Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166879, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37689158

RESUMO

The endogenous inhibitor of mitochondrial F1Fo-ATPase (ATP synthase), IF1, has been shown to exert pro-oncogenic actions, including reprogramming of cellular energy metabolism (Warburg effect). The latter action of IF1 has been reported to be hampered by its PKA-dependent phosphorylation, but both reprogramming of metabolism and PKA-dependent phosphorylation are intensely debated. To clarify these critical issues, we prepared stably IF1-silenced clones and compared their bioenergetics with that of the three parental IF1-expressing cancer cell lines. All functional parameters: respiration rate, ATP synthesis rate (OXPHOS), and mitochondrial membrane potential were similar in IF1-silenced and control cells, clearly indicating that IF1 cannot inhibit the ATP synthase in cancer cells when the enzyme works physiologically. Furthermore, all cell types exposed to PKA modulators and energized with NAD+-dependent substrates or succinate showed similar OXPHOS rate regardless of the presence or absence of IF1. Therefore, our results rule out that IF1 action is modulated by its PKA-dependent phosphorylated/dephosphorylated state. Notably, cells exposed to a negative PKA modulator and energized with NAD+-dependent substrates showed a significant decrease of the OXPHOS rate matching previously reported inactivation of complex I. Overall, this study definitively demonstrates that IF1 inhibits neither mitochondrial ATP synthase nor OXPHOS in normoxic cancer cells and does not contribute to the Warburg effect. Thus, currently the protection of cancer cells from severe hypoxia/anoxia and apoptosis remain the only unquestionable actions of IF1 as pro-oncogenic factor that may be exploited to develop therapeutic approaches.


Assuntos
NAD , Neoplasias , Humanos , Trifosfato de Adenosina/metabolismo , Mitocôndrias/metabolismo , NAD/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação , Proteína Inibidora de ATPase
2.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834071

RESUMO

Cancer cells overexpress IF1, the endogenous protein that inhibits the hydrolytic activity of ATP synthase when mitochondrial membrane potential (ΔµH+) falls, as in ischemia. Other roles have been ascribed to IF1, but the associated molecular mechanisms are still under debate. We investigated the ability of IF1 to promote survival and proliferation in osteosarcoma and colon carcinoma cells exposed to conditions mimicking ischemia and reperfusion, as occurs in vivo, particularly in solid tumors. IF1-silenced and parental cells were exposed to the FCCP uncoupler to collapse ΔµH+ and the bioenergetics of cell models were validated. All the uncoupled cells preserved mitochondrial mass, but the implemented mechanisms differed in IF1-expressing and IF1-silenced cells. Indeed, the membrane potential collapse and the energy charge preservation allowed an increase in both mitophagy and mitochondrial biogenesis in IF1-expressing cells only. Interestingly, the presence of IF1 also conferred a proliferative advantage to cells highly dependent on oxidative phosphorylation when the uncoupler was washed out, mimicking cell re-oxygenation. Overall, our results indicate that IF1, by allowing energy preservation and promoting mitochondrial renewal, can favor proliferation of anoxic cells and tumor growth. Therefore, hindering the action of IF1 may be promising for the therapy of tumors that rely on oxidative phosphorylation for energy production.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Mitocôndrias/metabolismo , Hipóxia/metabolismo , Osteossarcoma/metabolismo , Neoplasias Ósseas/metabolismo , Isquemia/metabolismo , Proliferação de Células , Trifosfato de Adenosina/metabolismo
3.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37569836

RESUMO

The Food and Drug Administration has recently classified the IQOS electronic cigarette as a modified-risk tobacco product. However, IQOS cigarettes still release various harmful constituents typical of conventional cigarettes (CCs), although the concentrations are markedly lower. Here, we investigated the damaging effects of IQOS smoking on the liver. Male Sprague Dawley rats were exposed, whole body, 5 days/week for 4 weeks to IQOS smoke (4 sticks/day), and hepatic xenobiotic metabolism, redox homeostasis and lipidomic profile were investigated. IQOS boosted reactive radicals and generated oxidative stress. Exposure decreased cellular reserves of total glutathione (GSH) but not GSH-dependent antioxidant enzymes. Catalase and xanthine oxidase were greater in the exposed group, as were various hepatic CYP-dependent monooxygenases (CYP2B1/2, CYP1A1, CYP2A1, CYP2E1-linked). Respiratory chain activity was unaltered, while the number of liver mitochondria was increased. IQOS exposure had an impact on the hepatic lipid profile. With regard to the expression of some MAP kinases commonly activated by CC smoking, IQOS increased the p-p38/p38 ratio, while erythroid nuclear transcription factor 2 (Nrf2) was negatively affected. Our data suggest that IQOS significantly impairs liver function, supporting the precautionary stance taken by the WHO toward the use of these devices, especially by young people and pregnant women.


Assuntos
Sistemas Eletrônicos de Liberação de Nicotina , Produtos do Tabaco , Gravidez , Ratos , Animais , Masculino , Feminino , Humanos , Fumaça , Ratos Sprague-Dawley , Produtos do Tabaco/efeitos adversos , Fígado
4.
Cell Death Dis ; 14(1): 54, 2023 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-36690622

RESUMO

The mitochondrial protein IF1 binds to the catalytic domain of the ATP synthase and inhibits ATP hydrolysis in ischemic tissues. Moreover, IF1 is overexpressed in many tumors and has been shown to act as a pro-oncogenic protein, although its mechanism of action is still debated. Here, we show that ATP5IF1 gene disruption in HeLa cells decreases colony formation in soft agar and tumor mass development in xenografts, underlining the role of IF1 in cancer. Notably, the lack of IF1 does not affect proliferation or oligomycin-sensitive mitochondrial respiration, but it sensitizes the cells to the opening of the permeability transition pore (PTP). Immunoprecipitation and proximity ligation analysis show that IF1 binds to the ATP synthase OSCP subunit in HeLa cells under oxidative phosphorylation conditions. The IF1-OSCP interaction is confirmed by NMR spectroscopy analysis of the recombinant soluble proteins. Overall, our results suggest that the IF1-OSCP interaction protects cancer cells from PTP-dependent apoptosis under normoxic conditions.


Assuntos
ATPases Mitocondriais Próton-Translocadoras , Neoplasias , Humanos , Células HeLa , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Mitocôndrias/metabolismo , Óxido Nítrico Sintase/metabolismo , Apoptose , Trifosfato de Adenosina/metabolismo , Neoplasias/patologia
5.
J Comp Physiol B ; 192(2): 349-360, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35001173

RESUMO

The ability to induce a hypothermia resembling that of natural torpor would be greatly beneficial in medical and non-medical fields. At present, two procedures based on central nervous pharmacological manipulation have been shown to be effective in bringing core body temperature well below 30 °C in the rat, a non-hibernator: the first, based on the inhibition of a key relay in the central thermoregulatory pathway, the other, based on the activation of central adenosine A1 receptors. Although the role of mitochondria in the activation and maintenance of torpor has been extensively studied, no data are available for centrally induced hypothermia in non-hibernators. Thus, in the present work the respiration rate of mitochondria in the liver and in the kidney of rats following the aforementioned hypothermia-inducing treatments was studied. Moreover, to have an internal control, the same parameters were assessed in a well-consolidated model, i.e., mice during fasting-induced torpor. Our results show that state 3 respiration rate, which significantly decreased in the liver of mice, was unchanged in rats. An increase of state 4 respiration rate was observed in both species, although it was not statistically significant in rats under central adenosine stimulation. Also, a significant decrease of the respiratory control ratio was detected in both species. Finally, no effects were detected in kidney mitochondria in both species. Overall, in these hypothermic conditions liver mitochondria of rats remained active and apparently ready to be re-activated to produce energy and warm up the cells. These findings can be interpreted as encouraging in view of the finalization of a translational approach to humans.


Assuntos
Hipotermia , Torpor , Animais , Respiração Celular , Camundongos , Mitocôndrias/metabolismo , Ratos , Receptor A1 de Adenosina/fisiologia , Torpor/fisiologia
6.
Biochem Soc Trans ; 49(2): 815-827, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33929490

RESUMO

In the last two decades, IF1, the endogenous inhibitor of the mitochondrial F1Fo-ATPase (ATP synthase) has assumed greater and ever greater interest since it has been found to be overexpressed in many cancers. At present, several findings indicate that IF1 is capable of playing a central role in cancer cells by promoting metabolic reprogramming, proliferation and resistance to cell death. However, the mechanism(s) at the basis of this pro-oncogenic action of IF1 remains elusive. Here, we recall the main features of the mechanism of the action of IF1 when the ATP synthase works in reverse, and discuss the experimental evidence that support its relevance in cancer cells. In particular, a clear pro-oncogenic action of IF1 is to avoid wasting of ATP when cancer cells are exposed to anoxia or near anoxia conditions, therefore favoring cell survival and tumor growth. However, more recently, various papers have described IF1 as an inhibitor of the ATP synthase when it is working physiologically (i.e. synthethizing ATP), and therefore reprogramming cell metabolism to aerobic glycolysis. In contrast, other studies excluded IF1 as an inhibitor of ATP synthase under normoxia, providing the basis for a hot debate. This review focuses on the role of IF1 as a modulator of the ATP synthase in normoxic cancer cells with the awareness that the knowledge of the molecular action of IF1 on the ATP synthase is crucial in unravelling the molecular mechanism(s) responsible for the pro-oncogenic role of IF1 in cancer and in developing related anticancer strategies.


Assuntos
Metabolismo Energético/genética , ATPases Mitocondriais Próton-Translocadoras/genética , Neoplasias/genética , Proteínas/genética , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , ATPases Mitocondriais Próton-Translocadoras/química , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Modelos Moleculares , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Proteínas/química , Proteínas/metabolismo , Homologia de Sequência de Aminoácidos , Proteína Inibidora de ATPase
7.
Cell Death Dis ; 11(11): 1012, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33243973

RESUMO

Nutrient utilization and reshaping of metabolism in cancer cells is a well-known driver of malignant transformation. Less clear is the influence of the local microenvironment on metastasis formation and choice of the final organ to invade. Here we show that the level of the amino acid serine in the cytosol affects the migratory properties of lung adenocarcinoma (LUAD) cells. Inhibition of serine or glycine uptake from the extracellular milieu, as well as knockdown of the cytosolic one-carbon metabolism enzyme serine hydroxymethyltransferase (SHMT1), abolishes migration. Using rescue experiments with a brain extracellular extract, and direct measurements, we demonstrate that cytosolic serine starvation controls cell movement by increasing reactive oxygen species formation and decreasing ATP levels, thereby promoting activation of the AMP sensor kinase (AMPK) by phosphorylation. Activation of AMPK induces remodeling of the cytoskeleton and finally controls cell motility. These results highlight that cytosolic serine metabolism plays a key role in controlling motility, suggesting that cells are able to dynamically exploit the compartmentalization of this metabolism to adapt their metabolic needs to different cell functions (movement vs. proliferation). We propose a model to explain the relevance of serine/glycine metabolism in the preferential colonization of the brain by LUAD cells and suggest that the inhibition of serine/glycine uptake and/or cytosolic SHMT1 might represent a successful strategy to limit the formation of brain metastasis from primary tumors, a major cause of death in these patients.


Assuntos
Adenocarcinoma de Pulmão/genética , Adenilato Quinase/metabolismo , Glicina Hidroximetiltransferase/metabolismo , Adenocarcinoma de Pulmão/patologia , Movimento Celular , Humanos
8.
Cells ; 8(11)2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31752092

RESUMO

Changes of quantity and/or morphology of cell mitochondria are often associated with metabolic modulation, pathology, and apoptosis. Exogenous fluorescent probes used to investigate changes in mitochondrial content and dynamics are strongly dependent, for their internalization, on the mitochondrial membrane potential and composition, thus limiting the reliability of measurements. To overcome this limitation, genetically encoded recombinant fluorescent proteins, targeted to different cellular districts, were used as reporters. Here, we explored the potential use of mitochondrially targeted red fluorescent probe (mtRFP) to quantify, by flow cytometry, mitochondrial mass changes in cells exposed to different experimental conditions. We first demonstrated that the mtRFP fluorescence intensity is stable during cell culture and it is related with the citrate synthase activity, an established marker of the mitochondrial mass. Incidentally, the expression of mtRFP inside mitochondria did not alter the oxygen consumption rate under both state 3 and 4 respiration conditions. In addition, using this method, we showed for the first time that different inducers of mitochondrial mass change, such as hypoxia exposure or resveratrol treatment of cells, could be consistently detected. We suggest that transfection and selection of stable clones expressing mtRFP is a reliable method to monitor mitochondrial mass changes, particularly when pathophysiological or experimental conditions change ΔΨm, as it occurs during mitochondrial uncoupling or hypoxia/anoxia conditions.


Assuntos
Neoplasias Ósseas/metabolismo , Mitocôndrias/metabolismo , Osteossarcoma/metabolismo , Apoptose , Linhagem Celular Tumoral , Citrato (si)-Sintase/metabolismo , Citometria de Fluxo , Humanos , Potencial da Membrana Mitocondrial , Consumo de Oxigênio
9.
J Nutr Biochem ; 62: 9-17, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30216747

RESUMO

Dysfunctions caused by genetic defects in the mitochondrial DNA (mtDNA) of humans are called mitochondrial diseases; however, mtDNA mutations are also associated with aging and age-related diseases. Here, we present an original cellular model that allows gathering information on molecules that might contrast or prevent mitochondrial dysfunctions and their related diseases. This model allowed us to show that resveratrol (RSV), a phytochemical present in food, exerts protective effects at low concentrations on resting human fibroblasts carrying dysfunctional respiratory chain Complex I. Cells were maintained both in resting condition, to mimic the high energy demanding post-mitotic tissues (serum absence and gramicidin presence), and under glucose deficiency to push the synthesis of ATP via oxidative phosphorylation. Pre-incubation with RSV prolonged the viability of the fibroblasts exposed to rotenone, a well-known specific inhibitor of the respiratory chain Complex I, and decreased mitochondrial fragmentation. It significantly prevented the oxidative phosphorylation impairment indirectly caused by the rotenone-mediated Complex I inhibition, allowing for an almost complete preservation of the cellular ATP level. Indeed, RSV limited the rotenone-induced reactive oxygen species increase, allowing for the maintenance of a functional mitochondrial membrane potential. These findings indicate the potential usage of resveratrol to prevent or possibly treat many disorders, in which the bioenergetic defects and oxidative stress are the primary (mitochondrial encephalomyopathy), or the secondary (age-related diseases) causes of the pathology; and to also assist cell senescence during aging.


Assuntos
Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Resveratrol/farmacologia , Trifosfato de Adenosina/biossíntese , Células Cultivadas , Complexo I de Transporte de Elétrons/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Fosforilação Oxidativa/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Rotenona/toxicidade
10.
Cells ; 7(7)2018 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-29933600

RESUMO

The role of reactive oxygen species (ROS) in the metabolic reprogramming of cells adapted to hypoxia and the interplay between ROS and hypoxia in malignancy is under debate. Here, we examined how ROS levels are modulated by hypoxia in human cancer compared to untransformed cells. Short time exposure (20 min) of either fibroblasts or 143B osteosarcoma cells to low oxygen tension down to 0.5% induced a significant decrease of the cellular ROS level, as detected by the CellROX fluorescent probe (−70%). Prolonging the cells’ exposure to hypoxia for 24 h, ROS decreased further, reaching nearly 20% of the normoxic value. In this regard, due to the debated role of the endogenous inhibitor protein (IF1) of the ATP synthase complex in cancer cell bioenergetics, we investigated whether IF1 is involved in the control of ROS generation under severe hypoxic conditions. A significant ROS content decrease was observed in hypoxia in both IF1-expressing and IF1- silenced cells compared to normoxia. However, IF1-silenced cells showed higher ROS levels compared to IF1-containing cells. In addition, the MitoSOX Red-measured superoxide level of all the hypoxic cells was significantly lower compared to normoxia; however, the decrease was milder than the marked drop of ROS content. Accordingly, the difference between IF1-expressing and IF1-silenced cells was smaller but significant in both normoxia and hypoxia. In conclusion, the interplay between ROS and hypoxia and its modulation by IF1 have to be taken into account to develop therapeutic strategies against cancer.

11.
Cell Physiol Biochem ; 47(3): 1230-1243, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29913456

RESUMO

BACKGROUND/AIMS: Dietary polyphenols from green tea have been shown to possess cardio-protective activities in different experimental models of heart diseases and age-related ventricular dysfunction. The present study was aimed at evaluating whether long term in vivo administration of green tea extracts (GTE), can exert positive effects on the normal heart, with focus on the underlying mechanisms. METHODS: The study population consisted of 20 male adult Wistar rats. Ten animals were given 40 mL/day tap water solution of GTE (concentration 0.3%) for 4 weeks (GTE group). The same volume of water was administered to the 10 remaining control rats (CTRL). Then, in vivo and ex vivo measurements of cardiac function were performed in the same animal, at the organ (hemodynamics) and cellular (cardiomyocyte mechanical properties and intracellular calcium dynamics) levels. On cardiomyocytes and myocardial tissue samples collected from the same in vivo studied animals, we evaluated: (1) the intracellular content of ATP, (2) the endogenous mitochondrial respiration, (3) the expression levels of the Sarcoplasmic Reticulum Ca2+-dependent ATPase 2a (SERCA2), the Phospholamban (PLB) and the phosphorylated form of PLB, the L-type Ca2+ channel, the Na+-Ca2+ exchanger, and the ryanodine receptor 2. RESULTS: GTE cardiomyocytes exhibited a hyperdynamic contractility compared with CTRL (the rate of shortening and re-lengthening, the fraction of shortening, the amplitude of calcium transient, and the rate of cytosolic calcium removal were significantly increased). A faster isovolumic relaxation was also observed at the organ level. Consistent with functional data, we measured a significant increase in the intracellular ATP content supported by enhanced endogenous mitochondrial respiration in GTE cardiomyocytes, as well as higher values of the ratios phosphorylated-PLB/PLB and SERCA2/PLB. CONCLUSIONS: Long-term in vivo administration of GTE improves cell mechanical properties and intracellular calcium dynamics in normal cardiomyocytes, by increasing energy availability and removing the inhibitory effect of PLB on SERCA2.


Assuntos
Trifosfato de Adenosina/biossíntese , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Metabolismo Energético/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Polifenóis/farmacologia , Chá/química , Administração Oral , Animais , Masculino , Miócitos Cardíacos/citologia , Fosforilação/efeitos dos fármacos , Polifenóis/química , Ratos , Ratos Wistar , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
12.
Autophagy ; 12(11): 2098-2112, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27627835

RESUMO

Pathological mutations in the mitochondrial DNA (mtDNA) produce a diverse range of tissue-specific diseases and the proportion of mutant mitochondrial DNA can increase or decrease with time via segregation, dependent on the cell or tissue type. Previously we found that adenocarcinoma (A549.B2) cells favored wild-type (WT) mtDNA, whereas rhabdomyosarcoma (RD.Myo) cells favored mutant (m3243G) mtDNA. Mitochondrial quality control (mtQC) can purge the cells of dysfunctional mitochondria via mitochondrial dynamics and mitophagy and appears to offer the perfect solution to the human diseases caused by mutant mtDNA. In A549.B2 and RD.Myo cybrids, with various mutant mtDNA levels, mtQC was explored together with macroautophagy/autophagy and bioenergetic profile. The 2 types of tumor-derived cell lines differed in bioenergetic profile and mitophagy, but not in autophagy. A549.B2 cybrids displayed upregulation of mitophagy, increased mtDNA removal, mitochondrial fragmentation and mitochondrial depolarization on incubation with oligomycin, parameters that correlated with mutant load. Conversely, heteroplasmic RD.Myo lines had lower mitophagic markers that negatively correlated with mutant load, combined with a fully polarized and highly fused mitochondrial network. These findings indicate that pathological mutant mitochondrial DNA can modulate mitochondrial dynamics and mitophagy in a cell-type dependent manner and thereby offer an explanation for the persistence and accumulation of deleterious variants.


Assuntos
DNA Mitocondrial/genética , Mitocôndrias/metabolismo , Mutação/genética , Células A549 , Autofagia/efeitos dos fármacos , Autofagia/genética , Citoplasma/metabolismo , Metabolismo Energético , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Mitofagia/genética , Oligomicinas/farmacologia , Proteína Sequestossoma-1/metabolismo
13.
Oncotarget ; 7(5): 5521-37, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26575168

RESUMO

PEL is a B-cell non-Hodgkin lymphoma, occurring predominantly as a lymphomatous effusion in body cavities, characterized by aggressive clinical course, with no standard therapy. Based on previous reports that PEL cells display a Warburg phenotype, we hypothesized that the highly hypoxic environment in which they grow in vivo makes them more reliant on glycolysis, and more vulnerable to drugs targeting this pathway. We established here that indeed PEL cells in hypoxia are more sensitive to glycolysis inhibition. Furthermore, since PI3K/Akt/mTOR has been proposed as a drug target in PEL, we ascertained that pathway-specific inhibitors, namely the dual PI3K and mTOR inhibitor, PF-04691502, and the Akt inhibitor, Akti 1/2, display improved cytotoxicity to PEL cells in hypoxic conditions. Unexpectedly, we found that these drugs reduce lactate production/extracellular acidification rate, and, in combination with the glycolysis inhibitor 2-deoxyglucose (2-DG), they shift PEL cells metabolism from aerobic glycolysis towards oxidative respiration. Moreover, the associations possess strong synergistic cytotoxicity towards PEL cells, and thus may reduce adverse reaction in vivo, while displaying very low toxicity to normal lymphocytes. Finally, we showed that the association of 2-DG and PF-04691502 maintains its cytotoxic and proapoptotic effect also in PEL cells co-cultured with human primary mesothelial cells, a condition known to mimic the in vivo environment and to exert a protective and pro-survival action. All together, these results provide a compelling rationale for the clinical development of new therapies for the treatment of PEL, based on combined targeting of glycolytic metabolism and constitutively activated signaling pathways.


Assuntos
Glicólise/efeitos dos fármacos , Linfoma de Efusão Primária/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Desoxiglucose/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Citometria de Fluxo , Humanos , Linfoma de Efusão Primária/tratamento farmacológico , Linfoma de Efusão Primária/patologia , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Análise Serial de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridonas/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
14.
J Biol Chem ; 290(10): 6338-48, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25605724

RESUMO

The bioenergetics of IF1 transiently silenced cancer cells has been extensively investigated, but the role of IF1 (the natural inhibitor protein of F1F0-ATPase) in cancer cell metabolism is still uncertain. To shed light on this issue, we established a method to prepare stably IF1-silenced human osteosarcoma clones and explored the bioenergetics of IF1 null cancer cells. We showed that IF1-silenced cells proliferate normally, consume glucose, and release lactate as controls do, and contain a normal steady-state ATP level. However, IF1-silenced cells displayed an enhanced steady-state mitochondrial membrane potential and consistently showed a reduced ADP-stimulated respiration rate. In the parental cells (i.e. control cells containing IF1) the inhibitor protein was found to be associated with the dimeric form of the ATP synthase complex, therefore we propose that the interaction of IF1 with the complex either directly, by increasing the catalytic activity of the enzyme, or indirectly, by improving the structure of mitochondrial cristae, can increase the oxidative phosphorylation rate in osteosarcoma cells grown under normoxic conditions.


Assuntos
Neoplasias Ósseas/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , Osteossarcoma/metabolismo , Proteínas/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Metabolismo Energético/genética , Inativação Gênica , Humanos , Potencial da Membrana Mitocondrial/genética , ATPases Mitocondriais Próton-Translocadoras/antagonistas & inibidores , Osteossarcoma/genética , Osteossarcoma/patologia , Fosforilação Oxidativa , Proteínas/antagonistas & inibidores , RNA Interferente Pequeno , Proteína Inibidora de ATPase
15.
Oncotarget ; 5(7): 1779-92, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24732040

RESUMO

The increasing importance of hypoxia-inducible factor-1α (HIF-1α) in tumorigenesis raises the possibility that agents which specifically inhibit this transcription factor, would provide significant therapeutic benefit. The constitutive expression of HIF-1α in about 35% of Multiple Myeloma (MM) patients suggests HIF-1α suppression might be part of a therapeutic strategy. Accordingly, we explored the effect of EZN-2968, a small 3rd generation antisense oligonucleotide against HIF-1α, in a panel of MM cell lines and primary patients samples. Here, we demonstrated that EZN-2968 is highly specific for HIF-1α mRNA and that exposure of MM cells to EZN-2968 resulted in an efficient and homogeneous loading of the cells showing a long lasting low HIF-1α protein level. In MM cells, HIF-1α suppression induced a permanent cell cycle arrest by prolonging S-phase through cyclin A modulation and in addition it induced a mild apoptotic cell death. Moreover, HIF-1α suppression caused a metabolic shift that leaded to increased production of ATP by oxidative phosphorylation (i.e. Warburg effect reversion), that was confirmed by the observed mitochondrial membrane potential decrease. These results show that HIF-1α is an important player in MM homeostasis and that its inhibition by small antisense oligonucleotides provides a rationale for novel therapeutic strategy to improving MM treatment.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Oligonucleotídeos/farmacologia , Trifosfato de Adenosina/biossíntese , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina A/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mieloma Múltiplo/ultraestrutura , Fosforilação Oxidativa/efeitos dos fármacos , RNA Mensageiro/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Int J Biochem Cell Biol ; 45(7): 1356-65, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23538299

RESUMO

Hypoxia induces severe changes in cell biology, particularly affecting energy production pathways. Although the theme has been widely investigated, particularly in transformed cells, studies of the mitochondrial bioenergetics of normal cells exposed to both prolonged hypoxic periods and low or null glucose concentration have been scarcely addressed. To evaluate the mitochondrial changes of cells exposed to the latter conditions, we set experiments in which the contribution of the mitochondrial energy production was maximized at the expenses of low glycolysis. Human fibroblasts were exposed to 1% oxygen tension, a hypoxic condition experimentally established for these cells, and grown in glucose deficient media. At variance with fibroblasts grown in 5-25 mM glucose, fibroblasts grown in glucose deficiency adapted to hypoxia by reducing only slightly the mitochondrial mass and preserving a well structured network as it occurs in normoxia. Moreover, the oxidative phosphorylation (OXPHOS) rate of the mitochondria was enhanced, due to increased OXPHOS complexes level. The master transcriptional modulator induced by hypoxia HIF-1α and BNIP3, a factor activating mitochondrial autophagy, were expressed both in the presence and in the absence of glucose, but to a lower level in the latter condition. Similarly, the microtubule-associated protein light chain 3 active fragment (LC3-II), a typical marker of autophagy, was found less expressed in glucose-free medium than in the presence of glucose. Therefore, our data show for the first time that glucose availability significantly affects the hypoxia-induced HIF-1/BNIP3 response, and in particular glucose absence results in enhancing the OXPHOS rate.


Assuntos
Hipóxia Celular/fisiologia , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adolescente , Adulto , Autofagia/fisiologia , Proliferação de Células , Células Cultivadas , Criança , Metabolismo Energético , Fibroblastos , Glicólise , Humanos , Hipóxia/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Fosforilação Oxidativa , Oxigênio , Adulto Jovem
17.
ScientificWorldJournal ; 2012: 573410, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22593698

RESUMO

PURPOSE: Hypothermic machine perfusion systems seem more effective than the current static storage to prevent cold ischemic liver injury. Thus, we test an innovative hyperbaric hypothermic machine perfusion (HHMP), which combines hyperbaric oxygenation of the preservation solution and continuous perfusion of the graft. METHODS: Rat livers were preserved with Celsior solution according to 4 different modalities: normobaric static preservation; hyperbaric static preservation at 2 atmosphere absolute (ATA); normobaric dynamic preservation, with continuous perfusion; hyperbaric dynamic preservation, with continuous perfusion at 2 ATA. After 24 h cold preservation, we assessed different parameters. RESULTS: Compared to baseline, livers preserved with the current static storage showed severe ultrastructural damage, glycogen depletion and an increased oxidative stress. Normobaric perfused livers showed improved hepatocyte ultrastructure and ameliorated glycogen stores, but they still suffered a significant oxidative damage. The addition of hyperbaric oxygen produces an extra benefit by improving oxidative injury and by inducing endothelial NO synthase (eNOS) gene expression. CONCLUSIONS: Preservation by means of the present innovative HHMP reduced the liver injury occurring after the current static cold storage by lowering glycogen depletion and oxidative damage. Interestingly, only the use of hyperbaric oxygen was associated to a blunted oxidative stress and an increased eNOS gene expression.


Assuntos
Oxigenoterapia Hiperbárica/métodos , Hipotermia Induzida/métodos , Fígado , Preservação de Órgãos/métodos , Animais , Dissacarídeos/farmacologia , Eletrólitos/farmacologia , Regulação Enzimológica da Expressão Gênica , Glutamatos/farmacologia , Glutationa/metabolismo , Glutationa/farmacologia , Dissulfeto de Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Histidina/farmacologia , Hipotermia Induzida/instrumentação , Glicogênio Hepático/metabolismo , Manitol/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Soluções para Preservação de Órgãos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Perfusão , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Sulfidrila/metabolismo
18.
Biochim Biophys Acta ; 1807(6): 534-42, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20849810

RESUMO

Evidence suggests that mitochondrial metabolism may play a key role in controlling cancer cells life and proliferation. Recent evidence also indicates how the altered contribution of these organelles to metabolism and the resistance of cancer mitochondria against apoptosis-associated permeabilization are closely related. The hallmarks of cancer growth, increased glycolysis and lactate production in tumours, have raised attention due to recent observations suggesting a wide spectrum of oxidative phosphorylation deficit and decreased availability of ATP associated with malignancies and tumour cell expansion. More specifically, alteration in signal transduction pathways directly affects mitochondrial proteins playing critical roles in controlling the membrane potential as UCP2 and components of both MPTP and oxphos complexes, or in controlling cells life and death as the Bcl-2 proteins family. Moreover, since mitochondrial bioenergetics and dynamics, are also involved in processes of cells life and death, proper regulation of these mitochondrial functions is crucial for tumours to grow. Therefore a better understanding of the key pathophysiological differences between mitochondria in cancer cells and in their non-cancer surrounding tissue is crucial to the finding of tools interfering with these peculiar tumour mitochondrial functions and will disclose novel approaches for the prevention and treatment of malignant diseases. Here, we review the peculiarity of tumour mitochondrial bioenergetics and the mode it is linked to the cell metabolism, providing a short overview of the evidence accumulated so far, but highlighting the more recent advances.


Assuntos
Neoplasias/metabolismo , Fosforilação Oxidativa , Animais , Humanos , Redes e Vias Metabólicas/fisiologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitocôndrias/fisiologia , Modelos Biológicos , Neoplasias/patologia , Neoplasias/fisiopatologia , Consumo de Oxigênio/fisiologia
19.
Biochim Biophys Acta ; 1797(2): 314-23, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19931505

RESUMO

Many cancer cells are characterized by high rate of glycolysis and reduced rate of aerobic respiration, whose mechanism is still elusive. Here we investigate the down-regulation of oxidative phosphorylation (OXPHOS) in K-ras transformed mouse fibroblasts as compared to a control counterpart. Transcriptional analysis showed different expression levels of several OXPHOS nuclear genes in the two cell lines. In particular, during the exponential growth phase most genes encoding proteins of Complex I were expressed at lower levels in transformed cells. Consistently, a significant decrease of Complex I content was found in transformed cells. Moreover, analysis of NAD-dependent respiration and ATP synthesis indicated a strong decrease of Complex I activity in the mitochondria from neoplastic cells, that was confirmed by direct assay of the enzyme redox activity. At variance, succinate-dependent respiration and ATP synthesis were not significantly affected. Taken together, our results provide the new insight that the reduction of respiration observed in K-ras transformed cells is specifically due to a Complex I activity decrease.


Assuntos
Transformação Celular Neoplásica , Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/metabolismo , Metabolismo Energético , Fibroblastos/metabolismo , Genes ras , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Biomarcadores/metabolismo , Proliferação de Células , Respiração Celular/fisiologia , Células Cultivadas , Perfilação da Expressão Gênica , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , Fosforilação Oxidativa , Oxigênio/metabolismo
20.
Arch Neurol ; 66(8): 951-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19667215

RESUMO

OBJECTIVE: To verify whether enhanced substrate-level phosphorylation increases viability and adenosine 5'-triphosphate (ATP) content of cells with neuropathy, ataxia, and retinitis pigmentosa/maternally inherited Leigh syndrome (NARP/MILS) mitochondrial DNA mutations and ATP synthase dysfunction. DESIGN: We used cell lines "poisoned" with oligomycin, the specific inhibitor of ATP synthase, and "natural" models, including transmitochondrial human cell lines (cybrids) harboring 2 different pathogenic mutations associated with the NARP/MILS phenotypes. MAIN OUTCOME MEASURES: Cell survival, morphology, and ATP content. RESULTS: When normal human fibroblasts cultured in glucose-free medium were forced to increase energy consumption by exposure to the ionophore gramicidin or were energy challenged by oligomycin inhibition, their survival at 72 hours was 5%, but this increased to 70% when the medium was supplemented with alpha-ketoglutarate/aspartate to boost mitochondrial substrate-level phosphorylation. Homoplasmic cybrids harboring the 8993T-->G NARP mutation were also protected from death (75% vs 15% survival at 72 hours) by the supplemented medium and their ATP content was similar to controls. CONCLUSIONS: These results show that ATP synthase-deficient cells can be rescued by increasing mitochondrial substrate-level phosphorylation and suggest potential dietary or pharmacological therapeutic approaches based on the supplementation of alpha-ketoglutarate/aspartate to patients with impaired ATP synthase activity.


Assuntos
Trifosfato de Adenosina/metabolismo , Ácido Aspártico/farmacologia , Proteína C-Reativa/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , DNA Mitocondrial/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Ácidos Cetoglutáricos/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , ATPases Mitocondriais Próton-Translocadoras/antagonistas & inibidores , ATPases Mitocondriais Próton-Translocadoras/genética , Mutação , Proteínas do Tecido Nervoso/genética , Oligomicinas/farmacologia , Anti-Infecciosos Locais/farmacologia , Linhagem Celular , Gramicidina/farmacologia , Humanos , Doença de Leigh/genética , Fosforilação Oxidativa/efeitos dos fármacos , Retinose Pigmentar/genética , Degenerações Espinocerebelares/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA