Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
CA Cancer J Clin ; 71(4): 333-358, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33982817

RESUMEN

Cancer has myriad effects on metabolism that include both rewiring of intracellular metabolism to enable cancer cells to proliferate inappropriately and adapt to the tumor microenvironment, and changes in normal tissue metabolism. With the recognition that fluorodeoxyglucose-positron emission tomography imaging is an important tool for the management of many cancers, other metabolites in biological samples have been in the spotlight for cancer diagnosis, monitoring, and therapy. Metabolomics is the global analysis of small molecule metabolites that like other -omics technologies can provide critical information about the cancer state that are otherwise not apparent. Here, the authors review how cancer and cancer therapies interact with metabolism at the cellular and systemic levels. An overview of metabolomics is provided with a focus on currently available technologies and how they have been applied in the clinical and translational research setting. The authors also discuss how metabolomics could be further leveraged in the future to improve the management of patients with cancer.


Asunto(s)
Metabolómica , Neoplasias/metabolismo , Investigación Biomédica , Humanos , Oncología Médica , Terapia Molecular Dirigida , Neoplasias/terapia
2.
Commun Biol ; 7(1): 608, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769385

RESUMEN

Diverse tumor metabolic phenotypes are influenced by the environment and genetic lesions. Whether these phenotypes extend to rhabdomyosarcoma (RMS) and how they might be leveraged to design new therapeutic approaches remains an open question. Thus, we utilized a Pax7Cre-ER-T2/+; NrasLSL-G12D/+; p53fl/fl (P7NP) murine model of sarcoma with mutations that most frequently occur in human embryonal RMS. To study metabolism, we infuse 13C-labeled glucose or glutamine into mice with sarcomas and show that sarcomas consume more glucose and glutamine than healthy muscle tissue. However, we reveal a marked shift from glucose consumption to glutamine metabolism after radiation therapy (RT). In addition, we show that inhibiting glutamine, either through genetic deletion of glutaminase (Gls1) or through pharmacological inhibition of glutaminase, leads to significant radiosensitization in vivo. This causes a significant increase in overall survival for mice with Gls1-deficient compared to Gls1-proficient sarcomas. Finally, Gls1-deficient sarcomas post-RT elevate levels of proteins involved in natural killer cell and interferon alpha/gamma responses, suggesting a possible role of innate immunity in the radiosensitization of Gls1-deficient sarcomas. Thus, our results indicate that glutamine contributes to radiation response in a mouse model of RMS.


Asunto(s)
Glutaminasa , Glutamina , Sarcoma , Animales , Glutamina/metabolismo , Ratones , Glutaminasa/metabolismo , Glutaminasa/genética , Glutaminasa/antagonistas & inhibidores , Sarcoma/metabolismo , Sarcoma/radioterapia , Sarcoma/genética , Glucosa/metabolismo , Modelos Animales de Enfermedad , Tolerancia a Radiación
3.
bioRxiv ; 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37333373

RESUMEN

The main deterrent to long-term space travel is the risk of Radiation Exposure Induced Death (REID). The National Aeronautics and Space Administration (NASA) has adopted Permissible Exposure Levels (PELs) to limit the probability of REID to 3% for the risk of death due to radiation-induced carcinogenesis. The most significant contributor to current REID estimates for astronauts is the risk of lung cancer. Recently updated lung cancer estimates from Japan's atomic bomb survivors showed that the excess relative risk of lung cancer by age 70 is roughly four-fold higher in females compared to males. However, whether sex differences may impact the risk of lung cancer due to exposure to high charge and energy (HZE) radiation is not well studied. Thus, to evaluate the impact of sex differences on the risk of solid cancer development post-HZE radiation exposure, we irradiated Rb fl/fl ; Trp53 fl/+ male and female mice infected with Adeno-Cre with various doses of 320 kVp X-rays or 600 MeV/n 56 Fe ions and monitored them for any radiation-induced malignancies. We observed that lung adenomas/carcinomas and esthesioneuroblastomas (ENBs) were the most common primary malignancies in X-ray and 56 Fe ion-exposed mice, respectively. In addition, 1 Gy 56 Fe ion exposure compared to X-rays led to a significantly higher incidence of lung adenomas/carcinomas (p=0.02) and ENBs (p<0.0001). However, we did not find a significantly higher incidence of any solid malignancies in female mice as compared to male mice, regardless of radiation quality. Furthermore, gene expression analysis of ENBs suggested a distinct gene expression pattern with similar hallmark pathways altered, such as MYC targets and MTORC1 signaling, in X-ray and 56 Fe ion-induced ENBs. Thus, our data revealed that 56 Fe ion exposure significantly accelerated the development of lung adenomas/carcinomas and ENBs compared to X-rays, but the rate of solid malignancies was similar between male and female mice, regardless of radiation quality.

4.
Mol Cancer Ther ; 22(1): 112-122, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36162051

RESUMEN

This study aims to investigate whether adding neoadjuvant radiotherapy (RT), anti-programmed cell death protein-1 (PD-1) antibody (anti-PD-1), or RT + anti-PD-1 to surgical resection improves disease-free survival for mice with soft tissue sarcomas (STS). We generated a high mutational load primary mouse model of STS by intramuscular injection of adenovirus expressing Cas9 and guide RNA targeting Trp53 and intramuscular injection of 3-methylcholanthrene (MCA) into the gastrocnemius muscle of wild-type mice (p53/MCA model). We randomized tumor-bearing mice to receive isotype control or anti-PD-1 antibody with or without radiotherapy (20 Gy), followed by hind limb amputation. We used micro-CT to detect lung metastases with high spatial resolution, which was confirmed by histology. We investigated whether sarcoma metastasis was regulated by immunosurveillance by lymphocytes or tumor cell-intrinsic mechanisms. Compared with surgery with isotype control antibody, the combination of anti-PD-1, radiotherapy, and surgery improved local recurrence-free survival (P = 0.035) and disease-free survival (P = 0.005), but not metastasis-free survival. Mice treated with radiotherapy, but not anti-PD-1, showed significantly improved local recurrence-free survival and metastasis-free survival over surgery alone (P = 0.043 and P = 0.007, respectively). The overall metastasis rate was low (∼12%) in the p53/MCA sarcoma model, which limited the power to detect further improvement in metastasis-free survival with addition of anti-PD-1 therapy. Tail vein injections of sarcoma cells into immunocompetent mice suggested that impaired metastasis was due to inability of sarcoma cells to grow in the lungs rather than a consequence of immunosurveillance. In conclusion, neoadjuvant radiotherapy improves metastasis-free survival after surgery in a primary model of STS.


Asunto(s)
Sarcoma , Neoplasias de los Tejidos Blandos , Ratones , Animales , Terapia Neoadyuvante , Proteína p53 Supresora de Tumor/genética , Sarcoma/radioterapia , Supervivencia sin Progresión , Supervivencia sin Enfermedad , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/cirugía , Estudios Retrospectivos , Radioterapia Adyuvante , Recurrencia Local de Neoplasia/patología
5.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37200088

RESUMEN

ATRX is one of the most frequently altered genes in solid tumors, and mutation is especially frequent in soft tissue sarcomas. However, the role of ATRX in tumor development and response to cancer therapies remains poorly understood. Here, we developed a primary mouse model of soft tissue sarcoma and showed that Atrx-deleted tumors were more sensitive to radiation therapy and to oncolytic herpesvirus. In the absence of Atrx, irradiated sarcomas had increased persistent DNA damage, telomere dysfunction, and mitotic catastrophe. Our work also showed that Atrx deletion resulted in downregulation of the CGAS/STING signaling pathway at multiple points in the pathway and was not driven by mutations or transcriptional downregulation of the CGAS/STING pathway components. We found that both human and mouse models of Atrx-deleted sarcoma had a reduced adaptive immune response, markedly impaired CGAS/STING signaling, and increased sensitivity to TVEC, an oncolytic herpesvirus that is currently FDA approved for the treatment of aggressive melanomas. Translation of these results to patients with ATRX-mutant cancers could enable genomically guided cancer therapy approaches to improve patient outcomes.


Asunto(s)
Herpesviridae , Sarcoma , Animales , Ratones , Humanos , Transducción de Señal , Sarcoma/genética , Sarcoma/radioterapia , Proteína Nuclear Ligada al Cromosoma X/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Inmunidad Innata
6.
Tomography ; 7(3): 358-372, 2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-34449750

RESUMEN

We are developing imaging methods for a co-clinical trial investigating synergy between immunotherapy and radiotherapy. We perform longitudinal micro-computed tomography (micro-CT) of mice to detect lung metastasis after treatment. This work explores deep learning (DL) as a fast approach for automated lung nodule detection. We used data from control mice both with and without primary lung tumors. To augment the number of training sets, we have simulated data using real augmented tumors inserted into micro-CT scans. We employed a convolutional neural network (CNN), trained with four competing types of training data: (1) simulated only, (2) real only, (3) simulated and real, and (4) pretraining on simulated followed with real data. We evaluated our model performance using precision and recall curves, as well as receiver operating curves (ROC) and their area under the curve (AUC). The AUC appears to be almost identical (0.76-0.77) for all four cases. However, the combination of real and synthetic data was shown to improve precision by 8%. Smaller tumors have lower rates of detection than larger ones, with networks trained on real data showing better performance. Our work suggests that DL is a promising approach for fast and relatively accurate detection of lung tumors in mice.


Asunto(s)
Aprendizaje Profundo , Neoplasias Pulmonares , Animales , Pulmón , Neoplasias Pulmonares/diagnóstico por imagen , Ratones , Redes Neurales de la Computación , Microtomografía por Rayos X
7.
Cancer Discov ; 11(8): 2072-2093, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33757970

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is characterized by accumulation of neutral lipids and adipogenic transdifferentiation. We assessed adipokine expression in ccRCC and found that tumor tissues and patient plasma exhibit obesity-dependent elevations of the adipokine chemerin. Attenuation of chemerin by several approaches led to significant reduction in lipid deposition and impairment of tumor cell growth in vitro and in vivo. A multi-omics approach revealed that chemerin suppresses fatty acid oxidation, preventing ferroptosis, and maintains fatty acid levels that activate hypoxia-inducible factor 2α expression. The lipid coenzyme Q and mitochondrial complex IV, whose biogeneses are lipid-dependent, were found to be decreased after chemerin inhibition, contributing to lipid reactive oxygen species production. Monoclonal antibody targeting chemerin led to reduced lipid storage and diminished tumor growth, demonstrating translational potential of chemerin inhibition. Collectively, the results suggest that obesity and tumor cells contribute to ccRCC through the expression of chemerin, which is indispensable in ccRCC biology. SIGNIFICANCE: Identification of a hypoxia-inducible factor-dependent adipokine that prevents fatty acid oxidation and causes escape from ferroptosis highlights a critical metabolic dependency unique in the clear cell subtype of kidney cancer. Targeting lipid metabolism via inhibition of a soluble factor is a promising pharmacologic approach to expand therapeutic strategies for patients with ccRCC.See related commentary by Reznik et al., p. 1879.This article is highlighted in the In This Issue feature, p. 1861.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Obesidad/complicaciones , Animales , Carcinoma de Células Renales/complicaciones , Línea Celular Tumoral/efectos de los fármacos , Ácidos Grasos/metabolismo , Femenino , Ferroptosis/efectos de los fármacos , Humanos , Neoplasias Renales/complicaciones , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Desnudos
8.
Int J Radiat Oncol Biol Phys ; 108(4): 1091-1102, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32629081

RESUMEN

PURPOSE: Humans are exposed to charged particles in different scenarios. The use of protons and high-linear energy transfer (LET) in cancer treatment is steadily growing. In outer space, astronauts will be exposed to a mixed radiation field composed of both protons and heavy ions, in particularly the long-term space missions outside of earth's magnetosphere. Thus, understanding the radiobiology and transforming potential of these types of ionizing radiation are of paramount importance. METHODS AND MATERIALS: We examined the effect of 10 or 100 cGy of whole-body doses of protons or 28Si ions on the hematopoietic system of a genetic model of aging based on recent studies that showed selective loss of the MLH1 protein in human hematopoietic stems with age. RESULTS: We found that Mlh1 deficient animals are highly prone to develop lymphomas when exposed to either low doses of protons or 28Si ions. The lymphomas that develop are genetically indistinguishable, in spite of different types of damage elicited by low- and high-LET radiation. RNA sequencing analyses reveal similar gene expression patterns, similar numbers of altered genes, similar numbers of single nucleotide variants and insertions and deletions, and similar activation of known leukemogenic loci. CONCLUSIONS: Although the incidence of malignancy is related to radiation quality, and increased due to loss of Mlh1, the phenotype of the tumors is independent of LET.


Asunto(s)
Sistema Hematopoyético/efectos de la radiación , Transferencia Lineal de Energía , Linfoma/genética , Homólogo 1 de la Proteína MutL/deficiencia , Neoplasias Inducidas por Radiación/genética , Protones/efectos adversos , Silicio/efectos adversos , Envejecimiento , Animales , Reparación de la Incompatibilidad de ADN , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Sistema Hematopoyético/fisiología , Humanos , Linfoma/patología , Masculino , Ratones , Homólogo 1 de la Proteína MutL/genética , Neoplasias Inducidas por Radiación/patología , Penetrancia , Exposición a la Radiación/efectos adversos , Análisis de Secuencia de ARN/métodos , Vuelo Espacial , Irradiación Corporal Total/efectos adversos , Irradiación Corporal Total/métodos
9.
Nat Commun ; 11(1): 6410, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335088

RESUMEN

Immunotherapy fails to cure most cancer patients. Preclinical studies indicate that radiotherapy synergizes with immunotherapy, promoting radiation-induced antitumor immunity. Most preclinical immunotherapy studies utilize transplant tumor models, which overestimate patient responses. Here, we show that transplant sarcomas are cured by PD-1 blockade and radiotherapy, but identical treatment fails in autochthonous sarcomas, which demonstrate immunoediting, decreased neoantigen expression, and tumor-specific immune tolerance. We characterize tumor-infiltrating immune cells from transplant and primary tumors, revealing striking differences in their immune landscapes. Although radiotherapy remodels myeloid cells in both models, only transplant tumors are enriched for activated CD8+ T cells. The immune microenvironment of primary murine sarcomas resembles most human sarcomas, while transplant sarcomas resemble the most inflamed human sarcomas. These results identify distinct microenvironments in murine sarcomas that coevolve with the immune system and suggest that patients with a sarcoma immune phenotype similar to transplant tumors may benefit most from PD-1 blockade and radiotherapy.


Asunto(s)
Sarcoma/terapia , Análisis de la Célula Individual/métodos , Microambiente Tumoral/inmunología , Animales , Antineoplásicos Inmunológicos/farmacología , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Ratones Endogámicos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Sarcoma/genética , Sarcoma/inmunología , Escape del Tumor , Microambiente Tumoral/genética , Secuenciación del Exoma
10.
Leukemia ; 33(5): 1135-1147, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30275527

RESUMEN

Cancer-causing genome instability is a major concern during space travel due to exposure of astronauts to potent sources of high-linear energy transfer (LET) ionizing radiation. Hematopoietic stem cells (HSCs) are particularly susceptible to genotoxic stress, and accumulation of damage can lead to HSC dysfunction and oncogenesis. Our group recently demonstrated that aging human HSCs accumulate microsatellite instability coincident with loss of MLH1, a DNA Mismatch Repair (MMR) protein, which could reasonably predispose to radiation-induced HSC malignancies. Therefore, in an effort to reduce risk uncertainty for cancer development during deep space travel, we employed an Mlh1+/- mouse model to study the effects high-LET 56Fe ion space-like radiation. Irradiated Mlh1+/- mice showed a significantly higher incidence of lymphomagenesis with 56Fe ions compared to γ-rays and unirradiated mice, and malignancy correlated with increased MSI in the tumors. In addition, whole-exome sequencing analysis revealed high SNVs and INDELs in lymphomas being driven by loss of Mlh1 and frequently mutated genes had a strong correlation with human leukemias. Therefore, the data suggest that age-related MMR deficiencies could lead to HSC malignancies after space radiation, and that countermeasure strategies will be required to adequately protect the astronaut population on the journey to Mars.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Neoplasias Hematológicas/etiología , Homólogo 1 de la Proteína MutL/deficiencia , Exposición a la Radiación/efectos adversos , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/efectos de la radiación , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Heterocigoto , Humanos , Mutación INDEL , Inmunohistoquímica , Incidencia , Masculino , Ratones , Ratones Noqueados , Inestabilidad de Microsatélites , Polimorfismo de Nucleótido Simple , Medición de Riesgo , Factores de Riesgo , Secuenciación del Exoma
11.
Stem Cells Transl Med ; 7(7): 513-520, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29656536

RESUMEN

One of the major health concerns on long-duration space missions will be radiation exposure to the astronauts. Outside the earth's magnetosphere, astronauts will be exposed to galactic cosmic rays (GCR) and solar particle events that are principally composed of protons and He, Ca, O, Ne, Si, Ca, and Fe nuclei. Protons are by far the most common species, but the higher atomic number particles are thought to be more damaging to biological systems. Evaluation and amelioration of risks from GCR exposure will be important for deep space travel. The hematopoietic system is one of the most radiation-sensitive organ systems, and is highly dependent on functional DNA repair pathways for survival. Recent results from our group have demonstrated an acquired deficiency in mismatch repair (MMR) in human hematopoietic stem cells (HSCs) with age due to functional loss of the MLH1 protein, suggesting an additional risk to astronauts who may have significant numbers of MMR deficient HSCs at the time of space travel. In the present study, we investigated the effects gamma radiation, proton radiation, and 56 Fe radiation on HSC function in Mlh1+/+ and Mlh1-/- marrow from mice in a variety of assays and have determined that while cosmic radiation is a major risk to the hematopoietic system, there is no dependence on MMR capacity. Stem Cells Translational Medicine 2018;7:513-520.


Asunto(s)
Reparación de la Incompatibilidad de ADN/efectos de la radiación , Rayos gamma , Células Madre Hematopoyéticas/metabolismo , Animales , Recuento de Células Sanguíneas , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Proliferación Celular/efectos de la radiación , Femenino , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Homólogo 1 de la Proteína MutL/deficiencia , Homólogo 1 de la Proteína MutL/genética , Dosis de Radiación
12.
Radiat Res ; 188(6): 672-680, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28961076

RESUMEN

Efforts to protect astronauts from harmful galactic cosmic radiation (GCR) require a better understanding of the effects of GCR on human health. In particular, little is known about the lasting effects of GCR on the central nervous system (CNS), which may lead to behavior performance deficits. Previous studies have shown that high-linear energy transfer (LET) radiation in rodents leads to short-term declines in a variety of behavior tests. However, the lasting impact of low-, medium- and high-LET radiation on behavior are not fully defined. Therefore, in this study C57BL/6 male mice were irradiated with 100 or 250 cGy of γ rays (LET ∼0.3 KeV/µm), 10 or 100 cGy of 1H at 1,000 MeV/n (LET ∼0.2 KeV/µm), 28Si at 300 MeV/n (LET ∼69 KeV/µm) or 56Fe at 600 MeV/n (LET of ∼180 KeV/µm), and behavior metrics were collected at 5 and 9 months postirradiation to analyze differences among radiation qualities and doses. A significant dose effect was observed on recognition memory and activity levels measured 9 months postirradiation, regardless of radiation source. In contrast, we observed that each ion species had a distinct effect on anxiety, motor coordination and spatial memory at extended time points. Although 28Si and 56Fe are both regarded as high-LET particles, they were shown to have different detrimental effects on behavior. In summary, our findings suggest that GCR not only affects the CNS in the short term, but also has lasting damaging effects on the CNS that can cause sustained declines in behavior performance.


Asunto(s)
Ansiedad/etiología , Conducta Exploratoria/efectos de la radiación , Rayos gamma/efectos adversos , Transferencia Lineal de Energía , Aprendizaje por Laberinto/efectos de la radiación , Trastornos de la Memoria/etiología , Protones/efectos adversos , Desempeño Psicomotor/efectos de la radiación , Traumatismos Experimentales por Radiación/psicología , Reconocimiento en Psicología/efectos de la radiación , Animales , Hidrógeno , Hierro , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/etiología , Prueba de Desempeño de Rotación con Aceleración Constante , Silicio , Factores de Tiempo
13.
Radiother Oncol ; 121(2): 335-341, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27838149

RESUMEN

BACKGROUND AND PURPOSE: The anti-folate pemetrexed is a radiosensitizer. In pre-clinical models, pemetrexed is more effective along with the base-excision-repair inhibitor methoxyamine. We tested whether methoxyamine enhances pemetrexed-mediated radiosensitization of lung adenocarcinoma cells and xenografts. MATERIALS AND METHODS: A549 and H1299 cells were evaluated for cell cycle distribution by flow cytometry, radiosensitization by clonogenic assay, and DNA repair by neutral comet assay and repair protein activation. H460 cells were included in some studies. Xenografts in nude mice received drug(s) and/or radiation, and tumor growth was monitored by caliper and in vivo toxicity by animal weight. RESULTS: Exposure to pemetrexed/methoxyamine for 24 (H1299, H460) or 48 (A549)hours before irradiation resulted in accumulation of cells near the radiosensitive G1/S border; dose-enhancement factors of 1.62±0.19, 1.97±0.25, and 1.67±0.30, respectively; reduction of mean inactivation dose by 32%, 30%, and 46%, respectively; and significant reductions of SF2 and SF4 (p<0.05). Radiosensitization was associated with rapid DNA double-strand-break rejoining and increased levels of DNA-PKcs. Both tumor-growth rate and tumor-growth delay were significantly improved by adding methoxyamine to pemetrexed pre-irradiation (p<0.0001); no mice lost weight during treatment. CONCLUSIONS: Addition of methoxyamine to pemetrexed and fractionated radiotherapy may improve outcome for patients with locally advanced non-squamous non-small-cell lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Hidroxilaminas/farmacología , Pemetrexed/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/radioterapia , Adenocarcinoma del Pulmón , Animales , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/patología , Carcinoma de Células Grandes/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/metabolismo , Fraccionamiento de la Dosis de Radiación , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Ratones Desnudos , Proteínas Nucleares/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS One ; 8(6): e65113, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762292

RESUMEN

BACKGROUND: Treatment of breast cancer patients with antiestrogens and aromatase inhibitor(s) or Herceptin have shown significant success in steroid receptor positive or Her-2+ breast cancers respectively. However, choice of treatments for breast cancer patients with negative status for estrogen, progesterone receptors and HER2/neu is limited. As a result, search for appropriate therapy regimen for these triple negative breast cancers (TNBC) has become a major focus of investigations for many laboratories. Recently, Deguelin, a natural product isolated from African plant Mundulea sericea (Leguminossae) has shown both antiproliferative actions in various cancers including breast as well as chemoprenventive activity against carcinogen induced experimental cancers. In this report we evaluated efficacy and mechanism of action of Deguelin in triple negative breast cancer cell lines. METHODS/FINDINGS: In vitro, Deguelin in a dose and time dependent manner inhibited the growth of MDA-MB-231, MDA-MB-468, BT-549 and BT-20 cells. Deguelin (2 or 4 mg/kg body weight), when injected intraperitoneally, reduced the in vivo tumor growth of MDA-MB-231 cells transplanted subcutaneously in athymic mice. Moreover it was nontoxic as evident from daily observations on mobility, food and water consumption and comparison of bodyweight and other visceral organ weights with those in control animals at the termination of the study. The western blot analyses and immunostaining studies indicated that the deguelin effects may be mediated through EGFR-PAKT/c-Met p-ERK and NF-κB by down regulating their downstream targets such as p-STAT3, c-Myc, Survivin. CONCLUSION/SIGNIFICANCE: These results suggest that Deguelin may have a significant therapeutic value for the treatment of TNBC patients.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Rotenona/análogos & derivados , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Rotenona/farmacología , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Clin Exp Metastasis ; 30(7): 855-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23645347

RESUMEN

Cancer related deaths in breast cancer patients are due to metastasis of the disease. Murine 4T1 cells (Murine mammary cancer cell line developed from 6-thioguanine resistant tumor) provide an excellent research tool for metastasis related studies because these cells are highly aggressive and readily metastasize to the lungs. In this study we determined the effect of Deguelin on in vivo/vitro growth and metastasis of 4T1 cells. Deguelin inhibited the in vitro growth of 4T1 cells in a time and dose dependent manner accompanied with reduced nuclear PCNA immunostaining. In cells treated with Deguelin, reduced expression of nuclear c-Met, and its downstream targets such p-ERK and p-AKT was observed. Deguelin reduced the cell migration in 4T1 cells as determined by scratch wound assay. Combined treatment with Deguelin + ERK or PI3K/AKT inhibitor had no additional effect on cell migration. These results indicated that the action of Deguelin on cell migration may be mediated by AKT and ERK mediated signaling pathways. In vivo, Deguelin treatment significantly inhibited growth of 4T1 cells. Deguelin also reduced the occurrence of metastatic lung lesions by 33 % when cells were injected intravenously into Balb/c female mice. There was no difference in the body weight, nor was there a difference in liver and spleen weights between vehicle treated-control and Deguelin-treated animals, which indicated that Deguelin was nontoxic at the dose used in the present study. These results provide rationale for developing Deguelin as a chemotherapeutic agent for triple negative breast cancer patients.


Asunto(s)
Metástasis de la Neoplasia/prevención & control , Rotenona/análogos & derivados , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Rotenona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA