valuable new therapeutic target for GBM management.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Glioblastoma/patología , Células Madre Neoplásicas/patología , Factores de Transcripción/biosíntesis , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes myc , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 2 , ARN Interferente Pequeño/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
15.
Cancer Res ; 69(5): 1776-81, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19208848

RESUMEN

Cancer stem cells that display tumor-initiating properties have recently been identified in several distinct types of malignancies, holding promise for more effective therapeutic strategies. However, evidence of such cells in sarcomas, which include some of the most aggressive and therapy-resistant tumors, has not been shown to date. Here, we identify and characterize cancer stem cells in Ewing's sarcoma family tumors (ESFT), a highly aggressive pediatric malignancy believed to be of mesenchymal stem cell (MSC) origin. Using magnetic bead cell separation of primary ESFT, we have isolated a subpopulation of CD133+ tumor cells that display the capacity to initiate and sustain tumor growth through serial transplantation in nonobese diabetic/severe combined immunodeficiency mice, re-establishing at each in vivo passage the parental tumor phenotype and hierarchical cell organization. Consistent with the plasticity of MSCs, in vitro differentiation assays showed that the CD133+ cell population retained the ability to differentiate along adipogenic, osteogenic, and chondrogenic lineages. Quantitative real-time PCR analysis of genes implicated in stem cell maintenance revealed that CD133+ ESFT cells express significantly higher levels of OCT4 and NANOG than their CD133- counterparts. Taken together, our observations provide the first identification of ESFT cancer stem cells and demonstration of their MSC properties, a critical step towards a better biological understanding and rational therapeutic targeting of these tumors.


Asunto(s)
Neoplasias Óseas/patología , Separación Inmunomagnética/métodos , Células Madre Neoplásicas/patología , Sarcoma de Ewing/patología , Antígeno AC133 , Animales , Antígenos CD/análisis , Línea Celular Tumoral , Glicoproteínas/análisis , Humanos , Ratones , Péptidos/análisis
16.
Cancer Res ; 68(18): 7323-31, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18794119

RESUMEN

Radiotherapy is widely used to treat human cancer. Patients locally recurring after radiotherapy, however, have increased risk of metastatic progression and poor prognosis. The clinical management of postradiation recurrences remains an unresolved issue. Tumors growing in preirradiated tissues have an increased fraction of hypoxic cells and are more metastatic, a condition known as tumor bed effect. The transcription factor hypoxia inducible factor (HIF)-1 promotes invasion and metastasis of hypoxic tumors, but its role in the tumor bed effect has not been reported. Here, we show that tumor cells derived from SCCVII and HCT116 tumors growing in a preirradiated bed, or selected in vitro through repeated cycles of severe hypoxia, retain invasive and metastatic capacities when returned to normoxia. HIF activity, although facilitating metastatic spreading of tumors growing in a preirradiated bed, is not essential. Through gene expression profiling and gain- and loss-of-function experiments, we identified the matricellular protein CYR61 and alphaVbeta5 integrin as proteins cooperating to mediate these effects. The anti-alphaV integrin monoclonal antibody 17E6 and the small molecular alphaVbeta3/alphaVbeta5 integrin inhibitor EMD121974 suppressed invasion and metastasis induced by CYR61 and attenuated metastasis of tumors growing within a preirradiated field. These results represent a conceptual advance to the understanding of the tumor bed effect and identify CYR61 and alphaVbeta5 integrin as proteins that cooperate to mediate metastasis. They also identify alphaV integrin inhibition as a potential therapeutic approach for preventing metastasis in patients at risk for postradiation recurrences.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Colorrectales/patología , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Traumatismos Experimentales por Radiación/patología , Receptores de Vitronectina/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Hipoxia de la Célula/efectos de la radiación , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteína 61 Rica en Cisteína , Perfilación de la Expresión Génica , Células HCT116 , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Invasividad Neoplásica , Traumatismos Experimentales por Radiación/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/biosíntesis , Células del Estroma/patología , Células del Estroma/efectos de la radiación
17.
Cancer Res ; 68(7): 2176-85, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381423

RESUMEN

Ewing's sarcoma family tumors (ESFT) express the EWS-FLI-1 fusion gene generated by the chromosomal translocation t(11;22)(q24;q12). Expression of the EWS-FLI-1 fusion protein in a permissive cellular environment is believed to play a key role in ESFT pathogenesis. However, EWS-FLI-1 induces growth arrest or apoptosis in differentiated primary cells, and the identity of permissive primary human cells that can support its expression and function has until now remained elusive. Here we show that expression of EWS-FLI-1 in human mesenchymal stem cells (hMSC) is not only stably maintained without inhibiting proliferation but also induces a gene expression profile bearing striking similarity to that of ESFT, including genes that are among the highest ESFT discriminators. Expression of EWS-FLI-1 in hMSCs may recapitulate the initial steps of Ewing's sarcoma development, allowing identification of genes that play an important role early in its pathogenesis. Among relevant candidate transcripts induced by EWS-FLI-1 in hMSCs, we found the polycomb group gene EZH2, which we show to play a critical role in Ewing's sarcoma growth. These observations are consistent with our recent findings using mouse mesenchymal progenitor cells and provide compelling evidence that hMSCs are candidate cells of origin of ESFT.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Proteínas de Fusión Oncogénica/biosíntesis , Proteína Proto-Oncogénica c-fli-1/biosíntesis , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Diferenciación Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Huésped Inmunocomprometido , Ratones , Proteínas de Fusión Oncogénica/genética , Fenotipo , Complejo Represivo Polycomb 2 , Proteínas/genética , Proteína Proto-Oncogénica c-fli-1/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteína EWS de Unión a ARN , Sarcoma de Ewing/genética , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología
18.
Infect Immun ; 76(5): 1940-51, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18332214

RESUMEN

Cervical cancer, the second leading cause of cancer deaths in women, is the consequence of high-risk human papillomavirus (HPV) infections. Toward the development of therapeutic vaccines that can induce both innate and adaptive mucosal immune responses, we analyzed intravaginal (ivag) vaccine delivery of live attenuated Salmonella enterica serovar Typhimurium expressing HPV16L1 as a model antigen. Innate immune responses were examined in cervicovaginal tissues by determining gene expression patterns by microarray analysis using nylon membranes imprinted with cDNA fragments coding for inflammation-associated genes. At 24 h, a wide range of genes, including those for chemokines and Th1- and Th2-type cytokine and chemokine receptors were up-regulated in mice ivag immunized with Salmonella compared to control mice. However, the majority of transcripts returned to their steady-state levels 1 week after immunization, suggesting a transient inflammatory response. Indeed, cervicovaginal histology of immunized mice showed a massive, but transient, infiltration of macrophages and neutrophils, while T cells were still increased after 7 days. Ivag immunization also induced humoral and antitumor immune responses, i.e., serum and vaginal anti-HPV16VLP antibody titers similar to those induced by oral immunization, and significant protection in tumor protection experiments using HPV16-expressing C3 tumor cells. These results show that ivag immunization with live attenuated Salmonella expressing HPV16 antigens modulates the local mucosal gene expression pattern into a transient proinflammatory profile, elicits strong systemic and mucosal immunity against HPV16, and confers protection against HPV16 tumor cells subcutaneously implanted in mice. Examination of the efficacy with which ivag HPV16E7E6 Salmonella induces regression of tumors located in cervicovaginal tissue is warranted.


Asunto(s)
Administración Intravaginal , Proteínas de la Cápside/inmunología , Papillomavirus Humano 16/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/inmunología , Salmonella typhimurium/genética , Animales , Anticuerpos Antineoplásicos , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/genética , Cuello del Útero/inmunología , Citocinas/biosíntesis , Citocinas/genética , Femenino , Perfilación de la Expresión Génica , Papillomavirus Humano 16/genética , Macrófagos/inmunología , Ratones , Neoplasias/inmunología , Neoplasias/prevención & control , Neutrófilos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas Virales/genética , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/genética , Salmonella typhimurium/crecimiento & desarrollo , Linfocitos T/inmunología , Factores de Tiempo , Vagina/inmunología
19.
PLoS One ; 1: e32, 2006 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-17183660

RESUMEN

Primary and metastatic tumor growth induces host tissue responses that are believed to support tumor progression. Understanding the molecular changes within the tumor microenvironment during tumor progression may therefore be relevant not only for discovering potential therapeutic targets, but also for identifying putative molecular signatures that may improve tumor classification and predict clinical outcome. To selectively address stromal gene expression changes during cancer progression, we performed cDNA microarray analysis of laser-microdissected stromal cells derived from prostate intraepithelial neoplasia (PIN) and invasive cancer in a multistage model of prostate carcinogenesis. Human orthologs of genes identified in the stromal reaction to tumor progression in this mouse model were observed to be expressed in several human cancers, and to cluster prostate and breast cancer patients into groups with statistically different clinical outcomes. Univariate Cox analysis showed that overexpression of these genes is associated with shorter survival and recurrence-free periods. Taken together, our observations provide evidence that the expression signature of the stromal response to tumor invasion in a mouse tumor model can be used to probe human cancer, and to provide a powerful prognostic indicator for some of the most frequent human malignancies.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Catepsina D/genética , Movimiento Celular , Proliferación Celular , Supervivencia sin Enfermedad , Femenino , Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/patología , Securina , Células del Estroma/metabolismo , Células del Estroma/patología
20.
Cancer Res ; 66(14): 7016-23, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16849546

RESUMEN

A subset of sarcomas is associated with specific chromosomal translocations that give rise to fusion genes believed to participate in transformation and oncogenesis. Identification of the primary cell environment that provides permissiveness for the oncogenic potential of these fusion genes is essential to understand sarcoma pathogenesis. We have recently shown that expression of the EWS-FLI-1 fusion protein in primary mesenchymal progenitor cells (MPCs) suffices to develop Ewing's sarcoma-like tumors in mice. Because most sarcomas bearing unique chromosomal translocations are believed to originate from common progenitor cells, and because MPCs populate most organs, we expressed the sarcoma-associated fusion proteins FUS/TLS-CHOP, EWS-ATF1, and SYT-SSX1 in MPCs and tested the tumorigenic potential of these cells in vivo. Whereas expression of EWS-ATF1 and SYT-SSX1 failed to transform MPCs, FUS-CHOP-expressing cells formed tumors resembling human myxoid liposarcoma. Transcription profile analysis of these tumors revealed induction of transcripts known to be associated with myxoid liposarcoma and novel candidate genes, including PDGFA, whose expression was confirmed in human tumor samples. MPC(FUS-CHOP) and the previously described MPC(EWS-FLI-1) tumors displayed distinct transcription profiles, consistent with the different target gene repertoires of their respective fusion proteins. Unexpectedly, a set of genes implicated in cell survival and adhesion displayed similar behavior in the two tumors, suggesting events that may be common to primary MPC transformation. Taken together, our observations suggest that expression of FUS-CHOP may be the initiating event in myxoid liposarcoma pathogenesis, and that MPCs may constitute one cell type from which these tumors originate.


Asunto(s)
Transformación Celular Neoplásica/genética , Liposarcoma Mixoide/metabolismo , Liposarcoma Mixoide/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Proteínas de Fusión Oncogénica/biosíntesis , Proteína FUS de Unión a ARN/biosíntesis , Factor de Transcripción CHOP/biosíntesis , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células de la Médula Ósea/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Liposarcoma Mixoide/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteínas de Fusión Oncogénica/genética , Proteína FUS de Unión a ARN/genética , Factor de Transcripción CHOP/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA