Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 23(4): e53746, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35199910

RESUMEN

Cachexia is a wasting syndrome characterized by devastating skeletal muscle atrophy that dramatically increases mortality in various diseases, most notably in cancer patients with a penetrance of up to 80%. Knowledge regarding the mechanism of cancer-induced cachexia remains very scarce, making cachexia an unmet medical need. In this study, we discovered strong alterations of iron metabolism in the skeletal muscle of both cancer patients and tumor-bearing mice, characterized by decreased iron availability in mitochondria. We found that modulation of iron levels directly influences myotube size in vitro and muscle mass in otherwise healthy mice. Furthermore, iron supplementation was sufficient to preserve both muscle function and mass, prolong survival in tumor-bearing mice, and even rescues strength in human subjects within an unexpectedly short time frame. Importantly, iron supplementation refuels mitochondrial oxidative metabolism and energy production. Overall, our findings provide new mechanistic insights in cancer-induced skeletal muscle wasting, and support targeting iron metabolism as a potential therapeutic option for muscle wasting diseases.


Asunto(s)
Caquexia , Neoplasias , Animales , Caquexia/etiología , Caquexia/metabolismo , Suplementos Dietéticos , Humanos , Hierro/metabolismo , Ratones , Músculo Esquelético/metabolismo , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
2.
Ann Surg ; 278(6): e1313-e1326, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37450698

RESUMEN

OBJECTIVES: To test whether mitochondrial transplantation (MITO) mitigates damage in 2 models of acute kidney injury (AKI). BACKGROUND: MITO is a process where exogenous isolated mitochondria are taken up by cells. As virtually any morbid clinical condition is characterized by mitochondrial distress, MITO may find a role as a treatment modality in numerous clinical scenarios including AKI. METHODS: For the in vitro experiments, human proximal tubular cells were damaged and then treated with mitochondria or placebo. For the ex vivo experiments, we developed a non-survival ex vivo porcine model mimicking the donation after cardiac death renal transplantation scenario. One kidney was treated with mitochondria, although the mate organ received placebo, before being perfused at room temperature for 24 hours. Perfusate samples were collected at different time points and analyzed with Raman spectroscopy. Biopsies taken at baseline and 24 hours were analyzed with standard pathology, immunohistochemistry, and RNA sequencing analysis. RESULTS: In vitro, cells treated with MITO showed higher proliferative capacity and adenosine 5'-triphosphate production, preservation of physiological polarization of the organelles and lower toxicity and reactive oxygen species production. Ex vivo, kidneys treated with MITO shed fewer molecular species, indicating stability. In these kidneys, pathology showed less damage whereas RNAseq analysis showed modulation of genes and pathways most consistent with mitochondrial biogenesis and energy metabolism and downregulation of genes involved in neutrophil recruitment, including IL1A, CXCL8, and PIK3R1. CONCLUSIONS: MITO mitigates AKI both in vitro and ex vivo.


Asunto(s)
Lesión Renal Aguda , Trasplante de Riñón , Daño por Reperfusión , Humanos , Porcinos , Animales , Riñón/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo
3.
Br J Cancer ; 124(1): 207-216, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33257841

RESUMEN

BACKGROUND: Tumour acidosis is considered to play a central role in promoting cancer invasion and migration, but few studies have investigated in vivo how tumour pH correlates with cancer invasion. This study aims to determine in vivo whether tumour acidity is associated with cancer metastatic potential. METHODS: Breast cancer cell lines with different metastatic potentials have been characterised for several markers of aggressiveness and invasiveness. Murine tumour models have been developed and assessed for lung metastases and tumour acidosis has been assessed in vivo by a magnetic resonance imaging-based chemical exchange saturation transfer (CEST) pH imaging approach. RESULTS: The higher metastatic potential of 4T1 and TS/A primary tumours, in comparison to the less aggressive TUBO and BALB-neuT ones, was confirmed by the highest expression of cancer cell stem markers (CD44+CD24-), highlighting their propensity to migrate and invade, coinciding with the measurement obtained by in vitro assays. MRI-CEST pH imaging successfully discriminated the more aggressive 4T1 and TS/A tumours that displayed a more acidic pH. Moreover, the observed higher tumour acidity was significantly correlated with an increased number of lung metastases. CONCLUSIONS: The findings of this study indicate that the extracellular acidification is associated with the metastatic potential.


Asunto(s)
Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Invasividad Neoplásica/patología , Animales , Línea Celular Tumoral , Femenino , Concentración de Iones de Hidrógeno , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos BALB C
4.
Cancer Metastasis Rev ; 38(1-2): 189-203, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30820778

RESUMEN

In cancer, mitochondrial functions are commonly altered. Directly involved in metabolic reprogramming, mitochondrial plasticity confers to cancer cells a high degree of adaptability to a wide range of stresses and to the harsh tumor microenvironment. Lack of nutrients or oxygen caused by altered perfusion, metabolic needs of proliferating cells, co-option of the microenvironment, control of the immune system, cell migration and metastasis, and evasion of exogenous stress (e.g., chemotherapy) are all, at least in part, influenced by mitochondria. Mitochondria are undoubtedly one of the key contributors to cancer development and progression. Understanding their protumoral (dys)functions may pave the way to therapeutic strategies capable of turning them into innocent entities. Here, we will focus on the production and detoxification of mitochondrial reactive oxygen species (mtROS), on their impact on tumorigenesis (genetic, prosurvival, and microenvironmental effects and their involvement in autophagy), and on tumor metastasis. We will also summarize the latest therapeutic approaches involving mtROS.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Humanos , Mitocondrias/patología , Neoplasias/patología , Fosforilación Oxidativa
5.
Int J Mol Sci ; 21(22)2020 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-33202621

RESUMEN

Cachexia is a complication of dismal prognosis, which often represents the last step of several chronic diseases. For this reason, the comprehension of the molecular drivers of such a condition is crucial for the development of management approaches. Importantly, cachexia is a syndrome affecting various organs, which often results in systemic complications. To date, the majority of the research on cachexia has been focused on skeletal muscle, muscle atrophy being a pivotal cause of weight loss and the major feature associated with the steep reduction in quality of life. Nevertheless, defining the impact of cachexia on other organs is essential to properly comprehend the complexity of such a condition and potentially develop novel therapeutic approaches.


Asunto(s)
Caquexia , Músculo Esquelético , Atrofia Muscular , Calidad de Vida , Caquexia/metabolismo , Caquexia/patología , Caquexia/terapia , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Atrofia Muscular/terapia
6.
Circulation ; 138(7): 696-711, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-29348263

RESUMEN

BACKGROUND: Anthracyclines, such as doxorubicin (DOX), are potent anticancer agents for the treatment of solid tumors and hematologic malignancies. However, their clinical use is hampered by cardiotoxicity. This study sought to investigate the role of phosphoinositide 3-kinase γ (PI3Kγ) in DOX-induced cardiotoxicity and the potential cardioprotective and anticancer effects of PI3Kγ inhibition. METHODS: Mice expressing a kinase-inactive PI3Kγ or receiving PI3Kγ-selective inhibitors were subjected to chronic DOX treatment. Cardiac function was analyzed by echocardiography, and DOX-mediated signaling was assessed in whole hearts or isolated cardiomyocytes. The dual cardioprotective and antitumor action of PI3Kγ inhibition was assessed in mouse mammary tumor models. RESULTS: PI3Kγ kinase-dead mice showed preserved cardiac function after chronic low-dose DOX treatment and were protected against DOX-induced cardiotoxicity. The beneficial effects of PI3Kγ inhibition were causally linked to enhanced autophagic disposal of DOX-damaged mitochondria. Consistently, either pharmacological or genetic blockade of autophagy in vivo abrogated the resistance of PI3Kγ kinase-dead mice to DOX cardiotoxicity. Mechanistically, PI3Kγ was triggered in DOX-treated hearts, downstream of Toll-like receptor 9, by the mitochondrial DNA released by injured organelles and contained in autolysosomes. This autolysosomal PI3Kγ/Akt/mTOR/Ulk1 signaling provided maladaptive feedback inhibition of autophagy. PI3Kγ blockade in models of mammary gland tumors prevented DOX-induced cardiac dysfunction and concomitantly synergized with the antitumor action of DOX by unleashing anticancer immunity. CONCLUSIONS: Blockade of PI3Kγ may provide a dual therapeutic advantage in cancer therapy by simultaneously preventing anthracyclines cardiotoxicity and reducing tumor growth.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Cardiopatías/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Quinoxalinas/farmacología , Tiazolidinedionas/farmacología , Carga Tumoral/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/toxicidad , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cardiotoxicidad , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Femenino , Genes erbB-2 , Cardiopatías/inducido químicamente , Cardiopatías/enzimología , Cardiopatías/patología , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutación , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
7.
Biochim Biophys Acta Bioenerg ; 1858(8): 556-572, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28167100

RESUMEN

Altered metabolism in cancer cells is pivotal for tumor growth, most notably by providing energy, reducing equivalents and building blocks while several metabolites exert a signaling function promoting tumor growth and progression. A cancer tissue cannot be simply reduced to a bulk of proliferating cells. Tumors are indeed complex and dynamic structures where single cells can heterogeneously perform various biological activities with different metabolic requirements. Because tumors are composed of different types of cells with metabolic activities affected by different spatial and temporal contexts, it is important to address metabolism taking into account cellular and biological heterogeneity. In this review, we describe this heterogeneity also in metabolic fluxes, thus showing the relative contribution of different metabolic activities to tumor progression according to the cellular context. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Asunto(s)
Metabolismo Energético , Neoplasias/metabolismo , Animales , Muerte Celular , División Celular , Glucólisis , Humanos , Redes y Vías Metabólicas , Mitocondrias/metabolismo , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Células del Estroma/metabolismo
8.
Br J Cancer ; 117(9): 1336-1340, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28873082

RESUMEN

BACKGROUND: Activation of free fatty acid receptor 2 (FFAR2) by microbiota-derived metabolites (e.g., propionate) reduces leukaemic cell proliferation in vitro. This study aims to test whether Ffar2 expression per se also influences leukaemia cell growth in vivo. METHODS: Bcr-Abl-expressing BaF cells were used as a leukaemia model and the role of Ffar2 was evaluated in Balb/c mice after lentiviral shRNA transduction. RESULTS: Our data formally establish that reduced leukaemic cell proliferation is associated with increased Ffar2 expression in vivo and in vitro. Going beyond association, we point out that decreasing Ffar2 expression fosters cancer cell growth in vitro and in vivo. CONCLUSIONS: Our data demonstrate the role of Ffar2 in the control of leukaemic cell proliferation in vivo and indicate that a modulation of Ffar2 expression through nutritional tools or pharmacological agents may constitute an attractive therapeutic approach to tackle leukaemia progression in humans.


Asunto(s)
Proliferación Celular , Leucemia Experimental/patología , Receptores Acoplados a Proteínas G/fisiología , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Femenino , Leucemia Experimental/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3254-3264, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28866133

RESUMEN

Recent progress in dissecting the molecular paracrine circuits of cancer and stromal cells in bone metastases (BM) are offering new options to improve current merely palliative approach. The study of tumor-stroma metabolic interplay may further ameliorate this scenario. In this context, we demonstrated that highly glycolytic MDA-MB-231 cancer cells, that form osteolytic BM in vivo, release a large amount of lactate at a significantly higher level than MCF7 cells. Thus, we speculated that lactate released from carcinoma cells is uptaken and metabolically used by osteoclasts, the key players of osteolysis associated with BM. First, we demonstrated that the release of lactate at the bone site is mediated by monocarboxylate transporter 4 (MCT4), as revealed by immunostaining and MCT4 localization at the plasma membrane of tumor cells in mouse model of BM and in human tissue sections of BM. Then, we showed that in vitro lactate is uptaken by osteoclasts to be used as a fuel for the oxidative metabolism of osteoclasts, ultimately enhancing Type I collagen resorption. The passive transport of lactate into osteoclasts was mediated by MCT1: MCT1 expression is significantly upregulated during osteoclast differentiation and Type I collagen resorption is significantly impaired when osteoclasts are treated with 7-(N-benzyl-N-methylamino)-2-oxo-2H-chromene-3-carboxylic acid, an MCT-1 inhibitor. Together, these data demonstrate that lactate released by glycolytic breast carcinoma cells in the bone microenvironment promotes the formation of osteolytic lesions, and provide the rationale for further studies on the use of MCT1 targeting as a novel therapeutic approach in advanced cancer patients with BM.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Osteoclastos/metabolismo , Animales , Línea Celular Tumoral/metabolismo , Cumarinas/antagonistas & inhibidores , Femenino , Glucosa/metabolismo , Glucólisis , Humanos , Lactatos/metabolismo , Células MCF-7 , Ratones , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Osteoclastos/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Simportadores/metabolismo
10.
Cell Mol Life Sci ; 73(7): 1333-48, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26626411

RESUMEN

Metabolic adaptations are intimately associated with changes in cell behavior. Cancers are characterized by a high metabolic plasticity resulting from mutations and the selection of metabolic phenotypes conferring growth and invasive advantages. While metabolic plasticity allows cancer cells to cope with various microenvironmental situations that can be encountered in a primary tumor, there is increasing evidence that metabolism is also a major driver of cancer metastasis. Rather than a general switch promoting metastasis as a whole, a succession of metabolic adaptations is more likely needed to promote different steps of the metastatic process. This review addresses the contribution of pH, glycolysis and the pentose phosphate pathway, and a companion paper summarizes current knowledge regarding the contribution of mitochondria, lipids and amino acid metabolism. Extracellular acidification, intracellular alkalinization, the glycolytic enzyme phosphoglucose isomerase acting as an autocrine cytokine, lactate and the pentose phosphate pathway are emerging as important factors controlling cancer metastasis.


Asunto(s)
Neoplasias/patología , Vía de Pentosa Fosfato/fisiología , Transición Epitelial-Mesenquimal , Glucosa-6-Fosfato Isomerasa/metabolismo , Glucólisis , Humanos , Ácido Láctico/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo
11.
Cell Mol Life Sci ; 73(7): 1349-63, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26646069

RESUMEN

Metabolic alterations are a hallmark of cancer controlling tumor progression and metastasis. Among the various metabolic phenotypes encountered in tumors, this review focuses on the contributions of mitochondria, lipid and amino acid metabolism to the metastatic process. Tumor cells require functional mitochondria to grow, proliferate and metastasize, but shifts in mitochondrial activities confer pro-metastatic traits encompassing increased production of mitochondrial reactive oxygen species (mtROS), enhanced resistance to apoptosis and the increased or de novo production of metabolic intermediates of the TCA cycle behaving as oncometabolites, including succinate, fumarate, and D-2-hydroxyglutarate that control energy production, biosynthesis and the redox state. Lipid metabolism and the metabolism of amino acids, such as glutamine, glutamate and proline are also currently emerging as focal control points of cancer metastasis.


Asunto(s)
Aminoácidos/metabolismo , Metabolismo de los Lípidos/fisiología , Mitocondrias/metabolismo , Neoplasias/patología , Ciclo del Ácido Cítrico , Humanos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo
12.
Int J Cancer ; 138(8): 2043-9, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26595604

RESUMEN

The cholinic phenotype, characterized by elevated phosphocholine and a high production of total-choline (tCho)-containing metabolites, is a metabolic hallmark of cancer. It can be exploited for targeted therapy. Non-invasive imaging biomarkers are required to evaluate an individual's response to targeted anticancer agents that usually do not rapidly cause tumor shrinkage. Because metabolic changes can manifest at earlier stages of therapy than changes in tumor size, the aim of the current study was to evaluate (1)H-MRS and diffusion-weighted MRI (DW-MRI) as markers of tumor response to the modulation of the choline pathway in mammary tumor xenografts. Inhibition of choline kinase activity was achieved with the direct pharmacological inhibitor H-89, indirect inhibitor sorafenib and down-regulation of choline-kinase α (ChKA) expression using specific short-hairpin RNA (shRNA). While all three strategies significantly decreased tCho tumor content in vivo, only sorafenib and anti-ChKA shRNA significantly repressed tumor growth. The increase of apparent-diffusion-coefficient of water (ADCw) measured by DW-MRI, was predictive of the induced necrosis and inhibition of the tumor growth in sorafenib treated mice, while the absence of change in ADC values in H89 treated mice predicted the absence of effect in terms of tumor necrosis and tumor growth. In conclusion, (1)H-choline spectroscopy can be useful as a pharmacodynamic biomarker for choline targeted agents, while DW-MRI can be used as an early marker of effective tumor response to choline targeted therapies. DW-MRI combined to choline spectroscopy may provide a useful non-invasive marker for the early clinical assessment of tumor response to therapies targeting choline signaling.


Asunto(s)
Colina Quinasa/antagonistas & inhibidores , Imagen de Difusión por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Neoplasias Mamarias Experimentales/patología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/farmacología , Femenino , Xenoinjertos , Humanos , Isoquinolinas/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Protones , Sorafenib , Sulfonamidas/farmacología
13.
Adv Exp Med Biol ; 876: 209-214, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26782214

RESUMEN

The aim of the study was to assess the link between the metabolic profile and the proliferation capacity of a range of human and murine cancer cell lines. First, the combination of mitochondrial respiration and glycolytic efficiency measurements allowed the determination of different metabolic profiles among the cell lines, ranging from a mostly oxidative to a mostly glycolytic phenotype. Second, the study revealed that cell proliferation, evaluated by DNA synthesis measurements, was statistically correlated to glycolytic efficiency. This indicated that glycolysis is the key energetic pathway linked to cell proliferation rate. Third, to validate this hypothesis and exclude non-metabolic factors, mitochondria-depleted were compared to wild-type cancer cells, and the data showed that enhanced glycolysis observed in mitochondria-depleted cells is also associated with an increase in proliferation capacity.


Asunto(s)
Metabolismo Energético , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Glucólisis , Humanos , Ratones , Mitocondrias/metabolismo , Neoplasias/patología , Consumo de Oxígeno
14.
NMR Biomed ; 28(3): 367-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25611487

RESUMEN

Cell tracking could be useful to elucidate fundamental processes of cancer biology such as metastasis. The aim of this study was to visualize, using MRI, and to quantify, using electron paramagnetic resonance (EPR), the entrapment of murine breast cancer cells labeled with superparamagnetic iron oxide particles (SPIOs) in the mouse brain after intracardiac injection. For this purpose, luciferase-expressing murine 4 T1-luc breast cancer cells were labeled with fluorescent Molday ION Rhodamine B SPIOs. Following intracardiac injection, SPIO-labeled 4 T1-luc cells were imaged using multiple gradient-echo sequences. Ex vivo iron oxide quantification in the mouse brain was performed using EPR (9 GHz). The long-term fate of 4 T1-luc cells after injection was characterized using bioluminescence imaging (BLI), brain MRI and immunofluorescence. We observed hypointense spots due to SPIO-labeled cells in the mouse brain 4 h after injection on T2 *-weighted images. Histology studies showed that SPIO-labeled cancer cells were localized within blood vessels shortly after delivery. Ex vivo quantification of SPIOs showed that less than 1% of the injected cells were taken up by the mouse brain after injection. MRI experiments did not reveal the development of macrometastases in the mouse brain several days after injection, but immunofluorescence studies demonstrated that these cells found in the brain established micrometastases. Concerning the metastatic patterns of 4 T1-luc cells, an EPR biodistribution study demonstrated that SPIO-labeled 4 T1-luc cells were also entrapped in the lungs of mice after intracardiac injection. BLI performed 6 days after injection of 4 T1-luc cells showed that this cell line formed macrometastases in the lungs and in the bones. Conclusively, EPR and MRI were found to be complementary for cell tracking applications. MRI cell tracking at 11.7 T allowed sensitive detection of isolated SPIO-labeled cells in the mouse brain, whereas EPR allowed the assessment of the number of SPIO-labeled cells in organs shortly after injection.


Asunto(s)
Encéfalo/patología , Rastreo Celular/métodos , Espectroscopía de Resonancia por Spin del Electrón/métodos , Imagen por Resonancia Magnética/métodos , Neoplasias Mamarias Animales/patología , Animales , Línea Celular Tumoral , Dextranos/metabolismo , Femenino , Inyecciones , Mediciones Luminiscentes , Pulmón/metabolismo , Nanopartículas de Magnetita , Ratones Endogámicos BALB C , Miocardio/metabolismo , Especificidad de Órganos , Rodaminas/metabolismo , Coloración y Etiquetado , Factores de Tiempo , Distribución Tisular
15.
Acta Oncol ; 54(2): 266-74, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25007226

RESUMEN

BACKGROUND: The Warburg phenotype identified decades ago describes tumor cells with increased glycolysis and decreased mitochondrial respiration even in the presence of oxygen. This particular metabolism also termed 'aerobic glycolysis' reflects an adaptation of tumor cells to proliferation in a heterogeneous tumor microenvironment. Although metabolic alterations in cancer cells are common features, their impact on the response to radiotherapy is not yet fully elucidated. This study investigated the impact of cellular oxygen consumption inhibition on the tumor response to radiotherapy. MATERIAL AND METHODS: Warburg-phenotype tumor cells with impaired mitochondrial respiration (MD) were produced and compared in respect to their metabolism to the genetically matched parental cells (WT). After characterization of their metabolism we compared the response of MD cells to irradiation in vivo and in vitro to the genetically matched parental cells (WT). RESULTS: We first confirmed that MD cells were exclusively glycolytic while WT cells exhibited mitochondrial respiration. We then used these cells for assessing the response of WT and MD tumors to a single dose of radiation and showed that the in vivo tumor growth delay of the MD group was increased, indicating an increased radiosensitivity compared to WT while the in vitro ability of both cell lines to repair radiation-induced DNA damage was similar. CONCLUSION: Taken together, these results indicate that in addition to intrinsic radiosensitivity parameters the tumor response to radiation will also depend on their metabolic rate of oxygen consumption.


Asunto(s)
Hipoxia de la Célula/fisiología , Glucólisis/fisiología , Mitocondrias/fisiología , Neoplasias/metabolismo , Neoplasias/radioterapia , Consumo de Oxígeno/fisiología , Tolerancia a Radiación/fisiología , Aerobiosis , Animales , Línea Celular Tumoral , Supervivencia Celular , Reparación del ADN , Femenino , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Ratones Desnudos , Neoplasias/patología , Fenotipo
16.
Mol Ther ; 22(4): 734-42, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24394186

RESUMEN

Host defense peptides, in particular LL-37, are emerging as potential therapeutics for promoting wound healing and inhibiting bacterial growth. However, effective delivery of the LL-37 peptide remains limiting. We hypothesized that skin-targeted electroporation of a plasmid encoding hCAP-18/LL-37 would promote the healing of wounds. The plasmid was efficiently delivered to full-thickness skin wounds by electroporation and it induced expression of LL-37 in the epithelium. It significantly accelerated reepithelialization of nondiabetic and diabetic wounds and caused a significant VEGFa and interleukin (IL)-6 induction. IL-6 was involved in LL-37-mediated keratinocyte migration in vitro and IL-6 neutralizing antibodies delivered to mice were able to suppress the wound healing activity of the hCAP-18/LL-37 plasmid. In a hindlimb ischemia model, electroporation of the hCAP-18/LL-37 plasmid increased blood perfusion, reduced muscular atrophy, and upregulated the angiogenic chemokines VEGFa and SDF-1a, and their receptors VEGF-R and CXCR-4. These findings demonstrate that a localized gene therapy with LL-37 is a promising approach for the treatment of wounds.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Electroquimioterapia , Terapia Genética , Cicatrización de Heridas/genética , Animales , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Células Cultivadas , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Plásmidos/administración & dosificación , Plásmidos/genética , Catelicidinas
17.
J Immunol ; 187(11): 5941-51, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22048771

RESUMEN

Diacylglycerol kinases (DGKs) metabolize diacylglycerol to phosphatidic acid. In T lymphocytes, DGKα acts as a negative regulator of TCR signaling by decreasing diacylglycerol levels and inducing anergy. In this study, we show that upon costimulation of the TCR with CD28 or signaling lymphocyte activation molecule (SLAM), DGKα, but not DGKζ, exits from the nucleus and undergoes rapid negative regulation of its enzymatic activity. Inhibition of DGKα is dependent on the expression of SAP, an adaptor protein mutated in X-linked lymphoproliferative disease, which is essential for SLAM-mediated signaling and contributes to TCR/CD28-induced signaling and T cell activation. Accordingly, overexpression of SAP is sufficient to inhibit DGKα, whereas SAP mutants unable to bind either phospho-tyrosine residues or SH3 domain are ineffective. Moreover, phospholipase C activity and calcium, but not Src-family tyrosine kinases, are also required for negative regulation of DGKα. Finally, inhibition of DGKα in SAP-deficient cells partially rescues defective TCR/CD28 signaling, including Ras and ERK1/2 activation, protein kinase C membrane recruitment, induction of NF-AT transcriptional activity, and IL-2 production. Thus SAP-mediated inhibition of DGKα sustains diacylglycerol signaling, thereby regulating T cell activation, and it may represent a novel pharmacological strategy for X-linked lymphoproliferative disease treatment.


Asunto(s)
Diacilglicerol Quinasa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Western Blotting , Diglicéridos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/inmunología , Células Jurkat , Transporte de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transfección
18.
Proc Natl Acad Sci U S A ; 107(9): 4182-7, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160093

RESUMEN

Diacylglycerol kinases (DGKs) convert diacylglycerol (DAG) into phosphatidic acid (PA), acting as molecular switches between DAG- and PA-mediated signaling. We previously showed that Src-dependent activation and plasma membrane recruitment of DGKalpha are required for growth-factor-induced cell migration and ruffling, through the control of Rac small-GTPase activation and plasma membrane localization. Herein we unveil a signaling pathway through which DGKalpha coordinates the localization of Rac. We show that upon hepatocyte growth-factor stimulation, DGKalpha, by producing PA, provides a key signal to recruit atypical PKCzeta/iota (aPKCzeta/iota) in complex with RhoGDI and Rac at ruffling sites of colony-growing epithelial cells. Then, DGKalpha-dependent activation of aPKCzeta/iota mediates the release of Rac from the inhibitory complex with RhoGDI, allowing its activation and leading to formation of membrane ruffles, which constitute essential requirements for cell migration. These findings highlight DGKalpha as the central element of a lipid signaling pathway linking tyrosine kinase growth-factor receptors to regulation of aPKCs and RhoGDI, and providing a positional signal regulating Rac association to the plasma membrane.


Asunto(s)
Diacilglicerol Quinasa/metabolismo , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Factor de Crecimiento de Hepatocito/fisiología , Proteína Quinasa C/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Línea Celular , Membrana Celular/fisiología , Perros , Técnica del Anticuerpo Fluorescente , Fosforilación , Transducción de Señal , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
19.
Proc Natl Acad Sci U S A ; 107(47): 20477-82, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059928

RESUMEN

Hyperthermia (HT) is a strong adjuvant treatment with radiotherapy and chemotherapy because it causes tumor reoxygenation. However, the detailed molecular mechanisms of how HT enhances tumor oxygenation have not been elucidated. Here we report that 1 h of HT activates hypoxia-inducible factor-1 (HIF-1) in tumors and its downstream targets, vascular endothelial growth factor (VEGF) and pyruvate dehydrogenase kinase 1 (PDK1). Consistent with HIF-1 activation and up-regulation of its downstream genes, HT also enhances tumor perfusion/vascularization and decreases oxygen consumption. As a result, tumor hypoxia is reduced after HT, suggesting that these physiological changes contribute to HT-induced tumor reoxygenation. Because HIF-1 is a potent regulator of tumor vascularization and metabolism, our findings suggest that HIF-1 plays a role in HT-induced tumor reoxygenation by transactivating its downstream targets. We demonstrate that NADPH oxidase-mediated reactive oxygen species production, as a mechanism, up-regulates HIF-1 after HT. Furthermore, we determine that this pathway is initiated by increased transcription of NADPH oxidase-1 through the ERK pathway. In conclusion, this study determines that, although HIF-1 is a good therapeutic target, the timing of its inhibition needs to be optimized to achieve the most beneficial outcome when it is combined with other treatments of HT, radiation, and chemotherapy.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipertermia Inducida , Factor 1 Inducible por Hipoxia/metabolismo , NADPH Oxidasas/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Especies Reactivas de Oxígeno/metabolismo , Análisis de Varianza , Animales , Western Blotting , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Ácido Láctico/sangre , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Nat Commun ; 14(1): 1849, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37012289

RESUMEN

Cachexia is a debilitating wasting syndrome and highly prevalent comorbidity in cancer patients. It manifests especially with energy and mitochondrial metabolism aberrations that promote tissue wasting. We recently identified nicotinamide adenine dinucleotide (NAD+) loss to associate with muscle mitochondrial dysfunction in cancer hosts. In this study we confirm that depletion of NAD+ and downregulation of Nrk2, an NAD+ biosynthetic enzyme, are common features of severe cachexia in different mouse models. Testing NAD+ repletion therapy in cachectic mice reveals that NAD+ precursor, vitamin B3 niacin, efficiently corrects tissue NAD+ levels, improves mitochondrial metabolism and ameliorates cancer- and chemotherapy-induced cachexia. In a clinical setting, we show that muscle NRK2 is downregulated in cancer patients. The low expression of NRK2 correlates with metabolic abnormalities underscoring the significance of NAD+ in the pathophysiology of human cancer cachexia. Overall, our results propose NAD+ metabolism as a therapy target for cachectic cancer patients.


Asunto(s)
Neoplasias , Niacina , Humanos , Ratones , Animales , Niacina/farmacología , Niacina/uso terapéutico , Niacina/metabolismo , NAD/metabolismo , Caquexia/tratamiento farmacológico , Caquexia/etiología , Caquexia/metabolismo , Niacinamida/metabolismo , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Músculo Esquelético/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA