Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Theranostics ; 13(2): 438-457, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36632231

RESUMEN

Rationale: Despite growing evidence for mitochondria's involvement in cancer, the roles of specific metabolic components outside the respiratory complex have been little explored. We conducted metabolomic studies on mitochondrial DNA (mtDNA)-deficient (ρ0) cancer cells with lower proliferation rates to clarify the undefined roles of mitochondria in cancer growth. Methods and results: Despite extensive metabolic downregulation, ρ0 cells exhibited high glycerol-3-phosphate (G3P) level, due to low activity of mitochondrial glycerol-3-phosphate dehydrogenase (GPD2). Knockout (KO) of GPD2 resulted in cell growth suppression as well as inhibition of tumor progression in vivo. Surprisingly, this was unrelated to the conventional bioenergetic function of GPD2. Instead, multi-omics results suggested major changes in ether lipid metabolism, for which GPD2 provides dihydroxyacetone phosphate (DHAP) in ether lipid biosynthesis. GPD2 KO cells exhibited significantly lower ether lipid level, and their slower growth was rescued by supplementation of a DHAP precursor or ether lipids. Mechanistically, ether lipid metabolism was associated with Akt pathway, and the downregulation of Akt/mTORC1 pathway due to GPD2 KO was rescued by DHAP supplementation. Conclusion: Overall, the GPD2-ether lipid-Akt axis is newly described for the control of cancer growth. DHAP supply, a non-bioenergetic process, may constitute an important role of mitochondria in cancer.


Asunto(s)
Glicerolfosfato Deshidrogenasa , Mitocondrias , Neoplasias , Proteínas Proto-Oncogénicas c-akt , Metabolismo Energético , Éteres/metabolismo , Glicerolfosfato Deshidrogenasa/genética , Glicerolfosfato Deshidrogenasa/metabolismo , Mitocondrias/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Ratones , Neoplasias/enzimología , Neoplasias/patología , Humanos
2.
Cell Death Dis ; 11(2): 110, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034120

RESUMEN

p53-mutated tumors often exhibit increased resistance to standard chemotherapy and enhanced metastatic potential. Here we demonstrate that inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme of the de novo pyrimidine synthesis pathway, effectively decreases proliferation of cancer cells via induction of replication and ribosomal stress in a p53- and checkpoint kinase 1 (Chk1)-dependent manner. Mechanistically, a block in replication and ribosomal biogenesis result in p53 activation paralleled by accumulation of replication forks that activate the ataxia telangiectasia and Rad3-related kinase/Chk1 pathway, both of which lead to cell cycle arrest. Since in the absence of functional p53 the cell cycle arrest fully depends on Chk1, combined DHODH/Chk1 inhibition in p53-dysfunctional cancer cells induces aberrant cell cycle re-entry and erroneous mitosis, resulting in massive cell death. Combined DHODH/Chk1 inhibition effectively suppresses p53-mutated tumors and their metastasis, and therefore presents a promising therapeutic strategy for p53-mutated cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Puntos de Control del Ciclo Celular , Proliferación Celular , Pirimidinas/biosíntesis , Ribosomas/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Dihidroorotato Deshidrogenasa , Femenino , Regulación Neoplásica de la Expresión Génica , Genes erbB-2 , Células HCT116 , Humanos , Leflunamida/farmacología , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Ribosomas/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
3.
Cell Metab ; 29(2): 399-416.e10, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30449682

RESUMEN

Cancer cells without mitochondrial DNA (mtDNA) do not form tumors unless they reconstitute oxidative phosphorylation (OXPHOS) by mitochondria acquired from host stroma. To understand why functional respiration is crucial for tumorigenesis, we used time-resolved analysis of tumor formation by mtDNA-depleted cells and genetic manipulations of OXPHOS. We show that pyrimidine biosynthesis dependent on respiration-linked dihydroorotate dehydrogenase (DHODH) is required to overcome cell-cycle arrest, while mitochondrial ATP generation is dispensable for tumorigenesis. Latent DHODH in mtDNA-deficient cells is fully activated with restoration of complex III/IV activity and coenzyme Q redox-cycling after mitochondrial transfer, or by introduction of an alternative oxidase. Further, deletion of DHODH interferes with tumor formation in cells with fully functional OXPHOS, while disruption of mitochondrial ATP synthase has little effect. Our results show that DHODH-driven pyrimidine biosynthesis is an essential pathway linking respiration to tumorigenesis, pointing to inhibitors of DHODH as potential anti-cancer agents.


Asunto(s)
ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Neoplasias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/fisiología , Pirimidinas/metabolismo , Animales , Línea Celular Tumoral , Respiración de la Célula , Dihidroorotato Deshidrogenasa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación Oxidativa , Ubiquinona/metabolismo
4.
iScience ; 3: 192-207, 2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-30428319

RESUMEN

We synthesized a mitochondria-targeted honokiol (Mito-HNK) that facilitates its mitochondrial accumulation; this dramatically increases its potency and efficacy against highly metastatic lung cancer lines in vitro, and in orthotopic lung tumor xenografts and brain metastases in vivo. Mito-HNK is >100-fold more potent than HNK in inhibiting cell proliferation, inhibiting mitochondrial complex ?, stimulating reactive oxygen species generation, oxidizing mitochondrial peroxiredoxin-3, and suppressing the phosphorylation of mitoSTAT3. Within lung cancer brain metastases in mice, Mito-HNK induced the mediators of cell death and decreased the pathways that support invasion and proliferation. In contrast, in the non-malignant stroma, Mito-HNK suppressed pathways that support metastatic lesions, including those involved in inflammation and angiogenesis. Mito-HNK showed no toxicity and targets the metabolic vulnerabilities of primary and metastatic lung cancers. Its pronounced anti-invasive and anti-metastatic effects in the brain are particularly intriguing given the paucity of treatment options for such patients either alone or in combination with standard chemotherapeutics.

5.
Elife ; 62017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28195532

RESUMEN

Recently, we showed that generation of tumours in syngeneic mice by cells devoid of mitochondrial (mt) DNA (ρ0 cells) is linked to the acquisition of the host mtDNA. However, the mechanism of mtDNA movement between cells remains unresolved. To determine whether the transfer of mtDNA involves whole mitochondria, we injected B16ρ0 mouse melanoma cells into syngeneic C57BL/6Nsu9-DsRed2 mice that express red fluorescent protein in their mitochondria. We document that mtDNA is acquired by transfer of whole mitochondria from the host animal, leading to normalisation of mitochondrial respiration. Additionally, knockdown of key mitochondrial complex I (NDUFV1) and complex II (SDHC) subunits by shRNA in B16ρ0 cells abolished or significantly retarded their ability to form tumours. Collectively, these results show that intact mitochondria with their mtDNA payload are transferred in the developing tumour, and provide functional evidence for an essential role of oxidative phosphorylation in cancer.


Asunto(s)
ADN Mitocondrial/genética , Transferencia de Gen Horizontal , Melanoma/patología , Animales , Línea Celular Tumoral , Respiración de la Célula , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
6.
Curr Opin Genet Dev ; 38: 75-82, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27219870

RESUMEN

Current dogma holds that genes are the property of individual mammalian cells and partition between daughter cells during cell division. However, and rather unexpectedly, recent research has demonstrated horizontal cell-to-cell transfer of mitochondria and mitochondrial DNA in several mammalian cell culture systems. Furthermore, unequivocal evidence that mitochondrial DNA transfer occurs in vivo has now been published. While these studies show horizontal transfer of mitochondrial DNA in pathological settings, it is also possible that intercellular mitochondrial transfer is a fundamental physiological process with a role in development and tissue homeostasis.


Asunto(s)
Comunicación Celular/genética , ADN Mitocondrial/genética , Transferencia de Gen Horizontal/genética , Mitocondrias/genética , Animales , División Celular/genética , Humanos
8.
Methods Mol Biol ; 1265: 181-94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25634276

RESUMEN

Respiration is one of the major functions of mitochondria, whereby these vital organelles use oxygen to produce energy. Many agents that may be of potential clinical relevance act by targeting mitochondria, where they may suppress mitochondrial respiration. It is therefore important to evaluate this process and understand how this is modulated by small molecules. Here, we describe the general methodology to assess respiration in cultured cells, followed by the evaluation of the effect of one anticancer agent targeted to mitochondria on this process, and also how to assess this in tumor tissue.


Asunto(s)
Antineoplásicos/farmacología , Respiración de la Célula/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Animales , Línea Celular Tumoral , Humanos , Oxidación-Reducción/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
9.
PLoS One ; 10(5): e0119549, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25932953

RESUMEN

Malignant mesothelioma (MM) is an aggressive type of tumour causing high mortality. One reason for this paradigm may be the existence of a subpopulation of tumour-initiating cells (TICs) that endow MM with drug resistance and recurrence. The objective of this study was to identify and characterise a TIC subpopulation in MM cells, using spheroid cultures, mesospheres, as a model of MM TICs. Mesospheres, typified by the stemness markers CD24, ABCG2 and OCT4, initiated tumours in immunodeficient mice more efficiently than adherent cells. CD24 knock-down cells lost the sphere-forming capacity and featured lower tumorigenicity. Upon serial transplantation, mesospheres were gradually more efficiently tumrigenic with increased level of stem cell markers. We also show that mesospheres feature mitochondrial and metabolic properties similar to those of normal and cancer stem cells. Finally, we show that mesothelioma-initiating cells are highly susceptible to mitochondrially targeted vitamin E succinate. This study documents that mesospheres can be used as a plausible model of mesothelioma-initiating cells and that they can be utilised in the search for efficient agents against MM.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/patología , Mesotelioma/patología , Células Madre Neoplásicas/patología , Animales , Biomarcadores de Tumor/metabolismo , Antígeno CD24/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Mesotelioma Maligno , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fenotipo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Tocoferoles/farmacología
10.
Cell Metab ; 21(1): 81-94, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25565207

RESUMEN

We report that tumor cells without mitochondrial DNA (mtDNA) show delayed tumor growth, and that tumor formation is associated with acquisition of mtDNA from host cells. This leads to partial recovery of mitochondrial function in cells derived from primary tumors grown from cells without mtDNA and a shorter lag in tumor growth. Cell lines from circulating tumor cells showed further recovery of mitochondrial respiration and an intermediate lag to tumor growth, while cells from lung metastases exhibited full restoration of respiratory function and no lag in tumor growth. Stepwise assembly of mitochondrial respiratory (super)complexes was correlated with acquisition of respiratory function. Our findings indicate horizontal transfer of mtDNA from host cells in the tumor microenvironment to tumor cells with compromised respiratory function to re-establish respiration and tumor-initiating efficacy. These results suggest pathophysiological processes for overcoming mtDNA damage and support the notion of high plasticity of malignant cells.


Asunto(s)
Mitocondrias/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citrato (si)-Sintasa/metabolismo , ADN Mitocondrial/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/genética , Mitocondrias/ultraestructura , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trasplante Homólogo
11.
Redox Rep ; 19(1): 16-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24225203

RESUMEN

UNLABELLED: Malignant mesothelioma (MM) is a fatal neoplastic disease with no therapeutic option. Therefore, the search for novel therapies is of paramount importance. METHODS: Since mitochondrial targeting of α-tocopheryl succinate (α-TOS) by its tagging with triphenylphosphonium enhances its cytotoxic effects to cancer cells, we tested its effect on MM cells and experimental mesotheliomas. RESULTS: Mitochondrially targeted vitamin E succinate (MitoVES) was more efficient in killing MM cells than α-TOS with IC50 lower by up to two orders of magnitude. Mitochondrial association of MitoVES in MM cells was documented using its fluorescently tagged analogue. MitoVES caused apoptosis in MM cells by mitochondrial destabilization, resulting in the loss of mitochondrial membrane potential, generation of reactive oxygen species, and destabilization of respiratory supercomplexes. The role of the mitochondrial complex II in the activity of MitoVES was confirmed by the finding that MM cells with suppressed succinate quinone reductase were resistant to MitoVES. MitoVES suppressed mesothelioma growth in nude mice with high efficacy. DISCUSSION: MitoVES is more efficient in killing MM cells and suppressing experimental mesotheliomas compared with the non-targeted α-TOS, giving it a potential clinical benefit.


Asunto(s)
Antineoplásicos/uso terapéutico , Mesotelioma/tratamiento farmacológico , alfa-Tocoferol/análogos & derivados , alfa-Tocoferol/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/genética , Humanos , Concentración 50 Inhibidora , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mesotelioma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Estructura Molecular , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-Tocoferol/farmacocinética
12.
Antioxid Redox Signal ; 21(15): 2109-25, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24444362

RESUMEN

AIMS: MiR126 was found to be frequently lost in many types of cancer, including malignant mesothelioma (MM), which represents one of the most challenging neoplastic diseases. In this study, we investigated the potential tumor suppressor function of MiR126 in MM cells. The effect of MiR126 was examined in response to oxidative stress, aberrant mitochondrial function induced by inhibition of complex I, mitochondrial DNA (mtDNA) depletion, and hypoxia. RESULTS: MiR126 was up-regulated by oxidative stress in nonmalignant mesothelial (Met5A) and MM (H28) cell lines. In Met5A cells, rotenone inhibited MiR126 expression, but mtDNA depletion and hypoxia up-regulated MiR126. However, these various stimuli suppressed the levels of MiR126 in H28 cells. MiR126 affected mitochondrial energy metabolism, reduced mitochondrial respiration, and promoted glycolysis in H28 cells. This metabolic shift, associated with insulin receptor substrate-1 (IRS1)-modulated ATP-citrate lyase deregulation, resulted in higher ATP and citrate production. These changes were linked to the down-regulation of IRS1 by ectopic MiR126, reducing Akt signaling and inhibiting cytosolic sequestration of Forkhead box O1 (FoxO1), which promoted the expression of genes involved in gluconeogenesis and oxidative stress defense. These metabolic changes induced hypoxia-inducible factor-1α (HIF1α) stabilization. Consequently, MiR126 suppressed the malignancy of MM cells in vitro, a notion corroborated by the failure of H28(MiR126) cells to form tumors in nude mice. INNOVATION AND CONCLUSION: MiR126 affects mitochondrial energy metabolism, resulting in MM tumor suppression. Since MM is a fatal neoplastic disease with a few therapeutic options, this finding is of potential translational importance.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/genética , Neoplasias Pulmonares/genética , Mesotelioma/genética , MicroARNs/genética , Mitocondrias/metabolismo , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neoplasias Pulmonares/patología , Mesotelioma/patología , Mesotelioma Maligno , Ratones Desnudos , Trasplante de Neoplasias , Estrés Oxidativo , Consumo de Oxígeno , Interferencia de ARN , Transducción de Señal
13.
Free Radic Biol Med ; 67: 41-50, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24145120

RESUMEN

Tumor-initiating cells (TICs) often survive therapy and give rise to second-line tumors. We tested the plausibility of sphere cultures as models of TICs. Microarray data and microRNA data analysis confirmed the validity of spheres as models of TICs for breast and prostate cancer as well as mesothelioma cell lines. Microarray data analysis revealed the Trp pathway as the only pathway upregulated significantly in all types of studied TICs, with increased levels of indoleamine-2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme of Trp metabolism along the kynurenine pathway. All types of TICs also expressed higher levels of the Trp uptake system consisting of CD98 and LAT1 with functional consequences. IDO1 expression was regulated via both transcriptional and posttranscriptional mechanisms, depending on the cancer type. Serial transplantation of TICs in mice resulted in gradually increased IDO1. Mitocans, represented by α-tocopheryl succinate and mitochondrially targeted vitamin E succinate (MitoVES), suppressed IDO1 in TICs. MitoVES suppressed IDO1 in TICs with functional mitochondrial complex II, involving transcriptional and posttranscriptional mechanisms. IDO1 increase and its suppression by VE analogues were replicated in TICs from primary human glioblastomas. Our work indicates that IDO1 is increased in TICs and that mitocans suppress the protein.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Regulación Neoplásica de la Expresión Génica , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Mitocondrias/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , alfa-Tocoferol/farmacología , Línea Celular Tumoral , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Femenino , Proteína-1 Reguladora de Fusión/genética , Proteína-1 Reguladora de Fusión/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Mitocondrias/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal , Triptófano/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA