Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Cell Mol Med ; 24(2): 1934-1944, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31833658

RESUMEN

Nearly all melanoma patients with a BRAF-activating mutation will develop resistance after an initial clinical benefit from BRAF inhibition (BRAFi). The aim of this work is to evaluate whether metabolic imaging using hyperpolarized (HP) 13 C pyruvate can serve as a metabolic marker of early response to BRAFi in melanoma, by exploiting the metabolic effects of BRAFi. Mice bearing human melanoma xenografts were treated with the BRAFi vemurafenib or vehicle. In vivo HP 13 C magnetic resonance spectroscopy was performed at baseline and 24 hours after treatment to evaluate changes in pyruvate-to-lactate conversion. Oxygen partial pressure was measured via electron paramagnetic resonance oximetry. Ex vivo qRT-PCR, immunohistochemistry and WB analysis were performed on tumour samples collected at the same time-points selected for in vivo experiments. Similar approaches were applied to evaluate the effect of BRAFi on sensitive and resistant melanoma cells in vitro, excluding the role of tumour microenvironment. BRAF inhibition induced a significant increase in the HP pyruvate-to-lactate conversion in vivo, followed by a reduction of hypoxia. Conversely, the conversion was inhibited in vitro, which was consistent with BRAFi-mediated impairment of glycolysis. The paradoxical increase of pyruvate-to-lactate conversion in vivo suggests that such conversion is highly influenced by the tumour microenvironment.


Asunto(s)
Isótopos de Carbono/metabolismo , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Ácido Pirúvico/metabolismo , Vemurafenib/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Espectroscopía de Resonancia por Spin del Electrón , Femenino , Glucólisis/efectos de los fármacos , Glucólisis/genética , Humanos , Melanoma/patología , Ratones Desnudos , Oximetría , Consumo de Oxígeno/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transcripción Genética/efectos de los fármacos
2.
J Cell Mol Med ; 24(24): 14195-14204, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33107196

RESUMEN

Acetate is reported as a regulator of fat mass but also as lipogenic source for cancer cells. Breast cancer is surrounded by adipose tissue and has been associated with obesity. However, whether acetate contributes to cancer cell metabolism as lipogenic substrate and/or by changing fat storage and eventually obesity-induced breast cancer progression remains unknown. Therefore, we studied the contribution of acetate to breast cancer metabolism and progression. In vitro, we found that acetate is not a bioenergetic substrate under normoxia and did not result in a significant change of growth. However, by using lipidomic approaches, we discovered that acetate changes the lipid profiles of the cells under hypoxia. Moreover, while mice fed a high-fat diet (HFD) developed bigger tumours than their lean counterparts, exogenous acetate supplementation leads to a complete abolishment of fat mass gain without reverting the HFD-induced obesity-driven tumour progression. In conclusion, although acetate protects against diet-induced obesity, our data suggest that it is not affecting HFD-driven tumour progression.


Asunto(s)
Acetatos/metabolismo , Acetatos/farmacología , Neoplasias de la Mama/metabolismo , Obesidad/metabolismo , Adipogénesis , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Lipidómica/métodos , Ratones , Oxígeno/metabolismo , Carga Tumoral/efectos de los fármacos
3.
J Cell Mol Med ; 24(17): 10233-10244, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32681609

RESUMEN

Epidemiological studies have shown that obese subjects have an increased risk of developing triple-negative breast cancer (TNBC) and an overall reduced survival. However, the relation between obesity and TNBC remains difficult to understand. We hypothesize that apelin, an adipokine whose levels are increased in obesity, could be a major factor contributing to both tumour growth and metastatization in TNBC obese patients. We observed that development of obesity under high-fat diet in TNBC tumour-bearing mice significantly increased tumour growth. By showing no effect of high-fat diet in obesity-resistant mice, we demonstrated the necessity to develop obesity-related disorders to increase tumour growth. Apelin mRNA expression was also increased in the subcutaneous adipose tissue and tumours of obese mice. We further highlighted that the reproduction of obesity-related levels of apelin in lean mice led to an increased TNBC growth and brain metastases formation. Finally, injections of the apelinergic antagonist F13A to obese mice significantly reduced TNBC growth, suggesting that apelinergic system interference could be an interesting therapeutic strategy in the context of obesity and TNBC.


Asunto(s)
Apelina/metabolismo , Obesidad/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/fisiología , Dieta Alta en Grasa/efectos adversos , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/patología , Obesidad/patología , ARN Mensajero/metabolismo , Grasa Subcutánea/metabolismo , Neoplasias de la Mama Triple Negativas/patología
4.
Sci Rep ; 11(1): 9922, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972642

RESUMEN

Obesity is a known factor increasing the risk of developing breast cancer and reducing disease free survival. In addition to these well-documented effects, recent studies have shown that obesity is also affecting response to chemotherapy. Among the multiple dysregulations associated with obesity, increased level of the apelin adipokine has been recently shown to be directly involved in the association between obesity and increased breast cancer progression. In this study, we analyzed in a retrospective cohort of 62 breast cancer patients the impact of obesity and tumoral apelin expression on response to neoadjuvant chemotherapy. In the multivariate logistic regression, obesity and high tumoral apelin expression were associated with a reduced response to NAC in our cohort. However, obesity and high tumoral apelin expression were not correlated, suggesting that those two parameters could be independently associated with reduced NAC response. These findings should be confirmed in independent cohorts.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apelina/metabolismo , Neoplasias de la Mama/terapia , Terapia Neoadyuvante/estadística & datos numéricos , Obesidad/epidemiología , Adulto , Anciano , Apelina/análisis , Índice de Masa Corporal , Mama/patología , Mama/cirugía , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Quimioterapia Adyuvante/estadística & datos numéricos , Femenino , Humanos , Mastectomía , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Obesidad/complicaciones , Estudios Retrospectivos , Resultado del Tratamiento
5.
Clin Cancer Res ; 26(8): 1932-1943, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31831557

RESUMEN

PURPOSE: Optimal head and neck squamous cell carcinoma (HNSCC) patient selection for anti-EGFR-based therapy remains an unmet need since only a minority of patients derive long-term benefit from cetuximab treatment. We assessed the ability of state-of-the-art noninvasive in vivo metabolic imaging to probe metabolic shift in cetuximab-sensitive and -resistant HNSCC patient-derived tumor xenografts (PDTXs). EXPERIMENTAL DESIGN: Three models selected based on their known sensitivity to cetuximab in patients (cetuximab-sensitive or acquired-resistant HNC007 PDTXs, cetuximab-naïve UCLHN4 PDTXs, and cetuximab-resistant HNC010 PDTXs) were inoculated in athymic nude mice. RESULTS: Cetuximab induced tumor size stabilization in mice for 4 weeks in cetuximab-sensitive and -naïve models treated with weekly injections (30 mg/kg) of cetuximab. Hyperpolarized 13C-pyruvate-13C-lactate exchange was significantly decreased in vivo in cetuximab-sensitive xenograft models 8 days after treatment initiation, whereas it was not modified in cetuximab-resistant xenografts. Ex vivo analysis of sensitive tumors resected at day 8 after treatment highlighted specific metabolic changes, likely to participate in the decrease in the lactate to pyruvate ratio in vivo. Diffusion MRI showed a decrease in tumor cellularity in the HNC007-sensitive tumors, but failed to show sensitivity to cetuximab in the UCLHN4 model. CONCLUSIONS: This study constitutes the first in vivo demonstration of cetuximab-induced metabolic changes in cetuximab-sensitive HNSCC PDTXs that were not present in resistant tumors. Using metabolic imaging, we were able to identify hyperpolarized 13C-pyruvate as a potential marker for response and resistance to the EGFR inhibitor in HNSCC.


Asunto(s)
Isótopos de Carbono/análisis , Carcinoma de Células Escamosas/patología , Cetuximab/farmacología , Resistencia a Antineoplásicos , Neoplasias de Cabeza y Cuello/patología , Lactatos/metabolismo , Piruvatos/metabolismo , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/antagonistas & inhibidores , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Free Radic Res ; 52(2): 256-266, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29320894

RESUMEN

The ability of certain cancer cells to maintain a highly reduced intracellular environment is correlated with aggressiveness and drug resistance. Since the glutathione (GSH) and thioredoxin (TRX) systems cooperate to a tight regulation of ROS in cell physiology, and to a stimulation of tumour initiation and progression, modulation of the GSH and TRX pathways are emerging as new potential targets in cancer. In vivo methods to assess changes in tumour redox status are critically needed to assess the relevance of redox-targeted agents. The current study assesses in vitro and in vivo biomarkers of tumour redox status in response to treatments targeting the GSH and TRX pathways, by comparing cytosolic and mitochondrial redox nitroxide electron paramagnetic resonance (EPR) probes, and cross-validation with redox dynamic fluorescent measurement. For that purpose, the effect of the GSH modulator buthionine sulfoximine (BSO) and of the TRX reductase inhibitor auranofin were measured in vitro using both cytosolic and mitochondrial EPR and roGFP probes in breast and cervical cancer cells. In vivo, mice bearing breast or cervical cancer xenografts were treated with the GSH or TRX modulators and monitored using the mito-TEMPO spin probe. Our data highlight the importance of using mitochondria-targeted spin probes to assess changes in tumour redox status induced by redox modulators. Further in vivo validation of the mito-tempo spin probe with alternative in vivo methods should be considered, yet the spin probe used in vivo in xenografts demonstrated sensitivity to the redox status modulators.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glutatión/efectos de los fármacos , Estrés Oxidativo , Tiorredoxinas/antagonistas & inhibidores , Neoplasias del Cuello Uterino/metabolismo , Animales , Biomarcadores/análisis , Línea Celular Tumoral , Femenino , Humanos , Ratones , Oxidación-Reducción , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Curr Pathobiol Rep ; 5(4): 315-322, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29188139

RESUMEN

PURPOSE OF REVIEW: Obesity is strongly associated with the development of several types of cancers. This review aims to discuss the recent key mechanisms and actors underlying the link between adipose tissue metabolism and cancer, and the unequivocal common mechanisms connecting gut microbes to adipose tissue and eventually cancer development. RECENT FINDINGS: Complex interactions among systemic and tissue-specific pathways are suggested to link obesity and cancer, involving endocrine hormones, adipokines, fatty acids, inflammation, metabolic alterations, and hypoxia. Emerging evidence also suggests that the gut microbiota, another key environmental factor, may be considered as a converging element. Studies have shown that cancer susceptibility may be induced in germ-free mice colonized with the gut microbiota from high-fat diet-fed mice. Suggested mechanisms may involve inflammation, immunity changes, lipogenic substrates, and adipogenesis. SUMMARY: Cancer development is a complex process that may be under the control of previously unthought factors such as the gut microbiota. Whether specific intervention targeting the gut microbiota may reduce adipose tissue-driven cancer is an interesting strategy that remains to be proven.

8.
Sci Rep ; 7(1): 370, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28337033

RESUMEN

The amyloid precursor protein (APP) modulates synaptic activity, resulting from the fine tuning of excitatory and inhibitory neurotransmission. GABAergic inhibitory neurotransmission is affected by modifications in intracellular chloride concentrations regulated by Na+-K+-2Cl- cotransporter 1 (NKCC1) and neuronal K+-Cl- cotransporter 2 (KCC2), allowing entrance and efflux of chloride, respectively. Modifications in NKCC1 and KCC2 expression during maturation of cortical cells induce a shift in GABAergic signaling. Here, we demonstrated that APP affects this GABA shift. Expression of APP in cortical cells decreased the expression of KCC2, without modifying NKCC1, eliciting a less inhibitory GABA response. Downregulation of KCC2 expression by APP was independent of the APP intracellular domain, but correlated with decreased expression of upstream stimulating factor 1 (USF1), a potent regulator of Slc12a5 gene expression (encoding KCC2). KCC2 was also downregulated in vivo following APP expression in neonatal mouse brain. These results argue for a key role of APP in the regulation of GABAergic neurotransmission.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Corteza Cerebral/fisiología , Neuronas GABAérgicas/fisiología , Transmisión Sináptica , Ácido gamma-Aminobutírico/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Señalización del Calcio , Corteza Cerebral/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Ratas Wistar , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Simportadores/metabolismo , Cotransportadores de K Cl
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA