Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 222(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36795453

ABSTRACT

Mammalian genes were long thought to be constrained within somatic cells in most cell types. This concept was challenged recently when cellular organelles including mitochondria were shown to move between mammalian cells in culture via cytoplasmic bridges. Recent research in animals indicates transfer of mitochondria in cancer and during lung injury in vivo, with considerable functional consequences. Since these pioneering discoveries, many studies have confirmed horizontal mitochondrial transfer (HMT) in vivo, and its functional characteristics and consequences have been described. Additional support for this phenomenon has come from phylogenetic studies. Apparently, mitochondrial trafficking between cells occurs more frequently than previously thought and contributes to diverse processes including bioenergetic crosstalk and homeostasis, disease treatment and recovery, and development of resistance to cancer therapy. Here we highlight current knowledge of HMT between cells, focusing primarily on in vivo systems, and contend that this process is not only (patho)physiologically relevant, but also can be exploited for the design of novel therapeutic approaches.


Subject(s)
Mitochondria , Neoplasms , Animals , Phylogeny , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Energy Metabolism , Mammals
2.
Front Pharmacol ; 12: 586715, 2021.
Article in English | MEDLINE | ID: mdl-33762933

ABSTRACT

Background: Circulating bilirubin is associated with reduced adiposity in human and animal studies. A possible explanation is provided by in vitro data that demonstrates that bilirubin inhibits mitochondrial function and decreases efficient energy production. However, it remains unclear whether hyperbilirubinemic animals have similar perturbed mitochondrial function and whether this is important for regulation of energy homeostasis. Aim: To investigate the impact of unconjugated hyperbilirubinemia on body composition, and mitochondrial function in hepatic tissue and skeletal muscle. Materials and Methods: 1) Food intake and bodyweight gain of 14-week old hyperbilirubinemic Gunn (n = 19) and normobilirubinemic littermate (control; n = 19) rats were measured over a 17-day period. 2) Body composition was determined using dual-energy X-ray absorptiometry and by measuring organ and skeletal muscle masses. 3) Mitochondrial function was assessed using high-resolution respirometry of homogenized liver and intact permeabilized extensor digitorum longus and soleus fibers. 4) Liver tissue was flash frozen for later gene (qPCR), protein (Western Blot and citrate synthase activity) and lipid analysis. Results: Female hyperbilirubinemic rats had significantly reduced fat mass (Gunn: 9.94 ± 5.35 vs. Control: 16.6 ± 6.90 g, p < 0.05) and hepatic triglyceride concentration (Gunn: 2.39 ± 0.92 vs. Control: 4.65 ± 1.67 mg g-1, p < 0.01) compared to normobilirubinemic controls. Furthermore, hyperbilirubinemic rats consumed fewer calories daily (p < 0.01) and were less energetically efficient (Gunn: 8.09 ± 5.75 vs. Control: 14.9 ± 5.10 g bodyweight kcal-1, p < 0.05). Hepatic mitochondria of hyperbilirubinemic rats demonstrated increased flux control ratio (FCR) via complex I and II (CI+II) (Gunn: 0.78 ± 0.16 vs. Control: 0.62 ± 0.09, p < 0.05). Similarly, exogenous addition of 31.3 or 62.5 µM unconjugated bilirubin to control liver homogenates significantly increased CI+II FCR (p < 0.05). Hepatic PGC-1α gene expression was significantly increased in hyperbilirubinemic females while FGF21 and ACOX1 was significantly greater in male hyperbilirubinemic rats (p < 0.05). Finally, hepatic mitochondrial complex IV subunit 1 protein expression was significantly increased in female hyperbilirubinemic rats (p < 0.01). Conclusions: This is the first study to comprehensively assess body composition, fat metabolism, and mitochondrial function in hyperbilirubinemic rats. Our findings show that hyperbilirubinemia is associated with reduced fat mass, and increased hepatic mitochondrial biogenesis, specifically in female animals, suggesting a dual role of elevated bilirubin and reduced UGT1A1 function on adiposity and body composition.

3.
Theranostics ; 10(12): 5259-5275, 2020.
Article in English | MEDLINE | ID: mdl-32373211

ABSTRACT

Purpose: Lacking effective targeted therapies, triple-negative breast cancer (TNBCs) is highly aggressive and metastatic disease, and remains clinically challenging breast cancer subtype to treat. Despite the survival dependency on the proteasome pathway genes, FDA-approved proteasome inhibitors induced minimal clinical response in breast cancer patients due to weak proteasome inhibition. Hence, developing effective targeted therapy using potent proteasome inhibitor is required. Methods: We evaluated anti-cancer activity of a potent proteasome inhibitor, marizomib, in vitro using breast cancer lines and in vivo using 4T1.2 murine syngeneic model, MDA-MB-231 xenografts, and patient-derived tumor xenografts. Global proteome profiling, western blots, and RT-qPCR were used to investigate the mechanism of action for marizomib. Effect of marizomib on lung and brain metastasis was evaluated using syngeneic 4T1BR4 murine TNBC model in vivo. Results: We show that marizomib inhibits multiple proteasome catalytic activities and induces a better anti-tumor response in TNBC cell lines and patient-derived xenografts alone and in combination with the standard-of-care chemotherapy. Mechanistically, we show that marizomib is a dual inhibitor of proteasome and oxidative phosphorylation (OXPHOS) in TNBCs. Marizomib reduces lung and brain metastases by reducing the number of circulating tumor cells and the expression of genes involved in the epithelial-to-mesenchymal transition. We demonstrate that marizomib-induced OXPHOS inhibition upregulates glycolysis to meet the energetic demands of TNBC cells and combined inhibition of glycolysis with marizomib leads to a synergistic anti-cancer activity. Conclusions: Our data provide a strong rationale for a clinical evaluation of marizomib in primary and metastatic TNBC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Lactones/therapeutic use , Proteasome Endopeptidase Complex/metabolism , Pyrroles/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice , Oxidative Phosphorylation/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
4.
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
5.
Antioxid Redox Signal ; 26(2): 84-103, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27392540

ABSTRACT

AIMS: Expression of the HER2 oncogene in breast cancer is associated with resistance to treatment, and Her2 may regulate bioenergetics. Therefore, we investigated whether disruption of the electron transport chain (ETC) is a viable strategy to eliminate Her2high disease. RESULTS: We demonstrate that Her2high cells and tumors have increased assembly of respiratory supercomplexes (SCs) and increased complex I-driven respiration in vitro and in vivo. They are also highly sensitive to MitoTam, a novel mitochondrial-targeted derivative of tamoxifen. Unlike tamoxifen, MitoTam efficiently suppresses experimental Her2high tumors without systemic toxicity. Mechanistically, MitoTam inhibits complex I-driven respiration and disrupts respiratory SCs in Her2high background in vitro and in vivo, leading to elevated reactive oxygen species production and cell death. Intriguingly, higher sensitivity of Her2high cells to MitoTam is dependent on the mitochondrial fraction of Her2. INNOVATION: Oncogenes such as HER2 can restructure ETC, creating a previously unrecognized therapeutic vulnerability exploitable by SC-disrupting agents such as MitoTam. CONCLUSION: We propose that the ETC is a suitable therapeutic target in Her2high disease. Antioxid. Redox Signal. 26, 84-103.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Electron Transport Chain Complex Proteins/metabolism , Receptor, ErbB-2/metabolism , Antineoplastic Agents/chemistry , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Respiration/drug effects , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/chemistry , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Female , Humans , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Molecular , Molecular Conformation , Molecular Targeted Therapy , Protein Binding , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Tamoxifen/pharmacology
7.
Oncotarget ; 7(24): 36338-36352, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27119351

ABSTRACT

Autophagy favors both cell survival and cancer suppression, and increasing evidence reveals that microRNAs (MIRs) regulate autophagy. Previously we reported that MIR126 is downregulated in malignant mesothelioma (MM). Therefore, we investigated the role of MIR126 in the regulation of cell metabolism and autophagy in MM models. We report that MIR126 induces autophagic flux in MM cells by downregulating insulin receptor substrate-1 (IRS1) and disrupting the IRS1 signaling pathway. This was specific to MM cells, and was not observed in non-malignant cells of mesothelial origin or in MM cells expressing MIR126-insensitive IRS1 transcript. The MIR126 effect on autophagy in MM cells was recapitulated by IRS1 silencing, and antagonized by IRS1 overexpression or antisense MIR126 treatment. The MIR126-induced loss of IRS1 suppressed glucose uptake, leading to energy deprivation and AMPK-dependent phosphorylation of ULK1. In addition, MIR126 stimulated lipid droplet accumulation in a hypoxia-inducible factor-1α (HIF1α)-dependent manner. MIR126 also reduced pyruvate dehydrogenase kinase (PDK) and acetyl-CoA-citrate lyase (ACL) expression, leading to the accumulation of cytosolic citrate and paradoxical inhibition of pyruvate dehydrogenase (PDH) activity. Simultaneous pharmacological and genetic intervention with PDK and ACL activity phenocopied the effects of MIR126. This suggests that in MM MIR126 initiates a metabolic program leading to high autophagic flux and HIF1α stabilization, incompatible with tumor progression of MM. Consistently, MIR126-expressing MM cells injected into immunocompromised mice failed to progress beyond the initial stage of tumor formation, showing that increased autophagy has a protective role in MM.


Subject(s)
Autophagy/genetics , Gene Expression Regulation, Neoplastic , Insulin Receptor Substrate Proteins/genetics , Lung Neoplasms/genetics , Mesothelioma/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling/methods , Humans , Insulin Receptor Substrate Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mesothelioma/metabolism , Mesothelioma/pathology , Mice, Inbred BALB C , Mice, Nude , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Transplantation, Heterologous
8.
Lung Cancer ; 90(3): 457-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26431916

ABSTRACT

OBJECTIVES: Malignant mesothelioma (MM) is a highly aggressive tumor with poor prognosis. A major challenge is the development and application of early and highly reliable diagnostic marker(s). Serum biomarkers, such as 'soluble mesothelin-related proteins' (SMRPs), is the most studied and frequently used in MM. However, the low sensitivity of SMRPs for early MM limits its value; therefore, additional biomarkers are required. In this study, two epigenetically regulated markers in MM (microRNA-126, miR-126, and methylated thrombomodulin promoter, Met-TM) were combined with SMRPs and evaluated as a potential strategy to detect MM at an early stage. MATERIALS AND METHODS: A total of 188 subjects, including 45 MM patients, 99 asbestos-exposed subjects, and 44 healthy controls were prospectively enrolled, serum samples collected, and serum levels of SMRPs, miR-126 and Met-TM evaluated. Logistic regression analysis was performed to evaluate the diagnostic value of the three biomarkers. Using this approach, the performance of the '3-biomarker classifier' was tested by calculating the overall probability score of the MM and control samples, respectively, and the ROC curve was generated. RESULTS AND CONCLUSION: The combination of the three biomarkers was the best predictor to differentiate MM patients from asbestos-exposed subjects and healthy controls. The accuracy and cancer specificity was confirmed in a second validation cohort and lung cancer population. We propose that the combination of the two epigenetic biomarkers with SMRPs as a diagnosis for early MM overcomes the limitations of using SMRPs alone.


Subject(s)
Biomarkers, Tumor , Epigenesis, Genetic , GPI-Linked Proteins/genetics , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Mesothelioma/diagnosis , Mesothelioma/genetics , Aged , DNA Methylation , Female , GPI-Linked Proteins/blood , Humans , Lung Neoplasms/blood , Lung Neoplasms/etiology , Lung Neoplasms/therapy , Male , Mesothelin , Mesothelioma/blood , Mesothelioma/etiology , Mesothelioma/therapy , Mesothelioma, Malignant , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Multidrug Resistance-Associated Proteins/blood , Prognosis , Reproducibility of Results
9.
Placenta ; 36(8): 863-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26154583

ABSTRACT

INTRODUCTION: Placental oxidative stress has been implicated in pregnancy complications and previous work has shown that selenium can protect trophoblast mitochondria from oxidative stress. This report examines mitochondrial function and content in trophoblasts supplemented with selenium. METHODS: Swan-71, JEG-3 and BeWo cells and placental tissue were incubated with sodium selenite or selenomethionine. Mitochondrial function was examined in a respirometer. Mitochondrial content was determined using RT-PCR. The levels of the mitochondrial biogenesis markers selenoprotein H, PGC-1α and NRF-1 was examined by western blotting. RESULTS: Mitochondrial respiration was significantly enhanced post selenium supplementation in cells and tissues. Selenium supplementation increased mitochondrial content and up-regulated mitochondrial biogenesis mediators in cells. DISCUSSION: These results emphasise the importance of selenium in mitochondrial regeneration in trophoblasts.


Subject(s)
Mitochondria/drug effects , Organelle Biogenesis , Placenta/drug effects , Selenium/administration & dosage , Trophoblasts/drug effects , Cell Line , DNA-Binding Proteins/metabolism , Female , Humans , Mitochondria/metabolism , NF-E2-Related Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Placenta/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Selenoproteins/metabolism , Transcription Factors/metabolism , Trophoblasts/metabolism , Up-Regulation/drug effects
10.
PLoS One ; 10(5): e0119549, 2015.
Article in English | MEDLINE | ID: mdl-25932953

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/pathology , Mesothelioma/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Gene Knockdown Techniques , Humans , Inhibitory Concentration 50 , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tocopherols/pharmacology
11.
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
12.
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
13.
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
14.
Curr Med Chem ; 22(5): 552-68, 2015.
Article in English | MEDLINE | ID: mdl-25245377

ABSTRACT

Tumours exhibit higher basal levels of reactive oxygen species (ROS) and altered redox environment compared to normal cells. Excessive level of ROS can be toxic to these cells, thus they become more vulnerable to damage by further ROS insults induced by pharmacological agents. However, the upregulation of antioxidant capacity in adaptation to intrinsic oxidative stress in cancer cells can confer drug resistance. Therefore, abrogation of such drug-resistant mechanisms by redox modulation could have significant therapeutic implications. Many redox-modulating agents have been developed. The redox-active system epitomised by ascorbate-driven quinone redox cycling, and the group of redox-silent vitamin E analogues represented by α-tocopheryl succinate have been shown to induce selective cancer cell death in different types of cancer. These compounds synergistically act by destabilising organelles like mitochondria, unleashing their apoptogenic potential, which results in efficient death of malignant cells and suppression of tumour growth. Consistent with this notion, clinical trials that aim to examine the therapeutic performance of novel redox-modulating drugs in cancer patients are currently under way.


Subject(s)
Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Apoptosis , Drug Synergism , Humans , Oxidation-Reduction
15.
Prog Mol Biol Transl Sci ; 127: 211-27, 2014.
Article in English | MEDLINE | ID: mdl-25149219

ABSTRACT

Cancer can be characterized as a state of multifaceted cellular deregulation including control of proliferation and bioenergetics. The latter involves in particular mitochondria, the site of the generation of ATP, essential for the proper cellular function (including proliferation). Mitochondria also contain a variety of proteins that are necessary for the induction/promotion, as well as for the prevention of cell death. Therefore, mitochondria are pivotal in deciding the fate of a cell. In cancer, mitochondria are dysfunctional, which was observed as early as in the 1930s by Otto Warburg. Due to the central role of mitochondria, these organelles, endowed with its own DNA, are a focus of research as possible "culprits" for the malignancy of cancer cells (or at least contributing to this phenotype) and, importantly, as emerging targets for anticancer therapy.


Subject(s)
Mitochondria/pathology , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Mitochondria/drug effects
16.
Mitochondrion ; 19 Pt A: 20-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24933571

ABSTRACT

Complex II of the respiratory chain (RC) recently emerged as a prominent regulator of cell death. In both cancer cells as well as neurodegenerative diseases, mutations in subunits have been found along with other genetic alterations indirectly affecting this complex. Anticancer compounds were developed that target complex II and cause cell death in a tumor-specific way. Our mechanistic understanding of how complex II is activated for cell death induction has recently been made clearer in recent studies, the results of which are covered in this review. This protein assembly is specifically activated for cell death via the dissociation of its SDHA and SDHB subunits from the membrane-anchoring proteins through pH change or mitochondrial Ca(2+) influx. The SDH activity contained in the SDHA/SDHB subcomplex remains intact and then generates, in an uncontrolled fashion, excessive amounts of reactive oxygen species (ROS) for cell death. Future studies on this mitochondrial complex will further elucidate it as a target for cancer treatments and reveal its role as a nexus for many diverse stimuli in cell death signaling.


Subject(s)
Electron Transport Chain Complex Proteins/physiology , Mitochondria/physiology , Animals , Cell Death/physiology , Energy Metabolism/physiology , Gene Expression Regulation/physiology
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.
Redox Rep ; 19(1): 16-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24225203

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Mesothelioma/drug therapy , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Delivery Systems , Drug Screening Assays, Antitumor , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex II/genetics , Humans , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Mesothelioma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Molecular Structure , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , alpha-Tocopherol/pharmacokinetics
19.
Free Radic Biol Med ; 67: 41-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24145120

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Neoplastic , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mitochondria/drug effects , Neoplastic Stem Cells/drug effects , alpha-Tocopherol/pharmacology , Cell Line, Tumor , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Female , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Tryptophan/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...