Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Onco Targets Ther ; 16: 371-383, 2023.
Article in English | MEDLINE | ID: mdl-37309471

ABSTRACT

MYC can be considered to be one of the most pressing and important targets for the development of novel anti-cancer therapies. This is due to its frequent dysregulation in tumors and due to the wide-ranging impact this dysregulation has on gene expression and cellular behavior. As a result, there have been numerous attempts to target MYC over the last few decades, both directly and indirectly, with mixed results. This article reviews the biology of MYC in the context of cancers and drug development. It discusses strategies aimed at targeting MYC directly, including those aimed at reducing its expression and blocking its function. In addition, the impact of MYC dysregulation on cellular biology is outlined, and how understanding this can underpin the development of approaches aimed at molecules and pathways regulated by MYC. In particular, the review focuses on the role that MYC plays in the regulation of metabolism, and the therapeutic avenues offered by inhibiting the metabolic pathways that are essential for the survival of MYC-transformed cells.

3.
Cancers (Basel) ; 14(22)2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36428647

ABSTRACT

There have been significant recent advances in the understanding of the role of metabolism in normal and malignant B-cell biology. Previous research has focused on the role of MYC and mammalian target of rapamycin (mTOR) and how these interact with B-cell receptor signaling and hypoxia to regulate glycolysis, glutaminolysis, oxidative phosphorylation (OXPHOS) and related metabolic pathways in germinal centers. Many of the commonest forms of lymphoma arise from germinal center B-cells, reflecting the physiological attenuation of normal DNA damage checkpoints to facilitate somatic hypermutation of the immunoglobulin genes. As a result, these lymphomas can inherit the metabolic state of their cell-of-origin. There is increasing interest in the potential of targeting metabolic pathways for anti-cancer therapy. Some metabolic inhibitors such as methotrexate have been used to treat lymphoma for decades, with several new agents being recently licensed such as inhibitors of phosphoinositide-3-kinase. Several other inhibitors are in development including those blocking mTOR, glutaminase, OXPHOS and monocarboxylate transporters. In addition, recent work has highlighted the importance of the interaction between diet and cancer, with particular focus on dietary modifications that restrict carbohydrates and specific amino acids. This article will review the current state of this field and discuss future developments.

4.
Hemasphere ; 6(6): e722, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35747847

ABSTRACT

It has been unclear what role metabolism is playing in the pathophysiology of chronic lymphocytic leukemia (CLL). One reason is that the study of CLL metabolism is challenging due to the resting nature of circulating CLL cells. Also, it is not clear if any of the genomic aberrations observed in this disease have any impact on metabolism. Here, we demonstrate that CLL cells in proliferation centers exhibit upregulation of several molecules involved in glycolysis and mitochondrial metabolism. Comparison of CXCR4/CD5 intraclonal cell subpopulations showed that these changes are paralleled by increases in the metabolic activity of the CXCR4lowCD5high fraction that have recently egressed from the lymph nodes. Notably, anti-IgM stimulation of CLL cells recapitulates many of these metabolic alterations, including increased glucose uptake, increased lactate production, induction of glycolytic enzymes, and increased respiratory reserve. Treatment of CLL cells with inhibitors of B-cell receptor (BCR) signaling blocked these anti-IgM-induced changes in vitro, which was mirrored by decreases in hexokinase 2 expression in CLL cells from ibrutinib-treated patients in vivo. Interestingly, several samples from patients with 17p-deletion manifested increased spontaneous aerobic glycolysis in the unstimulated state suggestive of a BCR-independent metabolic phenotype. We conclude that the proliferative fraction of CLL cells found in lymphoid tissues or the peripheral blood of CLL patients exhibit increased metabolic activity when compared with the bulk CLL-cell population. Although this is due to microenvironmental stimulatory signals such as BCR-engagement in most cases, increases in resting metabolic activity can be observed in cases with 17p-deletion.

5.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35316216

ABSTRACT

The synthesis of serine from glucose is a key metabolic pathway supporting cellular proliferation in healthy and malignant cells. Despite this, the role that this aspect of metabolism plays in germinal center biology and pathology is not known. Here, we performed a comprehensive characterization of the role of the serine synthesis pathway in germinal center B cells and lymphomas derived from these cells. We demonstrate that upregulation of a functional serine synthesis pathway is a metabolic hallmark of B cell activation and the germinal center reaction. Inhibition of phosphoglycerate dehydrogenase (PHGDH), the first and rate-limiting enzyme in this pathway, led to defective germinal formation and impaired high-affinity antibody production. In addition, overexpression of enzymes involved in serine synthesis was a characteristic of germinal center B cell-derived lymphomas, with high levels of expression being predictive of reduced overall survival in diffuse large B cell lymphoma. Inhibition of PHGDH induced apoptosis in lymphoma cells, reducing disease progression. These findings establish PHGDH as a critical player in humoral immunity and a clinically relevant target in lymphoma.


Subject(s)
Lymphoma, B-Cell , Lymphoma , Cell Proliferation , Germinal Center , Humans , Lymphoma/genetics , Lymphoma, B-Cell/genetics , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Serine/metabolism
6.
Cancer Res ; 81(13): 3480-3494, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34127497

ABSTRACT

Succinate dehydrogenase is a key enzyme in the tricarboxylic acid cycle and the electron transport chain. All four subunits of succinate dehydrogenase are tumor suppressor genes predisposing to paraganglioma, but only mutations in the SDHB subunit are associated with increased risk of metastasis. Here we generated an Sdhd knockout chromaffin cell line and compared it with Sdhb-deficient cells. Both cell types exhibited similar SDH loss of function, metabolic adaptation, and succinate accumulation. In contrast, Sdhb-/- cells showed hallmarks of mesenchymal transition associated with increased DNA hypermethylation and a stronger pseudo-hypoxic phenotype compared with Sdhd-/- cells. Loss of SDHB specifically led to increased oxidative stress associated with dysregulated iron and copper homeostasis in the absence of NRF2 activation. High-dose ascorbate exacerbated the increase in mitochondrial reactive oxygen species, leading to cell death in Sdhb-/- cells. These data establish a mechanism linking oxidative stress to iron homeostasis that specifically occurs in Sdhb-deficient cells and may promote metastasis. They also highlight high-dose ascorbate as a promising therapeutic strategy for SDHB-related cancers. SIGNIFICANCE: Loss of different succinate dehydrogenase subunits can lead to different cell and tumor phenotypes, linking stronger 2-OG-dependent dioxygenases inhibition, iron overload, and ROS accumulation following SDHB mutation.


Subject(s)
Ascorbic Acid/pharmacology , Homeostasis , Iron/metabolism , Mutation , Oxidative Stress , Succinate Dehydrogenase/physiology , Animals , Antioxidants/pharmacology , Dioxygenases/antagonists & inhibitors , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Phenotype , Reactive Oxygen Species
7.
FASEB J ; 34(1): 303-315, 2020 01.
Article in English | MEDLINE | ID: mdl-31914648

ABSTRACT

Mutations in succinate dehydrogenase (SDH) lead to the development of tumors in a restricted subset of cell types, including chromaffin cells and paraganglia. The molecular basis for this specificity is currently unknown. We show that loss of SDH activity in a chromaffin cell model does not perturb complex I function, retaining the ability to oxidize NADH within the electron transport chain. This activity supports continued oxidation of substrates within the tricarboxylic acid (TCA) cycle. However, due to the block in the TCA cycle at SDH, the high glutamine oxidation activity is only maintained through an efflux of succinate. We also show that although the mitochondria of SDH-deficient cells are less active per se, their higher mass per cell results in an overall respiratory rate that is comparable with wild-type cells. Finally, we observed that when their mitochondria are uncoupled, SDH-deficient cells are unable to preserve their viability, suggesting that the mitochondrial metabolic network is unable to compensate when exposed to additional stress. We therefore show that in contrast to models of SDH deficiency based on epithelial cells, a chromaffin cell model retains aspects of metabolic "health," which could form the basis of cell specificity of this rare tumor type.


Subject(s)
Chromaffin Cells/metabolism , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Neoplasms/metabolism , Succinate Dehydrogenase/physiology , Animals , Chromaffin Cells/pathology , Humans , Male , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mutation , NAD/metabolism , Neoplasms/pathology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Transcriptome
8.
Int J Mol Sci ; 20(16)2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31430957

ABSTRACT

Sorafenib is a multi-kinase inhibitor and one of the few systemic treatment options for patients with advanced hepatocellular carcinomas (HCCs). Resistance to sorafenib develops frequently and could be mediated by the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin (SIRT)1. We aimed to test whether sorafenib efficacy is influenced by cellular NAD levels and NAD-dependent SIRT1 function. We analyzed sorafenib effects on apoptosis induction, NAD salvage, mitochondrial function, and related signaling pathways in HCC cell lines (HepG2, Hep3B, und HUH7) overexpressing SIRT1 or supplemented with the NAD metabolite nicotinamide mononucleotide (NMN) compared to controls. Treatment of HCC cell lines with sorafenib dose-dependently induced apoptosis and a significant decrease in cellular NAD concentrations. The SIRT1 protein was downregulated in HUH7 cells but not in Hep3B cells. After sorafenib treatment, mitochondrial respiration in permeabilized cells was lower, citrate synthase activity was attenuated, and cellular adenosine triphosphate (ATP) levels were decreased. Concomitant to increased phosphorylation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), sorafenib treatment led to decreased activity of the mechanistic target of rapamycin (mTOR), indicative of energy deprivation. Transient overexpression of SIRT1, as well as NAD repletion by NMN, decreased sorafenib-induced apoptosis. We can, therefore, conclude that sorafenib influences the NAD/SIRT1/AMPK axis. Overexpression of SIRT1 could be an underlying mechanism of resistance to sorafenib treatment in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Sirtuin 1/metabolism , Sorafenib/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Humans , Liver Neoplasms/metabolism , Nicotinamide Mononucleotide/metabolism
9.
Cell Rep ; 28(7): 1717-1728.e6, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412242

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is modulated by conditions of metabolic stress and has been reported to decline with aging in preclinical models, but human data are sparse. Nicotinamide riboside (NR) supplementation ameliorates metabolic dysfunction in rodents. We aimed to establish whether oral NR supplementation in aged participants can increase the skeletal muscle NAD+ metabolome and if it can alter muscle mitochondrial bioenergetics. We supplemented 12 aged men with 1 g NR per day for 21 days in a placebo-controlled, randomized, double-blind, crossover trial. Targeted metabolomics showed that NR elevated the muscle NAD+ metabolome, evident by increased nicotinic acid adenine dinucleotide and nicotinamide clearance products. Muscle RNA sequencing revealed NR-mediated downregulation of energy metabolism and mitochondria pathways, without altering mitochondrial bioenergetics. NR also depressed levels of circulating inflammatory cytokines. Our data establish that oral NR is available to aged human muscle and identify anti-inflammatory effects of NR.


Subject(s)
Aging/metabolism , Anti-Inflammatory Agents/blood , Cytokines/blood , Metabolome/drug effects , Muscle, Skeletal/metabolism , Niacinamide/analogs & derivatives , Transcriptome/drug effects , Aged , Aged, 80 and over , Aging/drug effects , Cross-Sectional Studies , Cytokines/drug effects , Double-Blind Method , Humans , Male , Muscle, Skeletal/drug effects , NAD/metabolism , Niacinamide/pharmacology , Pyridinium Compounds
10.
Cell Metab ; 29(2): 399-416.e10, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30449682

ABSTRACT

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.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Oxidoreductases Acting on CH-CH Group Donors/physiology , Pyrimidines/metabolism , Animals , Cell Line, Tumor , Cell Respiration , Dihydroorotate Dehydrogenase , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidative Phosphorylation , Ubiquinone/metabolism
11.
Cell Rep ; 22(12): 3107-3114, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562167

ABSTRACT

Since the discovery of mutations in isocitrate dehydrogenase 1 (IDH1) in gliomas and other tumors, significant efforts have been made to gain a deeper understanding of the consequences of this oncogenic mutation. One aspect of the neomorphic function of the IDH1 R132H enzyme that has received less attention is the perturbation of cellular redox homeostasis. Here, we describe a biosynthetic pathway exhibited by cells expressing mutant IDH1. By virtue of a change in cellular redox homeostasis, IDH1-mutated cells synthesize excess glutamine-derived proline through enhanced activity of pyrroline 5-carboxylate reductase 1 (PYCR1), coupled to NADH oxidation. Enhanced proline biosynthesis partially uncouples the electron transport chain from tricarboxylic acid (TCA) cycle activity through the maintenance of a lower NADH/NAD+ ratio and subsequent reduction in oxygen consumption. Thus, we have uncovered a mechanism by which tumor cell survival may be promoted in conditions associated with perturbed redox homeostasis, as occurs in IDH1-mutated glioma.


Subject(s)
Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mitochondria/metabolism , Mutation , Proline/biosynthesis , Pyrroline Carboxylate Reductases/metabolism , Cell Line, Tumor , Citric Acid Cycle , Gene Knockdown Techniques , Glutamine/metabolism , Homeostasis , Humans , Mitochondria/enzymology , Mitochondria/genetics , Oligodendroglioma , Oxidation-Reduction , Pyrroline Carboxylate Reductases/genetics , delta-1-Pyrroline-5-Carboxylate Reductase
12.
Cell Tissue Res ; 372(2): 367-378, 2018 05.
Article in English | MEDLINE | ID: mdl-29450727

ABSTRACT

Hypoxia is a critical driver of cancer pathogenesis, directly inducing malignant phenotypes such as epithelial-mesenchymal transition, stem cell-like characteristics and metabolic transformation. However, hypoxia-associated phenotypes are often observed in cancer in the absence of hypoxia, a phenotype known as pseudohypoxia, which is very well documented in specific tumour types, including in paraganglioma/pheochromocytoma (PPGL). Approximately 40% of the PPGL tumours carry a germ line mutation in one of a number of susceptibility genes of which those that are found in succinate dehydrogenase (SDH) or in von Hippel-Lindau (VHL) genes manifest a strong pseudohypoxic phenotype. Mutations in SDH are oncogenic, forming tumours in a select subset of tissues, but the cause for this remains elusive. Although elevated succinate levels lead to increase in hypoxia-like signalling, there are other phenotypes that are being increasingly recognised in SDH-mutated PPGL, such as DNA hypermethylation. Further, recently unveiled changes in metabolic re-wiring of SDH-deficient cells might help to decipher cancer related roles of SDH in the future. In this review, we will discuss the various implications that the malfunctioning SDH can have and its impact on cancer development.


Subject(s)
Hypoxia/metabolism , Paraganglioma/metabolism , Pheochromocytoma/metabolism , Adrenal Gland Neoplasms , Animals , Humans , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Elife ; 62017 02 15.
Article in English | MEDLINE | ID: mdl-28195532

ABSTRACT

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.


Subject(s)
DNA, Mitochondrial/genetics , Gene Transfer, Horizontal , Melanoma/pathology , Animals , Cell Line, Tumor , Cell Respiration , Disease Models, Animal , Mice, Inbred C57BL
14.
Cell Metab ; 21(1): 81-94, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25565207

ABSTRACT

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.


Subject(s)
Mitochondria/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Citrate (si)-Synthase/metabolism , DNA, Mitochondrial/metabolism , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mitochondria/genetics , Mitochondria/ultrastructure , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transplantation, Homologous
15.
Methods Mol Biol ; 1265: 181-94, 2015.
Article in English | MEDLINE | ID: mdl-25634276

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Respiration/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Cell Line, Tumor , Humans , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism
16.
Antioxid Redox Signal ; 22(11): 883-900, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25578105

ABSTRACT

AIMS: To assess the effect of mitochondrially targeted vitamin E (VE) analogs on mitochondrial function and biogenesis. RESULTS: Mitochondrially targeted vitamin E succinate (MitoVES) is an efficient inducer of apoptosis in cancer cells. Here, we show that unlike its untargeted counterpart α-tocopheryl succinate, MitoVES suppresses proliferation of cancer cells at sub-apoptotic doses by way of affecting the mitochondrial DNA (mtDNA) transcripts. We found that MitoVES strongly suppresses the level of the displacement loop transcript followed by those of mtDNA genes coding for subunits of mitochondrial complexes. This process is coupled to the inhibition of mitochondrial respiration, dissipation of the mitochondrial membrane potential, and generation of reactive oxygen species. In addition, exposure of cancer cells to MitoVES led to decreased expression of TFAM and diminished mitochondrial biogenesis. The inhibition of mitochondrial transcription was replicated in vivo in a mouse model of HER2(high) breast cancer, where MitoVES lowered the level of mtDNA transcripts in cancer cells but not in normal tissue. INNOVATION: Our data show that mitochondrially targeted VE analogs represent a novel class of mitocans that not only induce apoptosis at higher concentrations but also block proliferation and suppress normal mitochondrial function and transcription at low, non-apoptogenic doses. CONCLUSIONS: Our data indicate a novel, selective anti-cancer activity of compounds that act by targeting mitochondria of cancer cells, inducing significant alterations in mitochondrial function associated with transcription of mtDNA-coded genes. These changes subsequently result in the arrest of cell proliferation.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/drug effects , Receptor, ErbB-2/genetics , alpha-Tocopherol/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Respiration/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Transgenic , Mitochondria/physiology , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
17.
Antioxid Redox Signal ; 21(15): 2109-25, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-24444362

ABSTRACT

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.


Subject(s)
Insulin Receptor Substrate Proteins/genetics , Lung Neoplasms/genetics , Mesothelioma/genetics , MicroRNAs/genetics , Mitochondria/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Receptor Substrate Proteins/metabolism , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Mice, Nude , Neoplasm Transplantation , Oxidative Stress , Oxygen Consumption , RNA Interference , Signal Transduction
18.
PLoS One ; 8(8): e71869, 2013.
Article in English | MEDLINE | ID: mdl-23967256

ABSTRACT

Mitochondrial respiratory chain is organised into supramolecular structures that can be preserved in mild detergent solubilisates and resolved by native electrophoretic systems. Supercomplexes of respiratory complexes I, III and IV as well as multimeric forms of ATP synthase are well established. However, the involvement of complex II, linking respiratory chain with tricarboxylic acid cycle, in mitochondrial supercomplexes is questionable. Here we show that digitonin-solubilised complex II quantitatively forms high molecular weight structures (CIIhmw) that can be resolved by clear native electrophoresis. CIIhmw structures are enzymatically active and differ in electrophoretic mobility between tissues (500 - over 1000 kDa) and cultured cells (400-670 kDa). While their formation is unaffected by isolated defects in other respiratory chain complexes, they are destabilised in mtDNA-depleted, rho0 cells. Molecular interactions responsible for the assembly of CIIhmw are rather weak with the complexes being more stable in tissues than in cultured cells. While electrophoretic studies and immunoprecipitation experiments of CIIhmw do not indicate specific interactions with the respiratory chain complexes I, III or IV or enzymes of the tricarboxylic acid cycle, they point out to a specific interaction between CII and ATP synthase.


Subject(s)
Electron Transport Complex II/chemistry , Animals , Cell Line , Electron Transport , Electron Transport Chain Complex Proteins/chemistry , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex II/metabolism , Humans , Metabolic Networks and Pathways , Mitochondria/genetics , Mitochondria/metabolism , Molecular Weight , Organ Specificity , Oxidative Phosphorylation , Protein Binding
19.
Apoptosis ; 18(3): 286-99, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23299931

ABSTRACT

α-Tocopheryl succinate (α-TOS) is a promising anti-cancer agent due to its selectivity for cancer cells. It is important to understand whether long-term exposure of tumour cells to the agent will render them resistant to the treatment. Exposure of the non-small cell lung carcinoma H1299 cells to escalating doses of α-TOS made them resistant to the agent due to the upregulation of the ABCA1 protein, which caused its efflux. Full susceptibility of the cells to α-TOS was restored by knocking down the ABCA1 protein. Similar resistance including ABCA1 gene upregulation was observed in the A549 lung cancer cells exposed to α-TOS. The resistance of the cells to α-TOS was overcome by its mitochondrially targeted analogue, MitoVES, that is taken up on the basis of the membrane potential, bypassing the enhanced expression of the ABCA1 protein. The in vitro results were replicated in mouse models of tumours derived from parental and resistant H1299 cells. We conclude that long-term exposure of cancer cells to α-TOS causes their resistance to the drug, which can be overcome by its mitochondrially targeted counterpart. This finding should be taken into consideration when planning clinical trials with vitamin E analogues.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Mitochondria/drug effects , alpha-Tocopherol/therapeutic use , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Knockdown Techniques , Mice
20.
Biochim Biophys Acta ; 1827(5): 552-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23142170

ABSTRACT

With the arrival of the third millennium, in spite of unprecedented progress in molecular medicine, cancer remains as untamed as ever. The complexity of tumours, dictating the potential response of cancer cells to anti-cancer agents, has been recently highlighted in a landmark paper by Weinberg and Hanahan on hallmarks of cancer [1]. Together with the recently published papers on the complexity of tumours in patients and even within the same tumour (see below), the cure for this pathology seems to be an elusive goal. Indisputably, the strategy ought to be changed, searching for targets that are generally invariant across the landscape of neoplastic diseases. One such target appears to be the mitochondrial complex II (CII) of the electron transfer chain, a recent focus of research. We document and highlight this particularly intriguing target in this review paper and give examples of drugs that use CII as their molecular target. This article is part of a Special Issue entitled: Respiratory complex II: Role in cellular physiology and disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Electron Transport Complex II/antagonists & inhibitors , Mitochondria/drug effects , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/classification , Apoptosis/drug effects , Electron Transport/drug effects , Electron Transport Complex II/metabolism , Humans , Mitochondria/metabolism , Models, Biological , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...