Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Breast Cancer Res ; 24(1): 75, 2022 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-36333737

RESUMEN

BACKGROUND: Breast cancer is a heterogenous disease with several histological and molecular subtypes. Models that represent these subtypes are essential for translational research aimed at improving clinical strategy for targeted therapeutics. METHODS: Different combinations of genetic aberrations (Brca1 and Trp53 loss, and inhibition of proteins of the Rb family) were induced in the mammary gland by injection of adenovirus expressing Cre recombinase into the mammary ducts of adult genetically engineered mice. Mammary tumors with different genetic aberrations were classified into molecular subtypes based on expression of molecular markers and RNAseq analysis. In vitro potency assays and Western blots were used to examine their drug sensitivities. RESULTS: Induction of Brca1 and Trp53 loss in mammary ductal epithelium resulted in development of basal-like hormone receptor (HR)-negative mammary tumors. Inhibition of Rb and Trp53 loss or the combination of Rb, Trp53 and Brca1 aberrations resulted in development of luminal ductal carcinoma positive for ER, PR, and Her2 expression. HR positivity in tumors with Rb, Trp53 and Brca1 aberrations indicated that functionality of the Rb pathway rather than Brca1 status affected HR status in these models. Mammary tumor gene expression profiles recapitulated human basal-like or luminal B breast cancer signatures, but HR-positive luminal cancer models were endocrine resistant and exhibited upregulation of PI3K signaling and sensitivity to this pathway inhibition. Furthermore, both tumor subtypes were resistant to CDK4/6 inhibition. CONCLUSIONS: Examination of molecular expression profiles and drug sensitivities of tumors indicate that these breast cancer models can be utilized as a translational platform for evaluation of targeted combinations to improve chemotherapeutic response in patients that no longer respond to hormone therapy or that are resistant to CDK4/6 inhibition.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Humanas , Neoplasias Mamarias Animales , Ratones , Animales , Humanos , Femenino , Glándulas Mamarias Humanas/metabolismo , Fosfatidilinositol 3-Quinasas , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Animales/patología , Epitelio/metabolismo , Hormonas , Proteína BRCA1/genética
2.
Cell Rep Med ; 5(9): 101696, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39178856

RESUMEN

Pancreatic adenocarcinoma (PDAC) is one the most intractable cancers, in part due to its highly inflammatory microenvironment and paucity of infiltrating dendritic cells (DCs). Here, we find that genetic ablation or antibody blockade of tumor necrosis factor receptor 1 (TNFR1) enhanced intratumor T cell activation and slowed PDAC growth. While anti-PD-1 checkpoint inhibition alone had little effect, it further enhanced intratumor T cell activation in combination with anti-TNFR1. The major cellular alteration in the tumor microenvironment in the absence of TNFR1 signaling was a large increase in DC number and immunostimulatory phenotype. This may reflect a direct effect on DCs, because TNF induced TNFR1-dependent apoptosis of bone-marrow-derived DCs. The therapeutic response to anti-TNFR1 alone was superior to the combination of DC-activating agonistic anti-CD40 and Flt3 ligand (Flt3L). These observations suggest that targeting TNFR1, perhaps in concert with other strategies that promote DC generation and mobilization, may have therapeutic benefits.


Asunto(s)
Células Dendríticas , Neoplasias Pancreáticas , Receptores Tipo I de Factores de Necrosis Tumoral , Transducción de Señal , Células Dendríticas/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Animales , Ratones , Humanos , Microambiente Tumoral , Ratones Endogámicos C57BL , Línea Celular Tumoral , Activación de Linfocitos/inmunología , Apoptosis , Proliferación Celular , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Mol Cancer Ther ; 20(10): 2082-2092, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34315768

RESUMEN

Antibody-based therapies designed for human use frequently fail to cross-react with the murine isoform of their target. Because of this problem, preclinical studies of antibody-based mesothelin (Msl)-targeted therapeutics in immunocompetent systems have been limited by the lack of suitable mouse models. Here, we describe two immunocompetent humanized mesothelin transgenic mouse lines that can act as tolerant hosts for C57Bl/6-syngeneic cell lines expressing the human isoform of mesothelin. Thyroid peroxidase (TPO) mice have thyroid-restricted human mesothelin expression. Mesothelin (Msl) mice express human mesothelin in the typical serosal membrane distribution and can additionally be utilized to assess on-target, off-tumor toxicity of human mesothelin-targeted therapeutics. Both transgenic strains shed human mesothelin into the serum like human mesothelioma and patients with ovarian cancer, and serum human mesothelin can be used as a blood-based surrogate of tumor burden. Using these models, we examined the on-target toxicity and antitumor activity of human mesothelin-targeted recombinant immunotoxins. We found that immunotoxin treatment causes acute and chronic histologic changes to serosal membranes in Msl mice, while human mesothelin-expressing thyroid follicular cells in TPO mice are resistant to immunotoxin despite excellent drug delivery. Furthermore, poor delivery of immunotoxin to syngeneic orthotopic human mesothelin-expressing pancreatic adenocarcinoma limits antitumor activity both alone and in combination with immune checkpoint inhibition. In summary, we have developed two high-fidelity, immunocompetent murine models for human cancer that allow for rigorous preclinical evaluation of human mesothelin-targeted therapeutics.


Asunto(s)
Adenocarcinoma/terapia , Mesotelina/administración & dosificación , Mesotelioma/terapia , Neoplasias Pancreáticas/terapia , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Proliferación Celular , Femenino , Ingeniería Genética , Humanos , Masculino , Mesotelina/genética , Mesotelina/metabolismo , Mesotelioma/genética , Mesotelioma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 20(10): 1926-1940, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376576

RESUMEN

The desmoplastic stroma of pancreatic cancers forms a physical barrier that impedes intratumoral drug delivery. Attempts to modulate the desmoplastic stroma to increase delivery of administered chemotherapy have not shown positive clinical results thus far, and preclinical reports in which chemotherapeutic drugs were coadministered with antistromal therapies did not universally demonstrate increased genotoxicity despite increased intratumoral drug levels. In this study, we tested whether TGFß antagonism can break the stromal barrier, enhance perfusion and tumoral drug delivery, and interrogated cellular and molecular mechanisms by which the tumor prevents synergism with coadministered gemcitabine. TGFß inhibition in genetically engineered murine models (GEMM) of pancreas cancer enhanced tumoral perfusion and increased intratumoral gemcitabine levels. However, tumors rapidly adapted to TGFß-dependent stromal modulation, and intratumoral perfusion returned to pre-treatment levels upon extended TGFß inhibition. Perfusion was governed by the phenotypic identity and distribution of cancer-associated fibroblasts (CAF) with the myelofibroblastic phenotype (myCAFs), and myCAFs which harbored unique genomic signatures rapidly escaped the restricting effects of TGFß inhibition. Despite the reformation of the stromal barrier and reversal of initially increased intratumoral exposure levels, TGFß inhibition in cooperation with gemcitabine effectively suppressed tumor growth via cooperative reprogramming of T regulatory cells and stimulation of CD8 T cell-mediated antitumor activity. The antitumor activity was further improved by the addition of anti-PD-L1 immune checkpoint blockade to offset adaptive PD-L1 upregulation induced by TGFß inhibition. These findings support the development of combined antistroma anticancer therapies capable of impacting the tumor beyond the disruption of the desmoplastic stroma as a physical barrier to improve drug delivery.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/inmunología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/inmunología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Células del Estroma/inmunología , Microambiente Tumoral , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Proliferación Celular , Terapia Combinada , Desoxicitidina/farmacología , Humanos , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
5.
Cancer Res ; 80(8): 1630-1643, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31911550

RESUMEN

Pancreatic cancer is a disease with limited therapeutic options. Resistance to chemotherapies poses a significant clinical challenge for patients with pancreatic cancer and contributes to a high rate of recurrence. Oncogenic KRAS, a critical driver of pancreatic cancer, promotes metabolic reprogramming and upregulates NRF2, a master regulator of the antioxidant network. Here, we show that NRF2 contributed to chemoresistance and was associated with a poor prognosis in patients with pancreatic cancer. NRF2 activation metabolically rewired and elevated pathways involved in glutamine metabolism. This curbed chemoresistance in KRAS-mutant pancreatic cancers. In addition, manipulating glutamine metabolism restrained the assembly of stress granules, an indicator of chemoresistance. Glutaminase inhibitors sensitized chemoresistant pancreatic cancer cells to gemcitabine, thereby improving the effectiveness of chemotherapy. This therapeutic approach holds promise as a novel therapy for patients with pancreatic cancer harboring KRAS mutation. SIGNIFICANCE: These findings illuminate the mechanistic features of KRAS-mediated chemoresistance and provide a rationale for exploiting metabolic reprogramming in pancreatic cancer cells to confer therapeutic opportunities that could be translated into clinical trials. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/8/1630/F1.large.jpg.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Glutamina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Glutaminasa/antagonistas & inhibidores , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Mutación , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Pronóstico , Distribución Aleatoria , Análisis de Matrices Tisulares , Regulación hacia Arriba , Gemcitabina
6.
J Pharm Biomed Anal ; 181: 113093, 2020 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-31931447

RESUMEN

Synthetic host defense peptides (HDP) are a new class of promising therapeutic agents with potential application in a variety of diseases. RP-182 is a 10mer synthetic HDP design, which selectively reduces M2-like tumor associated macrophages via engagement with the cell surface lectin receptor MRC1/CD206 and is currently being developed as an innate immune defense regulator to improve anti-tumor immunity in immunologically cold tumors. Herein, we describe a sensitive and specific liquid chromatography (LC) coupled to quadrupole electron spray tandem mass spectrometry method to measure positively charged HDPs and HDP peptide fragments in complex biological matrices. Carboxylic acid magnetic beads were used as an affinity-capturing agent to extract the positively charged RP-182 from both mouse plasma and tissue homogenates. Beads were eluted with 0.1% (v/v) formic acid and chromatographic separation was achieved on a Waters 2.1 × 100 mm, 3.5 µm XSelect Peptide CSH C18 column with a Vanguard pre-column of the same phase. MS/MS was performed on a Thermo TSQ Quantiva triple quadrupole mass spectrometer operating in Selected Reaction Monitoring (SRM) mode fragmenting the plus three parent ion 458.9+3 and monitoring ions 624.0+2, 550.5+2, and 597.3+1 for RP-182 and 462.4+3 > 629.1+2, 555.5+2, and 607.3+1 for isotopic RP-182 standard. The assay had good linearity ranging from 1 ng to 1000 ng in mouse plasma with the lower limit of detection for RP-182 at 1 ng in mouse plasma with good intra- and inter-sample precision and accuracy. Recovery ranged from 66% to 77% with minimum matrix effects. The method was successfully applied to an abbreviated pharmacokinetic study in mice after single IP injection of RP-182. The method was successfully tested on a second HDP, the 17mer D4E1, and the cationic human peptide hormone ghrelin suggesting that it might be a general sensitive method applicable to quantifying HDP peptides that are difficult to extract.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Animales , Péptidos Catiónicos Antimicrobianos/sangre , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacocinética , Ácidos Carboxílicos/química , Cromatografía de Afinidad/métodos , Cromatografía Líquida de Alta Presión/métodos , Evaluación Preclínica de Medicamentos/métodos , Ghrelina/sangre , Ghrelina/química , Ghrelina/aislamiento & purificación , Límite de Detección , Fenómenos Magnéticos , Ratones , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/métodos
7.
Sci Transl Med ; 12(530)2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32051227

RESUMEN

Solid tumors elicit a detectable immune response including the infiltration of tumor-associated macrophages (TAMs). Unfortunately, this immune response is co-opted into contributing toward tumor growth instead of preventing its progression. We seek to reestablish an antitumor immune response by selectively targeting surface receptors and endogenous signaling processes of the macrophage subtypes driving cancer progression. RP-182 is a synthetic 10-mer amphipathic analog of host defense peptides that selectively induces a conformational switch of the mannose receptor CD206 expressed on TAMs displaying an M2-like phenotype. RP-182-mediated activation of this receptor in human and murine M2-like macrophages elicits a program of endocytosis, phagosome-lysosome formation, and autophagy and reprograms M2-like TAMs to an antitumor M1-like phenotype. In syngeneic and autochthonous murine cancer models, RP-182 suppressed tumor growth, extended survival, and was an effective combination partner with chemo- or immune checkpoint therapy. Antitumor activity of RP-182 was also observed in CD206high patient-derived xenotransplantation models. Mechanistically, via selective reduction of immunosuppressive M2-like TAMs, RP-182 improved adaptive and innate antitumor immune responses, including increased cancer cell phagocytosis by reprogrammed TAMs.


Asunto(s)
Lectinas de Unión a Manosa , Macrófagos Asociados a Tumores , Animales , Línea Celular Tumoral , Humanos , Inmunidad Innata , Lectinas Tipo C , Receptor de Manosa , Ratones , Receptores de Superficie Celular
8.
Cancer Chemother Pharmacol ; 82(6): 1067-1080, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30306263

RESUMEN

PURPOSE: Metarrestin is a first-in-class small molecule clinical candidate capable of disrupting the perinucleolar compartment, a subnuclear structure unique to metastatic cancer cells. This study aims to define the pharmacokinetic (PK) profile of metarrestin and the pharmacokinetic/pharmacodynamic relationship of metarrestin-regulated markers. METHODS: PK studies included the administration of single or multiple dose of metarrestin at 3, 10, or 25 mg/kg via intravenous (IV) injection, gavage (PO) or with chow to wild-type C57BL/6 mice and KPC mice bearing autochthonous pancreatic tumors. Metarrestin concentrations were analyzed by UPLC-MS/MS. Pharmacodynamic assays included mRNA expression profiling by RNA-seq and qRT-PCR for KPC mice. RESULTS: Metarrestin had a moderate plasma clearance of 48 mL/min/kg and a large volume of distribution of 17 L/kg at 3 mg/kg IV in C57BL/6 mice. The oral bioavailability after single-dose (SD) treatment was > 80%. In KPC mice treated with SD 25 mg/kg PO, plasma AUC0-∞ of 14400 ng h/mL, Cmax of 810 ng/mL and half-life (t1/2) of 8.5 h were observed. At 24 h after SD of 25 mg/kg PO, the intratumor concentration of metarrestin was high with a mean value of 6.2 µg/g tissue (or 13 µM), well above the cell-based IC50 of 0.4 µM. At multiple dose (MD) 25 mg/kg/day PO in KPC mice, mean tissue/plasma AUC0-24h ratio for tumor, spleen and liver was 37, 30 and 31, respectively. There was a good linear relationship of dosage to AUC0-24h and C24h. AUC0-24h MD to AUC0-24h SD ratios ranged from two for liver to five for tumor indicating additional accumulation in tumors. Dose-dependent normalization of FOXA1 and FOXO6 mRNA expression was observed in KPC tumors. CONCLUSIONS: Metarrestin is an effective therapeutic candidate with a favorable PK profile achieving excellent intratumor tissue levels in a disease with known poor drug delivery.


Asunto(s)
Antineoplásicos/farmacocinética , Orgánulos/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Pirimidinas/farmacocinética , Pirroles/farmacocinética , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Antineoplásicos/uso terapéutico , Área Bajo la Curva , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Factores de Transcripción Forkhead/genética , Semivida , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Orgánulos/metabolismo , Orgánulos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pirimidinas/administración & dosificación , Pirimidinas/sangre , Pirimidinas/uso terapéutico , Pirroles/administración & dosificación , Pirroles/sangre , Pirroles/uso terapéutico , Distribución Tisular
9.
J Clin Invest ; 111(3): 389-97, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12569165

RESUMEN

Protease inhibitors decrease the viral load in HIV patients, however the patients develop hypertriglyceridemia, hypercholesterolemia, and atherosclerosis. It has been assumed that protease inhibitor-dependent increases in atherosclerosis are secondary to the dyslipidemia. Incubation of THP-1 cells or human PBMCs with protease inhibitors caused upregulation of CD36 and the accumulation of cholesteryl esters. The use of CD36-blocking antibodies, a CD36 morpholino, and monocytes isolated from CD36 null mice demonstrated that protease inhibitor-induced increases in cholesteryl esters were dependent on CD36 upregulation. These data led to the hypothesis that protease inhibitors induce foam cell formation and consequently atherosclerosis by upregulating CD36 and cholesteryl ester accumulation independent of dyslipidemia. Studies with LDL receptor null mice demonstrated that low doses of protease inhibitors induce an increase in the level of CD36 and cholesteryl ester in peritoneal macrophages and the development of atherosclerosis without altering plasma lipids. Furthermore, the lack of CD36 protected the animals from protease inhibitor-induced atherosclerosis. Finally, ritonavir increased PPAR-gamma and CD36 mRNA levels in a PKC- and PPAR-gamma-dependent manner. We conclude that protease inhibitors contribute to the formation of atherosclerosis by promoting the upregulation of CD36 and the subsequent accumulation of sterol in macrophages.


Asunto(s)
Arteriosclerosis/inducido químicamente , Antígenos CD36/biosíntesis , Ésteres del Colesterol/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Macrófagos/metabolismo , Animales , Antígenos CD36/metabolismo , Hiperlipidemias/tratamiento farmacológico , Inmunoglobulina M/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Proteína Quinasa C/metabolismo , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
10.
J Clin Invest ; 111(10): 1579-87, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12750408

RESUMEN

Cardiovascular diseases remain the leading cause of death in the United States. Two factors associated with a decreased risk of developing cardiovascular disease are elevated HDL levels and sex - specifically, a decreased risk is found in premenopausal women. HDL and estrogen stimulate eNOS and the production of nitric oxide, which has numerous protective effects in the vascular system including vasodilation, antiadhesion, and anti-inflammatory effects. We tested the hypothesis that HDL binds to its receptor, scavenger receptor class B type I (SR-BI), and delivers estrogen to eNOS, thereby stimulating the enzyme. HDL isolated from women stimulated eNOS, whereas HDL isolated from men had minimal activity. Studies with ovariectomized and ovariectomized/estrogen replacement mouse models demonstrated that HDL-associated estradiol stimulation of eNOS is SR-BI dependent. Furthermore, female HDL, but not male HDL, promoted the relaxation of muscle strips isolated from C57BL/6 mice but not SR-BI null mice. Finally, HDL isolated from premenopausal women or postmenopausal women receiving estradiol replacement therapy stimulated eNOS, whereas HDL isolated from postmenopausal women did not stimulate eNOS. We conclude that HDL-associated estrodial is capable of the stimulating eNOS. These studies establish a new paradigm for examining the cardiovascular effects of HDL and estrogen.


Asunto(s)
Antígenos CD36/metabolismo , Endotelio Vascular/efectos de los fármacos , Estradiol/farmacología , Lipoproteínas HDL/farmacología , Proteínas de la Membrana , Óxido Nítrico Sintasa/metabolismo , Receptores Inmunológicos , Receptores de Lipoproteína , Animales , Antígenos CD36/genética , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Estradiol/metabolismo , Femenino , Arteria Femoral/efectos de los fármacos , Arteria Femoral/fisiología , Terapia de Reemplazo de Hormonas , Humanos , Técnicas In Vitro , Lipoproteínas HDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Ovariectomía , Receptores Depuradores , Receptores Depuradores de Clase B , Factores Sexuales , Vasodilatación/efectos de los fármacos
11.
Cancer Res ; 72(16): 4141-53, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22617326

RESUMEN

The majority of human high-grade serous epithelial ovarian cancer (SEOC) is characterized by frequent mutations in p53 and alterations in the RB and FOXM1 pathways. A subset of human SEOC harbors a combination of germline and somatic mutations as well as epigenetic dysfunction for BRCA1/2. Using Cre-conditional alleles and intrabursal induction by Cre-expressing adenovirus in genetically engineered mice, we analyzed the roles of pathway perturbations in epithelial ovarian cancer initiation and progression. Inactivation of RB-mediated tumor suppression induced surface epithelial proliferation with progression to stage I carcinoma. Additional biallelic inactivation and/or missense p53 mutation in the presence or absence of Brca1/2 caused progression to stage IV disease. As in human SEOC, mice developed peritoneal carcinomatosis, ascites, and distant metastases. Unbiased gene expression and metabolomic profiling confirmed that Rb, p53, and Brca1/2-triple mutant tumors aligned with human SEOC, and not with other intraperitoneal cancers. Together, our findings provide a novel resource for evaluating disease etiology and biomarkers, therapeutic evaluation, and improved imaging strategies in epithelial ovarian cancer.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Epitelial de Ovario , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mutación , Metástasis de la Neoplasia , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Proteína p53 Supresora de Tumor/genética
12.
Cancer Res ; 72(22): 5921-33, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22969147

RESUMEN

Patients with lung cancer with activating mutations in the EGF receptor (EGFR) kinase, who are treated long-term with tyrosine kinase inhibitors (TKI), often develop secondary mutations in EGFR associated with resistance. Mice engineered to develop lung adenocarcinomas driven by the human EGFR T790M resistance mutation are similarly resistant to the EGFR TKI erlotinib. By tumor volume endpoint analysis, these mouse tumors respond to BIBW 2992 (an irreversible EGFR/HER2 TKI) and rapamycin combination therapy. To correlate EGFR-driven changes in the lung with response to drug treatment, we conducted an integrative analysis of global transcriptome and metabolite profiling compared with quantitative imaging and histopathology at several time points during tumor progression and treatment. Responses to single-drug treatments were temporary, whereas combination therapy elicited a sustained response. During tumor development, metabolomic signatures indicated a shift to high anabolic activity and suppression of antitumor programs with 11 metabolites consistently present in both lung tissue and blood. Combination drug treatment reversed many of the molecular changes found in tumored lung. Data integration linking cancer signaling networks with metabolic activity identified key pathways such as glutamine and glutathione metabolism that signified response to single or dual treatments. Results from combination drug treatment suggest that metabolic transcriptional control through C-MYC and SREBP, as well as ELK1, NRF1, and NRF2, depends on both EGFR and mTORC1 signaling. Our findings establish the importance of kinetic therapeutic studies in preclinical assessment and provide in vivo evidence that TKI-mediated antiproliferative effects also manifest in specific metabolic regulation.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/farmacología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Afatinib , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Procesos de Crecimiento Celular/fisiología , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/administración & dosificación , Sirolimus/administración & dosificación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Cell Metab ; 11(3): 194-205, 2010 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-20197052

RESUMEN

Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for triacylglycerol (TAG) hydrolysis in adipocytes. The precise mechanisms whereby ATGL is regulated remain uncertain. Here, we demonstrate that a protein encoded by G(0)/G(1) switch gene 2 (G0S2) is a selective regulator of ATGL. G0S2 is highly expressed in adipose tissue and differentiated adipocytes. When overexpressed in HeLa cells, G0S2 localizes to lipid droplets and prevents their degradation mediated by ATGL. Moreover, G0S2 specifically interacts with ATGL through the hydrophobic domain of G0S2 and the patatin-like domain of ATGL. More importantly, interaction with G0S2 inhibits ATGL TAG hydrolase activity. Knockdown of endogenous G0S2 accelerates basal and stimulated lipolysis in adipocytes, whereas overexpression of G0S2 diminishes the rate of lipolysis in both adipocytes and adipose tissue explants. Thus, G0S2 functions to attenuate ATGL action both in vitro and in vivo and by this mechanism regulates TAG hydrolysis.


Asunto(s)
Adipocitos/enzimología , Tejido Adiposo/enzimología , Hidrolasas de Éster Carboxílico/metabolismo , Proteínas de Ciclo Celular/genética , Lipasa/metabolismo , Lipólisis/genética , Adipocitos/ultraestructura , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Lípidos , Ratones , Dominios y Motivos de Interacción de Proteínas , Vacuolas/metabolismo
14.
Am J Physiol Cell Physiol ; 294(1): C295-305, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17977947

RESUMEN

In the current study, we examined whether diabetes affected the ability of HDL to stimulate nitric oxide (NO) production. Using HDL isolated from both diabetic humans and diabetic mouse models, we found that female HDL no longer induced NO synthesis, despite containing equivalent amounts of estrogen as nondiabetic controls. Furthermore, HDL isolated from diabetic females and males prevented acetylcholine-induced stimulation of NO generation. Analyses of both the human and mouse diabetic HDL particles showed that the HDLs contained increased levels of myristic acid. To determine whether myristic acid associated with HDL particles was responsible for the decrease in NO generation, myristic acid was added to HDL isolated from nondiabetic humans and mice. Myristic acid-associated HDL inhibited the generation of NO in a dose-dependent manner. Importantly, diabetic HDL did not alter the levels of endothelial NO synthase or acetylcholine receptors associated with the cells. Surprisingly, diabetic HDL inhibited ionomycin-induced stimulation of NO production without affecting ionomycin-induced increases in intracellular calcium. Further analysis indicated that diabetic HDL prevented calmodulin from interacting with endothelial NO synthase (eNOS) but did not affect the activation of calmodulin kinase or calcium-independent mechanisms for stimulating eNOS. These studies are the first to show that a specific fatty acid associated with HDL inhibits the stimulation of NO generation. These findings have important implications regarding cardiovascular disease in diabetic patients.


Asunto(s)
Acetilcolina/metabolismo , Calmodulina/metabolismo , Diabetes Mellitus/metabolismo , Células Endoteliales/metabolismo , Lipoproteínas HDL/metabolismo , Ácido Mirístico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Estrógenos/metabolismo , Femenino , Humanos , Ionomicina/farmacología , Ionóforos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Mirístico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Receptores Colinérgicos/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Transducción de Señal
15.
Am J Physiol Cell Physiol ; 291(6): C1271-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16822947

RESUMEN

HIV protease inhibitors are important pharmacological agents used in the treatment of HIV-infected patients. One of the major disadvantages of HIV protease inhibitors is that they increase several cardiovascular risk factors, including the expression of CD36 in macrophages. The expression of CD36 in macrophages promotes the accumulation of cholesterol, the development of foam cells, and ultimately atherosclerosis. Recent studies have suggested that alpha-tocopherol can prevent HIV protease inhibitor-induced increases in macrophage CD36 levels. Because of the potential clinical utility of using alpha-tocopherol to limit some of the side effects of HIV protease inhibitors, we tested the ability of alpha-tocopherol to prevent ritonavir, a common HIV protease inhibitor, from inducing atherosclerosis in the LDL receptor (LDLR) null mouse model. Surprisingly, alpha-tocopherol did not prevent ritonavir-induced atherosclerosis. However, cotreatment with the nucleoside reverse transcriptase inhibitors (NRTIs), didanosine or D4T, did prevent ritonavir-induced atherosclerosis. Using macrophages isolated from LDLR null mice, we demonstrated that the NRTIs prevented the upregulation of CD36 and cholesterol accumulation in macrophages. Treatment of LDLR null mice with NRTIs promoted the ubiquitination and downregulation of protein kinase Calpha (PKC). Previous studies demonstrated that HIV protease inhibitor activation of PKC was necessary for the upregulation of CD36. Importantly, the in vivo inhibition of PKC with chelerythrine prevented ritonavir-induced upregulation of CD36, accumulation of cholesterol, and the formation of atherosclerotic lesions. These novel mechanistic studies suggest that NRTIs may provide protection from one of the negative side effects associated with HIV protease inhibitors, namely the increase in CD36 levels and subsequent cholesterol accumulation and atherogenesis.


Asunto(s)
Aterosclerosis/inducido químicamente , Aterosclerosis/prevención & control , Inhibidores de la Proteasa del VIH/efectos adversos , Proteína Quinasa C-alfa/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo , Ritonavir/efectos adversos , Ubiquitina/metabolismo , Animales , Antioxidantes/administración & dosificación , Antioxidantes/metabolismo , Aterosclerosis/metabolismo , Antígenos CD36/metabolismo , Colesterol/metabolismo , Didanosina/metabolismo , Activación Enzimática , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Humanos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa C-alfa/antagonistas & inhibidores , Receptores de LDL/genética , Receptores de LDL/metabolismo , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Ritonavir/metabolismo , Ritonavir/uso terapéutico , Estavudina/metabolismo , alfa-Tocoferol/administración & dosificación , alfa-Tocoferol/metabolismo
16.
J Biol Chem ; 277(26): 23525-33, 2002 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-11976335

RESUMEN

Numerous studies have implicated either the presence or absence of CD36 in the development of hypertension. In addition, hypercholesterolemia is associated with the loss of nitric oxide-induced vasodilation and the subsequent increase in blood pressure. In the current study, we tested the hypothesis that diet-induced hypercholesterolemia promotes the disruption of agonist-stimulated nitric oxide generation and vasodilation in a CD36-dependent manner. To test this, C57BL/6, apoE null, CD36 null, and apoE/CD36 null mice were maintained on chow or high fat diets. In contrast to apoE null mice fed a chow diet, apoE null mice fed a high fat diet did not respond to acetylcholine with a decrease in blood pressure. Caveolae isolated from in vivo vessels did not contain endothelial nitric-oxide synthase and were depleted of cholesterol. Age-matched apoE/CD36 null mice fed a chow or high fat diet responded to acetylcholine with a decrease in blood pressure. The mechanism underlying the vascular dysfunction was reversible because vessels isolated from apoE null high fat-fed mice regained responsiveness to acetylcholine when incubated with plasma obtained from chow-fed mice. Further analysis demonstrated that the plasma low density lipoprotein fraction was responsible for depleting caveolae of cholesterol, removing endothelial nitric-oxide synthase from caveolae, and preventing nitric oxide production. In addition, the pharmacological removal of caveola cholesterol with cyclodextrin mimicked the effects caused by the low density lipoprotein fraction. We conclude that the ablation of CD36 prevented the negative impact of hypercholesterolemia on agonist-stimulated nitric oxide-mediated vasodilation in apoE null mice. These studies provide a direct link between CD36 and the early events that underlie hypercholesterolemia-mediated hypertension and mechanistic linkages between CD36 function, nitric-oxide synthase activation, caveolae integrity, and blood pressure regulation.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Antígenos CD36/fisiología , Hipercolesterolemia/fisiopatología , Óxido Nítrico Sintasa/fisiología , Acetilcolina/farmacología , Animales , Caveolas/fisiología , Ciclodextrinas/toxicidad , Hipercolesterolemia/complicaciones , Hipertensión/etiología , Lipoproteínas LDL/toxicidad , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa/análisis , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Fosforilación , Vasodilatación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA